1
|
Schoukroun F, Befort K, Bourdy R. The rostromedial tegmental nucleus gates fat overconsumption through ventral tegmental area output in male rats. Neuropsychopharmacology 2024; 49:1569-1579. [PMID: 38570645 PMCID: PMC11319719 DOI: 10.1038/s41386-024-01855-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/27/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Excessive consumption of palatable foods that are rich in fats and sugars has contributed to the increasing prevalence of obesity worldwide. Similar to addictive drugs, such foods activate the brain's reward circuit, involving mesolimbic dopaminergic projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAc) and the prefrontal cortex. Neuroadaptations occurring in this circuit are hypothesized to contribute to uncontrolled consumption of such foods, a common feature of most of eating disorders and obesity. The rostromedial tegmental nucleus (RMTg), also named tail of the VTA (tVTA), is an inhibitory structure projecting to the VTA and the lateral hypothalamus (LH), two key brain regions in food intake regulation. Prior research has demonstrated that the RMTg responds to addictive drugs and influences their impact on mesolimbic activity and reward-related behaviors. However, the role of the RMTg in food intake regulation remains largely unexplored. The present study aimed to investigate the role of the RMTg and its projections to the VTA and the LH in regulating food intake in rats. To do so, we examined eating patterns of rats with either bilateral excitotoxic lesions of the RMTg or specific lesions of RMTg-VTA and RMTg-LH pathways. Rats were exposed to a 6-week 'free choice high-fat and high-sugar' diet, followed by a 4-week palatable food forced abstinence and a 24 h re-access period. Our results indicate that an RMTg-VTA pathway lesion increases fat consumption following 6 weeks of diet and at time of re-access. The RMTg-LH pathway lesion produces a milder effect with a decrease in global calorie intake. These findings suggest that the RMTg influences palatable food consumption and relapse through its projections to the VTA.
Collapse
Affiliation(s)
- Florian Schoukroun
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, UMR7364, CNRS, 12 Rue Goethe, 67000, Strasbourg, France
| | - Katia Befort
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, UMR7364, CNRS, 12 Rue Goethe, 67000, Strasbourg, France.
| | - Romain Bourdy
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Université de Strasbourg, UMR7364, CNRS, 12 Rue Goethe, 67000, Strasbourg, France.
| |
Collapse
|
2
|
Gomes-Ribeiro J, Martins J, Sereno J, Deslauriers-Gauthier S, Summavielle T, Coelho JE, Remondes M, Castelo-Branco M, Lopes LV. Mapping functional traces of opioid memories in the rat brain. Brain Commun 2024; 6:fcae281. [PMID: 39229487 PMCID: PMC11369824 DOI: 10.1093/braincomms/fcae281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 07/04/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
Addiction to psychoactive substances is a maladaptive learned behaviour. Contexts surrounding drug use integrate this aberrant mnemonic process and hold strong relapse-triggering ability. Here, we asked where context and salience might be concurrently represented in the brain during retrieval of drug-context paired associations. For this, we developed a morphine-conditioned place preference protocol that allows contextual stimuli presentation inside a magnetic resonance imaging scanner and investigated differences in activity and connectivity at context recall. We found context-specific responses to stimulus onset in multiple brain regions, namely, limbic, sensory and striatal. Differences in functional interconnectivity were found among amygdala, lateral habenula, and lateral septum. We also investigated alterations to resting-state functional connectivity and found increased centrality of the lateral septum in a proposed limbic network, as well as increased functional connectivity of the lateral habenula and hippocampal 'cornu ammonis' 1 region, after a protocol of associative drug-context. Finally, we found that pre- conditioned place preference resting-state connectivity of the lateral habenula and amygdala was predictive of inter-individual conditioned place preference score differences. Overall, our findings show that drug and saline-paired contexts establish distinct memory traces in overlapping functional brain microcircuits and that intrinsic connectivity of the habenula, septum, and amygdala likely underlies the individual maladaptive contextual learning to opioid exposure. We have identified functional maps of acquisition and retrieval of drug-related memory that may support the relapse-triggering ability of opioid-associated sensory and contextual cues. These findings may clarify the inter-individual sensitivity and vulnerability seen in addiction to opioids found in humans.
Collapse
Affiliation(s)
- Joana Gomes-Ribeiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - João Martins
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| | - José Sereno
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- CQC, Chemistry Department, University of Coimbra, 3004-535 Coimbra, Portugal
| | | | - Teresa Summavielle
- Addiction Biology Group, i3S- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- ESS, Polytechnic of Porto, 4200-072 Porto, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Miguel Remondes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Faculdade de Medicina Veterinária, Universidade Lusófona, 1749-024 Lisboa, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Luísa V Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
3
|
Wu J, Li X, Zhang Q, Li J, Cui R, Li X. Differential effects of intra-RMTg infusions of pilocarpine or 4-DAMP on regulating depression- and anxiety-like behaviors. Behav Brain Res 2024; 462:114833. [PMID: 38220059 DOI: 10.1016/j.bbr.2023.114833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
Depression and anxiety are associated with dysfunction of the mesolimbic dopamine system. The rostromedial tegmental nucleus (RMTg) is predominantly composed of GABAergic neurons that exhibit dense projections and strongly inhibit mesolimbic dopaminergic neurons, proposed as a major "brake" for the system. Consequently, the RMTg may be a crucial brain region for regulating these emotions. The central cholinergic system, particularly the muscarinic receptors, plays an important regulatory role in depression and anxiety. M3 muscarinic receptors are distributed on GABAergic neurons in the RMTg, but their involvement in the regulation of depression and anxiety remains uncertain. This study aimed to examine the effects of RMTg M3 muscarinic receptors on regulating depression- and anxiety-like behaviors in adult male Wistar rats, as assessed through the forced swim, tail suspension, and elevated plus maze tests. The results showed that intra-RMTg injections of the M1/M3 muscarinic receptors agonist, pilocarpine (3, 10, and 30 μg/side), or the M3 muscarinic receptors antagonist, 4-DAMP (0.5, 1, and 2 μg/side), did not alter the immobility time in the forced swim and tail suspension tests. Additionally, pilocarpine (30 μg/side) decreased time spent in open arms and increased time in closed arms in the elevated plus maze; while 4-DAMP (1 and 2 μg/side) played the opposite role by increasing time spent in open arms and decreasing time in closed arms. These findings suggest that RMTg M3 muscarinic receptors have differential effects on regulating depression- and anxiety-like behaviors. Enhancing or inhibiting these receptors can produce anxiogenic or anxiolytic effects, but have no impact on depression-like behavior. Therefore, RMTg M3 muscarinic receptors are involved in regulating anxiety and may be a potential therapeutic target for anxiolytic drugs.
Collapse
Affiliation(s)
- Jing Wu
- Beijing Key Laboratory of Learning and Cognition, Capital Normal University, Beijing, China; Faculty of Education, Henan Normal University, Xinxiang, China
| | - Xuhong Li
- Department of Education, Lyuliang University, Lyuliang, China
| | - Qi Zhang
- Beijing Key Laboratory of Learning and Cognition, Capital Normal University, Beijing, China
| | - Jiaxiang Li
- Beijing Key Laboratory of Learning and Cognition, Capital Normal University, Beijing, China
| | - Ruisi Cui
- Beijing Key Laboratory of Learning and Cognition, Capital Normal University, Beijing, China
| | - Xinwang Li
- Beijing Key Laboratory of Learning and Cognition, Capital Normal University, Beijing, China.
| |
Collapse
|
4
|
Wilczkowski M, Karwowska K, Kielbinski M, Zajda K, Pradel K, Drwięga G, Rajfur Z, Blasiak T, Przewlocki R, Solecki WB. Recruitment of inhibitory neuronal pathways regulating dopaminergic activity for the control of cocaine seeking. Eur J Neurosci 2023; 58:4487-4501. [PMID: 36479859 DOI: 10.1111/ejn.15885] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
Drug seeking is associated with the ventral tegmental area (VTA) dopaminergic (DA) activity. Previously, we have shown that brief optogenetic inhibition of VTA DA neurons with 1 s pulses delivered every 9 s attenuates cocaine seeking under extinction conditions in rats without producing overt signs of dysphoria or locomotor sedation. Whether recruitment of neuronal pathways inhibiting VTA neuronal activity would suppress drug seeking remains unknown. Here, we asked if optogenetic stimulation of the lateral habenula (LHb) efferents in the rostromedial tegmental nucleus (RMTg) as well as RMTg efferents in VTA would reduce drug seeking. To investigate this, we measured how recruitment of elements of this inhibitory pathway affects cocaine seeking in male rats under extinction conditions. The effectiveness of brief optogenetic manipulations was confirmed electrophysiologically at the level of electrical activity of VTA DA neurons. Real-time conditioned place aversion (RT-CPA) and open field tests were performed to control for potential dysphoric/sedating effects of brief optogenetic stimulation of LHb-RMTg-VTA circuitry. Optogenetic stimulation of either RMTg or LHb inhibited VTA DAergic neuron firing, whereas similar stimulation of RMTg efferents in VTA or LHb efferents in RMTg reduced cocaine seeking under extinction conditions. Moreover, stimulation of LHb-RMTg efferents produced an effect that was maintained 24 h later, during cocaine seeking test without stimulation. This effect was specific, as brief optogenetic stimulation did not affect locomotor activity and was not aversive. Our results indicate that defined inhibitory pathways can be recruited to inhibit cocaine seeking, providing potential new targets for non-pharmacological treatment of drug craving.
Collapse
Affiliation(s)
- Michał Wilczkowski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
- Department of Brain Biochemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Karolina Karwowska
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | - Michal Kielbinski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | - Katarzyna Zajda
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Gniewosz Drwięga
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | - Zenon Rajfur
- Department of Biosystems Physics, Institute of Physics, Jagiellonian University, Krakow, Poland
| | - Tomasz Blasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Wojciech B Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
5
|
Ochandarena NE, Niehaus JK, Tassou A, Scherrer G. Cell-type specific molecular architecture for mu opioid receptor function in pain and addiction circuits. Neuropharmacology 2023; 238:109597. [PMID: 37271281 PMCID: PMC10494323 DOI: 10.1016/j.neuropharm.2023.109597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/13/2023] [Indexed: 06/06/2023]
Abstract
Opioids are potent analgesics broadly used for pain management; however, they can produce dangerous side effects including addiction and respiratory depression. These harmful effects have led to an epidemic of opioid abuse and overdose deaths, creating an urgent need for the development of both safer pain medications and treatments for opioid use disorders. Both the analgesic and addictive properties of opioids are mediated by the mu opioid receptor (MOR), making resolution of the cell types and neural circuits responsible for each of the effects of opioids a critical research goal. Single-cell RNA sequencing (scRNA-seq) technology is enabling the identification of MOR-expressing cell types throughout the nervous system, creating new opportunities for mapping distinct opioid effects onto newly discovered cell types. Here, we describe molecularly defined MOR-expressing neuronal cell types throughout the peripheral and central nervous systems and their potential contributions to opioid analgesia and addiction.
Collapse
Affiliation(s)
- Nicole E Ochandarena
- Neuroscience Curriculum, Biological and Biomedical Sciences Program, The University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Jesse K Niehaus
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
6
|
Morishita M, Kobayashi K, Mitsuzuka M, Takagi R, Ono K, Momma R, Tsuneoka Y, Horio S, Tsukahara S. Two-Step Actions of Testicular Androgens in the Organization of a Male-Specific Neural Pathway from the Medial Preoptic Area to the Ventral Tegmental Area for Modulating Sexually Motivated Behavior. J Neurosci 2023; 43:7322-7336. [PMID: 37722849 PMCID: PMC10621776 DOI: 10.1523/jneurosci.0361-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/16/2023] [Accepted: 09/13/2023] [Indexed: 09/20/2023] Open
Abstract
The medial preoptic area (MPOA) is a sexually dimorphic region of the brain that regulates social behaviors. The sexually dimorphic nucleus (SDN) of the MPOA has been studied to understand sexual dimorphism, although the anatomy and physiology of the SDN is not fully understood. Here, we characterized SDN neurons that contribute to sexual dimorphism and investigated the mechanisms underlying the emergence of such neurons and their roles in social behaviors. A target-specific neuroanatomical study using transgenic mice expressing Cre recombinase under the control of Calb1, a gene expressed abundantly in the SDN, revealed that SDN neurons are divided into two subpopulations, GABA neurons projecting to the ventral tegmental area (VTA), where they link to the dopamine system (CalbVTA neurons), and GABA neurons that extend axons in the MPOA or project to neighboring regions (CalbnonVTA neurons). CalbVTA neurons were abundant in males, but were scarce or absent in females. There was no difference in the number of CalbnonVTA neurons between sexes. Additionally, we found that emergence of CalbVTA neurons requires two testicular androgen actions that occur first in the postnatal period and second in the peripubertal period. Chemogenetic analyses of CalbVTA neurons indicated a role in modulating sexual motivation in males. Knockdown of Calb1 in the MPOA reduced the intromission required for males to complete copulation. These findings provide strong evidence that a male-specific neural pathway from the MPOA to the VTA is organized by the two-step actions of testicular androgens for the modulation of sexually motivated behavior.SIGNIFICANCE STATEMENT The MPOA is a sexually dimorphic region of the brain that regulates social behaviors, although its sexual dimorphism is not fully understood. Here, we describe a population of MPOA neurons that contribute to the sexual dimorphism. These neurons only exist in masculinized brains, and they project their axons to the ventral tegmental area, where they link to the dopamine system. Emergence of such neurons requires two testicular androgen actions that occur first in the postnatal period and second in the peripubertal period. These MPOA neurons endow masculinized brains with a neural pathway from the MPOA to the ventral tegmental area and modulate sexually motivated behavior in males.
