51
|
Abstract
Electro photonic imaging (EPI) is being researched relative to its application for yoga therapy. Three parameters of interest in EPI measurements are as follows: Communication energy (C), integral or normalized area (IA), and Entropy (E). It is important to note that C indicates the total energy of communication for the organ system; IA is an indication of total amount of energy that is available for the organ system while entropy is an indication of the amount of coherence of the energy. Coherence and entropy are inversely related; this means less the entropy, more the coherence and vice versa. Illustrative cases of successful therapy with yoga practices in a wide variety of abnormal conditions are examined, and in every case, entropy is shown to decrease for the affected organ system while communication energy stays within stable range. Relative to the electromagnetic (Rubik) and living matrix (Oschman) models, it is suggested that the regulation of energy, its coherence in the biological system and interaction with life processes provide the basis for model building and design of health-promoting procedures. Further, this approach is examined relative to yoga theory, traditional medicine systems, and scientific developments in the field of gene expression and neuroplasticity and a generalized model that we call Unified System of Medicine is proposed. This model has direct implications on methods used to control the environmental factors to get robust results from EPI application for therapeutic purposes. Implications for furthering research in yoga therapy using EPI and implications of EPI as a translational technology between traditional medicine systems and modern medicine is discussed.
Collapse
Affiliation(s)
| | - Konstantin Korotkov
- Department of Innovation Technologies, Research Institute of Physical Culture and Sport, St. Petersburg, Russia
| | | |
Collapse
|
52
|
Parrilla-Carrero J, Buchta WC, Goswamee P, Culver O, McKendrick G, Harlan B, Moutal A, Penrod R, Lauer A, Ramakrishnan V, Khanna R, Kalivas P, Riegel AC. Restoration of Kv7 Channel-Mediated Inhibition Reduces Cued-Reinstatement of Cocaine Seeking. J Neurosci 2018; 38:4212-4229. [PMID: 29636392 PMCID: PMC5963852 DOI: 10.1523/jneurosci.2767-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 12/16/2022] Open
Abstract
Cocaine addicts display increased sensitivity to drug-associated cues, due in part to changes in the prelimbic prefrontal cortex (PL-PFC). The cellular mechanisms underlying cue-induced reinstatement of cocaine seeking remain unknown. Reinforcement learning for addictive drugs may produce persistent maladaptations in intrinsic excitability within sparse subsets of PFC pyramidal neurons. Using a model of relapse in male rats, we sampled >600 neurons to examine spike frequency adaptation (SFA) and afterhyperpolarizations (AHPs), two systems that attenuate low-frequency inputs to regulate neuronal synchronization. We observed that training to self-administer cocaine or nondrug (sucrose) reinforcers decreased SFA and AHPs in a subpopulation of PL-PFC neurons. Only with cocaine did the resulting hyperexcitability persist through extinction training and increase during reinstatement. In neurons with intact SFA, dopamine enhanced excitability by inhibiting Kv7 potassium channels that mediate SFA. However, dopamine effects were occluded in neurons from cocaine-experienced rats, where SFA and AHPs were reduced. Pharmacological stabilization of Kv7 channels with retigabine restored SFA and Kv7 channel function in neuroadapted cells. When microinjected bilaterally into the PL-PFC 10 min before reinstatement testing, retigabine reduced cue-induced reinstatement of cocaine seeking. Last, using cFos-GFP transgenic rats, we found that the loss of SFA correlated with the expression of cFos-GFP following both extinction and re-exposure to drug-associated cues. Together, these data suggest that cocaine self-administration desensitizes inhibitory Kv7 channels in a subpopulation of PL-PFC neurons. This subpopulation of neurons may represent a persistent neural ensemble responsible for driving drug seeking in response to cues.SIGNIFICANCE STATEMENT Long after the cessation of drug use, cues associated with cocaine still elicit drug-seeking behavior, in part by activation of the prelimbic prefrontal cortex (PL-PFC). The underlying cellular mechanisms governing these activated neurons remain unclear. Using a rat model of relapse to cocaine seeking, we identified a population of PL-PFC neurons that become hyperexcitable following chronic cocaine self-administration. These neurons show persistent loss of spike frequency adaptation, reduced afterhyperpolarizations, decreased sensitivity to dopamine, and reduced Kv7 channel-mediated inhibition. Stabilization of Kv7 channel function with retigabine normalized neuronal excitability, restored Kv7 channel currents, and reduced drug-seeking behavior when administered into the PL-PFC before reinstatement. These data highlight a persistent adaptation in a subset of PL-PFC neurons that may contribute to relapse vulnerability.
Collapse
Affiliation(s)
- Jeffrey Parrilla-Carrero
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - William C Buchta
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Priyodarshan Goswamee
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Oliver Culver
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Greer McKendrick
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Benjamin Harlan
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, Arizona 85724, and
| | - Rachel Penrod
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Abigail Lauer
- Department of Public Health Sciences., Medical University of South Carolina, Charleston, SC 29425
| | - Viswanathan Ramakrishnan
- Department of Public Health Sciences., Medical University of South Carolina, Charleston, SC 29425
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, Arizona 85724, and
| | - Peter Kalivas
- Department of Neuroscience
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Arthur C Riegel
- Department of Neuroscience,
- Neurobiology of Addiction Research Center, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
53
|
Zhu R, Bu Q, Fu D, Shao X, Jiang L, Guo W, Chen B, Liu B, Hu Z, Tian J, Zhao Y, Cen X. Toll-like receptor 3 modulates the behavioral effects of cocaine in mice. J Neuroinflammation 2018; 15:93. [PMID: 29571298 PMCID: PMC5865345 DOI: 10.1186/s12974-018-1130-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 03/13/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The nucleus accumbens in the midbrain dopamine limbic system plays a key role in cocaine addiction. Toll-like receptors (TLRs) are important pattern-recognition receptors (PPRs) in the innate immune system that are also involved in drug dependence; however, the detailed mechanism is largely unknown. METHODS The present study was designed to investigate the potential role of TLR3 in cocaine addiction. Cocaine-induced conditioned place preference (CPP), locomotor activity, and self-administration were used to determine the effects of TLR3 in the rewarding properties of cocaine. Lentivirus-mediated re-expression of Tlr3 (LV-TLR3) was applied to determine if restoration of TLR3 expression in the NAc is sufficient to restore the cocaine effect in TLR3-/- mice. The protein levels of phospho-NF-κB p65, IKKβ, and p-IκBα both in the cytoplasm and nucleus of cocaine-induced CPP mice were detected by Western blot. RESULTS We showed that both TLR3 deficiency and intra-NAc injection of TLR3 inhibitors significantly attenuated cocaine-induced CPP, locomotor activity, and self-administration in mice. Importantly, the TLR3-/- mice that received intra-NAc injection of LV-TLR3 displayed significant increases in cocaine-induced CPP and locomotor activity. Finally, we found that TLR3 inhibitor reverted cocaine-induced upregulation of phospho-NF-κB p65, IKKβ, and p-IκBα. CONCLUSIONS Taken together, our results describe that TLR3 modulates cocaine-induced behaviors and provide further evidence supporting a role for central pro-inflammatory immune signaling in drug reward. We propose that TLR3 blockade could be a novel approach to treat cocaine addiction.
Collapse
Affiliation(s)
- Ruiming Zhu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Qian Bu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China.,Healthy Food Evaluation Research Center, Department of Food Science and Technology, College of Light Industry, Textile and Food Engineering, Sichuan University, Chengdu, 610065, China
| | - Dengqi Fu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Xue Shao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Linhong Jiang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Wei Guo
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China.,School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Bo Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Bin Liu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Zhengtao Hu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China
| | - Yinglan Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Lab of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, West China Medical School, Sichuan University, #28 Gaopeng Street, High Technological Development Zone, Chengdu, 610041, China.
| |
Collapse
|
54
|
Mukherjee D, Ignatowska-Jankowska BM, Itskovits E, Gonzales BJ, Turm H, Izakson L, Haritan D, Bleistein N, Cohen C, Amit I, Shay T, Grueter B, Zaslaver A, Citri A. Salient experiences are represented by unique transcriptional signatures in the mouse brain. eLife 2018; 7:e31220. [PMID: 29412137 PMCID: PMC5862526 DOI: 10.7554/elife.31220] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/05/2018] [Indexed: 12/11/2022] Open
Abstract
It is well established that inducible transcription is essential for the consolidation of salient experiences into long-term memory. However, whether inducible transcription relays information about the identity and affective attributes of the experience being encoded, has not been explored. To this end, we analyzed transcription induced by a variety of rewarding and aversive experiences, across multiple brain regions. Our results describe the existence of robust transcriptional signatures uniquely representing distinct experiences, enabling near-perfect decoding of recent experiences. Furthermore, experiences with shared attributes display commonalities in their transcriptional signatures, exemplified in the representation of valence, habituation and reinforcement. This study introduces the concept of a neural transcriptional code, which represents the encoding of experiences in the mouse brain. This code is comprised of distinct transcriptional signatures that correlate to attributes of the experiences that are being committed to long-term memory.
Collapse
Affiliation(s)
- Diptendu Mukherjee
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | | | - Eyal Itskovits
- Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
- School of Computer Science and EngineeringThe Hebrew UniversityJerusalemIsrael
| | - Ben Jerry Gonzales
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Hagit Turm
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Liz Izakson
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Doron Haritan
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Noa Bleistein
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Chen Cohen
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Ido Amit
- Department of ImmunologyWeizmann Institute of ScienceRehovotIsrael
| | - Tal Shay
- Department of Life SciencesBen-Gurion University of the NegevBeer-ShevaIsrael
| | - Brad Grueter
- Department of PsychiatryVanderbilt University School of MedicineNashvilleUnited States
| | - Alon Zaslaver
- Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Ami Citri
- Department of Biological Chemistry, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
- The Edmond and Lily Safra Center for Brain SciencesThe Hebrew UniversityJerusalemIsrael
- Child and Brain Development ProgramCanadian Institute for Advanced ResearchTorontoCanada
| |
Collapse
|
55
|
In silico identification and in vivo validation of miR-495 as a novel regulator of motivation for cocaine that targets multiple addiction-related networks in the nucleus accumbens. Mol Psychiatry 2018; 23:434-443. [PMID: 28044061 PMCID: PMC5495632 DOI: 10.1038/mp.2016.238] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 10/31/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are important post-transcriptional regulators of gene expression and are implicated in the etiology of several neuropsychiatric disorders, including substance use disorders (SUDs). Using in silico genome-wide sequence analyses, we identified miR-495 as a miRNA whose predicted targets are significantly enriched in the Knowledgebase for Addiction Related Genes (ARG) database (KARG; http://karg.cbi.pku.edu.cn). This small non-coding RNA is also highly expressed within the nucleus accumbens (NAc), a pivotal brain region underlying reward and motivation. Using luciferase reporter assays, we found that miR-495 directly targeted the 3'UTRs of Bdnf, Camk2a and Arc. Furthermore, we measured miR-495 expression in response to acute cocaine in mice and found that it is downregulated rapidly and selectively in the NAc, along with concomitant increases in ARG expression. Lentiviral-mediated miR-495 overexpression in the NAc shell (NAcsh) not only reversed these cocaine-induced effects but also downregulated multiple ARG mRNAs in specific SUD-related biological pathways, including those that regulate synaptic plasticity. miR-495 expression was also downregulated in the NAcsh of rats following cocaine self-administration. Most importantly, we found that NAcsh miR-495 overexpression suppressed the motivation to self-administer and seek cocaine across progressive ratio, extinction and reinstatement testing, but had no effect on food reinforcement, suggesting that miR-495 selectively affects addiction-related behaviors. Overall, our in silico search for post-transcriptional regulators identified miR-495 as a novel regulator of multiple ARGs that have a role in modulating motivation for cocaine.
