151
|
Serotonergic afferents from the dorsal raphe decrease the excitability of pyramidal neurons in the anterior piriform cortex. Proc Natl Acad Sci U S A 2020; 117:3239-3247. [PMID: 31992641 DOI: 10.1073/pnas.1913922117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The olfactory system receives extensive serotonergic inputs from the dorsal raphe, a nucleus involved in control of behavior, regulation of mood, and modulation of sensory processing. Although many studies have investigated how serotonin modulates the olfactory bulb, few have focused on the anterior piriform cortex (aPC), a region important for olfactory learning and encoding of odor identity and intensity. Specifically, the mechanism and functional significance of serotonergic modulation of the aPC remain largely unknown. Here we used pharmacologic, optogenetic, and fiber photometry techniques to examine the serotonergic modulation of neural activity in the aPC in vitro and in vivo. We found that serotonin (5-HT) reduces the excitability of pyramidal neurons directly via 5-HT2C receptors, phospholipase C, and calcium-activated potassium (BK) channels. Furthermore, endogenous serotonin attenuates odor-evoked calcium responses in aPC pyramidal neurons. These findings identify the mechanism underlying serotonergic modulation of the aPC and shed light on its potential role.
Collapse
|
152
|
Prakash N, Stark CJ, Keisler MN, Luo L, Der-Avakian A, Dulcis D. Serotonergic Plasticity in the Dorsal Raphe Nucleus Characterizes Susceptibility and Resilience to Anhedonia. J Neurosci 2020; 40:569-584. [PMID: 31792153 PMCID: PMC6961996 DOI: 10.1523/jneurosci.1802-19.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/04/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic stress induces anhedonia in susceptible but not resilient individuals, a phenomenon observed in humans as well as animal models, but the molecular mechanisms underlying susceptibility and resilience are not well understood. We hypothesized that the serotonergic system, which is implicated in stress, reward, and antidepressant therapy, may play a role. We found that plasticity of the serotonergic system contributes to the differential vulnerability to stress displayed by susceptible and resilient animals. Stress-induced anhedonia was assessed in adult male rats using social defeat and intracranial self-stimulation, while changes in serotonergic phenotype were investigated using immunohistochemistry and in situ hybridization. Susceptible, but not resilient, rats displayed an increased number of neurons expressing the biosynthetic enzyme for serotonin, tryptophan-hydroxylase-2 (TPH2), in the ventral subnucleus of the dorsal raphe nucleus (DRv). Further, a decrease in the number of DRv glutamatergic (VGLUT3+) neurons was observed in all stressed rats. This neurotransmitter plasticity is activity-dependent, as was revealed by chemogenetic manipulation of the central amygdala, a stress-sensitive nucleus that forms a major input to the DR. Activation of amygdalar corticotropin-releasing hormone (CRH)+ neurons abolished the increase in DRv TPH2+ neurons and ameliorated stress-induced anhedonia in susceptible rats. These findings show that activation of amygdalar CRH+ neurons induces resilience, and suppresses the gain of serotonergic phenotype in the DRv that is characteristic of susceptible rats. This molecular signature of vulnerability to stress-induced anhedonia and the active nature of resilience could be targeted to develop new treatments for stress-related disorders like depression.SIGNIFICANCE STATEMENT Depression and other mental disorders can be induced by chronic or traumatic stressors. However, some individuals are resilient and do not develop depression in response to chronic stress. A complete picture of the molecular differences between susceptible and resilient individuals is necessary to understand how plasticity of limbic circuits is associated with the pathophysiology of stress-related disorders. Using a rodent model, our study identifies a novel molecular marker of susceptibility to stress-induced anhedonia, a core symptom of depression, and a means to modulate it. These findings will guide further investigation into cellular and circuit mechanisms of resilience, and the development of new treatments for depression.
Collapse
Affiliation(s)
- Nandkishore Prakash
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Christiana J Stark
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Maria N Keisler
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Lily Luo
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Andre Der-Avakian
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| | - Davide Dulcis
- Department of Psychiatry, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
153
|
Lu L, Ren Y, Yu T, Liu Z, Wang S, Tan L, Zeng J, Feng Q, Lin R, Liu Y, Guo Q, Luo M. Control of locomotor speed, arousal, and hippocampal theta rhythms by the nucleus incertus. Nat Commun 2020; 11:262. [PMID: 31937768 PMCID: PMC6959274 DOI: 10.1038/s41467-019-14116-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 12/13/2019] [Indexed: 01/06/2023] Open
Abstract
Navigation requires not only the execution of locomotor programs but also high arousal and real-time retrieval of spatial memory that is often associated with hippocampal theta oscillations. However, the neural circuits for coordinately controlling these important processes remain to be fully dissected. Here we show that the activity of the neuromedin B (NMB) neurons in the nucleus incertus (NI) is tightly correlated with mouse locomotor speed, arousal level, and hippocampal theta power. These processes are reversibly suppressed by optogenetic inhibition and rapidly promoted by optogenetic stimulation of NI NMB neurons. These neurons form reciprocal connections with several subcortical areas associated with arousal, theta oscillation, and premotor processing. Their projections to multiple downstream stations regulate locomotion and hippocampal theta, with the projection to the medial septum being particularly important for promoting arousal. Therefore, NI NMB neurons functionally impact the neural circuit for navigation control according to particular brains states. In addition to activation of locomotor circuits, navigation also requires regulation of arousal and spatial memory processes. Here the authors identify neuromedin B neurons in the nucleus incertus and their subcortical projections in controlling these various processes during navigation.
Collapse
Affiliation(s)
- Lihui Lu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Yuqi Ren
- School of Life Sciences, Peking University, Beijing, 100871, China.,Peking University-Tsinghua University-NIBS Joint Graduate Program, Beijing, 102206, China
| | - Tao Yu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,National Institute of Biological Sciences (NIBS), Beijing, 102206, China.,Peking University-Tsinghua University-NIBS Joint Graduate Program, Beijing, 102206, China
| | - Zhixiang Liu
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Sice Wang
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China.,School of Life Sciences, Peking University, Beijing, 100871, China.,Peking University-Tsinghua University-NIBS Joint Graduate Program, Beijing, 102206, China
| | - Lubin Tan
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Jiawei Zeng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Qiru Feng
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,National Institute of Biological Sciences (NIBS), Beijing, 102206, China.,Peking University-Tsinghua University-NIBS Joint Graduate Program, Beijing, 102206, China
| | - Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Yang Liu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Minmin Luo
- School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China. .,National Institute of Biological Sciences (NIBS), Beijing, 102206, China. .,Peking University-Tsinghua University-NIBS Joint Graduate Program, Beijing, 102206, China. .,Chinese Institute for Brain Research, Beijing, 102206, China.
| |
Collapse
|
154
|
Task-Demand-Dependent Neural Representation of Odor Information in the Olfactory Bulb and Posterior Piriform Cortex. J Neurosci 2019; 39:10002-10018. [PMID: 31672791 DOI: 10.1523/jneurosci.1234-19.2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/16/2019] [Accepted: 10/19/2019] [Indexed: 02/03/2023] Open
Abstract
In awake rodents, the neural representation of olfactory information in the olfactory bulb is largely dependent on brain state and behavioral context. Learning-modified neural plasticity has been observed in mitral/tufted cells, the main output neurons of the olfactory bulb. Here, we propose that the odor information encoded by mitral/tufted cell responses in awake mice is highly dependent on the behavioral task demands. We used fiber photometry to record calcium signals from the mitral/tufted cell population in awake, head-fixed male mice under different task demands. We found that the mitral/tufted cell population showed similar responses to two distinct odors when the odors were presented in the context of a go/go task, in which the mice received a water reward regardless of the identity of the odor presented. However, when the same odors were presented in a go/no-go task, in which one odor was rewarded and the other was not, then the mitral cell population responded very differently to the two odors, characterized by a robust reduction in the response to the nonrewarded odor. Thus, the representation of odors in the mitral/tufted cell population depends on whether the task requires discrimination of the odors. Strikingly, downstream of the olfactory bulb, pyramidal neurons in the posterior piriform cortex also displayed a task-demand-dependent neural representation of odors, but the anterior piriform cortex did not, indicating that these two important higher olfactory centers use different strategies for neural representation.SIGNIFICANCE STATEMENT The most important task of the olfactory system is to generate a precise representation of odor information under different brain states. Whether the representation of odors by neurons in olfactory centers such as the olfactory bulb and the piriform cortex depends on task demands remains elusive. We find that odor representation in the mitral/tufted cells of the olfactory bulb depends on whether the task requires odor discrimination. A similar neural representation is found in the posterior piriform cortex but not the anterior piriform cortex, indicating that these higher olfactory centers use different representational strategies. The task-demand-dependent representational strategy is likely important for facilitating information processing in higher brain centers responsible for decision making and encoding of salience.