Collapse
Affiliation(s)
- Masahiro Morishita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Kaito Kobayashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Moeri Mitsuzuka
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Ryo Takagi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Kota Ono
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Rami Momma
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo 43-8540, Japan
| | - Shuhei Horio
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| |
Collapse
|
7
|
Esposito-Zapero C, Fernández-Rodríguez S, Sánchez-Catalán MJ, Zornoza T, Cano-Cebrián MJ, Granero L. The rostromedial tegmental nucleus RMTg is not a critical site for ethanol-induced motor activation in rats. Psychopharmacology (Berl) 2023; 240:2071-2080. [PMID: 37474756 PMCID: PMC10506920 DOI: 10.1007/s00213-023-06425-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/07/2023] [Indexed: 07/22/2023]
Abstract
RATIONALE Opioid drugs indirectly activate dopamine (DA) neurons in the ventral tegmental area (VTA) through a disinhibition mechanism mediated by mu opioid receptors (MORs) present both on the GABA projection neurons located in the medial tegmental nucleus/tail of the VTA (RMTg/tVTA) and on the VTA GABA interneurons. It is well demonstrated that ethanol, like opioid drugs, provokes VTA DA neuron disinhibition by interacting (through its secondary metabolite, salsolinol) with MORs present in VTA GABA interneurons, but it is not known whether ethanol could disinhibit VTA DA neurons through the MORs present in the RMTg/tVTA. OBJECTIVES The objective of the present study was to determine whether ethanol, directly microinjected into the tVTA/RMTg, is also able to induce VTA DA neurons disinhibition. METHODS Disinhibition of VTA DA neurons was indirectly assessed through the analysis of the motor activity of rats. Cannulae were placed into the tVTA/RMTg to perform microinjections of DAMGO (0.13 nmol), ethanol (150 or 300 nmol) or acetaldehyde (250 nmol) in animals pre-treated with either aCSF or the irreversible antagonist of MORs, beta-funaltrexamine (beta-FNA; 2.5 nmol). After injections, spontaneous activity was monitored for 30 min. RESULTS Neither ethanol nor acetaldehyde directly administered into the RMTg/tVTA were able to increase the locomotor activity of rats at doses that, in previous studies performed in the posterior VTA, were effective in increasing motor activities. However, microinjections of 0.13 nmol of DAMGO into the tVTA/RMTg significantly increased the locomotor activity of rats. These activating effects were reduced by local pre-treatment of rats with beta-FNA (2.5 nmol). CONCLUSIONS The tVTA/RMTg does not appear to be a key brain region for the disinhibiting action of ethanol on VTA DA neurons. The absence of dopamine in the tVTA/RMTg extracellular medium, the lack of local ethanol metabolism or both could explain the present results.
Collapse
Affiliation(s)
- Claudia Esposito-Zapero
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Facultat de Farmàcia, Universitat de València, Burjassot, Spain
| | - Sandra Fernández-Rodríguez
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Facultat de Farmàcia, Universitat de València, Burjassot, Spain
| | - María José Sánchez-Catalán
- Lab of Functional Neuroanatomy (NeuroFun-UJI-UV), Unitat Predepartamental de Medicina, Faculty of Health Sciences, Universitat Jaume I, Castellón de la Plana, Spain
| | - Teodoro Zornoza
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Facultat de Farmàcia, Universitat de València, Burjassot, Spain
| | - María José Cano-Cebrián
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Facultat de Farmàcia, Universitat de València, Burjassot, Spain.
| | - Luis Granero
- Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Facultat de Farmàcia, Universitat de València, Burjassot, Spain.
| |
Collapse
|
8
|
Yaodong C, Zhang Y, Feng G, Lei Y, Liu Q, Liu Y. Light therapy for sleep disturbance comorbid depression in relation to neural circuits and interactive hormones-A systematic review. PLoS One 2023; 18:e0286569. [PMID: 37768984 PMCID: PMC10538739 DOI: 10.1371/journal.pone.0286569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 05/19/2023] [Indexed: 09/30/2023] Open
Abstract
AIM To provide an overview of the evidence on the effect of light therapy on sleep disturbance and depression, identify the light-active neural and hormonal correlates of the effect of light therapy on sleep disturbance comorbid depression (SDCD), and construct the mechanism by which light therapy alleviates SDCD. METHODS Articles published between 1981 and 2021 in English were accessed using Science Direct, Elsevier, and Google Scholar following a three-step searching process via evolved keywords. The evidence level, reliability, and credibility of the literature were evaluated using the evidence pyramid method, which considers the article type, impact factor, and journal citation report (JCR) partition. RESULTS A total of 372 articles were collected, of which 129 articles fit the inclusion criteria and 44% were at the top of the evidence pyramid hierarchy; 50% were in the first quarter of the JCR partitions. 114 articles provided specific neural and hormonal evidence of light therapy and were further divided into three groups: 37% were related to circadian regulation circuits, 27% were related to emotional regulation circuits, and 36% were related to hormones. CONCLUSIONS First, neural and hormonal light-active pathways for alleviating sleep disturbance or depression were identified, based on which the neural correlates of SDCD were located. Second, the light responses and interactions of hormones were reviewed and summarized, which also provided a way to alleviate SDCD. Finally, the light-active LHb and SCN exert extensive regulation impacts on the circadian and emotional circuits and hormones, forming a dual-core system for alleviating SDCD.
Collapse
Affiliation(s)
- Chen Yaodong
- School of Architecture, Southwest JiaoTong University, Chengdu, China
| | - Yingzi Zhang
- School of Architecture, Southwest JiaoTong University, Chengdu, China
| | - Guo Feng
- Psychological Research and Counseling Center, Southwest Jiaotong Univerisity, Chengdu, China
| | - Yuanfang Lei
- School of Architecture, Southwest JiaoTong University, Chengdu, China
| | - Qiuping Liu
- School of Architecture, Southwest JiaoTong University, Chengdu, China
| | - Yang Liu
- School of Architecture, Southwest JiaoTong University, Chengdu, China
| |
Collapse
|
9
|
Wang J, Li Z, Tu Y, Gao F. The Dopaminergic System in the Ventral Tegmental Area Contributes to Morphine Analgesia and Tolerance. Neuroscience 2023; 527:74-83. [PMID: 37286162 DOI: 10.1016/j.neuroscience.2023.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 05/17/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023]
Abstract
Morphine has a strong analgesic effect and is suitable for various types of pain, so it is widely used. But long-term usage of morphine can lead to drug tolerance, which limits its clinical application. The complex mechanisms underlying the development of morphine analgesia into tolerance involve multiple nuclei in the brain. Recent studies reveal the signaling at the cellular and molecular levels as well as neural circuits contributing to morphine analgesia and tolerance in the ventral tegmental area (VTA), which is traditionally considered a critical center of opioid reward and addiction. Existing studies show that dopamine receptors and μ-opioid receptors participate in morphine tolerance through the altered activities of dopaminergic and/or non-dopaminergic neurons in the VTA. Several neural circuits related to the VTA are also involved in the regulation of morphine analgesia and the development of drug tolerance. Reviewing specific cellular and molecular targets and related neural circuits may provide novel precautionary strategies for morphine tolerance.
Collapse
Affiliation(s)
- Jihong Wang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ye Tu
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
Oishi Y, Saito YC, Sakurai T. GABAergic modulation of sleep-wake states. Pharmacol Ther 2023; 249:108505. [PMID: 37541595 DOI: 10.1016/j.pharmthera.2023.108505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Benzodiazepine, a classical medication utilized in the treatment of insomnia, operates by augmenting the activity of the GABAA receptor. This underscores the significance of GABAergic neurotransmission in both the initiation and maintenance of sleep. Nevertheless, an increasing body of evidence substantiates the notion that GABA-mediated neurotransmission also assumes a vital role in promoting wakefulness in specific neuronal circuits. Despite the longstanding belief in the pivotal function of GABA in regulating the sleep-wake cycle, there exists a dearth of comprehensive documentation regarding the specific regions within the central nervous system where GABAergic neurons are crucial for these functions. In this review, we delve into the involvement of GABAergic neurons in the regulation of sleep-wake cycles, with particular focus on those located in the preoptic area (POA) and ventral tegmental area (VTA). Recent research, including our own, has further underscored the importance of GABAergic neurotransmission in these areas for the regulation of sleep-wake cycles.
Collapse
Affiliation(s)
- Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki C Saito
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan; Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
11
|
Gakare SG, Ugale RR. Pharmacological evaluation of lateral habenula and rostromedial tegmental nucleus in the expression of ethanol-induced place preference. Behav Pharmacol 2023; 34:225-235. [PMID: 37171461 DOI: 10.1097/fbp.0000000000000728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Although ethanol administration produces a range of physiological effects, the rewarding aspect associated with its consumption is a major contributory factor to its abuse liability. Recently, lateral habenula (LHb) has been shown to be engaged by both rewarding and aversive stimuli. Its major glutamatergic output, the fasciculus retroflexus, projects to the rostromedial tegmental nucleus (RMTg) and controls the activity of the ventral tegmental area (VTA) dopaminergic system to promote reward circuitry. While several attempts have been made to understand the relationship between LHb and addiction, there is still a lack of knowledge in relation to ethanol addiction. In the present study, by pharmacologically exacerbating or inhibiting the LHb or RMTg neuronal activity during a post-conditioning test, we investigated the role of LHb-RMTg fasciculus retroflexus in ethanol-induced reward behavior using the conditioned place preference (CPP) test. We found that activation of LHb glutamatergic system by intra-LHb administration of l-trans-2,4-pyrrolidine dicarboxylate (PDC) (glutamate transporter inhibitor) significantly decreased CPP score; on the contrary, lamotrigine (inhibits glutamate release) significantly increased CPP score and showed a rewarding effect in CPP. Instead, intra-RMTg administration of muscimol (GABAA receptor agonist) significantly increased CPP score, whereas bicuculline (GABAA antagonist) treatment decreased CPP score. In immunohistochemistry, we found that PDC administration significantly decreased, whereas lamotrigine treatment significantly increased tyrosine hydroxylase immunoreactivity (TH-ir) in VTA and nucleus accumbens (NAc). Furthermore, while intra-RMTg administration of muscimol increased, the bicuculline treatment significantly decreased the TH-ir in VTA and NAc. Together, our behavioral and immunohistochemical results signify the role of LHb and RMTg in the expression of ethanol-conditioned reward behavior.
Collapse
Affiliation(s)
- Sukanya G Gakare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, India
| | | |
Collapse
|
12
|
Yang SH, Yang E, Lee J, Kim JY, Yoo H, Park HS, Jung JT, Lee D, Chun S, Jo YS, Pyeon GH, Park JY, Lee HW, Kim H. Neural mechanism of acute stress regulation by trace aminergic signalling in the lateral habenula in male mice. Nat Commun 2023; 14:2435. [PMID: 37105975 PMCID: PMC10140019 DOI: 10.1038/s41467-023-38180-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Stress management is necessary for vertebrate survival. Chronic stress drives depression by excitation of the lateral habenula (LHb), which silences dopaminergic neurons in the ventral tegmental area (VTA) via GABAergic neuronal projection from the rostromedial tegmental nucleus (RMTg). However, the effect of acute stress on this LHb-RMTg-VTA pathway is not clearly understood. Here, we used fluorescent in situ hybridisation and in vivo electrophysiology in mice to show that LHb aromatic L-amino acid decarboxylase-expressing neurons (D-neurons) are activated by acute stressors and suppress RMTg GABAergic neurons via trace aminergic signalling, thus activating VTA dopaminergic neurons. We show that the LHb regulates RMTg GABAergic neurons biphasically under acute stress. This study, carried out on male mice, has elucidated a molecular mechanism in the efferent LHb-RMTg-VTA pathway whereby trace aminergic signalling enables the brain to manage acute stress by preventing the hypoactivity of VTA dopaminergic neurons.