Collapse
|
56
|
Wu Q, Cunningham JT, Mifflin S. Transcription factor ΔFosB acts within the nucleus of the solitary tract to increase mean arterial pressure during exposures to intermittent hypoxia. Am J Physiol Heart Circ Physiol 2017; 314:H270-H277. [PMID: 29101166 DOI: 10.1152/ajpheart.00268.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ΔFosB is a member of the activator protein-1 family of transcription factors. ΔFosB has low constitutive expression in the central nervous system and is induced after exposure of rodents to intermittent hypoxia (IH), a model of the arterial hypoxemia that accompanies sleep apnea. We hypothesized ΔFosB in the nucleus of the solitary tract (NTS) contributes to increased mean arterial pressure (MAP) during IH. The NTS of 11 male Sprague-Dawley rats was injected (3 sites, 100 nl/site) with a dominant negative construct against ΔFosB (ΔJunD) in an adeno-associated viral vector (AAV)-green fluorescent protein (GFP) reporter. The NTS of 10 rats was injected with AAV-GFP as sham controls. Two weeks after NTS injections, rats were exposed to IH for 8 h/day for 7 days, and MAP was recorded using telemetry. In the sham group, 7 days of IH increased MAP from 99.8 ± 1.1 to 107.3 ± 0.5 mmHg in the day and from 104.4 ± 1.1 to 109.8 ± 0.6 mmHg in the night. In the group that received ΔJunD, IH increased MAP during the day from 95.9 ± 1.7 to 101.3 ± 0.4 mmHg and from 100.9 ± 1.7 to 102.8 ± 0.5 mmHg during the night (both IH-induced changes in MAP were significantly lower than sham, P < 0.05). After injection of the dominant negative construct in the NTS, IH-induced ΔFosB immunoreactivity was decreased in the paraventricular nucleus ( P < 0.05); however, no change was observed in the rostral ventrolateral medulla. These data indicate that ΔFosB within the NTS contributes to the increase in MAP induced by IH exposure. NEW & NOTEWORTHY The results of this study provides new insights into the molecular mechanisms that mediate neuronal adaptations during exposures to intermittent hypoxia, a model of the hypoxemias that occur during sleep apnea. These adaptations are noteworthy as they contribute to the persistent increase in blood pressure induced by exposures to intermittent hypoxia.
Collapse
Affiliation(s)
- Qiong Wu
- Departments of Psychiatry and Biobehavioral Sciences and Neurobiology, The Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California
| | - J Thomas Cunningham
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas
| | - Steve Mifflin
- Institute for Cardiovascular and Metabolic Disease, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
57
|
Saxena A, Scaini G, Bavaresco DV, Leite C, Valvassori SS, Carvalho AF, Quevedo J. Role of Protein Kinase C in Bipolar Disorder: A Review of the Current Literature. MOLECULAR NEUROPSYCHIATRY 2017; 3:108-124. [PMID: 29230399 DOI: 10.1159/000480349] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/14/2017] [Indexed: 12/19/2022]
Abstract
Bipolar disorder (BD) is a major health problem. It causes significant morbidity and imposes a burden on the society. Available treatments help a substantial proportion of patients but are not beneficial for an estimated 40-50%. Thus, there is a great need to further our understanding the pathophysiology of BD to identify new therapeutic avenues. The preponderance of evidence pointed towards a role of protein kinase C (PKC) in BD. We reviewed the literature pertinent to the role of PKC in BD. We present recent advances from preclinical and clinical studies that further support the role of PKC. Moreover, we discuss the role of PKC on synaptogenesis and neuroplasticity in the context of BD. The recent development of animal models of BD, such as stimulant-treated and paradoxical sleep deprivation, and the ability to intervene pharmacologically provide further insights into the involvement of PKC in BD. In addition, the effect of PKC inhibitors, such as tamoxifen, in the resolution of manic symptoms in patients with BD further points in that direction. Furthermore, a wide variety of growth factors influence neurotransmission through several molecular pathways that involve downstream effects of PKC. Our current understanding identifies the PKC pathway as a potential therapeutic avenue for BD.
Collapse
Affiliation(s)
- Ashwini Saxena
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Giselli Scaini
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Daniela V Bavaresco
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - Camila Leite
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - Samira S Valvassori
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - André F Carvalho
- Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| |
Collapse
|
58
|
Fatsini E, Rey S, Ibarra-Zatarain Z, Mackenzie S, Duncan NJ. Dominance behaviour in a non-aggressive flatfish, Senegalese sole (Solea senegalensis) and brain mRNA abundance of selected transcripts. PLoS One 2017; 12:e0184283. [PMID: 28877259 PMCID: PMC5587333 DOI: 10.1371/journal.pone.0184283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/21/2017] [Indexed: 11/18/2022] Open
Abstract
Dominance is defined as the preferential access to limited resources. The present study aimed to characterise dominance in a non-aggressive flatfish species, the Senegalese sole (Solea senegalensis) by 1) identifying dominance categories and associated behaviours and 2) linking dominance categories (dominant and subordinate) with the abundance of selected mRNA transcripts in the brain. Early juveniles (n = 74, 37 pairs) were subjected to a dyadic dominance test, related to feeding, and once behavioural phenotypes had been described the abundance of ten selected mRNAs related to dominance and aggressiveness was measured in the brain. Late juveniles were subjected to two dyadic dominance tests (n = 34, 17 pairs), related to feeding and territoriality and one group test (n = 24, 4 groups of 6 fish). Sole feeding first were categorized as dominant and sole feeding second or not feeding as subordinate. Three social behaviours (i. "Resting the head" on another fish, ii. "Approaching" another fish, iii. "Swimming above another" fish) were associated with dominance of feeding. Two other variables (i. Total time occupying the preferred area during the last 2 hours of the 24 h test, ii. Organisms occupying the preferred area when the test ended) were representative of dominance in the place preference test. In all tests, dominant fish compared to subordinate fish displayed a significantly higher number of the behaviours "Rest the head" and "Approaches". Moreover, dominant sole dominated the sand at the end of the test, and in the group test dominated the area close to the feed delivery point before feed was delivered. The mRNA abundance of the selected mRNAs related to neurogenesis (nrd2) and neuroplasticity (c-fos) in dominant sole compared to subordinate were significantly different. This is the first study to characterise dominance categories with associated behaviours and mRNA abundance in Senegalese sole and provides tools to study dominance related problems in feeding and reproduction in aquaculture.
Collapse
Affiliation(s)
| | - Sonia Rey
- Institute of Aquaculture, Pathfoot Building, University of Stirling, Stirling, Scotland, United Kingdom
| | - Zohar Ibarra-Zatarain
- IRTA, Sant Carles de la Ràpita, Tarragona, Spain.,CONACYT-UAN-CENIT, Calle 3 S/N, Ciudad industrial, Tepic, Mexico
| | - Simon Mackenzie
- Institute of Aquaculture, Pathfoot Building, University of Stirling, Stirling, Scotland, United Kingdom
| | | |
Collapse
|
59
|
Legastelois R, Jeanblanc J, Vilpoux C, Bourguet E, Naassila M. [Epigenetic mechanisms and alcohol use disorders: a potential therapeutic target]. Biol Aujourdhui 2017; 211:83-91. [PMID: 28682229 DOI: 10.1051/jbio/2017014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Indexed: 01/27/2023]
Abstract
Alcohol use disorder is a devastating illness with a profound health impact, and its development is dependent on both genetic and environmental factors. This disease occurs over time and requires changes in brain gene expression. There is converging evidence suggesting that the epigenetic processes may play a role in the alcohol-induced gene regulations and behavior such as the intervention of DNA methylation and histone acetylation. Histone acetylation, like histone methylation, is a highly dynamic process regulated by two classes of enzymes: histone acetyltransferases and histone deacetylases (HDACs). To date, 18 human HDAC isoforms have been characterized, and based on their sequence homologies and cofactor dependencies, they have been phylogenetically categorized into 4 main classes: classes I, II (a and b), III, and IV. In the brain, expression of the different classes of HDACs varies between cell types and also in their subcellular localization (nucleus and/or cytosol). Furthermore, we recently showed that a single ethanol exposure inhibits HDAC activity and increases both H3 and H4 histone acetylation within the amygdala of rats. In the brain of alcoholic patients, ethanol has been shown to induce histone-related and DNA methylation epigenetic changes in several reward regions involved in reward processes such as hippocampus, prefrontal cortex, and amygdala. We recently demonstrated alteration of histone H3 acetylation levels in several brain regions from the reward circuit of rats made dependent to alcohol after chronic and intermittent exposure to ethanol vapor. In neuronal cell line culture, ethanol was shown to induce HDAC expression. In mouse and rat brain, numerous studies reported epigenetic alterations following ethanol exposure. We also demonstrated that both the expression of genes and the activity of enzymes involved in epigenetic mechanisms are changed after repeated administrations of ethanol in mice sensitized to the motor stimulant effect of ethanol (a model of drug-induced neuroplasticity). Numerous studies have shown that HDAC inhibitors are able to counter ethanol-induced behaviors and the ethanol-induced changes in the levels of HDAC and/or levels of acetylated HDAC. For example, trichostatin A (TSA) treatment caused the reversal of ethanol-induced tolerance, anxiety, and ethanol drinking by inhibiting HDAC activity, thereby increasing histone acetylation in the amygdala of rats. Another study demonstrated that TSA prevented the development of ethanol withdrawal induced anxiety in rats by rescuing deficits in histone acetylation induced by increased HDAC activity in the amygdala. We have demonstrated that treatment with the HDAC inhibitor sodium butyrate blocks both the development and the expression of ethanol-induced behavioral sensitization in mice. In this context, converging evidence indicates that HDAC inhibitors could be useful in counteracting ethanol-induced gene regulations via epigenetic mechanisms, that is, HDAC inhibitors could affect different acetylation sites and may also alter the expression of different genes that could in turn counteract the effect of ethanol. Recent work in rodents has shown that systemic administration of pan HDAC class I and II inhibitors, TSA and N-hydroxy-N-phenyl-octanediamide [SuberoylAnilide Hydroxamic Acid] (SAHA), and of the more selective inhibitor (mainly HDAC1 and HDAC9) MS-275, decrease binge-like alcohol drinking in mice. SAHA selectively reduced ethanol operant self-administration and seeking in rats. Our previous study revealed that MS-275 strongly decreased operant ethanol self-administration in alcohol-dependent rats when administered 30 minutes before the session at the second day of injection. We also demonstrated that intra-cerebro-ventricular infusion of MS-275 increases acetylation of Histone 4 within the nucleus accumbens and the dorsolateral striatum, associated to a decrease in ethanol self-administration by about 75%. MS-275 also diminished both the motivation to consume ethanol (25% decrease), relapse (by about 50%) and postponed reacquisition after abstinence. Both literature and several of our studies strongly support the potential therapeutic interest of targeting epigenetic mechanisms in excessive alcohol drinking and strengthen theinterest of focusing on specific isoforms of histone deacetylases.
Collapse
Affiliation(s)
- Rémi Legastelois
- INSERM ERi 24 Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP) - Université de Picardie Jules Verne, Amiens, France
| | - Jérôme Jeanblanc
- INSERM ERi 24 Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP) - Université de Picardie Jules Verne, Amiens, France
| | - Catherine Vilpoux
- INSERM ERi 24 Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP) - Université de Picardie Jules Verne, Amiens, France
| | | | - Mickael Naassila
- INSERM ERi 24 Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP) - Université de Picardie Jules Verne, Amiens, France
| |
Collapse
|
60
|
The Crucial Role of DNA Methylation and MeCP2 in Neuronal Function. Genes (Basel) 2017; 8:genes8050141. [PMID: 28505093 PMCID: PMC5448015 DOI: 10.3390/genes8050141] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/25/2017] [Accepted: 05/05/2017] [Indexed: 12/16/2022] Open
Abstract
A neuron is unique in its ability to dynamically modify its transcriptional output in response to synaptic activity while maintaining a core gene expression program that preserves cellular identity throughout a lifetime that is longer than almost every other cell type in the body. A contributing factor to the immense adaptability of a neuron is its unique epigenetic landscape that elicits locus-specific alterations in chromatin architecture, which in turn influences gene expression. One such epigenetic modification that is sensitive to changes in synaptic activity, as well as essential for maintaining cellular identity, is DNA methylation. The focus of this article is on the importance of DNA methylation in neuronal function, summarizing recent studies on critical players in the establishment of (the “writing”), the modification or erasure of (the “editing”), and the mediation of (the “reading”) DNA methylation in neurodevelopment and neuroplasticity. One “reader” of DNA methylation in particular, methyl-CpG-binding protein 2 (MeCP2), is highlighted, given its undisputed importance in neuronal function.