Collapse
|
155
|
Cardozo Pinto DF, Yang H, Pollak Dorocic I, de Jong JW, Han VJ, Peck JR, Zhu Y, Liu C, Beier KT, Smidt MP, Lammel S. Characterization of transgenic mouse models targeting neuromodulatory systems reveals organizational principles of the dorsal raphe. Nat Commun 2019; 10:4633. [PMID: 31604921 PMCID: PMC6789139 DOI: 10.1038/s41467-019-12392-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/09/2019] [Indexed: 11/17/2022] Open
Abstract
The dorsal raphe (DR) is a heterogeneous nucleus containing dopamine (DA), serotonin (5HT), γ-aminobutyric acid (GABA) and glutamate neurons. Consequently, investigations of DR circuitry require Cre-driver lines that restrict transgene expression to precisely defined cell populations. Here, we present a systematic evaluation of mouse lines targeting neuromodulatory cells in the DR. We find substantial differences in specificity between lines targeting DA neurons, and in penetrance between lines targeting 5HT neurons. Using these tools to map DR circuits, we show that populations of neurochemically distinct DR neurons are arranged in a stereotyped topographical pattern, send divergent projections to amygdala subnuclei, and differ in their presynaptic inputs. Importantly, targeting DR DA neurons using different mouse lines yielded both structural and functional differences in the neural circuits accessed. These results provide a refined model of DR organization and support a comparative, case-by-case evaluation of the suitability of transgenic tools for any experimental application.
Collapse
Affiliation(s)
- Daniel F Cardozo Pinto
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongbin Yang
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Iskra Pollak Dorocic
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Johannes W de Jong
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Vivian J Han
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - James R Peck
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Yichen Zhu
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Christine Liu
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Kevin T Beier
- Departments of Physiology and Biophysics, Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, 92697, USA
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, FNWI University of Amsterdam, Amsterdam, The Netherlands
| | - Stephan Lammel
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
156
|
Neurobehavioral effects of acute and chronic lead exposure in a desert rodent Meriones shawi: Involvement of serotonin and dopamine. J Chem Neuroanat 2019; 102:101689. [PMID: 31580902 DOI: 10.1016/j.jchemneu.2019.101689] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 11/22/2022]
Abstract
Lead (Pb) is a non physiological metal that has been implicated in toxic processes affecting several organs and biological systems, including the central nervous system. Several studies have focused on changes in lead-associated neurobehavioral and neurochemical alterations that occur due to Pb exposure. The present study evaluates the effects of acute and chronic Pb acetate exposure on serotoninergic and dopaminergic systems within the dorsal raphe nucleus, regarding motor activity and anxiety behaviours. Experiments were carried out on adult male Meriones shawi exposed to acute lead acetate intoxication (25 mg/kg b.w., 3 i.p. injections) or to a chronic lead exposure (0,5%) in drinking water from intrauterine age to adult age. Immunohistochemical staining demonstrated that both acute and chronic lead exposure increased anti-serotonin (anti-5HT) and tyrosine hydroxylase (anti-TH) immuno-reactivities in the dorsal raphe nucleus. In parallel, our results demonstrated that a long term Pb-exposure, but not an acute lead intoxication, induced behavioural alterations including, hyperactivity (open field test), and anxiogenic like-effects. Such neurobehavioral impairments induced by Pb-exposure in Meriones shawi may be related to dopaminergic and serotoninergic injuries identified in the dorsal raphe nucleus.
Collapse
|
157
|
Carbone C, Lo Russo SLM, Lacivita E, Frank A, Alleva E, Stark H, Saso L, Leopoldo M, Adriani W. Prior Activation of 5-HT7 Receptors Modulates the Conditioned Place Preference With Methylphenidate. Front Behav Neurosci 2019; 13:208. [PMID: 31619973 PMCID: PMC6759476 DOI: 10.3389/fnbeh.2019.00208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 08/29/2019] [Indexed: 11/13/2022] Open
Abstract
The serotonin receptor subtype 7 (5-HT7R) is clearly involved in behavioral functions such as learning/memory, mood regulation and circadian rhythm. Recent discoveries proposed modulatory physiological roles for serotonergic systems in reward-guided behavior. However, the interplay between serotonin (5-HT) and dopamine (DA) in reward-related behavioral adaptations needs to be further assessed. TP-22 is a recently developed arylpiperazine-based 5-HT7R agonist, which is also showing high affinity and selectivity towards D1 receptors. Here, we report that TP-22 displays D1 receptor antagonist activity. Moreover, we describe the first in vivo tests with TP-22: first, a pilot experiment (assessing dosage and timing of action) identified the 0.25 mg/kg i.v. dosage for locomotor stimulation of rats. Then, a conditioned place preference (CPP) test with the DA-releasing psychostimulant drug, methylphenidate (MPH), involved three rat groups: prior i.v. administration of TP-22 (0.25 mg/kg), or vehicle (VEH), 90 min before MPH (5 mg/kg), was intended for modulation of conditioning to the white chamber (saline associated to the black chamber); control group (SAL) was conditioned with saline in both chambers. Prior TP-22 further increased the stimulant effect of MPH on locomotor activity. During the place-conditioning test, drug-free activity of TP-22+MPH subjects remained steadily elevated, while VEH+MPH subjects showed a decline. Finally, after a priming injection of TP-22 in MPH-free conditions, rats showed a high preference for the MPH-associated white chamber, which conversely had vanished in VEH-primed MPH-conditioned subjects. Overall, the interaction between MPH and pre-treatment with TP-22 seems to improve both locomotor stimulation and the conditioning of motivational drives to environmental cues. Together with recent studies, a main modulatory role of 5-HT7R for the processing of rewards can be suggested. In the present study, TP-22 proved to be a useful psychoactive tool to better elucidate the role of 5-HT7R and its interplay with DA in reward-related behavior.