Collapse
Affiliation(s)
- Soo Hyun Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Esther Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Jaekwang Lee
- Division of Functional Food Research, Korea Food Research Institute, Wanju, 55365, South Korea
| | - Jin Yong Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Hyeijung Yoo
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Hyung Sun Park
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Jin Taek Jung
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Dongmin Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea
| | - Sungkun Chun
- Department of Physiology, Jeonbuk National University Medical School, Jeonju, 54907, South Korea
| | - Yong Sang Jo
- School of Psychology, Korea University, Seoul, 02841, South Korea
| | - Gyeong Hee Pyeon
- School of Psychology, Korea University, Seoul, 02841, South Korea
| | - Jae-Yong Park
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, 02841, South Korea
| | - Hyun Woo Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea.
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
13
|
Lyu J, Cai H, Chen Y, Chen G. Brain areas modulation in consciousness during sevoflurane anesthesia. Front Integr Neurosci 2022; 16:1031613. [PMID: 36619239 PMCID: PMC9811387 DOI: 10.3389/fnint.2022.1031613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Sevoflurane is presently one of the most used inhaled anesthetics worldwide. However, the mechanisms through which sevoflurane acts and the areas of the brain associated with changes in consciousness during anesthesia remain important and complex research questions. Sevoflurane is generally regarded as a volatile anesthetic that blindly targets neuronal (and sometimes astrocyte) GABAA receptors. This review focuses on the brain areas of sevoflurane action and their relation to changes in consciousness during anesthesia. We cover 20 years of history, from the bench to the bedside, and include perspectives on functional magnetic resonance, electroencephalogram, and pharmacological experiments. We review the interactions and neurotransmitters involved in brain circuits during sevoflurane anesthesia, improving the effectiveness and accuracy of sevoflurane's future application and shedding light on the mechanisms behind human consciousness.
Collapse
|
14
|
Kourosh-Arami M, Gholami M, Alavi-Kakhki SS, Komaki A. Neural correlates and potential targets for the contribution of orexin to addiction in cortical and subcortical areas. Neuropeptides 2022; 95:102259. [PMID: 35714437 DOI: 10.1016/j.npep.2022.102259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/01/2023]
Abstract
The orexin (hypocretin) is one of the hypothalamic neuropeptides that plays a critical role in some behaviors including feeding, sleep, arousal, reward processing, and drug addiction. This variety of functions can be described by a united function for orexins in translating states of heightened motivation, for example during physiological requirement states or following exposure to reward opportunities, into planned goal-directed behaviors. An addicted state is characterized by robust activation of orexin neurons from the environment, which triggers downstream circuits to facilitate behavior directed towards obtaining the drug. Two orexin receptors 1 (OX1R) and 2 (OX2R) are widely distributed in the brain. Here, we will introduce and describe the cortical and subcortical brain areas involved in addictive-like behaviors and the impact of orexin on addiction.
Collapse
Affiliation(s)
- Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Gholami
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran.
| | - Seyed Sajjad Alavi-Kakhki
- Student Research Committee, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
15
|
Solecki WB, Kielbinski M, Wilczkowski M, Zajda K, Karwowska K, Joanna B, Rajfur Z, Przewłocki R. Regulation of cocaine seeking behavior by locus coeruleus noradrenergic activity in the ventral tegmental area is time- and contingency-dependent. Front Neurosci 2022; 16:967969. [PMID: 35992934 PMCID: PMC9388848 DOI: 10.3389/fnins.2022.967969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Substance use disorder is linked to impairments in the ventral tegmental area (VTA) dopamine (DA) reward system. Noradrenergic (NA) inputs from locus coeruleus (LC) into VTA have been shown to modulate VTA neuronal activity, and are implicated in psychostimulant effects. Phasic LC activity controls time- and context-sensitive processes: decision making, cognitive flexibility, motivation and attention. However, it is not yet known how such temporally-distinct LC activity contributes to cocaine seeking. In a previous study we demonstrated that pharmacological inhibition of NA signaling in VTA specifically attenuates cocaine-seeking. Here, we used virally-delivered opsins to target LC neurons for inhibition or excitation, delivered onto afferents in VTA of male rats seeking cocaine under extinction conditions. Optogenetic stimulation or inhibition was delivered in distinct conditions: upon active lever press, contingently with discreet cues; or non-contingently, i.e., throughout the cocaine seeking session. Non-contingent inhibition of LC noradrenergic terminals in VTA attenuated cocaine seeking under extinction conditions. In contrast, contingent inhibition increased, while contingent stimulation reduced cocaine seeking. These findings were specific for cocaine, but not natural reward (food) seeking. Our results show that NA release in VTA drives behavior depending on timing and contingency between stimuli – context, discreet conditioned cues and reinforcer availability. We show that, depending on those factors, noradrenergic signaling in VTA has opposing roles, either driving CS-induced drug seeking, or contributing to behavioral flexibility and thus extinction.
Collapse
Affiliation(s)
- Wojciech B. Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
- *Correspondence: Wojciech B. Solecki,
| | - Michał Kielbinski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Michał Wilczkowski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
- Department of Brain Biochemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Zajda
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Karolina Karwowska
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Bernacka Joanna
- Laboratory of Pharmacology and Brain Biostructure, Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Zenon Rajfur
- Department of Biosystems Physics, Institute of Physics, Jagiellonian University, Kraków, Poland
| | - Ryszard Przewłocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
16
|
Fagiani F, Baronchelli E, Pittaluga A, Pedrini E, Scacchi C, Govoni S, Lanni C. The Circadian Molecular Machinery in CNS Cells: A Fine Tuner of Neuronal and Glial Activity With Space/Time Resolution. Front Mol Neurosci 2022; 15:937174. [PMID: 35845604 PMCID: PMC9283971 DOI: 10.3389/fnmol.2022.937174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/07/2022] [Indexed: 11/24/2022] Open
Abstract
The circadian molecular machinery is a fine timekeeper with the capacity to harmonize physiological and behavioral processes with the external environment. This tight-knit regulation is coordinated by multiple cellular clocks across the body. In this review, we focus our attention on the molecular mechanisms regulated by the clock in different brain areas and within different cells of the central nervous system. Further, we discuss evidence regarding the role of circadian rhythms in the regulation of neuronal activity and neurotransmitter systems. Not only neurons, but also astrocytes and microglia actively participate in the maintenance of timekeeping within the brain, and the diffusion of circadian information among these cells is fine-tuned by neurotransmitters (e.g., dopamine, serotonin, and γ-aminobutyric acid), thus impacting on the core clock machinery. The bidirectional interplay between neurotransmitters and the circadian clockwork is fundamental in maintaining accuracy and precision in daily timekeeping throughout different brain areas. Deepening the knowledge of these correlations allows us to define the basis of drug interventions to restore circadian rhythms, as well as to predict the onset of drug treatment/side effects that might promote daily desynchronization. Furthermore, it may lead to a deeper understanding of the potential impacts of modulations in rhythmic activities on the pace of aging and provide an insight in to the pathogenesis of psychiatric diseases and neurodegenerative disorders.
Collapse
Affiliation(s)
- Francesca Fagiani
- Institute of Experimental Neurology, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Eva Baronchelli
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Anna Pittaluga
- Department of Pharmacy (DiFar), School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- Center of Excellence for Biomedical Research, 3Rs Center, University of Genoa, Genoa, Italy
| | - Edoardo Pedrini
- Institute of Experimental Neurology, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Chiara Scacchi
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
| | - Cristina Lanni
- Department of Drug Sciences, Pharmacology Section, University of Pavia, Pavia, Italy
- Centro 3R (Inter-University Center for the Promotion of the 3Rs Principles in Teaching and Research), Italy
- *Correspondence: Cristina Lanni
| |
Collapse
|
17
|
Joshi A, Schott M, la Fleur SE, Barrot M. Role of the striatal dopamine, GABA and opioid systems in mediating feeding and fat intake. Neurosci Biobehav Rev 2022; 139:104726. [PMID: 35691472 DOI: 10.1016/j.neubiorev.2022.104726] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 12/08/2021] [Accepted: 06/05/2022] [Indexed: 10/18/2022]
Abstract
Food intake, which is a highly reinforcing behavior, provides nutrients required for survival in all animals. However, when fat and sugar consumption goes beyond the daily needs, it can favor obesity. The prevalence and severity of this health problem has been increasing with time. Besides covering nutrient and energy needs, food and in particular its highly palatable components, such as fats, also induce feelings of joy and pleasure. Experimental evidence supports a role of the striatal complex and of the mesolimbic dopamine system in both feeding and food-related reward processing, with the nucleus accumbens as a key target for reward or reinforcing-associated signaling during food intake behavior. In this review, we provide insights concerning the impact of feeding, including fat intake, on different types of receptors and neurotransmitters present in the striatal complex. Reciprocally, we also cover the evidence for a modulation of palatable food intake by different neurochemical systems in the striatal complex and in particular the nucleus accumbens, with a focus on dopamine, GABA and the opioid system.
Collapse
Affiliation(s)
- Anil Joshi
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France; Amsterdam UMC, University of Amsterdam, Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Gastroenterology & Metabolism, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Endocrinology & Metabolism, Amsterdam Neuroscience, Amsterdam, the Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands
| | - Marion Schott
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Susanne Eva la Fleur
- Amsterdam UMC, University of Amsterdam, Laboratory of Endocrinology, Department of Clinical Chemistry, Amsterdam Gastroenterology & Metabolism, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Endocrinology & Metabolism, Amsterdam Neuroscience, Amsterdam, the Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, the Netherlands.
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France.
| |
Collapse
|
18
|
Godfrey N, Qiao M, Borgland SL. Activation of LH GABAergic inputs counteracts fasting-induced changes in tVTA/RMTG neurons. J Physiol 2022; 600:2203-2224. [PMID: 35338656 DOI: 10.1113/jp282653] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS While dopamine neuronal activity changes with motivational state, it is unknown if fasting influences tVTA/RMTg GABAergic neurons, a major inhibitory input to VTA dopamine neurons. In unfasted mice, there were sex differences in inhibitory synaptic transmission onto tVTA/RMTg GABAergic neurons. Activation of LH GABAergic neurons decreases firing of tVTA/RMTg GABAergic neurons through a monosynaptic input. An acute fast decreased the excitability of tVTA/RMTg GABAergic neurons. An acute fast decreases inhibitory synaptic transmission of the LH GABA input to tVTA/RMTg GABAergic neurons in both male and female mice. ABSTRACT Dopamine neurons in the ventral tegmental area (VTA) are strongly innervated by GABAergic neurons in the 'tail of the VTA' (tVTA), also known as the rostralmedial tegmental nucleus (RMTg). Disinhibition of dopamine neurons through firing of the GABAergic neurons projecting from the lateral hypothalamus (LH) leads to reward seeking and consumption through dopamine release in the nucleus accumbens. VTA dopamine neurons respond to changes in motivational state, yet less is known of whether tVTA/RMTg GABAergic neurons or the LH GABAergic neurons that project to them are also affected by changes in motivational state, such as fasting. An acute 16 h overnight fast decreased the excitability of tVTA/RMTg GABAergic neurons of male and female mice. In addition, fasting decreased synaptic strength at LH GABA to tVTA/RMTg GABAergic synapses, indicated by reduced amplitude of optically evoked currents, decreased readily releasable pool (RRP) size and replenishment. Optical stimulation of LH GABA terminals suppressed evoked action potentials of tVTA/RMTg GABAergic neurons in unfasted mice, but this effect decreased following fasting. Furthermore, during fasting, LH GABA inputs to tVTA/RMTg neurons maintained functional connectivity during depolarization, as depolarization block was reduced following fasting. Taken together, inhibitory synaptic transmission from LH GABA inputs onto tVTA/RMTg GABAergic neurons decreases following fasting, however ability to functionally inhibit tVTA/RMTg GABAergic neurons is preserved, allowing for possible disinhibition of dopamine neurons and subsequent foraging. Abstract figure legend The inhibitory synaptic input is represented by the downward arrows. Following fasting, there was a decrease in inhibitory synaptic strength in both males and females. The action potentials represent the excitability, which also decreases in both males and females following fasting. Because both the LH GABA input and excitability of tVTA/RMTg GABA neurons have reduced activity following fasting, we predict that disinhibition of dopamine neurons with stimulation of LH inputs is preserved. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nathan Godfrey
- University of Calgary, Department of Physiology and Pharmacology, Calgary, Alberta, T2N 4N1
| | - Min Qiao
- University of Calgary, Department of Physiology and Pharmacology, Calgary, Alberta, T2N 4N1
| | - Stephanie L Borgland
- University of Calgary, Department of Physiology and Pharmacology, Calgary, Alberta, T2N 4N1
| |
Collapse
|
19
|
Gil-Lievana E, Ramírez-Mejía G, Urrego-Morales O, Luis-Islas J, Gutierrez R, Bermúdez-Rattoni F. Photostimulation of Ventral Tegmental Area-Insular Cortex Dopaminergic Inputs Enhances the Salience to Consolidate Aversive Taste Recognition Memory via D1-Like Receptors. Front Cell Neurosci 2022; 16:823220. [PMID: 35360496 PMCID: PMC8962201 DOI: 10.3389/fncel.2022.823220] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Taste memory involves storing information through plasticity changes in the neural network of taste, including the insular cortex (IC) and ventral tegmental area (VTA), a critical provider of dopamine. Although a VTA-IC dopaminergic pathway has been demonstrated, its role to consolidate taste recognition memory remains poorly understood. We found that photostimulation of dopaminergic neurons in the VTA or VTA-IC dopaminergic terminals of TH-Cre mice improves the salience to consolidate a subthreshold novel taste stimulus regardless of its hedonic value, without altering their taste palatability. Importantly, the inhibition of the D1-like receptor into the IC impairs the salience to facilitate consolidation of an aversive taste recognition memory. Finally, our results showed that VTA photostimulation improves the salience to consolidate a conditioned taste aversion memory through the D1-like receptor into the IC. It is concluded that the dopamine activity from the VTA into IC is required to increase the salience enabling the consolidation of a taste recognition memory. Notably, the D1-like receptor activity into the IC is required to consolidate both innate and learned aversive taste memories but not appetitive taste memory.