Collapse
|
61
|
Lima IVDA, Almeida-Santos AF, Ferreira-Vieira TH, Aguiar DC, Ribeiro FM, Campos AC, de Oliveira ACP. Antidepressant-like effect of valproic acid-Possible involvement of PI3K/Akt/mTOR pathway. Behav Brain Res 2017; 329:166-171. [PMID: 28408298 DOI: 10.1016/j.bbr.2017.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 01/05/2023]
Abstract
RATIONALE Few studies suggest that antidepressants exert their effects by activating some signaling pathways, including the phosphatidylinositol 3-kinase (PI3K). Moreover, valproic acid (VPA) activates the PI3K pathway. Thus, here we investigated the antidepressant-like effect of VPA and if its effect is related to PI3K/Akt/mTOR activation. METHODS C57Bl/6 (WT) and PI3Kγ-/- mice received VPA injections (30, 100 or 300mg/kg, i.p.) and 30min after they were submitted to the forced swimming (FS), tail suspension (TS) and open field (OF) tests. Another group was pretreated with rapamycin (5mg/kg, i.p.) 150min before VPA administration. Akt phosphorylation levels were measured by Western blotting. RESULTS In WT mice, VPA (30mg/kg) reduced the immobility time in both FS and TS tests. However, VPA (300mg/kg) increased the immobility time in FS test. All doses of VPA did not alter locomotor activity. In PI3Kγ-/- mice, none of the doses revealed antidepressant-like effect. However, in the OF test, the lower dose of VPA increased the travelled distance in comparison with vehicle group. An increase in Akt phosphorylation levels was observed in WT, but not in PI3Kγ-/- mice. Finally, the pretreatment of WT mice with rapamycin abolished the antidepressant-like effect of VPA (30mg/kg) in FS test. CONCLUSION These data suggest that the antidepressant-like effects of VPA might depend on PI3K and mTOR activation. Thus, more studies are necessary to investigate the mechanisms involved in the antidepressant-like effect induced by VPA in order to investigate novel therapeutic targets for the treatment of depression.
Collapse
Affiliation(s)
| | - Ana Flávia Almeida-Santos
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Talita Hélen Ferreira-Vieira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Daniele Cristina Aguiar
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Fabíola Mara Ribeiro
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Alline Cristina Campos
- Department of Pharmacology, Universidade de São Paulo, Ribeirão Preto, 14049-900, Brazil.
| | | |
Collapse
|
62
|
Levin SG, Godukhin OV. Modulating Effect of Cytokines on Mechanisms of Synaptic Plasticity in the Brain. BIOCHEMISTRY (MOSCOW) 2017; 82:264-274. [PMID: 28320267 DOI: 10.1134/s000629791703004x] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
After accumulation of data showing that resident brain cells (neurons, astrocytes, and microglia) produce mediators of the immune system, such as cytokines and their receptors under normal physiological conditions, a critical need emerged for investigating the role of these mediators in cognitive processes. The major problem for understanding the functional role of cytokines in the mechanisms of synaptic plasticity, de novo neurogenesis, and learning and memory is the small number of investigated cytokines. Existing concepts are based on data from just three proinflammatory cytokines: interleukin-1 beta, interleukin-6, and tumor necrosis factor-alpha. The amount of information in the literature on the functional role of antiinflammatory cytokines in the mechanisms of synaptic plasticity and cognitive functions of mature mammalian brain is dismally low. However, they are of principle importance for understanding the mechanisms of local information processing in the brain, since they modulate the activity of individual cells and local neural networks, being able to reconstruct the processes of synaptic plasticity and intercellular communication, in general, depending on the local ratio of the levels of different cytokines in certain areas of the brain. Understanding the functional role of cytokines in cellular mechanisms of information processing and storage in the brain would allow developing preventive and therapeutic means for the treatment of neuropathologies related to impairment of these mechanisms.
Collapse
Affiliation(s)
- S G Levin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | | |
Collapse
|
63
|
Opiol H, de Zavalia N, Delorme T, Solis P, Rutherford S, Shalev U, Amir S. Exploring the role of locomotor sensitization in the circadian food entrainment pathway. PLoS One 2017; 12:e0174113. [PMID: 28301599 PMCID: PMC5354457 DOI: 10.1371/journal.pone.0174113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/03/2017] [Indexed: 01/23/2023] Open
Abstract
Food entrainment is the internal mechanism whereby the phase and period of circadian clock genes comes under the control of daily scheduled food availability. Food entrainment allows the body to efficiently realign the internal timing of behavioral and physiological functions such that they anticipate food intake. Food entrainment can occur with or without caloric restriction, as seen with daily schedules of restricted feeding (RF) or restricted treat (RT) that restrict food or treat intake to a single feeding time. However, the extent of clock gene control is more pronounced with caloric restriction, highlighting the role of energy balance in regulating clock genes. Recent studies have implicated dopamine (DA) to be involved in food entrainment and caloric restriction is known to affect dopaminergic pathways to enhance locomotor activity. Since food entrainment results in the development of a distinct behavioral component, called food anticipatory activity (FAA), we examined the role of locomotor sensitization (LS) in food entrainment by 1) observing whether amphetamine (AMPH) sensitization results in enhanced locomotor output of FAA and 2) measuring LS of circadian and non-circadian feeding paradigms to an acute injection of AMPH (AMPH cross-sensitization). Unexpectedly, AMPH sensitization did not show enhancement of FAA. On the contrary, LS did develop with sufficient exposure to RF. LS was present after 2 weeks of RF, but not after 1, 3 or 7 days into RF. When food was returned and rats regain their original body weight at 10-15 days post-RF, LS remained present. LS did not develop to RT, nor to feedings of a non-circadian schedule, e.g. variable restricted feeding (VRF) or variable RT (VRT). Further, when RF was timed to the dark period, LS was observed only when tested at night; RF timed to the light period resulted in LS that was present during day and night. Taken together our results show that LS develops with food entrainment to RF, an effect that is dependent on the chronicity and circadian phase of RF but independent of body weight. Given that LS involves reorganization of DA-regulated motor circuitry, our work provides indirect support for the role of DA in the food entrainment pathway of RF. The findings also suggest differences in neuronal pathways involved in LS from AMPH sensitization and LS from RF.
Collapse
Affiliation(s)
- Hanna Opiol
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Nuria de Zavalia
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Tara Delorme
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Pavel Solis
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Spencer Rutherford
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Uri Shalev
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
| | - Shimon Amir
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, QC, Canada
- * E-mail:
| |
Collapse
|
64
|
Berridge MJ. Vitamin D and Depression: Cellular and Regulatory Mechanisms. Pharmacol Rev 2017; 69:80-92. [DOI: 10.1124/pr.116.013227] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
65
|
Flores-Ramos M, Leff P, Fernández-Guasti A, Becerra Palars C. Is it important to consider the sex of the patient when using lithium or valproate to treat the bipolar disorder? Pharmacol Biochem Behav 2017; 152:105-111. [DOI: 10.1016/j.pbb.2016.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 02/03/2016] [Accepted: 02/09/2016] [Indexed: 01/12/2023]
|
66
|
Ricq EL, Hooker JM, Haggarty SJ. Toward development of epigenetic drugs for central nervous system disorders: Modulating neuroplasticity via H3K4 methylation. Psychiatry Clin Neurosci 2016; 70:536-550. [PMID: 27485392 PMCID: PMC5764164 DOI: 10.1111/pcn.12426] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2016] [Indexed: 12/19/2022]
Abstract
The mammalian brain dynamically activates or silences gene programs in response to environmental input and developmental cues. This neuroplasticity is controlled by signaling pathways that modify the activity, localization, and/or expression of transcriptional-regulatory enzymes in combination with alterations in chromatin structure in the nucleus. Consistent with this key neurobiological role, disruptions in the fine-tuning of epigenetic and transcriptional regulation have emerged as a recurrent theme in studies of the genetics of neurodevelopmental and neuropsychiatric disorders. Furthermore, environmental factors have been implicated in the increased risk of heterogeneous, multifactorial, neuropsychiatric disorders via epigenetic mechanisms. Aberrant epigenetic regulation of gene expression thus provides an attractive unifying model for understanding the complex risk architecture of mental illness. Here, we review emerging genetic evidence implicating dysregulation of histone lysine methylation in neuropsychiatric disease and outline advancements in small-molecule probes targeting this chromatin modification. The emerging field of neuroepigenetic research is poised to provide insight into the biochemical basis of genetic risk for diverse neuropsychiatric disorders and to develop the highly selective chemical tools and imaging agents necessary to dissect dynamic transcriptional-regulatory mechanisms in the nervous system. On the basis of these findings, continued advances may lead to the validation of novel, disease-modifying therapeutic targets for a range of disorders with aberrant chromatin-mediated neuroplasticity.
Collapse
Affiliation(s)
- Emily L. Ricq
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
- Chemical Neurobiology Laboratory, Center for Human Genetic Research, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Jacob M. Hooker
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Center for Human Genetic Research, Departments of Neurology & Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
67
|
Peregud DI, Panchenko LF, Gulyaeva NV. MicroRNA may regulate the content of the brain-derived neurotrophic factor in the frontal cortex of rats after spontaneous morphine withdrawal. NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416040085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
68
|
Hoffmann HM, Crouzin N, Moreno E, Raivio N, Fuentes S, McCormick PJ, Ortiz J, Vignes M. Long-Lasting Impairment of mGluR5-Activated Intracellular Pathways in the Striatum After Withdrawal of Cocaine Self-Administration. Int J Neuropsychopharmacol 2016; 20:72-82. [PMID: 27744406 PMCID: PMC5412585 DOI: 10.1093/ijnp/pyw086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/22/2016] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Cocaine addiction continues to be a major heath concern, and despite public health intervention there is a lack of efficient pharmacological treatment options. A newly identified potential target are the group I metabotropic glutamate receptors, with allosteric modulators showing particular promise. METHODS We evaluated the capacity of group I metabotropic glutamate receptors to induce functional responses in ex vivo striatal slices from rats with (1) acute cocaine self-administration, (2) chronic cocaine self-administration, and (3) 60 days cocaine self-administration withdrawal by Western blot and extracellular recordings of synaptic transmission. RESULTS We found that striatal group I metabotropic glutamate receptors are the principal mediator of the mGluR1/5 agonist (RS)-3,5-dihydroxyphenylglycine-induced cAMP responsive-element binding protein phosphorylation. Both acute and chronic cocaine self-administration blunted group I metabotropic glutamate receptor effects on cAMP responsive-element binding protein phosphorylation in the striatum, which correlated with the capacity to induce long-term depression, an effect that was maintained 60 days after chronic cocaine self-administration withdrawal. In the nucleus accumbens, the principal brain region mediating the rewarding effects of drugs, chronic cocaine self-administration blunted group I metabotropic glutamate receptor stimulation of extracellular signal-regulated protein kinases 1/2 and cAMP responsive-element binding protein. Interestingly, the group I metabotropic glutamate receptor antagonist/inverse-agonist, 2-methyl-6-(phenylethynyl)pyridine hydrochloride, led to a specific increase in cAMP responsive-element binding protein phosphorylation after chronic cocaine self-administration, specifically in the nucleus accumbens, but not in the striatum. CONCLUSIONS Prolonged cocaine self-administration, through withdrawal, leads to a blunting of group I metabotropic glutamate receptor responses in the striatum. In addition, specifically in the accumbens, group I metabotropic glutamate receptor signaling to cAMP responsive-element binding protein shifts from an agonist-induced to an antagonist-induced cAMP responsive-element binding protein phosphorylation.