Collapse
Affiliation(s)
- Cristiana Carbone
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | | | - Enza Lacivita
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, Bari, Italy
| | - Annika Frank
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Enrico Alleva
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Luciano Saso
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University, Rome, Italy
| | - Marcello Leopoldo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, Bari, Italy.,BIOFORDRUG s.r.l., Università degli Studi di Bari, Bari, Italy
| | - Walter Adriani
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
158
|
Alugubelly N, Mohammad AN, Edelmann MJ, Nanduri B, Sayed M, Park JW, Carr RL. Proteomic and transcriptional profiling of rat amygdala following social play. Behav Brain Res 2019; 376:112210. [PMID: 31493430 DOI: 10.1016/j.bbr.2019.112210] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
Social play is the most characteristic form of social interaction which is necessary for adolescents to develop proper cognitive, emotional, and social competency. The information available on neural substrates and the mechanism involved in social play is limited. This study characterized social play by proteomic and transcriptional profiling studies. Social play was performed on male Sprague Dawley rats on postnatal day 38 and protein and gene expression in the amygdala was determined following behavioral testing. The proteomic analysis led to the identification of 170 differentially expressed proteins (p ≤ 0.05) with 67 upregulated and 103 downregulated proteins. The transcriptomic analysis led to the identification of 188 genes (FDR ≤ 0.05) with 55 upregulated and 133 downregulated genes. DAVID analysis of gene/protein expression data revealed that social play altered GABAergic signaling, glutamatergic signaling, and G-protein coupled receptor (GPCR) signaling. These data suggest that the synaptic levels of GABA and glutamate increased during play. Ingenuity Pathway Analysis (IPA) confirmed these alterations. IPA also revealed that differentially expressed genes/proteins in our data had significant over representation of neurotransmitter signaling systems, including the opioid, serotonin, and dopamine systems, suggesting that play alters the systems involved in the regulation of reward. In addition, corticotropin-releasing hormone signaling was altered indicating that an increased level of stress occurs during play. Overall, our data suggest that increased inhibitory GPCR signaling in these neurotransmitter pathways occurs following social play as a physiological response to regulate the induced level of reward and stress and to maintain the excitatory-inhibitory balance in the neurotransmitter systems.
Collapse
Affiliation(s)
- Navatha Alugubelly
- Center for Environmental Health Sciences, MS, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA
| | - Afzaal N Mohammad
- Center for Environmental Health Sciences, MS, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA
| | - Mariola J Edelmann
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Bindu Nanduri
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA
| | - Mohammed Sayed
- Department of Computer Engineering and Computer Science, KY, USA
| | - Juw Won Park
- Department of Computer Engineering and Computer Science, KY, USA; KBRIN Bioinformatics Core, University of Louisville, KY, USA.
| | - Russell L Carr
- Center for Environmental Health Sciences, MS, USA; Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, MS, USA.
| |
Collapse
|
159
|
Huang KW, Ochandarena NE, Philson AC, Hyun M, Birnbaum JE, Cicconet M, Sabatini BL. Molecular and anatomical organization of the dorsal raphe nucleus. eLife 2019; 8:e46464. [PMID: 31411560 PMCID: PMC6726424 DOI: 10.7554/elife.46464] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
The dorsal raphe nucleus (DRN) is an important source of neuromodulators and has been implicated in a wide variety of behavioral and neurological disorders. The DRN is subdivided into distinct anatomical subregions comprised of multiple cell types, and its complex cellular organization has impeded efforts to investigate the distinct circuit and behavioral functions of its subdomains. Here we used single-cell RNA sequencing, in situ hybridization, anatomical tracing, and spatial correlation analysis to map the transcriptional and spatial profiles of cells from the mouse DRN. Our analysis of 39,411 single-cell transcriptomes revealed at least 18 distinct neuron subtypes and 5 serotonergic neuron subtypes with distinct molecular and anatomical properties, including a serotonergic neuron subtype that preferentially innervates the basal ganglia. Our study lays out the molecular organization of distinct serotonergic and non-serotonergic subsystems, and will facilitate the design of strategies for further dissection of the DRN and its diverse functions.
Collapse
Affiliation(s)
- Kee Wui Huang
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Nicole E Ochandarena
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Adrienne C Philson
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Minsuk Hyun
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Jaclyn E Birnbaum
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Marcelo Cicconet
- Image and Data Analysis CoreHarvard Medical SchoolBostonUnited States
| | - Bernardo L Sabatini
- Department of NeurobiologyHoward Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
160
|
Guo B, Chen J, Chen Q, Ren K, Feng D, Mao H, Yao H, Yang J, Liu H, Liu Y, Jia F, Qi C, Lynn-Jones T, Hu H, Fu Z, Feng G, Wang W, Wu S. Anterior cingulate cortex dysfunction underlies social deficits in Shank3 mutant mice. Nat Neurosci 2019; 22:1223-1234. [DOI: 10.1038/s41593-019-0445-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/03/2019] [Indexed: 02/06/2023]
|
161
|
Garcia-Garcia AL, Soiza-Reilly M. Serotonin Subsystems Modulate Diverse and Opposite Behavioral Functions. ACS Chem Neurosci 2019; 10:3061-3063. [PMID: 30338982 DOI: 10.1021/acschemneuro.8b00534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Pioneering work showed that serotonin (5-HT) neurons have the unique capacity to engage in different and opposed aspects of motivated behaviors such as reward and punishment responses. These findings provided strong evidence about the functional heterogeneity of 5-HT neurons, and their possible engagement in multiple and behaviorally distinct neural subsystems. A recent study provides further compelling evidence supporting this notion, in which two ascending 5-HT circuits modulate opposed aspects of motivated behaviors.
Collapse
Affiliation(s)
- Alvaro L. Garcia-Garcia
- Department of Psychiatry, Division of Systems Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, New York 10032, United States
| | - Mariano Soiza-Reilly
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET, Universidad de Buenos Aires, Buenos Aires Argentina
| |
Collapse
|
162
|
Chen P, Hong W. Neural Circuit Mechanisms of Social Behavior. Neuron 2019; 98:16-30. [PMID: 29621486 DOI: 10.1016/j.neuron.2018.02.026] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 02/11/2018] [Accepted: 02/26/2018] [Indexed: 12/31/2022]
Abstract
We live in a world that is largely socially constructed, and we are constantly involved in and fundamentally influenced by a broad array of complex social interactions. Social behaviors among conspecifics, either conflictive or cooperative, are exhibited by all sexually reproducing animal species and are essential for the health, survival, and reproduction of animals. Conversely, impairment in social function is a prominent feature of several neuropsychiatric disorders, such as autism spectrum disorders and schizophrenia. Despite the importance of social behaviors, many fundamental questions remain unanswered. How is social sensory information processed and integrated in the nervous system? How are different social behavioral decisions selected and modulated in brain circuits? Here we discuss conceptual issues and recent advances in our understanding of brain regions and neural circuit mechanisms underlying the regulation of social behaviors.
Collapse
Affiliation(s)
- Patrick Chen
- Department of Biological Chemistry and Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Weizhe Hong
- Department of Biological Chemistry and Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
163
|
Thiele A, Bellgrove MA. Neuromodulation of Attention. Neuron 2019; 97:769-785. [PMID: 29470969 PMCID: PMC6204752 DOI: 10.1016/j.neuron.2018.01.008] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/26/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023]
Abstract
Attention is critical to high-level cognition and attention deficits are a hallmark of neurologic and neuropsychiatric disorders. Although years of research indicates that distinct neuromodulators influence attentional control, a mechanistic account that traverses levels of analysis (cells, circuits, behavior) is missing. However, such an account is critical to guide the development of next-generation pharmacotherapies aimed at forestalling or remediating the global burden associated with disorders of attention. Here, we summarize current neuroscientific understanding of how attention affects single neurons and networks of neurons. We then review key results that have informed our understanding of how neuromodulation shapes these neuron and network properties and thereby enables the appropriate allocation of attention to relevant external or internal events. Finally, we highlight areas where we believe hypotheses can be formulated and tackled experimentally in the near future, thereby critically increasing our mechanistic understanding of how attention is implemented at the cellular and network levels.