Collapse
Affiliation(s)
- Elvi Gil-Lievana
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gerardo Ramírez-Mejía
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Oscar Urrego-Morales
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Luis-Islas
- Laboratory of Neurobiology of Appetitive, Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV, Mexico City, Mexico
| | - Ranier Gutierrez
- Laboratory of Neurobiology of Appetitive, Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV, Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: Federico Bermúdez-Rattoni,
| |
Collapse
|
20
|
Sánchez-Catalán MJ, Barrot M. Fos response of the tail of the ventral tegmental area to food restriction entails a prediction error processing. Behav Brain Res 2022; 425:113826. [PMID: 35247487 DOI: 10.1016/j.bbr.2022.113826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 11/25/2022]
Abstract
The tail of the ventral tegmental area (tVTA) or rostromedial tegmental nucleus (RMTg) receives lateral habenula inputs and projects heavily to midbrain dopamine neurons. Midbrain dopamine and lateral habenula neurons participate in learning processes predicting the outcomes of actions, placing the tVTA in a critical location into prediction error pathways. tVTA GABA neurons show electrophysiological inhibition or activation after reward and aversive stimuli, respectively, and their predictive cues. tVTA molecular recruitment, however, is not elicited by all aversive stimuli. Indeed, precipitated opioid withdrawal, repeated footshocks or food restriction raise tVTA Fos expression, whereas various other unpleasant, stressful or painful stimuli does not elicit that molecular response. However, the basis of that difference remains unknown. In the present study, we tried to disentangle whether the tVTA c-Fos induction observed after food restriction was due to the aversive state of food restriction or to procedure-related reward prediction error. To this end, male Sprague-Dawley rats were food-restricted for 7-8 days. During this period, animals were handled and weighed every day before feeding. On the test day, rats underwent several behavioral procedures to explore the impact of food restriction and food-predictive cue exposure on tVTA c-Fos expression. We showed that food restriction per se was not able to recruit c-Fos in the tVTA. On the contrary, the food-predicting cues induced c-Fos locally in the absence of feeding, whereas the food-predicting cues followed by feeding evoked lower c-Fos expression. Overall, our results support the proposed involvement of the tVTA in reward prediction error.
Collapse
Affiliation(s)
- María-José Sánchez-Catalán
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, F-67000 Strasbourg, France; Unitat Predepartamental de Medicina, Universitat Jaume I, Castelló de la Plana, Spain.
| | - Michel Barrot
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, F-67000 Strasbourg, France
| |
Collapse
|
21
|
Tong Y, Pfeiffer L, Serchov T, Coenen VA, Döbrössy MD. Optogenetic stimulation of ventral tegmental area dopaminergic neurons in a female rodent model of depression: The effect of different stimulation patterns. J Neurosci Res 2022; 100:897-911. [PMID: 35088434 DOI: 10.1002/jnr.25014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 11/25/2021] [Accepted: 01/02/2022] [Indexed: 12/27/2022]
Abstract
Major depressive disorder is one of the most common mental disorders, and more than 300 million of people suffer from depression worldwide. Recent clinical trials indicate that deep brain stimulation of the superolateral medial forebrain bundle (mfb) can have rapid and long-term antidepressant effects in patients with treatment-resistant depression. However, the mechanisms of action are elusive. In this study, using female rats, we demonstrate the antidepressant effects of selective optogenetic stimulation of the ventral tegmental area's dopaminergic (DA) neurons passing through the mfb and compare different stimulation patterns. Chronic mild unpredictable stress (CMUS) induced depressive-like, but not anxiety-like phenotype. Short-term and long-term stimulation demonstrated antidepressant effect (OSST) and improved anxiolytic effect (EPM), while long-term stimulation during CMUS induction prevented depressive-like behavior (OSST and USV) and improved anxiolytic effect (EPM). The results highlight that long-term accumulative stimulation on DA pathways is required for antidepressant and anxiolytic effect.
Collapse
Affiliation(s)
- Yixin Tong
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany
| | - Lisa Pfeiffer
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Tsvetan Serchov
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Volker A Coenen
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation, Freiburg University, Freiburg, Germany
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
22
|
Pulcu E, Guinea C, Cowen PJ, Murphy SE, Harmer CJ. A translational perspective on the anti-anhedonic effect of ketamine and its neural underpinnings. Mol Psychiatry 2022; 27:81-87. [PMID: 34158619 PMCID: PMC8960410 DOI: 10.1038/s41380-021-01183-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 05/15/2021] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Anhedonia, a pronounced reduction in interest or pleasure in any of life's daily activities, is a cardinal symptom of major depression. In this Perspective article, we synthesise the recent evidence from rodent, monkey and human neuroimaging literature to highlight how the habenula, a small evolutionarily conserved subcortical structure located in the midbrain, may orchestrate the behavioural expression of anhedonia across fronto-mesolimbic networks. We then review how this circuitry can be modulated by ketamine, an NMDA receptor antagonist with rapid antidepressant properties. We propose that experimental paradigms founded in reinforcement learning and value-based decision-making can usefully probe this network and thereby help elucidate the mechanisms underlying ketamine's rapid antidepressant action.
Collapse
Affiliation(s)
- Erdem Pulcu
- Department of Psychiatry, University of Oxford, Oxford, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK.
| | - Calum Guinea
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Philip J Cowen
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Susannah E Murphy
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Catherine J Harmer
- Department of Psychiatry, University of Oxford, Oxford, UK.
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, OX3 7JX, UK.
| |
Collapse
|
23
|
Upadhyay J, Verrico CD, Cay M, Kodele S, Yammine L, Koob GF, Schreiber R. Neurocircuitry basis of the opioid use disorder-post-traumatic stress disorder comorbid state: conceptual analyses using a dimensional framework. Lancet Psychiatry 2022; 9:84-96. [PMID: 34774203 DOI: 10.1016/s2215-0366(21)00008-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/11/2020] [Accepted: 01/06/2021] [Indexed: 12/17/2022]
Abstract
Understanding the interface between opioid use disorder (OUD) and post-traumatic stress disorder (PTSD) is challenging. By use of a dimensional framework, such as research domain criteria, convergent and targetable neurobiological processes in OUD-PTSD comorbidity can be identified. We hypothesise that, in OUD-PTSD, circuitry that is implicated in two research domain criteria systems (ie, negative valence and cognitive control) underpins dysregulation of incentive salience, negative emotionality, and executive function. We also propose that the OUD-PTSD state might be systematically investigated with approaches outlined within a neuroclinical assessment framework for addictions and PTSD. Our dimensional analysis of the OUD-PTSD state shows how first-line therapeutic approaches (ie, partial μ-type opioid receptor [MOR1] agonism) modulate overlapping neurobiological and clinical features and also provides mechanistic rationale for evaluating polytherapeutic strategies (ie, partial MOR1 agonism, κ-type opioid receptor [KOR1] antagonism, and α-2A adrenergic receptor [ADRA2A] agonism). A combination of these therapeutic mechanisms is projected to facilitate recovery in patients with OUD-PTSD by mitigating negative valence states and enhancing executive control.
Collapse
Affiliation(s)
- Jaymin Upadhyay
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA.
| | - Christopher D Verrico
- Department of Psychiatry and Behavioral Sciences and Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Mariesa Cay
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Sanda Kodele
- Faculty of Psychology and Neuroscience, Section Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands
| | - Luba Yammine
- Louis A Faillace Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - George F Koob
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Rudy Schreiber
- Faculty of Psychology and Neuroscience, Section Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
24
|
Graham DP, Harding MJ, Nielsen DA. Pharmacogenetics of Addiction Therapy. Methods Mol Biol 2022; 2547:437-490. [PMID: 36068473 DOI: 10.1007/978-1-0716-2573-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Drug addiction is a serious relapsing disease that has high costs to society and to the individual addicts. Treatment of these addictions is still in its nascency, with only a few examples of successful therapies. Therapeutic response depends upon genetic, biological, social, and environmental components. A role for genetic makeup in the response to treatment has been shown for several addiction pharmacotherapies with response to treatment based on individual genetic makeup. In this chapter, we will discuss the role of genetics in pharmacotherapies, specifically for cocaine, alcohol, and opioid dependences. The continued elucidation of the role of genetics should aid in the development of new treatments and increase the efficacy of existing treatments.
Collapse
Affiliation(s)
- David P Graham
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Mark J Harding
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - David A Nielsen
- Michael E. DeBakey Veterans Affairs Medical Center, and the Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
25
|
Laignier MR, Lopes-Júnior LC, Santana RE, Leite FMC, Brancato CL. Down Syndrome in Brazil: Occurrence and Associated Factors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182211954. [PMID: 34831710 PMCID: PMC8620277 DOI: 10.3390/ijerph182211954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/03/2021] [Accepted: 10/12/2021] [Indexed: 11/20/2022]
Abstract
Background: Down syndrome is the most frequent genetic cause of intellectual disability, with an estimated birth prevalence of 14 per 10,000 live births. In Brazil, statistical data on the occurrence of babies born with Down syndrome remain unclear. We aimed to estimate the occurrence of Down syndrome between 2012 and 2018, and to observe its association with maternal, gestational, paternal characteristics, and newborn vitality. Methods: A retrospective study was carried out using secondary data included in the Certificate of Live Birth in a state located in the southeastern region of Brazil. Data analysis was performed in the software Stata 14.1. Pearson’s chi-square test for bivariate analysis, and logistic regression for multivariate analysis were performed, with a 95% confidence interval (CI) and a significance of 5%. Results: We observed that 157 cases of Down syndrome were reported among 386,571 live births, representing an incidence of 4 in 10,000 live births. Down syndrome was associated with maternal age ≥ 35 years, paternal age ≥ 30 years, the performance of six or more prenatal consultations, prematurity, and low birth weight (p < 0.05). Conclusions: Women aged 35 and over were more likely to have children born with Down syndrome. In addition, there is an association of Down syndrome with premature birth, low birth weight, and the number of prenatal consultations (≥6).
Collapse
Affiliation(s)
- Mariana Rabello Laignier
- Nursing Department at the Health Sciences Center, Universidade Federal do Espírito Santo, Vitória 29075-910, Brazil; (L.C.L.-J.); (F.M.C.L.)