Collapse
Affiliation(s)
- Hanne Mette Hoffmann
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Nadine Crouzin
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Estefanía Moreno
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Noora Raivio
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Silvia Fuentes
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Peter J. McCormick
- Oxidative Stress and Neuroprotection, IBMM, CNRS UMR-5247, University of Montpellier II, Montpellier, France (Drs Hoffmann, Crouzin, and Vignes); Neuroscience Institute and Department of Biochemistry and Molecular Biology, School of Medicine, Universitat Autonoma de Barcelona, Bellaterra, Spain (Dr Hoffmann, Ms Raivio, Dr Fuentes, and Dr Ortiz); Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas. Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Spain (Drs Moreno and McCormick); University of East Anglia, School of Pharmacy, NR4 7TJ, Norwich, United Kingdom (Dr McCormick)
| | - Jordi Ortiz
- Present address (H.M.H.): Department of Reproductive Medicine, 349 Leichtag Biomedical Research Building, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | | |
Collapse
|
69
|
Ka M, Kook YH, Liao K, Buch S, Kim WY. Transactivation of TrkB by Sigma-1 receptor mediates cocaine-induced changes in dendritic spine density and morphology in hippocampal and cortical neurons. Cell Death Dis 2016; 7:e2414. [PMID: 27735948 PMCID: PMC5133986 DOI: 10.1038/cddis.2016.319] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/31/2016] [Accepted: 09/09/2016] [Indexed: 01/02/2023]
Abstract
Cocaine is a highly addictive narcotic associated with dendritic spine plasticity in the striatum. However, it remains elusive whether cocaine modifies spines in a cell type-specific or region-specific manner or whether it alters different types of synapses in the brain. In addition, there is a paucity of data on the regulatory mechanism(s) involved in cocaine-induced modification of spine density. In the current study, we report that cocaine exposure differentially alters spine density, spine morphology, and the types of synapses in hippocampal and cortical neurons. Cocaine exposure in the hippocampus resulted in increased spine density, but had no significant effect on cortical neurons. Although cocaine exposure altered spine morphology in both cell types, the patterns of spine morphology were distinct for each cell type. Furthermore, we observed that cocaine selectively affects the density of excitatory synapses. Intriguingly, in hippocampal neurons cocaine-mediated effects on spine density and morphology involved sigma-1 receptor (Sig-1 R) and its downstream TrkB signaling, which were not the case in cortical neurons. Furthermore, pharmacological inhibition of Sig-1 R prevented cocaine-induced TrkB activation in hippocampal neurons. Our findings reveal a novel mechanism by which cocaine induces selective changes in spine morphology, spine density, and synapse formation, and could provide insights into the cellular basis for the cognitive impairment observed in cocaine addicts.
Collapse
Affiliation(s)
- Minhan Ka
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yeon-Hee Kook
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Woo-Yang Kim
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
70
|
Zhou L, Sun WL, Weierstall K, Minerly AC, Weiner J, Jenab S, Quinones-Jenab V. Sex differences in behavioral and PKA cascade responses to repeated cocaine administration. Psychopharmacology (Berl) 2016; 233:3527-36. [PMID: 27553823 DOI: 10.1007/s00213-016-4387-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/13/2016] [Indexed: 11/26/2022]
Abstract
Previous studies have shown sex different patterns in behavioral responses to cocaine. Here, we used between-subject experiment design to study whether sex differences exist in the development of behavioral sensitization and tolerance to repeated cocaine, as well as the role of protein kinase A (PKA) signaling cascade in this process. Ambulatory and rearing responses were recorded in male and female rats after 1 to 14 days of administration of saline or cocaine (15 mg/kg; ip). Correspondent PKA-associated signaling in the nucleus accumbens (NAc) and caudate-putamen (CPu) was measured at each time point. Our results showed that females exhibited higher cocaine-induced behavioral responses and developed behavioral sensitization and tolerance faster than males. Whereas females developed behavioral sensitization to cocaine after 2 days and tolerance after 14 days, male rats developed sensitization after 5 days. In addition, cocaine induced a sexual dimorphic pattern in the progression of neuronal adaptations on the PKA cascade signaling in region (NAc vs. CPu) and time (days of cocaine administration)-dependent manners. In general, more PKA signaling cascade changes were found in the NAc of males on day 5 and in the CPu of females with repeated cocaine injection. In addition, in females, behavioral activities positively correlated with FosB levels in the NAc and CPu and negatively correlated with Cdk5 and p35 in the CPu, while no correlation was observed in males. Our studies suggest that repeated cocaine administration induced different patterns of behavioral and molecular responses in the PKA cascade in male and female rats.
Collapse
Affiliation(s)
- Luyi Zhou
- Department of Biological Sciences, Hunter College of the City University of New York, New York, NY, 10065, USA
- Department of Biology PhD Program, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, 29208, USA
| | - Wei-Lun Sun
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, 29208, USA
- Department of Psychology, Hunter College of the City University of New York, New York, NY, 10065, USA
- Department of Biopsychology and Behavioral Neuroscience PhD Subprogram, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA
| | - Karen Weierstall
- Department of Psychology, Hunter College of the City University of New York, New York, NY, 10065, USA
- Department of Biopsychology and Behavioral Neuroscience PhD Subprogram, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA
| | - Ana Christina Minerly
- Department of Psychology, Hunter College of the City University of New York, New York, NY, 10065, USA
- Department of Biopsychology and Behavioral Neuroscience PhD Subprogram, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA
| | - Jan Weiner
- Department of Psychology, Hunter College of the City University of New York, New York, NY, 10065, USA
- Department of Biopsychology and Behavioral Neuroscience PhD Subprogram, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA
| | - Shirzad Jenab
- Department of Psychology, Hunter College of the City University of New York, New York, NY, 10065, USA
- Department of Biopsychology and Behavioral Neuroscience PhD Subprogram, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA
| | - Vanya Quinones-Jenab
- Department of Biology PhD Program, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA.
- Department of Psychology, Hunter College of the City University of New York, New York, NY, 10065, USA.
- Department of Biopsychology and Behavioral Neuroscience PhD Subprogram, Graduate School and University Center of the City University of New York, New York, NY, 10016, USA.
| |
Collapse
|
71
|
Xue B, Fitzgerald CA, Jin DZ, Mao LM, Wang JQ. Amphetamine elevates phosphorylation of eukaryotic initiation factor 2α (eIF2α) in the rat forebrain via activating dopamine D1 and D2 receptors. Brain Res 2016; 1646:459-466. [PMID: 27338925 DOI: 10.1016/j.brainres.2016.06.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 06/16/2016] [Accepted: 06/18/2016] [Indexed: 10/21/2022]
Abstract
Psychostimulants have an impact on protein synthesis, although underlying molecular mechanisms are unclear. Eukaryotic initiation factor 2α-subunit (eIF2α) is a key player in initiation of protein translation and is regulated by phosphorylation. While this factor is sensitive to changing synaptic input and is critical for synaptic plasticity, its sensitivity to stimulants is poorly understood. Here we systematically characterized responses of eIF2α to a systemic administration of the stimulant amphetamine (AMPH) in dopamine responsive regions of adult rat brains. Intraperitoneal injection of AMPH at 5mg/kg increased eIF2α phosphorylation at serine 51 in the striatum. This increase was transient. In the medial prefrontal cortex (mPFC), AMPH induced a relatively delayed phosphorylation of the factor. Pretreatment with a dopamine D1 receptor antagonist SCH23390 blocked the AMPH-stimulated eIF2α phosphorylation in both the striatum and mPFC. Similarly, a dopamine D2 receptor antagonist eticlopride reduced the effect of AMPH in the two regions. Two antagonists alone did not alter basal eIF2α phosphorylation. AMPH and two antagonists did not change the amount of total eIF2α proteins in both regions. These results demonstrate the sensitivity of eIF2α to stimulant exposure. AMPH possesses the ability to stimulate eIF2α phosphorylation in striatal and mPFC neurons in vivo in a D1 and D2 receptor-dependent manner.
Collapse
Affiliation(s)
- Bing Xue
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, MO 64108, USA
| | - Cole A Fitzgerald
- Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Dao-Zhong Jin
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, MO 64108, USA
| | - Li-Min Mao
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
72
|
Meadows JP, Guzman-Karlsson MC, Phillips S, Brown JA, Strange SK, Sweatt JD, Hablitz JJ. Dynamic DNA methylation regulates neuronal intrinsic membrane excitability. Sci Signal 2016; 9:ra83. [PMID: 27555660 DOI: 10.1126/scisignal.aaf5642] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic modifications, such as DNA cytosine methylation, contribute to the mechanisms underlying learning and memory by coordinating adaptive gene expression and neuronal plasticity. Transcription-dependent plasticity regulated by DNA methylation includes synaptic plasticity and homeostatic synaptic scaling. Memory-related plasticity also includes alterations in intrinsic membrane excitability mediated by changes in the abundance or activity of ion channels in the plasma membrane, which sets the threshold for action potential generation. We found that prolonged inhibition of DNA methyltransferase (DNMT) activity increased intrinsic membrane excitability of cultured cortical pyramidal neurons. Knockdown of the cytosine demethylase TET1 or inhibition of RNA polymerase blocked the increased membrane excitability caused by DNMT inhibition, suggesting that this effect was mediated by subsequent cytosine demethylation and de novo transcription. Prolonged DNMT inhibition blunted the medium component of the after-hyperpolarization potential, an effect that would increase neuronal excitability, and was associated with reduced expression of the genes encoding small-conductance Ca(2+)-activated K(+) (SK) channels. Furthermore, the specific SK channel blocker apamin increased neuronal excitability but was ineffective after DNMT inhibition. Our results suggested that DNMT inhibition enables transcriptional changes that culminate in decreased expression of SK channel-encoding genes and decreased activity of SK channels, thus providing a mechanism for the regulation of neuronal intrinsic membrane excitability by dynamic DNA cytosine methylation. This study has implications for human neurological and psychiatric diseases associated with dysregulated intrinsic excitability.
Collapse
Affiliation(s)
- Jarrod P Meadows
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mikael C Guzman-Karlsson
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Scott Phillips
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jordan A Brown
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sarah K Strange
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J David Sweatt
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA. Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - John J Hablitz
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
73
|
Zhu J, Zhao N, Chen Y, Zhu L, Zhong Q, Liu J, Chen T. Sodium butyrate modulates a methamphetamine-induced conditioned place preference. J Neurosci Res 2016; 95:1044-1052. [PMID: 27426635 DOI: 10.1002/jnr.23835] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/21/2016] [Accepted: 06/22/2016] [Indexed: 12/13/2022]
Abstract
Previous studies demonstrated that histone acetylation modulated the transcription of associated gene expression and thus contributed to the persistence of addictive behaviors and neuroplasticity. Nonetheless, the roles of histone acetylation in distinct phases of methamphetamine (METH)-induced conditioned place preference (CPP) remain unclear. The current study examines the effects of the histone deacetylases (HDACs) inhibitor sodium butyrate (NaB) on the acquisition, extinction, and reinstatement of METH-induced CPP in mice. Our results showed that 1 mg/kg METH induced CPP in mice after four conditioning sessions. METH-induced CPP was extinguished after three extinction training sessions and could be triggered by the same dose (1 mg/kg) of METH on the reinstatement test day. Meanwhile, NaB (400 mg/kg) per se had no effect on the natural preference of mice, but injections of NaB during the conditioning and extinction phases facilitated the acquisition and extinction of METH-induced CPP, respectively. Additionally, although the effect of a single NaB injection prior to the trigger of CPP reinstatement was not observed, repeated NaB injections during the extinction phase totally blocked the reinstatement of METH-induced CPP. Taken together, our results suggested a specific effect of histone acetylation on modulating distinct phases of METH-induced CPP and that treatment of NaB during the extinction phase not only produced beneficial effects on eliminating already established CPP but also blocked the reinstatement of METH-induced CPP. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jie Zhu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Na Zhao
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China.,Northwest University of Politics and Law School of Police, Xi'an, Shaanxi, People's Republic of China
| | - Yanjiong Chen
- Departments of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Li Zhu
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Qing Zhong
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Jian Liu
- The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Teng Chen
- College of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China.,The Key Laboratory of Health Ministry for Forensic Science, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
74
|
Raab-Graham KF, Workman ER, Namjoshi S, Niere F. Pushing the threshold: How NMDAR antagonists induce homeostasis through protein synthesis to remedy depression. Brain Res 2016; 1647:94-104. [PMID: 27125595 DOI: 10.1016/j.brainres.2016.04.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/04/2016] [Accepted: 04/07/2016] [Indexed: 12/17/2022]
Abstract
Healthy neurons have an optimal operating range, coded globally by the frequency of action potentials or locally by calcium. The maintenance of this range is governed by homeostatic plasticity. Here, we discuss how new approaches to treat depression alter synaptic activity. These approaches induce the neuron to recruit homeostatic mechanisms to relieve depression. Homeostasis generally implies that the direction of activity necessary to restore the neuron's critical operating range is opposite in direction to its current activity pattern. Unconventional antidepressant therapies-deep brain stimulation and NMDAR antagonists-alter the neuron's "depressed" state by pushing the neuron's current activity in the same direction but to the extreme edge. These therapies rally the intrinsic drive of neurons in the opposite direction, thereby allowing the cell to return to baseline activity, form new synapses, and restore proper communication. In this review, we discuss seminal studies on protein synthesis dependent homeostatic plasticity and their contribution to our understanding of molecular mechanisms underlying the effectiveness of NMDAR antagonists as rapid antidepressants. Rapid antidepressant efficacy is likely to require a cascade of mRNA translational regulation. Emerging evidence suggests that changes in synaptic strength or intrinsic excitability converge on the same protein synthesis pathways, relieving depressive symptoms. Thus, we address the question: Are there multiple homeostatic mechanisms that induce the neuron and neuronal circuits to self-correct to regulate mood in vivo? Targeting alternative ways to induce homeostatic protein synthesis may provide, faster, safer, and longer lasting antidepressants. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Kimberly F Raab-Graham
- Center for Learning and Memory, Department of Neuroscience, Institute of Neuroscience, University of Texas at Austin, Austin, TX 78712, United States; Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, United States; Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX 78712, United States.