Collapse
Affiliation(s)
- Alexander Thiele
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK.
| | - Mark A Bellgrove
- Monash Institute of Cognitive and Clinical Neurosciences (MICCN) and School of Psychological Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
164
|
Drozd US, Shaburova EV, Dygalo NN. Genetic approaches to the investigation of serotonergic neuron functions in animals. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The serotonergic system is one of the most important neurotransmitter systems that take part in the regulation of vital CNS functions. The understanding of its mechanisms will help scientists create new therapeutic approaches to the treatment of mental and neurodegenerative diseases and find out how this neurotransmitter system interacts with other parts of the brain and regulates their activity. Since the serotonergic system anatomy and functionality are heterogeneous and complex, the best tools for studying them are based on manipulation of individual types of neurons without affecting neurons of other neurotransmitter systems. The selective cell control is possible due to the genetic determinism of their functions. Proteins that determine the uniqueness of the cell type are expressed under the regulation of cell-specific promoters. By using promoters that are specific for genes of the serotonin system, one can control the expression of a gene of interest in serotonergic neurons. Here we review approaches based on such promoters. The genetic models to be discussed in the article have already shed the light on the role of the serotonergic system in modulating behavior and processing sensory information. In particular, genetic knockouts of serotonin genes sert, pet1, and tph2 promoted the determination of their contribution to the development and functioning of the brain. In addition, the review describes inducible models that allow gene expression to be controlled at various developmental stages. Finally, the application of these genetic approaches in optogenetics and chemogenetics provided a new resource for studying the functions, discharge activity, and signal transduction of serotonergic neurons. Nevertheless, the advantages and limitations of the discussed genetic approaches should be taken into consideration in the course of creating models of pathological conditions and developing pharmacological treatments for their correction.
Collapse
Affiliation(s)
- U. S. Drozd
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| | - E. V. Shaburova
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| | - N. N. Dygalo
- Novosibirsk State University; Institute of Cytology and Genetics, SB RAS
| |
Collapse
|
165
|
Sengupta A, Holmes A. A Discrete Dorsal Raphe to Basal Amygdala 5-HT Circuit Calibrates Aversive Memory. Neuron 2019; 103:489-505.e7. [PMID: 31204082 DOI: 10.1016/j.neuron.2019.05.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/14/2019] [Accepted: 05/15/2019] [Indexed: 11/26/2022]
Abstract
Despite a wealth of clinical and preclinical data implicating the serotonin (5-HT) system in fear-related affective disorders, a precise definition of this neuromodulator's role in fear remains elusive. Using convergent anatomical and functional approaches, we interrogate the contribution to fear of basal amygdala (BA) 5-HT inputs from the dorsal raphe nucleus (DRN). We show the DRN→BA 5-HT pathway is engaged during fear memory formation and retrieval, and activity of these projections facilitates fear and impairs extinction. The DRN→BA 5-HT pathway amplifies fear-associated BA neuronal firing and theta power and phase-locking. Although fear recruits 5-HT and VGluT3 co-expressing DRN neurons, the fear-potentiating influence of the DRN→BA 5-HT pathway requires signaling at BA 5-HT1A/2A receptors. Input-output mapping illustrates how the DRN→BA 5-HT pathway is anatomically distinct and connected with other brain regions that mediate fear. These findings reveal how a discrete 5-HT circuit orchestrates a broader neural network to calibrate aversive memory.
Collapse
Affiliation(s)
- Ayesha Sengupta
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA.
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, USA.
| |
Collapse
|
166
|
Zhang S, Lv F, Yuan Y, Fan C, Li J, Sun W, Hu J. Whole-Brain Mapping of Monosynaptic Afferent Inputs to Cortical CRH Neurons. Front Neurosci 2019; 13:565. [PMID: 31213976 PMCID: PMC6558184 DOI: 10.3389/fnins.2019.00565] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/16/2019] [Indexed: 01/02/2023] Open
Abstract
Corticotropin-releasing hormone (CRH) is a critical neuropeptide modulating the mammalian stress response. It is involved in many functional activities within various brain regions, among which there is a subset of CRH neurons occupying a considerable proportion of the cortical GABAergic interneurons. Here, we utilized rabies virus-based monosynaptic retrograde tracing system to map the whole-brain afferent presynaptic partners of the CRH neurons in the anterior cingulate cortex (ACC). We find that the ACC CRH neurons integrate information from the cortex, thalamus, hippocampal formation, amygdala, and also several other midbrain and hindbrain nuclei. Furthermore, our results reveal that ACC CRH neurons receive direct inputs from two neuromodulatory systems, the basal forebrain cholinergic neurons and raphe serotoninergic neurons. These findings together expand our knowledge about the connectivity of the cortical GABAergic neurons and also provide a basis for further investigation of the circuit function of cortical CRH neurons.
Collapse
Affiliation(s)
- Shouhua Zhang
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Lv
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- iHuman Institute, ShanghaiTech University, Shanghai, China
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Yuan Yuan
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Chengyu Fan
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - Jiang Li
- Division of Physical Biology and Bioimaging Center, Shanghai Synchrotron Radiation Facility, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, China
| | - Wenzhi Sun
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
- iHuman Institute, ShanghaiTech University, Shanghai, China
- Chinese Institute for Brain Research, Beijing, China
| | - Ji Hu
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
167
|
Wolke SA, Mehta MA, O'Daly O, Zelaya F, Zahreddine N, Keren H, O'Callaghan G, Young AH, Leibenluft E, Pine DS, Stringaris A. Modulation of anterior cingulate cortex reward and penalty signalling in medication-naive young-adult subjects with depressive symptoms following acute dose lurasidone. Psychol Med 2019; 49:1365-1377. [PMID: 30606271 PMCID: PMC6518385 DOI: 10.1017/s0033291718003306] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Aberrations in reward and penalty processing are implicated in depression and putatively reflect altered dopamine signalling. This study exploits the advantages of a placebo-controlled design to examine how a novel D2 antagonist with adjunctive antidepressant properties modifies activity in the brain's reward network in depression. METHODS We recruited 43 medication-naïve subjects across the range of depression severity (Beck's Depression Inventory-II score range: 0-43), including healthy volunteers, as well as people meeting full-criteria for major depressive disorder. In a double-blind placebo-controlled cross-over design, all subjects received either placebo or lurasidone (20 mg) across two visits separated by 1 week. Functional magnetic resonance imaging with the Monetary Incentive Delay (MID) task assessed reward functions via neural responses during anticipation and receipt of gains and losses. Arterial spin labelling measured cerebral blood flow (CBF) at rest. RESULTS Lurasidone altered fronto-striatal activity during anticipation and outcome phases of the MID task. A significant three-way Medication-by-Depression severity-by-Outcome interaction emerged in the anterior cingulate cortex (ACC) after correction for multiple comparisons. Follow-up analyses revealed significantly higher ACC activation to losses in high- v. low depression participants in the placebo condition, with a normalisation by lurasidone. This effect could not be accounted for by shifts in resting CBF. CONCLUSIONS Lurasidone acutely normalises reward processing signals in individuals with depressive symptoms. Lurasidone's antidepressant effects may arise from reducing responses to penalty outcomes in individuals with depressive symptoms.
Collapse
Affiliation(s)
- Selina A. Wolke
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
- Mood Brain and Development Unit, Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, MD, USA
| | - Mitul A. Mehta
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Owen O'Daly
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Fernando Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Nada Zahreddine
- Department of Psychiatry, Saint-Joseph University, Beirut, Lebanon
| | - Hanna Keren
- Mood Brain and Development Unit, Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, MD, USA
| | - Georgia O'Callaghan
- Mood Brain and Development Unit, Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, MD, USA
| | - Allan H. Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Ellen Leibenluft
- Section on Mood Dysregulation and Neuroscience, Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, MD, USA
| | - Daniel S. Pine
- Section on Development and Affective Neuroscience, Emotion and Development Branch, National Institute of Mental Health, MD, USA
| | - Argyris Stringaris
- Mood Brain and Development Unit, Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, MD, USA
| |
Collapse
|
168
|
Long-term Fiber Photometry for Neuroscience Studies. Neurosci Bull 2019; 35:425-433. [PMID: 31062336 DOI: 10.1007/s12264-019-00379-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/25/2019] [Indexed: 01/21/2023] Open
Abstract
Fiber photometry is a sensitive and easy way to detect changes in fluorescent signals. The combination of fiber photometry with various fluorescent biomarkers has substantially advanced neuroscience research over the last decade. Despite the wide use of fiber photometry in biomedical fields, the lack of a detailed and comprehensive protocol has limited progress and sometimes complicated the interpretation of data. Here, we describe detailed procedures of fiber photometry for the long-term monitoring of neuronal activity in freely-behaving animals, including surgery, apparatus setup, data collection, and analysis.