- Correspondence:
| | - Luís Carlos Lopes-Júnior
- Nursing Department at the Health Sciences Center, Universidade Federal do Espírito Santo, Vitória 29075-910, Brazil; (L.C.L.-J.); (F.M.C.L.)
| | - Raquel Esperidon Santana
- Associação de Pais, Amigos e Pessoas com Síndrome de Down do Espírito Santo, Vitória 29075-910, Brazil; (R.E.S.); (C.L.B.)
| | - Franciéle Marabotti Costa Leite
- Nursing Department at the Health Sciences Center, Universidade Federal do Espírito Santo, Vitória 29075-910, Brazil; (L.C.L.-J.); (F.M.C.L.)
| | - Carolina Laura Brancato
- Associação de Pais, Amigos e Pessoas com Síndrome de Down do Espírito Santo, Vitória 29075-910, Brazil; (R.E.S.); (C.L.B.)
| |
Collapse
|
26
|
Mercer Lindsay N, Chen C, Gilam G, Mackey S, Scherrer G. Brain circuits for pain and its treatment. Sci Transl Med 2021; 13:eabj7360. [PMID: 34757810 DOI: 10.1126/scitranslmed.abj7360] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Nicole Mercer Lindsay
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Biology, CNC Program, Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Chong Chen
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gadi Gilam
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sean Mackey
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,New York Stem Cell Foundation-Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
27
|
Jhou TC. The rostromedial tegmental (RMTg) "brake" on dopamine and behavior: A decade of progress but also much unfinished work. Neuropharmacology 2021; 198:108763. [PMID: 34433088 PMCID: PMC8593889 DOI: 10.1016/j.neuropharm.2021.108763] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/05/2021] [Accepted: 08/20/2021] [Indexed: 01/07/2023]
Abstract
Between 2005 and 2009, several research groups identified a strikingly dense inhibitory input to midbrain dopamine neurons arising from a previously uncharted region posterior to the ventral tegmental area (VTA). This region is now denoted as either the rostromedial tegmental nucleus (RMTg) or the "tail of the VTA" (tVTA), and is recognized to express distinct genetic markers, encode negative "prediction errors" (inverse to dopamine neurons), and play critical roles in behavioral inhibition and punishment learning. RMTg neurons are also influenced by many categories of abused drugs, and may drive some aversive responses to such drugs, particularly cocaine and alcohol. However, despite much progress, many important questions remain about RMTg molecular/genetic properties, diversity of projection targets, and applications to addiction, depression, and other neuropsychiatric disorders. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
|
28
|
The Role of Mesostriatal Dopamine System and Corticostriatal Glutamatergic Transmission in Chronic Pain. Brain Sci 2021; 11:brainsci11101311. [PMID: 34679376 PMCID: PMC8533867 DOI: 10.3390/brainsci11101311] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/21/2022] Open
Abstract
There is increasing recognition of the involvement of the nigrostriatal and mesolimbic dopamine systems in the modulation of chronic pain. The first part of the present article reviews the evidence indicating that dopamine exerts analgesic effects during persistent pain by stimulating the D2 receptors in the dorsal striatum and nucleus accumbens (NAc). Thereby, dopamine inhibits striatal output via the D2 receptor-expressing medium spiny neurons (D2-MSN). Dopaminergic neurotransmission in the mesostriatal pathways is hampered in chronic pain states and this alteration maintains and exacerbates pain. The second part of this article focuses on the glutamatergic inputs from the medial prefrontal cortex to the NAc, their activity changes in chronic pain, and their role in pain modulation. Finally, interactions between dopaminergic and glutamatergic inputs to the D2-MSN are considered in the context of persistent pain. Studies using novel techniques indicate that pain is regulated oppositely by two independent dopaminergic circuits linking separate parts of the ventral tegmental area and of the NAc, which also interact with distinct regions of the medial prefrontal cortex.
Collapse
|
29
|
Hernández VS, Zetter MA, Guerra EC, Hernández-Araiza I, Karuzin N, Hernández-Pérez OR, Eiden LE, Zhang L. ACE2 expression in rat brain: Implications for COVID-19 associated neurological manifestations. Exp Neurol 2021; 345:113837. [PMID: 34400158 PMCID: PMC8361001 DOI: 10.1016/j.expneurol.2021.113837] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/12/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023]
Abstract
We examined cell type-specific expression and distribution of rat brain angiotensin-converting enzyme 2 (ACE2), the receptor for SARS-CoV-2, in the rodent brain. ACE2 is ubiquitously present in brain vasculature, with the highest density of ACE2 expressing capillaries found in the olfactory bulb, the hypothalamic paraventricular, supraoptic, and mammillary nuclei, the midbrain substantia nigra and ventral tegmental area, and the hindbrain pontine nucleus, the pre-Bötzinger complex, and nucleus of tractus solitarius. ACE2 was expressed in astrocytes and astrocytic foot processes, pericytes and endothelial cells, key components of the blood-brain barrier. We found discrete neuronal groups immunopositive for ACE2 in brainstem respiratory rhythm generating centers, including the pontine nucleus, the parafascicular/retrotrapezoid nucleus, the parabrachial nucleus, the Bötzinger, and pre-Bötzinger complexes and the nucleus of tractus solitarius; in the arousal-related pontine reticular nucleus and gigantocellular reticular nuclei; in brainstem aminergic nuclei, including substantia nigra, ventral tegmental area, dorsal raphe, and locus coeruleus; in the epithalamic habenula, hypothalamic paraventricular and supramammillary nuclei; and in the hippocampus. Identification of ACE2-expressing neurons in rat brain within well-established functional circuits facilitates prediction of possible neurological manifestations of brain ACE2 dysregulation during and after COVID-19 infection.
Collapse
Affiliation(s)
- Vito S Hernández
- Dept. Physiology, Laboratory of Systems Neuroscience, School of Medicine, National Autonomous University of Mexico (UNAM, Mexico City, Mexico)
| | - Mario A Zetter
- Dept. Physiology, Laboratory of Systems Neuroscience, School of Medicine, National Autonomous University of Mexico (UNAM, Mexico City, Mexico)
| | - Enrique C Guerra
- Dept. Physiology, Laboratory of Systems Neuroscience, School of Medicine, National Autonomous University of Mexico (UNAM, Mexico City, Mexico)
| | - Ileana Hernández-Araiza
- Dept. Physiology, Laboratory of Systems Neuroscience, School of Medicine, National Autonomous University of Mexico (UNAM, Mexico City, Mexico); School of Medicine University of Maryland, Baltimore, MD, USA
| | - Nikita Karuzin
- Dept. Physiology, Laboratory of Systems Neuroscience, School of Medicine, National Autonomous University of Mexico (UNAM, Mexico City, Mexico); School of Medicine, Pan-American University, Mexico City, Mexico
| | - Oscar R Hernández-Pérez
- Dept. Physiology, Laboratory of Systems Neuroscience, School of Medicine, National Autonomous University of Mexico (UNAM, Mexico City, Mexico)
| | - Lee E Eiden
- Section on Molecular Neuroscience, NIMH-IRP, NIH, Bethesda, MD, USA
| | - Limei Zhang
- Dept. Physiology, Laboratory of Systems Neuroscience, School of Medicine, National Autonomous University of Mexico (UNAM, Mexico City, Mexico).
| |
Collapse
|
30
|
Seo S, Sizemore RJ, Reader KL, Smither RA, Wicky HE, Hughes SM, Bilkey DK, Parr-Brownlie LC, Oorschot DE. A schizophrenia risk factor induces marked anatomical deficits at GABAergic-dopaminergic synapses in the rat ventral tegmental area: Essential evidence for new targeted therapies. J Comp Neurol 2021; 529:3946-3973. [PMID: 34338311 DOI: 10.1002/cne.25225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/07/2021] [Accepted: 07/26/2021] [Indexed: 11/09/2022]
Abstract
To develop new therapies for schizophrenia, evidence accumulated over decades highlights the essential need to investigate the GABAergic synapses that presynaptically influence midbrain dopaminergic neurons. Since current technology restricts these studies to animals, and evidence accumulated in recent decades indicates a developmental origin of schizophrenia, we investigated synaptic changes in male rat offspring exposed to maternal immune activation (MIA), a schizophrenia risk factor. Using a novel combination of lentiviruses, peroxidase-immunogold double labeling, three-dimensional serial section transmission electron microscopy and stereology, we observed clear anatomical alterations in synaptic inputs on dopaminergic neurons in the midbrain posterior ventral tegmental area (pVTA). These changes relate directly to a characteristic feature of schizophrenia: increased dopamine release. In 3-month-old and 14-month-old MIA rats, we found a marked decrease in the volume of presynaptic GABAergic terminals from the rostromedial tegmental nucleus (RMTg) and in the length of the synapses they made, when innervating pVTA dopaminergic neurons. In MIA rats in the long-term, we also discovered a decrease in the volume of the postsynaptic density (PSD) and in the maximum thickness of the PSD at the same synapses. These marked deficits were evident in conventional GABA-dopamine synapses and in synaptic triads that we discovered involving asymmetric synapses that innervated RMTg GABAergic presynaptic terminals, which in turn innervated pVTA dopaminergic neurons. In triads, the PSD thickness of asymmetric synapses was significantly decreased in MIA rats in the long-term cohort. The extensive anatomical deficits provide a potential basis for new therapies targeted at synaptic inputs on midbrain pVTA dopaminergic neurons, in contrast to current striatum-targeted antipsychotic drugs.
Collapse
Affiliation(s)
- Steve Seo
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Rachel J Sizemore
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Karen L Reader
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Roseanna A Smither
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand.,Brain Research, New Zealand
| | - Hollie E Wicky
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand.,Brain Research, New Zealand.,Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Stephanie M Hughes
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand.,Brain Research, New Zealand.,Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand.,Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Louise C Parr-Brownlie
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand.,Brain Research, New Zealand
| | - Dorothy E Oorschot
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
31
|
Smagin DA, Babenko VN, Redina OE, Kovalenko IL, Galyamina AG, Kudryavtseva NN. Reduced Expression of Slc Genes in the VTA and NAcc of Male Mice with Positive Fighting Experience. Genes (Basel) 2021; 12:genes12071099. [PMID: 34356115 PMCID: PMC8306410 DOI: 10.3390/genes12071099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/27/2021] [Accepted: 07/16/2021] [Indexed: 12/28/2022] Open
Abstract
A range of several psychiatric medications targeting the activity of solute carrier (SLC) transporters have proved effective for treatment. Therefore, further research is needed to elucidate the expression profiles of the Slc genes, which may serve as markers of altered brain metabolic processes and neurotransmitter activities in psychoneurological disorders. We studied the Slc differentially expressed genes (DEGs) using transcriptomic profiles in the ventral tegmental area (VTA), nucleus accumbens (NAcc), and prefrontal cortex (PFC) of control and aggressive male mice with psychosis-like behavior induced by repeated experience of aggression accompanied with wins in daily agonistic interactions. The majority of the Slc DEGs were shown to have brain region-specific expression profiles. Most of these genes in the VTA and NAcc (12 of 17 and 25 of 26, respectively) were downregulated, which was not the case in the PFC (6 and 5, up- and downregulated, respectively). In the VTA and NAcc, altered expression was observed for the genes encoding the transporters of neurotransmitters as well as inorganic and organic ions, amino acids, metals, glucose, etc. This indicates an alteration in transport functions for many substrates, which can lead to the downregulation or even disruption of cellular and neurotransmitter processes in the VTA and NAcc, which are attributable to chronic stimulation of the reward systems induced by positive fighting experience. There is not a single Slc DEG common to all three brain regions. Our findings show that in male mice with repeated experience of aggression, altered activity of neurotransmitter systems leads to a restructuring of metabolic and neurotransmitter processes in a way specific for each brain region. We assume that the scoring of Slc DEGs by the largest instances of significant expression co-variation with other genes may outline a candidate for new prognostic drug targets. Thus, we propose that the Slc genes set may be treated as a sensitive genes marker scaffold in brain RNA-Seq studies.
Collapse
Affiliation(s)
- Dmitry A. Smagin
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Vladimir N. Babenko
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
| | - Olga E. Redina
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
| | - Irina L. Kovalenko
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Anna G. Galyamina
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Natalia N. Kudryavtseva
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Correspondence:
| |
Collapse
|
32
|
Kibaly C, Alderete JA, Liu SH, Nasef HS, Law PY, Evans CJ, Cahill CM. Oxycodone in the Opioid Epidemic: High 'Liking', 'Wanting', and Abuse Liability. Cell Mol Neurobiol 2021; 41:899-926. [PMID: 33245509 PMCID: PMC8155122 DOI: 10.1007/s10571-020-01013-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022]
Abstract
It is estimated that nearly a third of people who abuse drugs started with prescription opioid medicines. Approximately, 11.5 million Americans used prescription drugs recreationally in 2016, and in 2018, 46,802 Americans died as the result of an opioid overdose, including prescription opioids, heroin, and illicitly manufactured fentanyl (National Institutes on Drug Abuse (2020) Opioid Overdose Crisis. https://www.drugabuse.gov/drugs-abuse/opioids/opioid-overdose-crisis . Accessed 06 June 2020). Yet physicians will continue to prescribe oral opioids for moderate-to-severe pain in the absence of alternative therapeutics, underscoring the importance in understanding how drug choice can influence detrimental outcomes. One of the opioid prescription medications that led to this crisis is oxycodone, where misuse of this drug has been rampant. Being one of the most highly prescribed opioid medications for treating moderate-to-severe pain as reflected in the skyrocketed increase in retail sales of 866% between 1997 and 2007, oxycodone was initially suggested to be less addictive than morphine. The false-claimed non-addictive formulation of oxycodone, OxyContin, further contributed to the opioid crisis. Abuse was often carried out by crushing the pills for immediate burst release, typically by nasal insufflation, or by liquefying the pills for intravenous injection. Here, we review oxycodone pharmacology and abuse liability as well as present the hypothesis that oxycodone may exhibit a unique pharmacology that contributes to its high likability and abuse susceptibility. We will discuss various mechanisms that likely contribute to the high abuse rate of oxycodone including clinical drug likability, pharmacokinetics, pharmacodynamics, differences in its actions within mesolimbic reward circuity compared to other opioids, and the possibility of differential molecular and cellular receptor interactions that contribute to its selective effects. We will also discuss marketing strategies and drug difference that likely contributes to the oxycodone opioid use disorders and addiction.