| | - Emily R Workman
- Center for Learning and Memory, Department of Neuroscience, Institute of Neuroscience, University of Texas at Austin, Austin, TX 78712, United States; Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, United States
| | - Sanjeev Namjoshi
- Center for Learning and Memory, Department of Neuroscience, Institute of Neuroscience, University of Texas at Austin, Austin, TX 78712, United States; Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX 78712, United States
| | - Farr Niere
- Center for Learning and Memory, Department of Neuroscience, Institute of Neuroscience, University of Texas at Austin, Austin, TX 78712, United States; Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX 78712, United States
| |
Collapse
|
75
|
Tapocik JD, Ceniccola K, Mayo CL, Schwandt ML, Solomon M, Wang BD, Luu TV, Olender J, Harrigan T, Maynard TM, Elmer GI, Lee NH. MicroRNAs Are Involved in the Development of Morphine-Induced Analgesic Tolerance and Regulate Functionally Relevant Changes in Serpini1. Front Mol Neurosci 2016; 9:20. [PMID: 27047334 PMCID: PMC4805586 DOI: 10.3389/fnmol.2016.00020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 02/29/2016] [Indexed: 12/23/2022] Open
Abstract
Long-term opioid treatment results in reduced therapeutic efficacy and in turn leads to an increase in the dose required to produce equivalent pain relief and alleviate break-through or insurmountable pain. Altered gene expression is a likely means for inducing long-term neuroadaptations responsible for tolerance. Studies conducted by our laboratory (Tapocik et al., 2009) revealed a network of gene expression changes occurring in canonical pathways involved in neuroplasticity, and uncovered miRNA processing as a potential mechanism. In particular, the mRNA coding the protein responsible for processing miRNAs, Dicer1, was positively correlated with the development of analgesic tolerance. The purpose of the present study was to test the hypothesis that miRNAs play a significant role in the development of analgesic tolerance as measured by thermal nociception. Dicer1 knockdown, miRNA profiling, bioinformatics, and confirmation of high value targets were used to test the proposition. Regionally targeted Dicer1 knockdown (via shRNA) had the anticipated consequence of eliminating the development of tolerance in C57BL/6J (B6) mice, thus supporting the involvement of miRNAs in the development of tolerance. MiRNA expression profiling identified a core set of chronic morphine-regulated miRNAs (miR's 27a, 9, 483, 505, 146b, 202). Bioinformatics approaches were implemented to identify and prioritize their predicted target mRNAs. We focused our attention on miR27a and its predicted target serpin peptidase inhibitor clade I (Serpini1) mRNA, a transcript known to be intricately involved in dendritic spine density regulation in a manner consistent with chronic morphine's consequences and previously found to be correlated with the development of analgesic tolerance. In vitro reporter assay confirmed the targeting of the Serpini1 3'-untranslated region by miR27a. Interestingly miR27a was found to positively regulate Serpini1 mRNA and protein levels in multiple neuronal cell lines. Lastly, Serpini1 knockout mice developed analgesic tolerance at a slower rate than wild-type mice thus confirming a role for the protein in analgesic tolerance. Overall, these results provide evidence to support a specific role for miR27a and Serpini1 in the behavioral response to chronic opioid administration (COA) and suggest that miRNA expression and mRNA targeting may underlie the neuroadaptations that mediate tolerance to the analgesic effects of morphine.
Collapse
Affiliation(s)
- Jenica D. Tapocik
- National Institute of Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - Kristin Ceniccola
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Cheryl L. Mayo
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of MedicineBaltimore, MD, USA
| | - Melanie L. Schwandt
- National Institute of Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - Matthew Solomon
- National Institute of Alcohol Abuse and Alcoholism, National Institutes of HealthBethesda, MD, USA
| | - Bi-Dar Wang
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Truong V. Luu
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Jacqueline Olender
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Thomas Harrigan
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Thomas M. Maynard
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| | - Greg I. Elmer
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of MedicineBaltimore, MD, USA
| | - Norman H. Lee
- Department of Pharmacology and Physiology, The George Washington UniversityWashington, DC, USA
| |
Collapse
|
76
|
Zalewska T, Bielawski A, Stanaszek L, Wieczerzak K, Ziemka-Nałęcz M, Nalepa I. Imipramine administration induces changes in the phosphorylation of FAK and PYK2 and modulates signaling pathways related to their activity. Biochim Biophys Acta Gen Subj 2015; 1860:424-33. [PMID: 26620976 DOI: 10.1016/j.bbagen.2015.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/26/2015] [Accepted: 11/22/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND Antidepressants can modify neuronal functioning by affecting many levels of signal transduction pathways that are involved in neuroplasticity. We investigated whether the phosphorylation status of focal adhesion kinase (FAK/PTK2) and its homolog, PYK2/PTK2B, and their complex with the downstream effectors (Src kinase, p130Cas, and paxillin) are affected by administration of the antidepressant drug, imipramine. The treatment influence on the levels of ERK1/2 kinases and their phosphorylated forms (pERK1/2) or the Gαq, Gα11 and Gα12 proteins were also assessed. METHODS Rats were injected with imipramine (10 mg/kg, twice daily) for 21 days. The levels of proteins investigated in their prefrontal cortices were measured by Western blotting. RESULTS Imipramine induced contrasting changes in the phosphorylation of FAK and PYK2 at Tyr397 and Tyr402, respectively. The decreased FAK phosphorylation and increased PYK2 phosphorylation were reflected by changes in the levels of their complex with Src and p130Cas, which was observed predominantly after chronic imipramine treatment. Similarly only chronic imipramine decreased the Gαq expression while Gα11 and Gα12 proteins were untouched. Acute and chronic treatment with imipramine elevated ERK1 and ERK2 total protein levels, whereas only the pERK1 was significantly affected by the drug. CONCLUSION The enhanced activation of PYK2 observed here could function as compensation for FAK inhibition. GENERAL SIGNIFICANCE These data demonstrate that treatment with imipramine, which is a routine in counteracting depressive disorders, enhances the phosphorylation of PYK2, a non-receptor kinase instrumental in promoting synaptic plasticity. This effect documents as yet not considered target in the mechanism of imipramine action.
Collapse
Affiliation(s)
- Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Adam Bielawski
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Luiza Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof Wieczerzak
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Małgorzata Ziemka-Nałęcz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Irena Nalepa
- Department of Brain Biochemistry, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| |
Collapse
|
77
|
The effect of different durations of morphine exposure on mesencephalic dopaminergic neurons in morphine dependent rats. Neurotoxicology 2015; 51:51-7. [DOI: 10.1016/j.neuro.2015.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 09/04/2015] [Accepted: 09/14/2015] [Indexed: 12/28/2022]
|
78
|
Rahman S, Engleman EA, Bell RL. Recent Advances in Nicotinic Receptor Signaling in Alcohol Abuse and Alcoholism. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:183-201. [PMID: 26810002 PMCID: PMC4754113 DOI: 10.1016/bs.pmbts.2015.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alcohol is the most commonly abused legal substance and alcoholism is a serious public health problem. It is a leading cause of preventable death in the world. The cellular and molecular mechanisms of alcohol reward and addiction are still not well understood. Emerging evidence indicates that unlike other drugs of abuse, such as nicotine, cocaine, or opioids, alcohol targets numerous channel proteins, receptor molecules, and signaling pathways in the brain. Previously, research has identified brain nicotinic acetylcholine receptors (nAChRs), a heterogeneous family of pentameric ligand-gated cation channels expressed in the mammalian brain, as critical molecular targets for alcohol abuse and dependence. Genetic variations encoding nAChR subunits have been shown to increase the vulnerability to develop alcohol dependence. Here, we review recent insights into the rewarding effects of alcohol, as they pertain to different nAChR subtypes, associated signaling molecules, and pathways that contribute to the molecular mechanisms of alcoholism and/or comorbid brain disorders. Understanding these cellular changes and molecular underpinnings may be useful for the advancement of brain nicotinic-cholinergic mechanisms, and will lead to a better translational and therapeutic outcome for alcoholism and/or comorbid conditions.
Collapse
Affiliation(s)
- Shafiqur Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, South Dakota, USA.
| | - Eric A Engleman
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Richard L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
79
|
Levran O, Peles E, Randesi M, Correa da Rosa J, Ott J, Rotrosen J, Adelson M, Kreek MJ. Synaptic Plasticity and Signal Transduction Gene Polymorphisms and Vulnerability to Drug Addictions in Populations of European or African Ancestry. CNS Neurosci Ther 2015; 21:898-904. [PMID: 26384852 PMCID: PMC4619135 DOI: 10.1111/cns.12450] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/01/2015] [Accepted: 08/14/2015] [Indexed: 12/23/2022] Open
Abstract
AIM Drug addiction is characterized, in part, by deregulation of synaptic plasticity in circuits involved in reward, stress, cue learning, and memory. This study was designed to assess whether 185 variants in 32 genes central to synaptic plasticity and signal transduction contribute to vulnerability to develop heroin and/or cocaine addiction. METHODS Analyses were conducted in a sample of 1860 subjects divided according to ancestry (African and European) and drug of abuse (heroin or cocaine). RESULTS Eighteen SNPs in 11 genes (CDK5R1, EPHA4, EPHA6, FOSL2, MAPK3, MBP, MPDZ, NFKB1, NTRK2, NTSR1, and PRKCE) showed significant associations (P < 0.01), but the signals did not survive correction for multiple testing. SNP rs230530 in the NFKB1 gene, encoding the transcription regulator NF-kappa-B, was the only SNP indicated in both ancestry groups and both addictions. This SNP was previously identified in association with alcohol addiction. SNP rs3915568 in NTSR1, which encodes neurotensin receptor, and SNP rs1389752 in MPDZ, which encodes the multiple PDZ domain protein, were previously associated with heroin addiction or alcohol addiction, respectively. CONCLUSIONS The study supports the involvement of genetic variation in signal transduction pathways in heroin and cocaine addiction and provides preliminary evidence suggesting several new risk or protective loci that may be relevant for diagnosis and treatment success.
Collapse
Affiliation(s)
- Orna Levran
- The Laboratory of the Biology of Addictive DiseasesThe Rockefeller UniversityNew YorkNYUSA
| | - Einat Peles
- Dr. Miriam and Sheldon G. Adelson Clinic for Drug Abuse Treatment and ResearchTel Aviv Elias Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Matthew Randesi
- The Laboratory of the Biology of Addictive DiseasesThe Rockefeller UniversityNew YorkNYUSA
| | - Joel Correa da Rosa
- Center for Clinical and Translational ScienceThe Rockefeller UniversityNew YorkNYUSA
| | - Jurg Ott
- Institute of PsychologyChinese Academy of SciencesBeijingChina
- The Laboratory of Statistical GeneticsThe Rockefeller UniversityNew YorkNYUSA
| | - John Rotrosen
- VA New York Harbor Healthcare SystemNYU School of MedicineNew YorkNYUSA
| | - Miriam Adelson
- The Laboratory of the Biology of Addictive DiseasesThe Rockefeller UniversityNew YorkNYUSA
- Dr. Miriam and Sheldon G. Adelson Clinic for Drug Abuse Treatment and ResearchTel Aviv Elias Sourasky Medical CenterTel AvivIsrael
- Dr. Miriam and Sheldon G. Adelson Clinic for Drug Abuse Treatment and ResearchLas VegasNVUSA
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive DiseasesThe Rockefeller UniversityNew YorkNYUSA
| |
Collapse
|
80
|
Erbs E, Faget L, Ceredig RA, Matifas A, Vonesch JL, Kieffer BL, Massotte D. Impact of chronic morphine on delta opioid receptor-expressing neurons in the mouse hippocampus. Neuroscience 2015; 313:46-56. [PMID: 26480813 DOI: 10.1016/j.neuroscience.2015.10.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 11/18/2022]
Abstract
Delta opioid (DOP) receptors participate to the control of chronic pain and emotional responses. Recent data also identified their implication in spatial memory and drug-context associations pointing to a critical role of hippocampal delta receptors. To better appreciate the impact of repeated drug exposure on their modulatory activity, we used fluorescent knock-in mice that express a functional delta receptor fused at its carboxy-terminus with the green fluorescent protein in place of the native receptor. We then tested the impact of chronic morphine treatment on the density and distribution of delta receptor-expressing cells in the hippocampus. A decrease in delta receptor-positive cell density was observed in the CA1, CA3 and dentate gyrus without alteration of the distribution across the different GABAergic populations that mainly express delta receptors. This effect partly persisted after four weeks of morphine abstinence. In addition, we observed increased DOP receptor expression at the cell surface compared to saline-treated animals. In the hippocampus, chronic morphine administration thus induces DOP receptor cellular redistribution and durably decreases delta receptor-expressing cell density. Such modifications are likely to alter hippocampal physiology, and to contribute to long-term cognitive deficits.