Collapse
|
169
|
Yoshida K, Drew MR, Mimura M, Tanaka KF. Serotonin-mediated inhibition of ventral hippocampus is required for sustained goal-directed behavior. Nat Neurosci 2019; 22:770-777. [PMID: 30988523 DOI: 10.1038/s41593-019-0376-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 03/06/2019] [Indexed: 11/08/2022]
Abstract
The ability to sustain goal-directed action is essential for success in many domains, but little is known about the corresponding neural substrates. Using fiber photometry to monitor population neural activity, we demonstrate that engagement in sustained food- or punishment-motivated behavior is associated with suppression of ventral but not dorsal hippocampal activity. Using optogenetic stimulation, we demonstrate that this suppression is required for goal-directed behavior, whereas optogenetic suppression of the ventral hippocampus (vHP) enhances the ability to sustain goal-directed behavior. Suppression of vHP during sustained goal-directed behavior was accompanied by increased activity in median but not dorsal raphe, implicating serotonergic signaling through Htr3a as a mechanism of vHP suppression during successful goal-directed behavior. Sustainment of goal-directed action may require suppression of vHP because of the structure's well-documented role in behavioral inhibition.
Collapse
Affiliation(s)
- Keitaro Yoshida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Michael R Drew
- Center for Learning and Memory, Department of Neuroscience, The University of Texas at Austin, Austin, TX, USA
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
170
|
Dong P, Wang H, Shen XF, Jiang P, Zhu XT, Li Y, Gao JH, Lin S, Huang Y, He XB, Xu FQ, Duan S, Lian H, Wang H, Chen J, Li XM. A novel cortico-intrathalamic circuit for flight behavior. Nat Neurosci 2019; 22:941-949. [DOI: 10.1038/s41593-019-0391-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/19/2019] [Indexed: 11/09/2022]
|
171
|
Su XY, Chen M, Yuan Y, Li Y, Guo SS, Luo HQ, Huang C, Sun W, Li Y, Zhu MX, Liu MG, Hu J, Xu TL. Central Processing of Itch in the Midbrain Reward Center. Neuron 2019; 102:858-872.e5. [PMID: 31000426 DOI: 10.1016/j.neuron.2019.03.030] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/28/2018] [Accepted: 03/19/2019] [Indexed: 10/27/2022]
Abstract
Itch is an aversive sensation that evokes a desire to scratch. Paradoxically, scratching the itch also produces a hedonic experience. The specific brain circuits processing these different aspects of itch, however, remain elusive. Here, we report that GABAergic (GABA) and dopaminergic (DA) neurons in the ventral tegmental area (VTA) are activated with different temporal patterns during acute and chronic itch. DA neuron activation lags behind GABA neurons and is dependent on scratching of the itchy site. Optogenetic manipulations of VTA GABA neurons rapidly modulated scratching behaviors through encoding itch-associated aversion. In contrast, optogenetic manipulations of VTA DA neurons revealed their roles in sustaining recurrent scratching episodes through signaling scratching-induced reward. A similar dichotomy exists for the role of VTA in chronic itch. These findings advance understanding of circuit mechanisms of the unstoppable itch-scratch cycles and shed important insights into chronic itch therapy.
Collapse
Affiliation(s)
- Xin-Yu Su
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ming Chen
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Yuan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ying Li
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Su-Shan Guo
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huo-Qing Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chen Huang
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenzhi Sun
- iHuman Institute, ShanghaiTech University, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Chinese Institute for Brain Research, Beijing 102206, China
| | - Yong Li
- Collaborative Innovation Center for Brain Science, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ming-Gang Liu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.
| | - Tian-Le Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China.
| |
Collapse
|
172
|
Yuan Y, Wu W, Chen M, Cai F, Fan C, Shen W, Sun W, Hu J. Reward Inhibits Paraventricular CRH Neurons to Relieve Stress. Curr Biol 2019; 29:1243-1251.e4. [DOI: 10.1016/j.cub.2019.02.048] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 11/16/2022]
|
173
|
Lee EH, Han PL. Reciprocal interactions across and within multiple levels of monoamine and cortico-limbic systems in stress-induced depression: A systematic review. Neurosci Biobehav Rev 2019; 101:13-31. [PMID: 30917923 DOI: 10.1016/j.neubiorev.2019.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022]
Abstract
The monoamine hypothesis of depression, namely that the reduction in synaptic serotonin and dopamine levels causes depression, has prevailed in past decades. However, clinical and preclinical studies have identified various cortical and subcortical regions whose altered neural activities also regulate depressive-like behaviors, independently from the monoamine system. Our systematic review indicates that neural activities of specific brain regions and associated neural circuitries are adaptively altered after chronic stress in a specific direction, such that the neural activity in the infralimbic cortex, lateral habenula and amygdala is upregulated, whereas the neural activity in the prelimbic cortex, hippocampus and monoamine systems is downregulated. The altered neural activity dynamics between monoamine systems and cortico-limbic systems are reciprocally interwoven at multiple levels. Furthermore, depressive-like behaviors can be experimentally reversed by counteracting the altered neural activity of a specific neural circuitry at multiple brain regions, suggesting the importance of the reciprocally interwoven neural networks in regulating depressive-like behaviors. These results promise for reshaping altered neural activity dynamics as a therapeutic strategy for treating depression.
Collapse
Affiliation(s)
- Eun-Hwa Lee
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Pyung-Lim Han
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Republic of Korea; Department of Chemistry and Nano Science, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
174
|
Dong H, Wang J, Yang YF, Shen Y, Qu WM, Huang ZL. Dorsal Striatum Dopamine Levels Fluctuate Across the Sleep-Wake Cycle and Respond to Salient Stimuli in Mice. Front Neurosci 2019; 13:242. [PMID: 30949023 PMCID: PMC6436203 DOI: 10.3389/fnins.2019.00242] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/01/2019] [Indexed: 01/07/2023] Open
Abstract
Dopamine is involved in numerous neurological processes, and its deficiency has been implicated in Parkinson’s disease, whose patients suffer from severe sleep disorders. Destruction of nigrostriatal dopaminergic neurons or dorsal striatum disrupts the sleep–wake cycle. However, whether striatal dopamine levels correlate with vigilance states still remains to be elucidated. Here, we employed an intensity-based genetically encoded dopamine indicator, dLight1.1, to track striatal dopamine levels across the spontaneous sleep–wake cycle and the dopaminergic response to external stimuli. We found that the striatal dLight1.1 signal was at its highest during wakefulness, lower during non-rapid eye movement (non-REM or NREM) sleep, and lowest during REM sleep. Moreover, the striatal dLight1.1 signal increased significantly during NREM sleep-to-wake transitions, while it decreased during wake-to-NREM sleep transitions. Furthermore, different external stimuli, such as sudden door-opening of the home cage or cage-change to a new environment, caused striatal dopamine release, whereas an unexpected auditory tone did not. Finally, despite both modafinil and caffeine being wake-promoting agents that increased wakefulness, modafinil increased striatal dopamine levels while caffeine did not. Taken together, our findings demonstrated that striatal dopamine levels correlated with the spontaneous sleep–wake cycle and responded to specific external stimuli as well as the stimulant modafinil.