Collapse
Affiliation(s)
- Cherkaouia Kibaly
- Department of Psychiatry and Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA.
| | - Jacob A Alderete
- Department of Psychiatry and Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Steven H Liu
- Department of Psychiatry and Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Hazem S Nasef
- Department of Psychiatry and Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Ping-Yee Law
- Department of Psychiatry and Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Christopher J Evans
- Department of Psychiatry and Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioral Sciences, Jane & Terry Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA.
| |
Collapse
|
33
|
Amohashemi E, Reisi P, Alaei H. Lateral habenula electrical stimulation with different intensities in combination with GABA B receptor antagonist reduces acquisition and expression phases of morphine-induced CPP. Neurosci Lett 2021; 759:135996. [PMID: 34062194 DOI: 10.1016/j.neulet.2021.135996] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022]
Abstract
The lateral habenula (LHb) plays a principal role in response to aversive stimuli and negative emotional states. In this study, we have evaluated the effects of unilateral electrical stimulation (e-stim) of the LHb on morphine-conditioned place preference (CPP), before or after bilateral injections of Gamma-aminobutyric acid-B receptor (GABABR) antagonist, phaclofen, in male rats. Morphine (5 mg/kg; s.c.) induced a significant CPP, using a 5-day CPP paradigm. Intra-LHb microinjection of phaclofen or the LHb e-stim decreased only the acquisition of CPP. The 150 μA stimulation plus phaclofen significantly suppressed the expression phase but induced aversion in the acquisition of CPP, and an e-stim of 25 μA in combination with the antagonist, significantly prevented only the acquisition phase. The findings of this study confirm the possible role of GABABRs in the LHb on the acquisition and the expression of CPP. These results show that e-stim of LHb alone or plus phaclofen may change the GABA transmission, involving into CPP. Therefore, the GABAergic system, especially through GABABRs, may play a prominent role in the behavioral responses to morphine-induced CPP by LHb stimulation.
Collapse
Affiliation(s)
- Elahe Amohashemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hojjatallah Alaei
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
34
|
Moody OA, Zhang ER, Vincent KF, Kato R, Melonakos ED, Nehs CJ, Solt K. The Neural Circuits Underlying General Anesthesia and Sleep. Anesth Analg 2021; 132:1254-1264. [PMID: 33857967 DOI: 10.1213/ane.0000000000005361] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
General anesthesia is characterized by loss of consciousness, amnesia, analgesia, and immobility. Important molecular targets of general anesthetics have been identified, but the neural circuits underlying the discrete end points of general anesthesia remain incompletely understood. General anesthesia and natural sleep share the common feature of reversible unconsciousness, and recent developments in neuroscience have enabled elegant studies that investigate the brain nuclei and neural circuits underlying this important end point. A common approach to measure cortical activity across the brain is electroencephalogram (EEG), which can reflect local neuronal activity as well as connectivity among brain regions. The EEG oscillations observed during general anesthesia depend greatly on the anesthetic agent as well as dosing, and only some resemble those observed during sleep. For example, the EEG oscillations during dexmedetomidine sedation are similar to those of stage 2 nonrapid eye movement (NREM) sleep, but high doses of propofol and ether anesthetics produce burst suppression, a pattern that is never observed during natural sleep. Sleep is primarily driven by withdrawal of subcortical excitation to the cortex, but anesthetics can directly act at both subcortical and cortical targets. While some anesthetics appear to activate specific sleep-active regions to induce unconsciousness, not all sleep-active regions play a significant role in anesthesia. Anesthetics also inhibit cortical neurons, and it is likely that each class of anesthetic drugs produces a distinct combination of subcortical and cortical effects that lead to unconsciousness. Conversely, arousal circuits that promote wakefulness are involved in anesthetic emergence and activating them can induce emergence and accelerate recovery of consciousness. Modern neuroscience techniques that enable the manipulation of specific neural circuits have led to new insights into the neural circuitry underlying general anesthesia and sleep. In the coming years, we will continue to better understand the mechanisms that generate these distinct states of reversible unconsciousness.
Collapse
Affiliation(s)
- Olivia A Moody
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Edlyn R Zhang
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Kathleen F Vincent
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Risako Kato
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Eric D Melonakos
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Christa J Nehs
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ken Solt
- From the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts.,Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Pradel K, Drwiȩga G, Błasiak T. Superior Colliculus Controls the Activity of the Rostromedial Tegmental Nuclei in an Asymmetrical Manner. J Neurosci 2021; 41:4006-4022. [PMID: 33741724 PMCID: PMC8176749 DOI: 10.1523/jneurosci.1556-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/12/2021] [Accepted: 02/20/2021] [Indexed: 12/04/2022] Open
Abstract
Dopaminergic (DA) neurons of the midbrain are involved in controlling orienting and approach of animals toward relevant external stimuli. The firing of DA neurons is regulated by many brain structures; however, the sensory input is provided predominantly by the ipsilateral superior colliculus (SC). It is suggested that SC also innervates the contralateral rostromedial tegmental nucleus (RMTg)-the main inhibitory input to DA neurons. Therefore, this study aimed to describe the physiology and anatomy of the SC-RMTg pathway. To investigate the anatomic connections within the circuit of interest, anterograde, retrograde, and transsynaptic tract-tracing studies were performed on male Sprague Dawley rats. We have observed that RMTg is monosynaptically innervated predominantly by the lateral parts of the intermediate layer of the contralateral SC. To study the physiology of this neuronal pathway, we conducted in vivo electrophysiological experiments combined with optogenetics; the activity of RMTg neurons was recorded using silicon probes, while either contralateral or ipsilateral SC was optogenetically stimulated. Obtained results revealed that activation of the contralateral SC excites the majority of RMTg neurons, while stimulation of the ipsilateral SC evokes similar proportions of excitatory or inhibitory responses. Consequently, single-unit recordings showed that the activation of RMTg neurons innervated by the contralateral SC, or stimulation of contralateral SC-originating axon terminals within the RMTg, inhibits midbrain DA neurons. Together, the anatomy and physiology of the discovered brain circuit suggest its involvement in the orienting and motivation-driven locomotion of animals based on the direction of external sensory stimuli.SIGNIFICANCE STATEMENT Dopaminergic neurons are the target of predominantly ipsilateral, excitatory innervation originating from the superior colliculus. However, we demonstrate in our study that SC inhibits the activity of dopaminergic neurons on the contralateral side of the brain via the rostromedial tegmental nucleus. In this way, sensory information received by the animal from one hemifield could induce opposite effects on both sides of the dopaminergic system. It was shown that the side to which an animal directs its behavior is a manifestation of asymmetry in dopamine release between left and right striatum. Animals tend to move oppositely to the hemisphere with higher striatal dopamine concentration. This explains how the above-described circuit might guide the behavior of animals according to the direction of incoming sensory stimuli.
Collapse
Affiliation(s)
- Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387, Krakow, Poland
| | - Gniewosz Drwiȩga
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387, Krakow, Poland
| | - Tomasz Błasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387, Krakow, Poland
| |
Collapse
|
36
|
Sun GL, Song ZJ, Peng XH, Chen PP, Song Y, Qin X, Hua R, Zhang YM. Projection-specific dopamine neurons in the ventral tegmental area participated in morphine-induced hyperalgesia and anti-nociceptive tolerance in male mice. J Psychopharmacol 2021; 35:591-605. [PMID: 33749357 DOI: 10.1177/0269881120985183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Long-term morphine use is associated with serious side effects, such as morphine-induced hyperalgesia and analgesic tolerance. Previous investigations have documented the association between dopamine (DA) neurons in the ventral tegmental area (VTA) and pain. However, whether VTA DA neurons are implicated in morphine-induced hyperalgesia and analgesic tolerance remains elusive. METHODS Initially, we observed behavioural effects of lidocaine administration into VTA or ablation of VTA DA neurons on morphine-induced hyperalgesia and anti-nociceptive tolerance. Subsequently, c-Fos expression in nucleus accumbens (NAc) shell-projecting and medial prefrontal cortex (mPFC)-projecting VTA DA neurons after chronic morphine treatment was respectively investigated. Afterwards, the effects of chemogenetic manipulation of NAc shell-projecting or mPFC-projecting DA neurons on morphine-induced hyperalgesia and anti-nociceptive tolerance were observed. Additionally, effects of chemogenetic manipulation of VTA GABA neurons on c-Fos expression in VTA DA neurons were investigated. RESULTS Lidocaine injection into VTA relieved established hyperalgesia and anti-nociceptive tolerance whereas ablation of VTA DA neurons prevented the development of morphine-induced hyperalgesia and anti-nociceptive tolerance. Chronic morphine treatment increased c-Fos expression in NAc shell-projecting DA neurons, rather than in mPFC-projecting DA neurons. Chemogenetic manipulation of NAc shell-projecting DA neurons had influence on morphine-induced hyperalgesia and tolerance. However, chemogenetic manipulation of mPFC-projecting DA neurons had no significant effects on morphine-induced hyperalgesia and anti-nociceptive tolerance. Chemogenetic manipulation of VTA GABA neurons affected the c-Fos expression in VTA DA neurons. CONCLUSIONS These findings revealed the involvement of NAc shell-projecting VTA DA neurons in morphine-induced hyperalgesia and anti-nociceptive tolerance, and may shed new light on the clinical management of morphine-induced hyperalgesia and analgesic tolerance. PERSPECTIVE This study demonstrated that NAc shell-projecting DA neurons rather than mPFC-projecting DA neurons in the VTA were implicated in morphine-induced hyperalgesia and anti-nociceptive tolerance. Our findings may pave the way for the discovery of novel therapies for morphine-induced hyperalgesia and analgesic tolerance.
Collapse
Affiliation(s)
- Guo-Lin Sun
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Zhi-Jing Song
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical University, Xuzhou, PR China.,Department of Anesthesiology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, PR China
| | - Xiao-Han Peng
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Pan-Pan Chen
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Ying Song
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Xia Qin
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical University, Xuzhou, PR China
| | - Rong Hua
- Emergency Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, PR China
| | - Yong-Mei Zhang
- Jiangsu Province Key Laboratory of Anaesthesiology, Xuzhou Medical University, Xuzhou, PR China
| |
Collapse
|
37
|
Iordanova MD, Yau JOY, McDannald MA, Corbit LH. Neural substrates of appetitive and aversive prediction error. Neurosci Biobehav Rev 2021; 123:337-351. [PMID: 33453307 PMCID: PMC7933120 DOI: 10.1016/j.neubiorev.2020.10.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/24/2020] [Accepted: 10/13/2020] [Indexed: 12/14/2022]
Abstract
Prediction error, defined by the discrepancy between real and expected outcomes, lies at the core of associative learning. Behavioural investigations have provided evidence that prediction error up- and down-regulates associative relationships, and allocates attention to stimuli to enable learning. These behavioural advances have recently been followed by investigations into the neurobiological substrates of prediction error. In the present paper, we review neuroscience data obtained using causal and recording neural methods from a variety of key behavioural designs. We explore the neurobiology of both appetitive (reward) and aversive (fear) prediction error with a focus on the mesolimbic dopamine system, the amygdala, ventrolateral periaqueductal gray, hippocampus, cortex and locus coeruleus noradrenaline. New questions and avenues for research are considered.