Collapse
Affiliation(s)
- E Erbs
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - L Faget
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - R A Ceredig
- Institut des Neurosciences Cellulaires et Intégratives, UPR 3212, 5 Rue Blaise Pascal, F-67084 Strasbourg, France
| | - A Matifas
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - J-L Vonesch
- Imaging Center IGBMC, CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France
| | - B L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France; Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875, Boulevard LaSalle, Montreal (Quebec) H4H 1R3, Canada
| | - D Massotte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS/INSERM/UdS, 1 Rue Laurent Fries, F-67404 Illkirch-Graffenstaden, France; Institut des Neurosciences Cellulaires et Intégratives, UPR 3212, 5 Rue Blaise Pascal, F-67084 Strasbourg, France.
| |
Collapse
|
81
|
Bacopa monniera (CDRI-08) Upregulates the Expression of Neuronal and Glial Plasticity Markers in the Brain of Scopolamine Induced Amnesic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:837012. [PMID: 26413129 PMCID: PMC4564643 DOI: 10.1155/2015/837012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/20/2015] [Indexed: 12/18/2022]
Abstract
Preclinical studies on animal models have discerned the antiamnesic and memory-enhancing potential of Bacopa monniera (Brahmi) crude extract and standardized extracts. These studies primarily focus on behavioral consequences. However, lack of information on molecular underpinnings has limited the clinical trials of the potent herb in human subjects. In recent years, researchers highlight plasticity markers as molecular correlates of amnesia and being crucial to design therapeutic targets. In the present report, we have investigated the effect of a special extract of B. monniera (CDRI-08) on the expression of key neuronal (BDNF and Arc) and glial (GFAP) plasticity markers in the cerebrum of scopolamine induced amnesic mice. Pre- and postadministration of CDRI-08 ameliorated amnesic effect of scopolamine by decreasing acetyl cholinesterase activity and drastically upregulating the mRNA and protein expression of BDNF, Arc, and GFAP in mouse cerebrum. Interestingly, the plant extract per se elevated BDNF and Arc expression as compared to control but GFAP was unaltered. In conclusion, our findings provide the first molecular evidence for antiamnesic potential of CDRI-08 via enhancement of both neuronal and glial plasticity markers. Further investigations on detailed molecular pathways would encourage therapeutic application of the extract in memory disorders.
Collapse
|
82
|
Tognini P, Napoli D, Pizzorusso T. Dynamic DNA methylation in the brain: a new epigenetic mark for experience-dependent plasticity. Front Cell Neurosci 2015; 9:331. [PMID: 26379502 PMCID: PMC4548453 DOI: 10.3389/fncel.2015.00331] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/10/2015] [Indexed: 02/04/2023] Open
Abstract
Experience-dependent plasticity is the ability of brain circuits to undergo molecular, structural and functional changes as a function of neural activity. Neural activity continuously shapes our brain during all the stages of our life, from infancy through adulthood and beyond. Epigenetic modifications of histone proteins and DNA seem to be a leading molecular mechanism to modulate the transcriptional changes underlying the fine-tuning of synaptic connections and circuitry rewiring during activity-dependent plasticity. The recent discovery that cytosine methylation is an epigenetic mark particularly dynamic in brain cells has strongly increased the interest of neuroscientists in understanding the role of covalent modifications of DNA in activity-induced remodeling of neuronal circuits. Here, we provide an overview of the role of DNA methylation and hydroxylmethylation in brain plasticity both during adulthood, with emphasis on learning and memory related processes, and during postnatal development, focusing specifically on experience-dependent plasticity in the visual cortex.
Collapse
Affiliation(s)
- Paola Tognini
- Department of Biological Chemistry, University of California, Irvine Irvine, CA, USA
| | - Debora Napoli
- BioSNS laboratory, Scuola Normale Superiore di Pisa Pisa, Italy
| | - Tommaso Pizzorusso
- Institute of Neuroscience CNR Pisa, Italy ; Department of Neuroscience, Psychology, Drug Research and Child Health Neurofarba, University of Florence Florence, Italy
| |
Collapse
|
83
|
Piechota M, Golda S, Ficek J, Jantas D, Przewlocki R, Korostynski M. Regulation of alternative gene transcription in the striatum in response to antidepressant drugs. Neuropharmacology 2015; 99:328-36. [PMID: 26254862 DOI: 10.1016/j.neuropharm.2015.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 05/21/2015] [Accepted: 08/03/2015] [Indexed: 12/13/2022]
Abstract
The mechanisms that control the selection of transcription initiation and termination sites in response to pharmacological stimulation of neuronal cells are poorly understood. We used next-generation sequencing and bioinformatics to construct a genome-wide inventory of protein-coding and non-coding transcripts altered by antidepressant treatment. We analyzed available ChIP-seq data to identify mechanisms that control drug-inducible expression of alternative gene variants in the brain. We identified 153 transcripts of various biotypes regulated in the mouse striatum in response to tranylcypromine or mianserin (at a 0.1% FDR threshold). Five drug-responsive gene patterns are enriched in protein-coding variants (77%), regulated by different sets of transcriptional factors (including SRF/CREB1 and GR/CTCF) and expressed in separate cellular compartments of the brain. We found that alterations mediated by proximal promoters in neurons are more specific in the selection of regulated transcriptional isoforms compared with enhancer-dependent alterations in glia. The change in transcriptional programs, from housekeeping to inducible, provides cells with the resource of functionally distinct proteins. We conclude that the regulation of drug-induced brain plasticity may occur at the level of transcripts rather than genes. The expression of specific isoforms in response to antidepressants may constitute a trigger that initiates the long-lasting effects of these drugs.
Collapse
Affiliation(s)
- Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Slawomir Golda
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Joanna Ficek
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland
| | - Danuta Jantas
- Department of Experimental Neuroendocrinology, Institute of Pharmacology PAS, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland; Department of Neurobiology and Neuropsychology, IPS, UJ, Krakow, Poland
| | - Michal Korostynski
- Department of Molecular Neuropharmacology, Institute of Pharmacology PAS, Krakow, Poland.
| |
Collapse
|
84
|
Wetzell BB, Muller MM, Flax SM, King HE, DeCicco-Skinner K, Riley AL. Effect of preexposure on methylphenidate-induced taste avoidance and related BDNF/TrkB activity in the insular cortex of the rat. Psychopharmacology (Berl) 2015; 232:2837-47. [PMID: 25893639 DOI: 10.1007/s00213-015-3924-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 03/16/2015] [Indexed: 12/19/2022]
Abstract
RATIONALE Exogenous brain-derived neurotrophic factor (BDNF) in the insular cortex (IC) is known to influence conditioned taste avoidance (CTA) learning, but little is known of its endogenous role in the phenomenon. Preexposure to many abusable compounds attenuates their ability to induce CTA, thus providing a possible platform from which to further elucidate the endogenous role of IC BDNF in CTA. OBJECTIVES The role of IC BDNF in CTA learning was examined by assessing the effect of preexposure to methylphenidate (MPH) on MPH-induced CTA, followed by expression between preexposure groups of BDNF in the IC, central nucleus of the amygdala (CeA), and the nucleus accumbens (NAc). METHODS Following preexposure to MPH (18 mg/kg), CTAs induced by MPH (0, 10, 18, and 32 mg/kg) were assessed in adult male Sprague-Dawley rats (n = 64). In separate groups (n = 31), differences in BDNF and tropomyosin-related kinase receptor-B (TrkB) were assessed using Western blots following similar preexposure and conditioning procedures. RESULTS Preexposure to MPH significantly blunted MPH-CTA compared to preexposure to vehicle. Unexpectedly, there were no significant effects of MPH on BDNF activity following CTA, but animals preexposed to MPH exhibited decreased activity in the CeA and enhanced activity in the IC and NAc. CONCLUSIONS Preexposure to MPH attenuates its aversive effects on subsequent presentations, and BDNF's impact on CTA learning may be dependent upon its temporal relation to other CTA-related intracellular cascades.
Collapse
Affiliation(s)
- B Bradley Wetzell
- Psychopharmacology Laboratory, Department of Psychology, American University, Washington, DC, 20016, USA,
| | | | | | | | | | | |
Collapse
|
85
|
Collins GT, Chen Y, Tschumi C, Rush EL, Mensah A, Koek W, France CP. Effects of consuming a diet high in fat and/or sugar on the locomotor effects of acute and repeated cocaine in male and female C57BL/6J mice. Exp Clin Psychopharmacol 2015; 23:228-37. [PMID: 26237320 PMCID: PMC4523901 DOI: 10.1037/pha0000019] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Drug abuse and obesity are serious public health problems. Dopamine plays a central role in mediating the reinforcing effects of drugs and food. Prolonged use of drugs is known to alter the function and/or sensitivity of many neurotransmitter systems, including dopamine; however, the impact of consuming foods high in fat and/or sugar is less clear. These studies characterized the locomotor effects of acute and repeated cocaine in male and female C57BL/6J mice consuming 1 of 4 diets: (a) standard chow + water; (b) standard chow + 10% sucrose solution; (c) high-fat chow + water; or (d) high-fat chow + 10% sucrose solution. The acute locomotor effects of cocaine (3.2-32.0 mg/kg) were evaluated 4 weeks after initiating dietary conditions; the effects of repeated cocaine administration were evaluated after 5, 6, 7, and 12 weeks. During acute tests, mice consuming a diet high in fat and/or sucrose exhibited greater locomotor responses to cocaine than mice consuming standard chow and water, regardless of sex. Although diet-induced enhancements persisted across repeated cocaine testing, locomotor sensitization developed more rapidly in females drinking sucrose (and consuming either standard or high-fat chow) than in females consuming standard chow and water. In addition to providing evidence that consuming a diet high in fat and/or sugar enhances abuse-related effects of cocaine in ways that might increase vulnerability to abuse cocaine, these studies identified a potentially important sex-related difference in the interaction between nutrition and cocaine effects, with the impacts of sucrose consumption being greater in females than in males.
Collapse
Affiliation(s)
- Gregory T Collins
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
| | - Yu Chen
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
| | - Chris Tschumi
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
| | - Elise L Rush
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
| | - Ayele Mensah
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
| | - Wouter Koek
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
| | - Charles P France
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
| |
Collapse
|
86
|
Lister R, Mukamel EA. Turning over DNA methylation in the mind. Front Neurosci 2015; 9:252. [PMID: 26283895 PMCID: PMC4519686 DOI: 10.3389/fnins.2015.00252] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/06/2015] [Indexed: 01/22/2023] Open
Abstract
Cytosine DNA methylation is a stable epigenetic modification with established roles in regulating transcription, imprinting, female X-chromosome inactivation, and silencing of transposons. Dynamic gain or loss of DNA methylation reshapes the genomic landscape of cells during early differentiation, and in post-mitotic mammalian brain cells these changes continue to accumulate throughout the phases of cortical maturation in childhood and adolescence. There is also evidence for dynamic changes in the methylation status of specific genomic loci during the encoding of new memories, and these epigenome dynamics could play a causal role in memory formation. However, the mechanisms that may dynamically regulate DNA methylation in neurons during memory formation and expression, and the function of such epigenomic changes in this context, are unclear. Here we discuss the possible roles of DNA methylation in encoding and retrieval of memory.