Collapse
Affiliation(s)
- Hui Dong
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Juan Wang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yan-Fei Yang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Yan Shen
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.,Department of Neurology and National Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
175
|
Nishitani N, Nagayasu K, Asaoka N, Yamashiro M, Andoh C, Nagai Y, Kinoshita H, Kawai H, Shibui N, Liu B, Hewinson J, Shirakawa H, Nakagawa T, Hashimoto H, Kasparov S, Kaneko S. Manipulation of dorsal raphe serotonergic neurons modulates active coping to inescapable stress and anxiety-related behaviors in mice and rats. Neuropsychopharmacology 2019; 44:721-732. [PMID: 30377380 PMCID: PMC6372597 DOI: 10.1038/s41386-018-0254-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 10/16/2018] [Accepted: 10/21/2018] [Indexed: 01/21/2023]
Abstract
Major depression and anxiety disorders are a social and economic burden worldwide. Serotonergic signaling has been implicated in the pathophysiology of these disorders and thus has been a crucial target for pharmacotherapy. However, the precise mechanisms underlying these disorders are still unclear. Here, we used species-optimized lentiviral vectors that were capable of efficient and specific transduction of serotonergic neurons in mice and rats for elucidation of serotonergic roles in anxiety-like behaviors and active coping behavior in both species. Immunohistochemical analyses revealed that lentiviral vectors with an upstream sequence of tryptophan hydroxylase 2 gene efficiently transduced serotonergic neurons with a specificity of approximately 95% in both mice and rats. Electrophysiological recordings showed that these lentiviral vectors induced sufficient expression of optogenetic tools for precise control of serotonergic neurons. Using these vectors, we demonstrate that acute activation of serotonergic neurons in the dorsal raphe nucleus increases active coping with inescapable stress in rats and mice in a time-locked manner, and that acute inhibition of these neurons increases anxiety-like behaviors specifically in rats. These findings further our understanding of the pathophysiological role of dorsal raphe serotonergic neurons in different species and the role of these neurons as therapeutic targets in major depression and anxiety disorders.
Collapse
Affiliation(s)
- Naoya Nishitani
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
- Drug Innovation Center, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Yoshidahommachi, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Nozomi Asaoka
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Mayumi Yamashiro
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Chihiro Andoh
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yuma Nagai
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Haruko Kinoshita
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hiroyuki Kawai
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Norihiro Shibui
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Beihui Liu
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - James Hewinson
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Takayuki Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hitoshi Hashimoto
- Drug Innovation Center, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Sergey Kasparov
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK.
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
176
|
Noriuchi M, Kikuchi Y, Mori K, Kamio Y. The orbitofrontal cortex modulates parenting stress in the maternal brain. Sci Rep 2019; 9:1658. [PMID: 30733605 PMCID: PMC6367346 DOI: 10.1038/s41598-018-38402-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022] Open
Abstract
Many mothers are adaptive, deploying successful coping strategies that mitigate the deleterious effects of parenting stress on caregiving, nevertheless, the neural mechanisms underlying these adaptive responses remain unclear. We utilized functional magnetic resonance imaging to investigate brain activity in 28 healthy mothers of typically developing, 2-to-3-year-old children in response to the feeding behavior of their own children versus that of other children. We then examined the correlation between maternal brain activation and subjective feelings of parenting stress. Brain regions associated with maternal motivation including the orbitofrontal cortex (OFC), ventral pallidum, periaqueductal gray (PAG), dorsal raphe nucleus (DRN), and anterior insular cortex (AIC)—as well as those associated with the recognition of one’s own child’s state (e.g., cerebellum)—exhibited significant activation in response to their own children. While mothers with higher activation in the OFC showed less parenting stress related to one’s sense of competence in the parental role, mothers with higher co-activation of the OFC with both of the AIC and PAG/DRN, and with the cerebellum showed less parenting stress caused by child characteristics. Our findings suggest that well-balanced maternal brain mechanisms integrated by the OFC may provide effective adaptive responses in daily parenting scenarios.
Collapse
Affiliation(s)
- Madoka Noriuchi
- Department of Frontier Health Science, Division of Human Health Science, Graduate School of Tokyo Metropolitan University, 7-2-10 Higashi-ogu, Arakawa, Tokyo, 116-8551, Japan.
| | - Yoshiaki Kikuchi
- Department of Frontier Health Science, Division of Human Health Science, Graduate School of Tokyo Metropolitan University, 7-2-10 Higashi-ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Kumiko Mori
- Department of Frontier Health Science, Division of Human Health Science, Graduate School of Tokyo Metropolitan University, 7-2-10 Higashi-ogu, Arakawa, Tokyo, 116-8551, Japan
| | - Yoko Kamio
- National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8553, Japan.,Institute of Education and Human Development, Ochanomizu Univesrsity, 2-1-1 Otsuka, Bunkyo, Tokyo, 112-8610, Japan
| |
Collapse
|
177
|
Krol A, Lopez-Huerta VG, Corey TEC, Deisseroth K, Ting JT, Feng G. Two eARCHT3.0 Lines for Optogenetic Silencing of Dopaminergic and Serotonergic Neurons. Front Neural Circuits 2019; 13:4. [PMID: 30774584 PMCID: PMC6367884 DOI: 10.3389/fncir.2019.00004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 01/14/2019] [Indexed: 11/13/2022] Open
Abstract
Dopaminergic and serotonergic neurons modulate and control processes ranging from reward signaling to regulation of motor outputs. Further, dysfunction of these neurons is involved in both degenerative and psychiatric disorders. Elucidating the roles of these neurons has been greatly facilitated by bacterial artificial chromosome (BAC) transgenic mouse lines expressing channelrhodopsin to readily enable cell-type specific activation. However, corresponding lines to silence these monoaminergic neurons have been lacking. We have generated two BAC transgenic mouse lines expressing the outward proton pump, enhanced ArchT3.0 (eArchT3.0), and GFP under control of the regulatory elements of either the dopamine transporter (DAT; Jax# 031663) or the tryptophan hydroxylase 2 (TPH2; Jax# 031662) gene locus. We demonstrate highly faithful and specific expression of these lines in dopaminergic and serotonergic neurons respectively. Additionally we validate effective and sensitive eArchT3.0-mediated silencing of these neurons using slice electrophysiology as well as with a well-established behavioral assay. These new transgenic tools will help expedite the study of dopaminergic and serotonergic system function in normal behavior and disease.
Collapse
Affiliation(s)
- Alexandra Krol
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Violeta G Lopez-Huerta
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States.,Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States.,Institute of Cellular Physiology, Department of Neurodevelopment and Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Taylor E C Corey
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, United States.,Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, United States
| | - Jonathan T Ting
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States.,Human Cell Types, Allen Institute for Brain Science, Seattle, WA, United States
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States.,Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| |
Collapse
|
178
|
Wang HL, Zhang S, Qi J, Wang H, Cachope R, Mejias-Aponte CA, Gomez JA, Mateo-Semidey GE, Beaudoin GMJ, Paladini CA, Cheer JF, Morales M. Dorsal Raphe Dual Serotonin-Glutamate Neurons Drive Reward by Establishing Excitatory Synapses on VTA Mesoaccumbens Dopamine Neurons. Cell Rep 2019; 26:1128-1142.e7. [PMID: 30699344 PMCID: PMC6489450 DOI: 10.1016/j.celrep.2019.01.014] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/12/2018] [Accepted: 01/03/2019] [Indexed: 11/26/2022] Open
Abstract
Dorsal raphe (DR) serotonin neurons provide a major input to the ventral tegmental area (VTA). Here, we show that DR serotonin transporter (SERT) neurons establish both asymmetric and symmetric synapses on VTA dopamine neurons, but most of these synapses are asymmetric. Moreover, the DR-SERT terminals making asymmetric synapses on VTA dopamine neurons coexpress vesicular glutamate transporter 3 (VGluT3; transporter for accumulation of glutamate for its synaptic release), suggesting the excitatory nature of these synapses. VTA photoactivation of DR-SERT fibers promotes conditioned place preference, elicits excitatory currents on mesoaccumbens dopamine neurons, increases their firing, and evokes dopamine release in nucleus accumbens. These effects are blocked by VTA inactivation of glutamate and serotonin receptors, supporting the idea of glutamate release in VTA from dual DR SERT-VGluT3 inputs. Our findings suggest a path-specific input from DR serotonergic neurons to VTA that promotes reward by the release of glutamate and activation of mesoaccumbens dopamine neurons.