Collapse
Affiliation(s)
- Mihaela D Iordanova
- Department of Psychology/Centre for Studies in Behavioral Neurobiology, Concordia University, 7141 Sherbrooke St, Montreal, QC, H4B 1R6, Canada.
| | - Joanna Oi-Yue Yau
- School of Psychology, The University of New South Wales, UNSW Sydney, NSW, 2052, Australia.
| | - Michael A McDannald
- Department of Psychology & Neuroscience, Boston College, 140 Commonwealth Avenue, 514 McGuinn Hall, Chestnut Hill, MA, 02467, USA.
| | - Laura H Corbit
- Departments of Psychology and Cell and Systems Biology, University of Toronto, 100 St. George Street, Toronto, ON, M5S 3G3, Canada.
| |
Collapse
|
38
|
Mollick JA, Chang LJ, Krishnan A, Hazy TE, Krueger KA, Frank GKW, Wager TD, O'Reilly RC. The Neural Correlates of Cued Reward Omission. Front Hum Neurosci 2021; 15:615313. [PMID: 33679345 PMCID: PMC7928384 DOI: 10.3389/fnhum.2021.615313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/19/2021] [Indexed: 11/13/2022] Open
Abstract
Compared to our understanding of positive prediction error signals occurring due to unexpected reward outcomes, less is known about the neural circuitry in humans that drives negative prediction errors during omission of expected rewards. While classical learning theories such as Rescorla-Wagner or temporal difference learning suggest that both types of prediction errors result from a simple subtraction, there has been recent evidence suggesting that different brain regions provide input to dopamine neurons which contributes to specific components of this prediction error computation. Here, we focus on the brain regions responding to negative prediction error signals, which has been well-established in animal studies to involve a distinct pathway through the lateral habenula. We examine the activity of this pathway in humans, using a conditioned inhibition paradigm with high-resolution functional MRI. First, participants learned to associate a sensory stimulus with reward delivery. Then, reward delivery was omitted whenever this stimulus was presented simultaneously with a different sensory stimulus, the conditioned inhibitor (CI). Both reward presentation and the reward-predictive cue activated midbrain dopamine regions, insula and orbitofrontal cortex. While we found significant activity at an uncorrected threshold for the CI in the habenula, consistent with our predictions, it did not survive correction for multiple comparisons and awaits further replication. Additionally, the pallidum and putamen regions of the basal ganglia showed modulations of activity for the inhibitor that did not survive the corrected threshold.
Collapse
Affiliation(s)
- Jessica A Mollick
- Department of Psychiatry, Yale University, New Haven, CT, United States
| | - Luke J Chang
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Anjali Krishnan
- Department of Psychology, Brooklyn College, City University of New York, Brooklyn, NY, United States
| | | | | | - Guido K W Frank
- UCSD Eating Disorder Center for Treatment and Research, University of California, San Diego, San Diego, CA, United States
| | - Tor D Wager
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Randall C O'Reilly
- Department of Psychology and Computer Science Center for Neuroscience, University of California, Davis, Davis, CA, United States
| |
Collapse
|
39
|
Castillo-Rolón D, Ramírez-Sánchez E, Arenas-López G, Garduño J, Hernández-González O, Mihailescu S, Hernández-López S. Nicotine Increases Spontaneous Glutamate Release in the Rostromedial Tegmental Nucleus. Front Neurosci 2021; 14:604583. [PMID: 33519359 PMCID: PMC7838497 DOI: 10.3389/fnins.2020.604583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/23/2020] [Indexed: 01/26/2023] Open
Abstract
The rostromedial tegmental nucleus (RMTg) is a bilateral structure localized in the brainstem and comprise of mainly GABAergic neurons. One of the main functions of the RMTg is to regulate the activity of dopamine neurons of the mesoaccumbens pathway. Therefore, the RMTg has been proposed as a modulator of the reward system and adaptive behaviors associated to reward learning. The RMTg receives an important glutamatergic input from the lateral habenula. Also, it receives cholinergic inputs from the laterodorsal and pedunculopontine tegmental nuclei. Previously, it was reported that nicotine increases glutamate release, evoked by electric stimulation, in the RMTg nucleus. However, the mechanisms by which nicotine induces this effect were not explored. In the present work, we performed electrophysiological experiments in brainstem slices to study the effect of nicotine on spontaneous excitatory postsynaptic currents recorded from immunocytochemically identified RMTg neurons. Also, we used calcium imaging techniques to explore the effects of nicotine on multiple RMTg neurons simultaneously. We found that nicotine promotes the persistent release of glutamate through the activation of α7 nicotinic acetylcholine receptors present on glutamatergic afferents and by a mechanism involving calcium release from intracellular stores. Through these mechanisms, nicotine increases the excitability and synchronizes the activity of RMTg neurons. Our results suggest that the RMTg nucleus mediates the noxious effects of the nicotine, and it could be a potential therapeutic target against tobacco addiction.
Collapse
Affiliation(s)
- Diego Castillo-Rolón
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Enrique Ramírez-Sánchez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gabina Arenas-López
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Julieta Garduño
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Omar Hernández-González
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Stefan Mihailescu
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Salvador Hernández-López
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
40
|
Robinson SL, Dornellas APS, Burnham NW, Houck CA, Luhn KL, Bendrath SC, Companion MA, Brewton HW, Thomas RD, Navarro M, Thiele TE. Distinct and Overlapping Patterns of Acute Ethanol-Induced C-Fos Activation in Two Inbred Replicate Lines of Mice Selected for Drinking to High Blood Ethanol Concentrations. Brain Sci 2020; 10:brainsci10120988. [PMID: 33333877 PMCID: PMC7765285 DOI: 10.3390/brainsci10120988] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/31/2022] Open
Abstract
The inbred high drinking in the dark (iHDID1 and iHDID2) strains are two replicate lines bred from the parent HS/Npt (HS) line for achieving binge levels of blood ethanol concentration (≥80 mg/dL BEC) in a four-hour period. In this work, we sought to evaluate differences in baseline and ethanol-induced c-Fos activation between the HS, iHDID1, and iHDID2 genetic lines in brain regions known to process the aversive properties of ethanol. Methods: Male and female HS, iHDID1, and iHDID2 mice underwent an IP saline 2 3 g/kg ethanol injection. Brain sections were then stained for c-Fos expression in the basolateral/central amygdala (BLA/CeA), bed nucleus of the stria terminals (BNST), A2, locus coeruleus (LC), parabrachial nucleus (PBN), lateral/medial habenula (LHb/MHb), paraventricular nucleus of the thalamus (PVT), periaqueductal gray (PAG), Edinger–Westphal nuclei (EW), and rostromedial tegmental nucleus (RMTg). Results: The iHDID1 and iHDID2 lines showed similar and distinct patterns of regional c-Fos; however, in no region did the two both significantly differ from the HS line together. Conclusions: Our findings lend further support to the hypothesis the iHDID1 and the iHDID2 lines arrive at a similar behavior phenotype through divergent genetic mechanisms.
Collapse
Affiliation(s)
- Stacey L. Robinson
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ana Paula S. Dornellas
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nathan W. Burnham
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, USA;
| | - Christa A. Houck
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kendall L. Luhn
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
| | - Sophie C. Bendrath
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michel A. Companion
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Honoreé W. Brewton
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rhiannon D. Thomas
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Montserrat Navarro
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Todd E. Thiele
- Department of Psychology & Neuroscience, The University of North Carolina, Chapel Hill, NC 27599, USA; (S.L.R.); (A.P.S.D.); (C.A.H.); (K.L.L.); (S.C.B.); (M.A.C.); (H.W.B.); (R.D.T.); (M.N.)
- Bowles Center for Alcohol Studies, The University of North Carolina, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +1-919-966-1519; Fax: +1-919-962-2537
| |
Collapse
|
41
|
Brizuela M, Ootsuka Y. Inhibition of the dorsomedial hypothalamus substantially decreases brown adipose tissue sympathetic discharge induced by activation of the lateral habenula. Auton Neurosci 2020; 230:102745. [PMID: 33220531 DOI: 10.1016/j.autneu.2020.102745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 11/28/2022]
Abstract
The lateral habenula (LHb) is an evolutionarily ancient nucleus that plays an important role in the detection of salient/adverse environmental events. We have previously shown that the LHb is involved in brown adipose tissue (BAT) thermogenesis elicited by stressful situations, and that the medullary raphé, a key lower brainstem sympathetic control centre, mediates BAT thermogenesis elicited by stimulating the LHb. Since there are no direct projections from the LHb to the medullary raphé, it is plausible that the dorsomedial hypothalamus (DMH), a brain region known to be important for thermoregulatory responses to stress, is involved in this thermogenic pathway. In this study we aimed to test this possibility. In anaesthetized Sprague-Dawley rats, we recorded electrical discharges directly from sympathetic fibres that innervate BAT, as well as BAT temperature. Injections of bicuculline (1 nmol in 100 nl), a neuronal activator by disinhibition, into the LHb increased BAT sympathetic nerve discharge by 4.9 ± 1.4dBμV (n = 7, P < 0.05) and BAT temperature by 1.0 ± 0.1 °C (n = 7, P < 0.01). Subsequent injections of muscimol (0.25 nmol in 100 nl), a neuronal inhibitor, into the DMH promptly reduced BAT sympathetic nerve discharge by 4.7 ± 1.3 dBμV (n = 7, P < 0.05) and BAT temperature by 0.3 ± 0.1 °C (n = 7, P < 0.05). Injections of a mixture of the ionotropic glutamate receptor antagonists, DL-2-Amino-5-phosphonopentanoic acid (AP5) and 6-cyano-7-nitroquinoxaline-2,3-dioneis (CNQX) into the DMH, after activation of the LHb, also significantly decreased BAT sympathetic nerve discharge and BAT temperature. These results suggest that, for sympathetically-mediated BAT thermogenesis, the DMH is part of the neural circuitry linking the LHb with the medullary raphé.
Collapse
Affiliation(s)
- Mariana Brizuela
- Flinders Health and Medical Research Institute, Discipline of Human Physiology, College of Medicine and Public Health, Flinders University, SA, Australia
| | - Youichirou Ootsuka
- Flinders Health and Medical Research Institute, Discipline of Human Physiology, College of Medicine and Public Health, Flinders University, SA, Australia.
| |
Collapse
|
42
|
Mollick JA, Hazy TE, Krueger KA, Nair A, Mackie P, Herd SA, O'Reilly RC. A systems-neuroscience model of phasic dopamine. Psychol Rev 2020; 127:972-1021. [PMID: 32525345 PMCID: PMC8453660 DOI: 10.1037/rev0000199] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We describe a neurobiologically informed computational model of phasic dopamine signaling to account for a wide range of findings, including many considered inconsistent with the simple reward prediction error (RPE) formalism. The central feature of this PVLV framework is a distinction between a primary value (PV) system for anticipating primary rewards (Unconditioned Stimuli [USs]), and a learned value (LV) system for learning about stimuli associated with such rewards (CSs). The LV system represents the amygdala, which drives phasic bursting in midbrain dopamine areas, while the PV system represents the ventral striatum, which drives shunting inhibition of dopamine for expected USs (via direct inhibitory projections) and phasic pausing for expected USs (via the lateral habenula). Our model accounts for data supporting the separability of these systems, including individual differences in CS-based (sign-tracking) versus US-based learning (goal-tracking). Both systems use competing opponent-processing pathways representing evidence for and against specific USs, which can explain data dissociating the processes involved in acquisition versus extinction conditioning. Further, opponent processing proved critical in accounting for the full range of conditioned inhibition phenomena, and the closely related paradigm of second-order conditioning. Finally, we show how additional separable pathways representing aversive USs, largely mirroring those for appetitive USs, also have important differences from the positive valence case, allowing the model to account for several important phenomena in aversive conditioning. Overall, accounting for all of these phenomena strongly constrains the model, thus providing a well-validated framework for understanding phasic dopamine signaling. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
Affiliation(s)
- Jessica A Mollick
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Thomas E Hazy
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Kai A Krueger
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Ananta Nair
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Prescott Mackie
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Seth A Herd
- Department of Psychology and Neuroscience, University of Colorado Boulder
| | - Randall C O'Reilly
- Department of Psychology and Neuroscience, University of Colorado Boulder
| |
Collapse
|
43
|
Solecki W, Wilczkowski M, Pradel K, Karwowska K, Kielbinski M, Drwięga G, Zajda K, Blasiak T, Soltys Z, Rajfur Z, Szklarczyk K, Przewłocki R. Effects of brief inhibition of the ventral tegmental area dopamine neurons on the cocaine seeking during abstinence. Addict Biol 2020; 25:e12826. [PMID: 31478293 DOI: 10.1111/adb.12826] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 07/30/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022]
Abstract
Preclinical studies strongly suggest that cocaine seeking depends on the neuronal activity of the ventral tegmental area (VTA) and phasic dopaminergic (DA) signaling. Notably, VTA pharmacological inactivation or dopamine receptor blockade in the forebrain may induce behavioral inhibition in general and acute aversive states in particular, thus reducing cocaine seeking indirectly. Such artifacts hinder successful translation of these findings in clinical studies and practice. Here, we aimed to evaluate if dynamic VTA manipulations effectively reduce cocaine seeking. We used male tyrosine hydroxylase (TH) IRES-Cre+ rats along with optogenetic tools to inhibit directly and briefly VTA DA neurons during conditioned stimulus (CS)-induced cocaine seeking under extinction conditions. The behavioral effects of optogenetic inhibition were also assessed in the real-time dynamic place aversion, conditioned place aversion, and CS-induced food-seeking tests. We found that brief and nondysphoric/nonsedative pulses of VTA photo-inhibition (1 s every 9 s, ie, for 10% of time) attenuated CS-induced cocaine seeking under extinction conditions in rats expressing archaerhodopsin selectively on the TH+ neurons. Furthermore, direct inhibition of the VTA DA activity reduced CS-induced cocaine seeking 24 hours after photo-modulation. Importantly, such effect appears to be selective for cocaine seeking as similar inhibition of the VTA DA activity had no effect on CS-induced food seeking. Thus, briefly inhibiting VTA DA activity during CS-induced cocaine seeking drastically and selectively reduces seeking without behavioral artifacts such as sedation or dysphoria. Our results point to the therapeutic possibilities of coupling nonpharmacologic treatments with extinction training in reducing cocaine addiction.