Collapse
Affiliation(s)
- Ryan Lister
- ARC Center of Excellence in Plant Energy Biology, The University of Western Australia Perth, WA, Australia ; The Harry Perkins Institute of Medical Research Perth, WA, Australia
| | - Eran A Mukamel
- Department of Cognitive Science, University of California San Diego La Jolla, CA, USA
| |
Collapse
|
87
|
Cattaneo A, Macchi F, Plazzotta G, Veronica B, Bocchio-Chiavetto L, Riva MA, Pariante CM. Inflammation and neuronal plasticity: a link between childhood trauma and depression pathogenesis. Front Cell Neurosci 2015; 9:40. [PMID: 25873859 PMCID: PMC4379909 DOI: 10.3389/fncel.2015.00040] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/27/2015] [Indexed: 12/13/2022] Open
Abstract
During the past two decades, there has been increasing interest in understanding and characterizing the role of inflammation in major depressive disorder (MDD). Indeed, several are the evidences linking alterations in the inflammatory system to Major Depression, including the presence of elevated levels of pro-inflammatory cytokines, together with other mediators of inflammation. However, it is still not clear whether inflammation represents a cause or whether other factors related to depression result in these immunological effects. Regardless, exposure to early life stressful events, which represent a vulnerability factor for the development of psychiatric disorders, act through the modulation of inflammatory responses, but also of neuroplastic mechanisms over the entire life span. Indeed, early life stressful events can cause, possibly through epigenetic changes that persist over time, up to adulthood. Such alterations may concur to increase the vulnerability to develop psychopathologies. In this review we will discuss the role of inflammation and neuronal plasticity as relevant processes underlying depression development. Moreover, we will discuss the role of epigenetics in inducing alterations in inflammation-immune systems as well as dysfunction in neuronal plasticity, thus contributing to the long-lasting negative effects of stressful life events early in life and the consequent enhanced risk for depression. Finally we will provide an overview on the potential role of inflammatory system to aid diagnosis, predict treatment response, enhance treatment matching, and prevent the onset or relapse of Major Depression.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London London, UK ; IRCCS Centro S Giovanni di Dio, Fatebenefratelli Brescia, Italy
| | - Flavia Macchi
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| | - Giona Plazzotta
- IRCCS Centro S Giovanni di Dio, Fatebenefratelli Brescia, Italy
| | - Begni Veronica
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| | - Luisella Bocchio-Chiavetto
- IRCCS Centro S Giovanni di Dio, Fatebenefratelli Brescia, Italy ; Faculty of Psychology, eCampus University Novedrate (Como), Italy
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan Milan, Italy
| | - Carmine Maria Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London London, UK
| |
Collapse
|
88
|
Kudryashova IV. Neurodegenerative changes in depression: Excitotoxicity or a deficit of trophic factors? NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415010043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
89
|
Rapid antidepressants stimulate the decoupling of GABA(B) receptors from GIRK/Kir3 channels through increased protein stability of 14-3-3η. Mol Psychiatry 2015; 20:298-310. [PMID: 25560757 PMCID: PMC4357863 DOI: 10.1038/mp.2014.165] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 10/20/2014] [Accepted: 10/22/2014] [Indexed: 12/28/2022]
Abstract
A single injection of N-methyl-D-aspartate receptor (NMDAR) antagonists produces a rapid antidepressant response. Lasting changes in the synapse structure and composition underlie the effectiveness of these drugs. We recently discovered that rapid antidepressants cause a shift in the γ-aminobutyric acid receptor (GABABR) signaling pathway, such that GABABR activation shifts from opening inwardly rectifiying potassium channels (Kir/GIRK) to increasing resting dendritic calcium signal and mammalian Target of Rapamycin activity. However, little is known about the molecular and biochemical mechanisms that initiate this shift. Herein, we show that GABABR signaling to Kir3 (GIRK) channels decreases with NMDAR blockade. Blocking NMDAR signaling stabilizes the adaptor protein 14-3-3η, which decouples GABABR signaling from Kir3 and is required for the rapid antidepressant efficacy. Consistent with these results, we find that key proteins involved in GABABR signaling bidirectionally change in a depression model and with rapid antidepressants. In socially defeated rodents, a model for depression, GABABR and 14-3-3η levels decrease in the hippocampus. The NMDAR antagonists AP5 and Ro-25-6981, acting as rapid antidepressants, increase GABABR and 14-3-3η expression and decrease Kir3.2. Taken together, these data suggest that the shift in GABABR function requires a loss of GABABR-Kir3 channel activity mediated by 14-3-3η. Our findings support a central role for 14-3-3η in the efficacy of rapid antidepressants and define a critical molecular mechanism for activity-dependent alterations in GABABR signaling.
Collapse
|
90
|
Differential and converging molecular mechanisms of antidepressants' action in the hippocampal dentate gyrus. Neuropsychopharmacology 2015; 40:338-49. [PMID: 25035085 PMCID: PMC4443946 DOI: 10.1038/npp.2014.176] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/19/2014] [Accepted: 07/11/2014] [Indexed: 01/07/2023]
Abstract
Major depression is a highly prevalent, multidimensional disorder. Although several classes of antidepressants (ADs) are currently available, treatment efficacy is limited, and relapse rates are high; thus, there is a need to find better therapeutic strategies. Neuroplastic changes in brain regions such as the hippocampal dentate gyrus (DG) accompany depression and its amelioration with ADs. In this study, the unpredictable chronic mild stress (uCMS) rat model of depression was used to determine the molecular mediators of chronic stress and the targets of four ADs with different pharmacological profiles (fluoxetine, imipramine, tianeptine, and agomelatine) in the hippocampal DG. All ADs, except agomelatine, reversed the depression-like behavior and neuroplastic changes produced by uCMS. Chronic stress induced significant molecular changes that were generally reversed by fluoxetine, imipramine, and tianeptine. Fluoxetine primarily acted on neurons to reduce the expression of pro-inflammatory response genes and increased a set of genes involved in cell metabolism. Similarities were found between the molecular actions and targets of imipramine and tianeptine that activated pathways related to cellular protection. Agomelatine presented a unique profile, with pronounced effects on genes related to Rho-GTPase-related pathways in oligodendrocytes and neurons. These differential molecular signatures of ADs studied contribute to our understanding of the processes implicated in the onset and treatment of depression-like symptoms.
Collapse
|
91
|
Forsyth R, Basu A. The promotion of recovery through rehabilitation after acquired brain injury in children. Dev Med Child Neurol 2015; 57:16-22. [PMID: 25200439 DOI: 10.1111/dmcn.12575] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/15/2014] [Indexed: 12/12/2022]
Abstract
A degree of motor recovery is typically seen after acquired brain injury in children. The extent to which rehabilitation efforts can claim credit for this is disputed. Strong correlations between late impairment outcomes and early severity and impairment indices are seen both in adults and children. These correlations have been interpreted by some as evidence that recovery is largely intrinsic and that any additional rehabilitation effects are small. Such views are belied by published animal studies demonstrating the possibility of large rehabilitation effects. Animal models suggest that to achieve similar rehabilitation treatment effect sizes in clinical practice, rehabilitation 'doses' should be greater, rehabilitation efforts should start sooner, and premature accommodation of impairment should be avoided.
Collapse
Affiliation(s)
- Rob Forsyth
- Institute of Neuroscience, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle University, Newcastle upon Tyne, Newcastle, UK
| | | |
Collapse
|
92
|
Musazzi L, Seguini M, Mallei A, Treccani G, Pelizzari M, Tornese P, Racagni G, Tardito D. Time-dependent activation of MAPK/Erk1/2 and Akt/GSK3 cascades: modulation by agomelatine. BMC Neurosci 2014; 15:119. [PMID: 25332063 PMCID: PMC4207903 DOI: 10.1186/s12868-014-0119-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/09/2014] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The novel antidepressant agomelatine, a melatonergic MT1/MT2 agonist combined with 5-HT2c serotonin antagonist properties, showed antidepressant action in preclinical and clinical studies. There is a general agreement that the therapeutic action of antidepressants needs the activation of slow-onset adaptations in downstream signalling pathways finally regulating neuroplasticity. In the last several years, particular attention was given to cAMP-responsive element binding protein (CREB)-related pathways, since it was shown that chronic antidepressants increase CREB phosphorylation and transcriptional activity, through the activation of calcium/calmodulin-dependent (CaM) and mitogen activated protein kinase cascades (MAPK/Erk1/2). Aim of this work was to analyse possible effects of chronic agomelatine on time-dependent changes of different intracellular signalling pathways in hippocampus and prefrontal/frontal cortex of male rats. To this end, measurements were performed 1 h or 16 h after the last agomelatine or vehicle injection. RESULTS We have found that in naïve rats chronic agomelatine, contrary to traditional antidepressants, did not increase CREB phosphorylation, but modulates the time-dependent regulation of MAPK/Erk1/2 and Akt/glycogen synthase kinase-3 (GSK-3) pathways. CONCLUSION Our results suggest that the intracellular molecular mechanisms modulated by chronic agomelatine may be partly different from those of traditional antidepressants and involve the time-dependent regulation of MAPK/Erk1/2 and Akt/GSK-3 signalling pathways. This could exert a role in the antidepressant efficacy of the drug.
Collapse
Affiliation(s)
- Laura Musazzi
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy.
| | - Mara Seguini
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy.
| | - Alessandra Mallei
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy.
| | - Giulia Treccani
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy.
| | - Mariagrazia Pelizzari
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy.
| | - Paolo Tornese
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy.
| | - Giorgio Racagni
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy. .,Istituto di Ricovero e Cura a Carattere Scientifico Centro S. Giovanni di Dio-Fatebenefratelli, Brescia, Italy.
| | - Daniela Tardito
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases (CEND), Università degli Studi di Milano, Via Balzaretti 9, Milano, 20133, Italy.
| |
Collapse
|
93
|
Kong LY, Li GP, Yang P, Wu W, Shi JH, Li XL, Wang WZ. Identification of gene expression profile in the rat brain resulting from acute alcohol intoxication. Mol Biol Rep 2014; 41:8303-17. [PMID: 25218841 DOI: 10.1007/s11033-014-3731-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
This study aimed to identify gene expression profile in the rat brain resulting from acute alcohol intoxication (AAI). Eighteen SD rats were divided into the alcohol-treated group (n = 9) and saline control group (n = 9). Periorbital blood samples were taken to determine their blood alcohol content by gas chromatography. Tissue sections were analyzed by H and E staining and biochemical assays. Real-time reverse transcription PCR was used to validate microarray data. Statistical analysis was carried out using SPSS18.0 software (Version 18.0, SPSS Inc., Chicago, IL, USA). H and E staining demonstrated that alcohol-treated rats showed no obvious pathological changes in nerve cells compared with those in the control group. Biochemical tests revealed that alcohol-treated rats had lower superoxide dismutase activity than those in the control group (167.3 ± 10.3 U/mg vs. 189.2 ± 5.9 U/mg, P < 0.05). Furthermore, the malondialdehyde levels in alcohol-treated rats were higher than those in the control group (3.48 ± 0.24 mmol/mg vs. 2.51 ± 0.23 mmol/mg, P < 0.05). Microarray data presented 366 up-regulated genes and 300 down-regulated genes in the AAI rat brain. Gene ontology analysis identified 31 genes up-regulated and 39 down-regulated among all differentially expressed genes. Twenty-four pathways showed significant differences, including 12 pathways involved with up-regulated genes and 12 pathways involved with down-regulated genes. Selected genes showed significantly different expression in both alcohol-treated and control groups (P < 0.05). Gene expression analysis enabled clustering of alcohol intoxication-related genes by function. These genes expression may be potential targets for treatment or drug screening for acute alcohol intoxication.