Collapse
Affiliation(s)
- Hui-Ling Wang
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA
| | - Shiliang Zhang
- National Institute on Drug Abuse, Electron Microscopy Core, NIH, Baltimore, MD, USA
| | - Jia Qi
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA
| | - Huikun Wang
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA
| | - Roger Cachope
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Jorge A Gomez
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | | | - Gerard M J Beaudoin
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Carlos A Paladini
- Department of Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Joseph F Cheer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marisela Morales
- National Institute on Drug Abuse, Neuronal Networks Section, NIH, Baltimore, MD, USA.
| |
Collapse
|
179
|
Klawonn AM, Malenka RC. Nucleus Accumbens Modulation in Reward and Aversion. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:119-129. [PMID: 30674650 PMCID: PMC6650377 DOI: 10.1101/sqb.2018.83.037457] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The nucleus accumbens (NAc) is a key node of the brain’s circuitry that is responsible for translating motivation into action. It has been implicated in playing critical roles in virtually all forms of adaptive and pathological motivated behaviors. It is subject to modulation by a broad array of inputs that influence NAc activity in complex ways that are still poorly understood. Here, we briefly review current knowledge about the behavioral consequences of NAc modulation, focusing on recent studies that use novel techniques developed and implemented over the last decade.
Collapse
Affiliation(s)
- Anna M Klawonn
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
180
|
Li H, Pullmann D, Cho JY, Eid M, Jhou TC. Generality and opponency of rostromedial tegmental (RMTg) roles in valence processing. eLife 2019; 8:41542. [PMID: 30667358 PMCID: PMC6361585 DOI: 10.7554/elife.41542] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/04/2019] [Indexed: 12/31/2022] Open
Abstract
The rostromedial tegmental nucleus (RMTg), a GABAergic afferent to midbrain dopamine (DA) neurons, has been hypothesized to be broadly activated by aversive stimuli. However, this encoding pattern has only been demonstrated for a limited number of stimuli, and the RMTg influence on ventral tegmental (VTA) responses to aversive stimuli is untested. Here, we found that RMTg neurons are broadly excited by aversive stimuli of different sensory modalities and inhibited by reward-related stimuli. These stimuli include visual, auditory, somatosensory and chemical aversive stimuli, as well as “opponent” motivational states induced by removal of sustained rewarding or aversive stimuli. These patterns are consistent with broad encoding of negative valence in a subset of RMTg neurons. We further found that valence-encoding RMTg neurons preferentially project to the DA-rich VTA versus other targets, and excitotoxic RMTg lesions greatly reduce aversive stimulus-induced inhibitions in VTA neurons, particularly putative DA neurons, while also impairing conditioned place aversion to multiple aversive stimuli. Together, our findings indicate a broad RMTg role in encoding aversion and driving VTA responses and behavior.
Collapse
Affiliation(s)
- Hao Li
- Department of Neuroscience, Medical University of South Carolina, Charleston, United States
| | - Dominika Pullmann
- Department of Neuroscience, Medical University of South Carolina, Charleston, United States
| | - Jennifer Y Cho
- Department of Neuroscience, Medical University of South Carolina, Charleston, United States
| | - Maya Eid
- Department of Neuroscience, Medical University of South Carolina, Charleston, United States
| | | |
Collapse
|
181
|
Li Y, Li CY, Xi W, Jin S, Wu ZH, Jiang P, Dong P, He XB, Xu FQ, Duan S, Zhou YD, Li XM. Rostral and Caudal Ventral Tegmental Area GABAergic Inputs to Different Dorsal Raphe Neurons Participate in Opioid Dependence. Neuron 2019; 101:748-761.e5. [PMID: 30638902 DOI: 10.1016/j.neuron.2018.12.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/26/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022]
Abstract
Both the ventral tegmental area (VTA) and dorsal raphe nucleus (DRN) are involved in affective control and reward-related behaviors. Moreover, the neuronal activities of the VTA and DRN are modulated by opioids. However, the precise circuits from the VTA to DRN and how opioids modulate these circuits remain unknown. Here, we found that neurons projecting from the VTA to DRN are primarily GABAergic. Rostral VTA (rVTA) GABAergic neurons preferentially innervate DRN GABAergic neurons, thus disinhibiting DRN serotonergic neurons. Optogenetic activation of this circuit induces aversion. In contrast, caudal VTA (cVTA) GABAergic neurons mainly target DRN serotonergic neurons, and activation of this circuit promotes reward. Importantly, μ-opioid receptors (MOPs) are selectively expressed at rVTA→DRN GABAergic synapses, and morphine depresses the synaptic transmission. Chronically elevating the activity of the rVTA→DRN pathway specifically interrupts morphine-induced conditioned place preference. This opioid-modulated inhibitory circuit may yield insights into morphine reward and dependence pathogenesis.
Collapse
Affiliation(s)
- Yue Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chun-Yue Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wang Xi
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Sen Jin
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Zuo-Hang Wu
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ping Jiang
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ping Dong
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao-Bin He
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Fu-Qiang Xu
- CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Wuhan Institute of Physics and Mathematics, Wuhan 430071, China
| | - Shumin Duan
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yu-Dong Zhou
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao-Ming Li
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
182
|
Chen P, Hei M, Kong L, Liu Y, Yang Y, Mu H, Zhang X, Zhao S, Duan J. One water-soluble polysaccharide from Ginkgo biloba leaves with antidepressant activities via modulation of the gut microbiome. Food Funct 2019; 10:8161-8171. [DOI: 10.1039/c9fo01178a] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
By performing a pyrosequencing-based analysis of bacterial community, we detected that one Ginkgo biloba polysaccharide reversed depression-associated gut dysbiosis and increased the richness of Lactobacillus species which has been proven to be a path to relieve depression.