Collapse
Affiliation(s)
- WojciechB. Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology Jagiellonian University Krakow Poland
| | - Michał Wilczkowski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology Jagiellonian University Krakow Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research Jagiellonian University Krakow Poland
| | - Karolina Karwowska
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology Jagiellonian University Krakow Poland
| | - Michał Kielbinski
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology Jagiellonian University Krakow Poland
| | - Gniewosz Drwięga
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology Jagiellonian University Krakow Poland
| | - Katarzyna Zajda
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology Jagiellonian University Krakow Poland
| | - Tomasz Blasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research Jagiellonian University Krakow Poland
| | - Zbigniew Soltys
- Department of Neuroanatomy, Institute of Zoology and Biomedical Research Jagiellonian University Krakow Poland
| | - Zenon Rajfur
- Department of Biosystems Physics, Institute of Physics Jagiellonian University Krakow Poland
| | - Klaudia Szklarczyk
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology Jagiellonian University Krakow Poland
| | - Ryszard Przewłocki
- Department of Molecular Neuropharmacology Institute of Pharmacology Polish Academy of Sciences Krakow Poland
| |
Collapse
|
44
|
Döbrössy MD, Ramanathan C, Ashouri Vajari D, Tong Y, Schlaepfer T, Coenen VA. Neuromodulation in Psychiatric disorders: Experimental and Clinical evidence for reward and motivation network Deep Brain Stimulation: Focus on the medial forebrain bundle. Eur J Neurosci 2020; 53:89-113. [PMID: 32931064 DOI: 10.1111/ejn.14975] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 07/24/2020] [Accepted: 08/27/2020] [Indexed: 12/28/2022]
Abstract
Deep brain stimulation (DBS) in psychiatric illnesses has been clinically tested over the past 20 years. The clinical application of DBS to the superolateral branch of the medial forebrain bundle in treatment-resistant depressed patients-one of several targets under investigation-has shown to be promising in a number of uncontrolled open label trials. However, there are remain numerous questions that need to be investigated to understand and optimize the clinical use of DBS in depression, including, for example, the relationship between the symptoms, the biological substrates/projections and the stimulation itself. In the context of precision and customized medicine, the current paper focuses on clinical and experimental research of medial forebrain bundle DBS in depression or in animal models of depression, demonstrating how clinical and scientific progress can work in tandem to test the therapeutic value and investigate the mechanisms of this experimental treatment. As one of the hypotheses is that depression engenders changes in the reward and motivational networks, the review looks at how stimulation of the medial forebrain bundle impacts the dopaminergic system.
Collapse
Affiliation(s)
- Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation, Freiburg University, Freiburg, Germany
| | - Chockalingam Ramanathan
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany
| | - Danesh Ashouri Vajari
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany
| | - Yixin Tong
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany
| | - Thomas Schlaepfer
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Interventional Biological Psychiatry, University Hospital Freiburg, Freiburg, Germany
| | - Volker A Coenen
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation, Freiburg University, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Stereotactic and Functional Neurosurgery, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
45
|
Abstract
A recently defined structure, the rostromedial tegmental nucleus (RMTg; aka tail of the ventral tegmental area [VTA]), has been proposed as an inhibitory control center for dopaminergic activity of the VTA. This region is composed of GABAergic cells that send afferent projections to the ventral midbrain and synapse onto dopaminergic cells in the VTA and substantia nigra. These cells exhibit µ-opioid receptor immunoreactivity, and in vivo, ex vivo, and optogenetic/electrophysiological approaches demonstrate that morphine excites dopamine neurons by targeting receptors on GABAergic neurons localized in the RMTg. This suggests that the RMTg may be a key modulator of opioid effects and a major brake regulating VTA dopamine systems. However, no study has directly manipulated RMTg GABAergic neurons in vivo and assessed the effect on nociception or opioid analgesia. In this study, multiplexing of GABAergic neurons in the RMTg was achieved using stimulatory Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) and inhibitory kappa-opioid receptor DREADDs (KORD). Our data show that locally infused RMTg morphine or selective RMTg GABAergic neuron inhibition produces 87% of the maximal antinociceptive effect of systemic morphine, and RMTg GABAergic neurons modulate dopamine release in the nucleus accumbens. In addition, chemoactivation of VTA dopamine neurons significantly reduced pain behaviors both in resting and facilitated pain states and reduced by 75% the dose of systemic morphine required to produce maximal antinociception. These results provide compelling evidence that RMTg GABAergic neurons are involved in processing of nociceptive information and are important mediators of opioid analgesia.
Collapse
|
46
|
M 3 but not M 4 muscarinic receptors in the rostromedial tegmental nucleus are involved in the acquisition of morphine-induced conditioned place preference. Eur J Pharmacol 2020; 882:173274. [PMID: 32534071 DOI: 10.1016/j.ejphar.2020.173274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 01/08/2023]
Abstract
Opioids strongly inhibit GABAergic neurons in the rostromedial tegmental nucleus (RMTg) that expresses μ-opioid receptors to induce rewarding and psychomotor effects. M3 and M4 muscarinic receptors are co-localized with μ-opioid receptors at these GABAergic neurons. This study explored whether RMTg M3 and M4 muscarinic receptors are involved in regulating opioid-induced reward and locomotion via a conditioned place preference (CPP) paradigm. Selective muscarinic receptor agonists and antagonists were both singly and combinatorically injected into the RMTg to examine their effects on the acquisition of systemic morphine-induced CPP and locomotor activity. The M3 muscarinic receptor agonist, pilocarpine, inhibited the acquisition of morphine-induced CPP, whereas its antagonist, 1,1-dimethyl-4-diphenylacetoxypiperidinium iodide (4-DAMP, 1 μg/side), reversed the inhibitory effect of pilocarpine (30 μg/side). Additionally, 4-DAMP increased locomotor activity while pilocarpine (30 μg/side) partially decreased locomotor activity when combined with morphine. In contrast, the M4 muscarinic receptor agonist, LY2033298 (0.1 and 0.2 μg/side), and antagonist, tropicamide (20 and 40 μM/side), did not affect the acquisition of morphine-induced CPP or locomotor activity. Taken together, our findings suggest that RMTg M3 muscarinic receptors are involved in opioid-induced rewarding and psychomotor effects. Therefore, RMTg M3 muscarinic receptors may represent a promising target for the treatment of opioid addiction.
Collapse
|
47
|
Fakhoury M, Hernandez G, Lévesque D, Rompré PP. Modulation of brain stimulation reward and locomotor activity by ionotropic glutamate receptors of the tail of the ventral tegmental area. Behav Brain Res 2020; 393:112785. [PMID: 32593543 DOI: 10.1016/j.bbr.2020.112785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 11/15/2022]
Abstract
The rostromedial tegmental nucleus also referred to as the tail of the ventral tegmental area (tVTA) contains a cluster of gamma-aminobutyric acid (GABA)ergic neurons that receive dense glutamatergic afferents from the lateral habenula (LHb), and project to dopamine (DA) neurons of the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc). In light of previous evidence implicating glutamate transmission in the regulation of midbrain DA neuronal activity, we first assessed the impact of intra-tVTA microinjection of NBQX (0.8 nmol/side) and PPPA (0.825 nmol/side), respectively AMPA and NMDA receptor antagonists, on reward induced by intracranial self-stimulation (ICSS) and on locomotor activity. Since the tVTA contains a large concentration of mu opioid receptors, additional measures were obtained following microinjection of endomorphin-1 (EM-1, 1 nmol/side) to confirm tVTA placements. Then, using small interfering RNAs (siRNAs), we tested the effect of tVTA downregulation of the GluN1 subunit of the NMDA receptor on reward and locomotor activity. Results show that NBQX, PPPA and EM-1 all enhance reward and locomotor activity, effects that were of different magnitude in rostral and intermediate parts of the tVTA. On the other hand, a reduction in GluN1 subunits used a marked decrease in operant responding for ICSS, but failed to alter ICSS reward and the reward-enhancing effect of PPPA. Our results support a role for the tVTA as a main inhibitory component of DA-dependent behavioral measures, and suggest that tVTA NMDA receptors that modulate reward are most likely expressed on tVTA afferent terminals.
Collapse
Affiliation(s)
- Marc Fakhoury
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| | - Giovanni Hernandez
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Daniel Lévesque
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Pierre-Paul Rompré
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
48
|
Sun Y, Cao J, Xu C, Liu X, Wang Z, Zhao H. Rostromedial tegmental nucleus-substantia nigra pars compacta circuit mediates aversive and despair behavior in mice. Exp Neurol 2020; 333:113433. [PMID: 32791155 DOI: 10.1016/j.expneurol.2020.113433] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/28/2022]
Abstract
GABAergic neurons in the rostromedial tegmental nucleus (RMTg) receive major input from the lateral habenula (LHb), which conveys negative reward and motivation related information, and project intensively to midbrain dopamine neurons, including those in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc). The RMTg-VTA circuit has been shown to be linked to the affective behavior, but the role of the RMTg-SNc circuit in aversion and depression has not been well understood. This study demonstrated that exciting or inhibiting VgatRMTg-SNc neurons was sufficient to increase or decrease immobility time in the forced swim test (FST), respectively. Furthermore, exciting the VgatRMTg-SNc pathway caused aversive behavior. Ninety percent of the SNc putative dopamine neurons were inhibited in extracellular recordings. Furthermore, inhibiting the VgatRMTg-SNc pathway reversed behavioral despair in chronic restraint stress (CRS) depression model mice. Manipulations of the pathway did not affect the hedonic value of the reward in the sucrose-preference test (SPT) or general motor function. In conclusion, these results indicate that the VgatRMTg-SNc pathway regulates aversive and despair behavior, which suggests that the RMTg may mediate the role of LHb in negative behaviors through regulating the activity of SNc neurons.
Collapse
Affiliation(s)
- Yanfei Sun
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China.
| | - Jing Cao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China
| | - Chunpeng Xu
- Shijiazhuang Fifth Hospital, Shijiazhuang 050000, PR China
| | - Xiaofeng Liu
- Neuroscience Research Center, First Hospital of Jilin University, Changchun 130021, PR China
| | - Zicheng Wang
- Norman Bethune Health Science Center of Jilin University, Changchun 130021, PR China
| | - Hua Zhao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130021, PR China; Neuroscience Research Center, First Hospital of Jilin University, Changchun 130021, PR China.
| |
Collapse
|
49
|
Faivre F, Sánchez-Catalán MJ, Dovero S, Bido S, Joshi A, Bezard E, Barrot M. Ablation of the tail of the ventral tegmental area compensates symptoms in an experimental model of Parkinson's disease. Neurobiol Dis 2020; 139:104818. [DOI: 10.1016/j.nbd.2020.104818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/31/2020] [Accepted: 02/18/2020] [Indexed: 12/22/2022] Open
|
50
|
St Laurent R, Martinez Damonte V, Tsuda AC, Kauer JA. Periaqueductal Gray and Rostromedial Tegmental Inhibitory Afferents to VTA Have Distinct Synaptic Plasticity and Opiate Sensitivity. Neuron 2020; 106:624-636.e4. [PMID: 32191871 DOI: 10.1016/j.neuron.2020.02.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/01/2019] [Accepted: 02/25/2020] [Indexed: 12/16/2022]
Abstract
The ventral tegmental area (VTA) is a major target of addictive drugs and receives multiple GABAergic projections originating outside the VTA. We describe differences in synaptic plasticity and behavior when optogenetically driving two opiate-sensitive GABAergic inputs to the VTA, the rostromedial tegmental nucleus (RMTg), and the periaqueductal gray (PAG). Activation of GABAergic RMTg terminals in the VTA in vivo is aversive, and low-frequency stimulation induces long-term depression in vitro. Low-frequency stimulation of PAG afferents in vitro unexpectedly causes long-term potentiation. Opioid receptor activation profoundly depresses PAG and RMTg inhibitory synapses but prevents synaptic plasticity only at PAG synapses. Activation of the GABAergic PAG terminals in the VTA promotes immobility, and optogenetically-driven immobility is blocked by morphine. Our data reveal the PAG as a source of highly opioid-sensitive GABAergic afferents and support the idea that different GABAergic pathways to the VTA control distinct behaviors.
Collapse
Affiliation(s)
- Robyn St Laurent
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Valentina Martinez Damonte
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Ayumi C Tsuda
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI 02912, USA
| | - Julie A Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94035, USA.
| |
Collapse
|