Collapse
Affiliation(s)
- Ling-Yu Kong
- Department of Emergency, The First Affiliated Hospital of Xinxiang Medical University, No. 88 Health Road, Weihui, 453100, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
94
|
dela Peña I, de la Peña JB, Kim BN, Han DH, Noh M, Cheong JH. Gene expression profiling in the striatum of amphetamine-treated spontaneously hypertensive rats which showed amphetamine conditioned place preference and self-administration. Arch Pharm Res 2014; 38:865-75. [PMID: 25163681 DOI: 10.1007/s12272-014-0470-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/19/2014] [Indexed: 12/11/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD), the most commonly diagnosed neurobehavioral disorder of childhood, is usually treated with psychostimulants (e.g., amphetamine). Little is known about the neuronal and behavioral consequences of chronic amphetamine use or abuse in individuals with ADHD. Of all ADHD animal models, the spontaneously hypertensive rat (SHR) is the most validated and widely used. Here, we analyzed striatal transcriptomes in amphetamine-pretreated SHRs (5 mg/kg, i.p. for 7 days [twice daily]), which showed a conditioned place preference to and self-administration of amphetamine. Microarray analyses revealed increased mRNA expression of 55 genes (>1.65-fold increase), while 17 genes were downregulated (<0.6-fold) in the striatum of SHRs. The main functional categories overrepresented among the differentially expressed genes in the striatum include those involved in transcription (e.g., Cebpb, Per2), genes associated with angiogenesis (e.g., Kdr, Klf5), cell adhesion (e.g., Col11a1, Ctgf), apoptosis (e.g., Nfkbia, Perp) and neuronal development (e.g., Egr2, Nr4a3). In conclusion, we dissected the striatal transcriptional responses to the reinforcing effects of repeated amphetamine treatment in the SHR model of ADHD. Future studies should determine the influence of these altered transcripts on amphetamine reinforcement in amphetamine-treated SHRs, and the clinical relevance of the present findings with regard to amphetamine use/abuse in ADHD individuals.
Collapse
Affiliation(s)
- Ike dela Peña
- Uimyung Research Institute for Neuroscience, Sahmyook University, 26-21 Kongreung-2-dong, Hwarangro-815, Nowon-gu, Seoul, 139-742, Korea
| | | | | | | | | | | |
Collapse
|
95
|
Roni MA, Rahman S. The effects of lobeline on nicotine withdrawal-induced depression-like behavior in mice. Psychopharmacology (Berl) 2014; 231:2989-98. [PMID: 24682499 DOI: 10.1007/s00213-014-3472-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 01/27/2014] [Indexed: 01/12/2023]
Abstract
RATIONALE Evidence suggests that neuronal nicotinic acetylcholine receptor (nAChR) ligand lobeline has antidepressant-like properties. OBJECTIVES The present study investigated the effects of lobeline on nicotine withdrawal-induced depression-like behavior. METHODS Adult C57BL/6J mice were exposed to nicotine (200 μg/ml) in drinking solution for 3 weeks. During withdrawal, depression-like behavior was measured by the forced swim test (FST). We also determined norepinephrine (NE) levels in the prefrontal cortex (PFC) and hippocampus during nicotine withdrawal. Furthermore, we determined the effects of repeated treatment with lobeline or a selective α4β2 nAChR ligand 3-(pyridine-3́-yl)-cytisine on brain-derived neurotrophic factor (BDNF) and phosphorylated cAMP-responsive element binding (p-CREB) protein expression in the hippocampus. RESULTS Withdrawal from chronic nicotine increased immobility time in the FST, a measure for depression-like behavior. Pretreatment with lobeline significantly decreased immobility time during nicotine withdrawal. In addition, pretreatment with lobeline attenuated nicotine withdrawal-induced increased NE levels in the PFC and hippocampus. Further, repeated treatment with lobeline or 3-(pyridine-3́-yl)-cytisine decreased immobility time in the FST and reduced withdrawal-induced increased BDNF and p-CREB expression in the hippocampus. CONCLUSIONS Taken together, our results indicate that lobeline attenuated nicotine withdrawal-induced depression-like behavior likely by targeting brain nAChRs, noradrenergic neurotransmission, and/or hippocampal BDNF. Thus, lobeline may have some potential to prevent smoking relapse by counteracting nicotine withdrawal-induced depression in humans.
Collapse
Affiliation(s)
- Monzurul Amin Roni
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Avera Health and Science Center, SAV 265, Brookings, SD, 57007, USA
| | | |
Collapse
|
96
|
Dinai Y, Wolf L, Assaf Y. Combined neuroimaging and gene expression analysis of the genetic basis of brain plasticity indicates across species homology. Hum Brain Mapp 2014; 35:5888-902. [PMID: 25053200 DOI: 10.1002/hbm.22592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/22/2014] [Accepted: 07/14/2014] [Indexed: 12/29/2022] Open
Abstract
Brain plasticity and memory formation depend on the expression of a large number of genes. This relationship had been studied using several experimental approaches and researchers have identified genes regulating plasticity through a variety of mechanisms. Despite this effort, a great deal remains unknown regarding the role of different genes in brain plasticity. Previous studies usually focused on specific brain structures and many of the genes influencing plasticity have yet to be identified. In this work, we integrate results of in vivo neuroimaging studies of plasticity with whole-brain gene expression data for the study of neuroplasticity. Brain regions, found in the imaging study to be involved in plasticity, are first spatially mapped to the anatomical framework of the genetic database. Feature ranking methods are then applied to identify genes that are differentially expressed in these regions. We find that many of our highly ranked genes are involved in synaptic transmission and that some of these genes have been previously associated with learning and memory. We show these results to be consistent when applying our method to gene expression data from four human subjects. Finally, by performing similar experiments in mice, we reveal significant cross species correlation in the ranking of genes. In addition to the identification of plasticity related candidate genes, our results also demonstrate the potential of data integration approaches as a tool to link high level phenomena such as learning and memory to underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yonatan Dinai
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | | |
Collapse
|
97
|
Abstract
The methyl-DNA binding protein MeCP2 is emerging as an important regulator of drug reinforcement processes. Psychostimulants induce phosphorylation of MeCP2 at Ser421; however, the functional significance of this posttranslational modification for addictive-like behaviors was unknown. Here we show that MeCP2 Ser421Ala knock-in mice display both a reduced threshold for the induction of locomotor sensitization by investigator-administered amphetamine and enhanced behavioral sensitivity to the reinforcing properties of self-administered cocaine. These behavioral differences were accompanied in the knock-in mice by changes in medium spiny neuron intrinsic excitability and nucleus accumbens gene expression typically observed in association with repeated exposure to these drugs. These data show that phosphorylation of MeCP2 at Ser421 functions to limit the circuit plasticities in the nucleus accumbens that underlie addictive-like behaviors.
Collapse
|
98
|
Damen D, Heumann R. MeCP2 phosphorylation in the brain: from transcription to behavior. Biol Chem 2014; 394:1595-605. [PMID: 23912219 DOI: 10.1515/hsz-2013-0193] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 07/30/2013] [Indexed: 11/15/2022]
Abstract
Methyl-CpG binding protein 2 (MeCP2), a nuclear protein highly expressed in neurons, was identified because of its ability to bind methylated DNA. In association with the transcriptional corepressor proteins Sin3a and histone deacetylases, it represses gene transcription. However, it has since become clear that MeCP2 is a multifunctional protein involved not only in transcriptional silencing but also in transcriptional activation, chromatin remodeling, and RNA splicing. Especially, its involvement in the X-linked neurologic disorder Rett syndrome emphasizes the importance of MeCP2 for normal development and maturation of the central nervous system. A number of animal models with complete or partial lack of MeCP2 functions have been generated to correlate the clinical phenotype of Rett syndrome, and studies involving different mutations of MeCP2 have shown similar effects. Animal model studies have further demonstrated that even the loss of a specific phosphorylation site of MeCP2 (S80, S421, and S424) disturbs normal maturation of the mammalian brain. This review covers recent findings regarding MeCP2 functions and its regulation by posttranslational modification, particularly MeCP2 phosphorylation and its effects on mammalian brain maturation, learning, and plasticity.
Collapse
|
99
|
Nesbitt AMI, McCurdy RD, Bryant SM, Alter MD. Total levels of hippocampal histone acetylation predict normal variability in mouse behavior. PLoS One 2014; 9:e94224. [PMID: 24788142 PMCID: PMC4008481 DOI: 10.1371/journal.pone.0094224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 03/13/2014] [Indexed: 12/01/2022] Open
Abstract
Background Genetic, pharmacological, and environmental interventions that alter total levels of histone acetylation in specific brain regions can modulate behaviors and treatment responses. Efforts have been made to identify specific genes that are affected by alterations in total histone acetylation and to propose that such gene specific modulation could explain the effects of total histone acetylation levels on behavior — the implication being that under naturalistic conditions variability in histone acetylation occurs primarily around the promoters of specific genes. Methods/Results Here we challenge this hypothesis by demonstrating with a novel flow cytometry based technique that normal variability in open field exploration, a hippocampus-related behavior, was associated with total levels of histone acetylation in the hippocampus but not in other brain regions. Conclusions Results suggest that modulation of total levels of histone acetylation may play a role in regulating biological processes. We speculate in the discussion that endogenous regulation of total levels of histone acetylation may be a mechanism through which organisms regulate cellular plasticity. Flow cytometry provides a useful approach to measure total levels of histone acetylation at the single cell level. Relating such information to behavioral measures and treatment responses could inform drug delivery strategies to target histone deacetylase inhibitors and other chromatin modulators to places where they may be of benefit while avoiding areas where correction is not needed and could be harmful.
Collapse
Affiliation(s)
- Addie May I. Nesbitt
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Richard D. McCurdy
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sharell M. Bryant
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mark D. Alter
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Psychiatry and Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
100
|
García-Pérez D, López-Bellido R, Rodríguez RE, Laorden ML, Núñez C, Milanés MV. Dysregulation of dopaminergic regulatory mechanisms in the mesolimbic pathway induced by morphine and morphine withdrawal. Brain Struct Funct 2014; 220:1901-19. [PMID: 24706046 DOI: 10.1007/s00429-014-0761-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/19/2014] [Indexed: 12/16/2022]
Abstract
Dopamine (DA) is thought to represent a teaching signal and has been implicated in the induction of addictive behaviours. Previously, it has been proposed that the transcription factors Nurr1 and Pitx3, which are critical for transcription of a set of genes involved in DA metabolism in the mesolimbic pathway, are associated with addiction pathology. The aim of our study was to investigate abnormalities in the mesolimbic pathway associated with morphine dependence and withdrawal. Using quantitative real-time PCR, immunofluorescence, HPLC and Western blotting, here we studied the effects of single morphine administration, morphine dependence and morphine withdrawal on Nurr1 and Pitx3 expression as well as on the DA marker tyrosine hydroxylase (TH) and the turnover of DA in the ventral tegmental area (VTA) and/or nucleus accumbens. We showed that the three experimental conditions caused induction of Nurr1 and Pitx3 in the VTA, which correlated with changes in TH expression during chronic morphine administration. Present data also confirmed the colocalization of Nurr1 and Pitx3 with TH-positive neurons in the posterior VTA. Furthermore, during morphine dependence, Nurr1 was detected in the nucleus compartment of VTA TH-positive neurons, whereas Pitx3 was strongly detected in the nucleus of TH-positive neurons after single morphine administration and during morphine withdrawal. The number of TH neurons, number of Nurr1 or Pitx3-positive cells, and the number of TH neurons expressing Nurr1 or Pitx3 were not modified in the subpopulations of DA neurons. Present data provide novel insight into the potential correlation between Nurr1 and Pitx3 and DA neurons plasticity during opiate addiction in the mesolimbic pathway.
Collapse
MESH Headings
- 3,4-Dihydroxyphenylacetic Acid/metabolism
- Analysis of Variance
- Animals
- Disease Models, Animal
- Dopamine/genetics
- Dopamine/metabolism
- Gene Expression Regulation/drug effects
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Imaging, Three-Dimensional
- Male
- Microscopy, Confocal
- Morphine/pharmacology
- Morphine Dependence/pathology
- Naloxone/pharmacology
- Narcotic Antagonists/pharmacology
- Narcotics/pharmacology
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Tyrosine 3-Monooxygenase/genetics
- Tyrosine 3-Monooxygenase/metabolism
- Ventral Tegmental Area/metabolism
Collapse
Affiliation(s)
- Daniel García-Pérez
- Group of Molecular and Cellular Pharmacology, Medical Faculty of Murcia, University of Murcia, Campus de Espinardo, 30100, Murcia, Spain
| | | | | | | | | | | |
Collapse
|