Collapse
Affiliation(s)
- Peng Chen
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling 712100
- China
| | - Maofang Hei
- State Key Laboratory of Crop Stress Biology for Arid Areas
- College of Veterinary Medicine
- Northwest A&F University
- Yangling 712100
- China
| | - Lili Kong
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling 712100
- China
| | - Yinyin Liu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling 712100
- China
| | - Yu Yang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling 712100
- China
| | - Haibo Mu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling 712100
- China
| | - Xiuyun Zhang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling 712100
- China
| | - Shanting Zhao
- State Key Laboratory of Crop Stress Biology for Arid Areas
- College of Veterinary Medicine
- Northwest A&F University
- Yangling 712100
- China
| | - Jinyou Duan
- Shaanxi Key Laboratory of Natural Products & Chemical Biology
- College of Chemistry & Pharmacy
- Northwest A&F University
- Yangling 712100
- China
| |
Collapse
|
183
|
Di Miceli M, Omoloye A, Gronier B. Chronic methylphenidate treatment during adolescence has long-term effects on monoaminergic function. J Psychopharmacol 2019; 33:109-121. [PMID: 30334678 DOI: 10.1177/0269881118805494] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Psychostimulants like methylphenidate or D-amphetamine are often prescribed for attention deficit and hyperactivity disorders in children. Whether such drugs can be administered into a developing brain without consequences in adulthood is still an open question. METHODS Here, using in vivo extracellular electrophysiology in anesthetised preparations, combined with behavioural assays, we have examined the long-term consequences in adulthood of a chronic methylphenidate oral administration (5 mg/kg/day, 15 days) in early adolescent (post-natal day 28) and late adolescent (post-natal day 42) rats, by evaluating body weight change, sucrose preference (indicator of anhedonia), locomotor sensitivity to D-amphetamine and electrical activities of ventral tegmental area dopamine and dorsal raphe nucleus serotonin neurons. RESULTS Chronic methylphenidate treatment during early or late adolescence did not induce weight deficiencies and anhedonia-like behaviours at adulthood. However, it increased bursting activities of dorsal raphe nucleus serotonin neurons. Furthermore, chronic methylphenidate treatment during early but not during late adolescence enhanced D-amphetamine-induced rearing activity, as well as ventral tegmental area dopamine cell excitability (firing, burst and population activity), associated with a partial desensitisation of dopamine D2 auto-receptors. CONCLUSIONS We have demonstrated here that early, but not late, adolescent exposure to oral methylphenidate may induce long-lasting effects on monoamine neurotransmission. The possible clinical implication of these data will be discussed.
Collapse
Affiliation(s)
- Mathieu Di Miceli
- Pharmacology and Neuroscience Research Group, Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Adesina Omoloye
- Pharmacology and Neuroscience Research Group, Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | - Benjamin Gronier
- Pharmacology and Neuroscience Research Group, Leicester School of Pharmacy, De Montfort University, Leicester, UK
| |
Collapse
|
184
|
Rhoades JL, Nelson JC, Nwabudike I, Yu SK, McLachlan IG, Madan GK, Abebe E, Powers JR, Colón-Ramos DA, Flavell SW. ASICs Mediate Food Responses in an Enteric Serotonergic Neuron that Controls Foraging Behaviors. Cell 2018; 176:85-97.e14. [PMID: 30580965 DOI: 10.1016/j.cell.2018.11.023] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/27/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Abstract
Animals must respond to the ingestion of food by generating adaptive behaviors, but the role of gut-brain signaling in behavioral regulation is poorly understood. Here, we identify conserved ion channels in an enteric serotonergic neuron that mediate its responses to food ingestion and decipher how these responses drive changes in foraging behavior. We show that the C. elegans serotonergic neuron NSM acts as an enteric sensory neuron that acutely detects food ingestion. We identify the novel and conserved acid-sensing ion channels (ASICs) DEL-7 and DEL-3 as NSM-enriched channels required for feeding-dependent NSM activity, which in turn drives slow locomotion while animals feed. Point mutations that alter the DEL-7 channel change NSM dynamics and associated behavioral dynamics of the organism. This study provides causal links between food ingestion, molecular and physiological properties of an enteric serotonergic neuron, and adaptive feeding behaviors, yielding a new view of how enteric neurons control behavior.
Collapse
Affiliation(s)
- Jeffrey L Rhoades
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jessica C Nelson
- Department of Neuroscience and Department of Cell Biology, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Ijeoma Nwabudike
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephanie K Yu
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ian G McLachlan
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gurrein K Madan
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eden Abebe
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua R Powers
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel A Colón-Ramos
- Department of Neuroscience and Department of Cell Biology, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT 06536, USA
| | - Steven W Flavell
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
185
|
Stochastic synaptic plasticity underlying compulsion in a model of addiction. Nature 2018; 564:366-371. [PMID: 30568192 DOI: 10.1038/s41586-018-0789-4] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/13/2018] [Indexed: 01/08/2023]
Abstract
Activation of the mesolimbic dopamine system reinforces goal-directed behaviours. With repetitive stimulation-for example, by chronic drug abuse-the reinforcement may become compulsive and intake continues even in the face of major negative consequences. Here we gave mice the opportunity to optogenetically self-stimulate dopaminergic neurons and observed that only a fraction of mice persevered if they had to endure an electric shock. Compulsive lever pressing was associated with an activity peak in the projection terminals from the orbitofrontal cortex (OFC) to the dorsal striatum. Although brief inhibition of OFC neurons temporarily relieved compulsive reinforcement, we found that transmission from the OFC to the striatum was permanently potentiated in persevering mice. To establish causality, we potentiated these synapses in vivo in mice that stopped optogenetic self-stimulation of dopamine neurons because of punishment; this led to compulsive lever pressing, whereas depotentiation in persevering mice had the converse effect. In summary, synaptic potentiation of transmission from the OFC to the dorsal striatum drives compulsive reinforcement, a defining symptom of addiction.
Collapse
|
186
|
Luo T, Yu S, Cai S, Zhang Y, Jiao Y, Yu T, Yu W. Parabrachial Neurons Promote Behavior and Electroencephalographic Arousal From General Anesthesia. Front Mol Neurosci 2018; 11:420. [PMID: 30564094 PMCID: PMC6288364 DOI: 10.3389/fnmol.2018.00420] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022] Open
Abstract
General anesthesia has been used clinically for more than 170 years, yet its underlying mechanisms are still not fully understood. The parabrachial nucleus (PBN) in the brainstem has been known to be crucial for regulating wakefulness and signs of arousal on the cortical electroencephalogram (EEG). Lesions of the parabrachial complex lead to unresponsiveness and a monotonous high-voltage, and a slow-wave EEG, which are the two main features of general anesthesia. However, it is unclear whether and how the PBN functions in the process of general anesthesia. By recording the levels of calcium in vivo in real-time, we found that the neural activity in PBN is suppressed during anesthesia, while it is robustly activated during recovery from propofol and isoflurane anesthesia. The activation of PBN neurons by “designer receptors exclusively activated by designer drugs” (DREADDs) shortened the recovery time but did not change the induction time. Cortical EEG recordings revealed that the neural activation of PBN specifically affected the recovery period, with a decrease of δ-band power or an increase in β-band power; no EEG changes were seen in the anesthesia period. Furthermore, the activation of PBN elicited neural activation in the prefrontal cortex, basal forebrain, lateral hypothalamus, thalamus, and supramammillary nucleus. Thus, PBN is critical for behavioral and electroencephalographic arousal without affecting the induction of general anesthesia.
Collapse
Affiliation(s)
- Tianyuan Luo
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Shouyang Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Shuang Cai
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Yu Zhang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
187
|
Fractionating Blunted Reward Processing Characteristic of Anhedonia by Over-Activating Primate Subgenual Anterior Cingulate Cortex. Neuron 2018; 101:307-320.e6. [PMID: 30528065 PMCID: PMC6344231 DOI: 10.1016/j.neuron.2018.11.021] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/10/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022]
Abstract
Anhedonia is a core symptom of depression, but the underlying neurobiological mechanisms are unknown. Correlative neuroimaging studies implicate dysfunction within ventromedial prefrontal cortex, but the causal roles of specific subregions remain unidentified. We addressed these issues by combining intracerebral microinfusions with cardiovascular and behavioral monitoring in marmoset monkeys to show that over-activation of primate subgenual anterior cingulate cortex (sgACC, area 25) blunts appetitive anticipatory, but not consummatory, arousal, whereas manipulations of adjacent perigenual ACC (pgACC, area 32) have no effect. sgACC/25 over-activation also reduces the willingness to work for reward. 18F-FDG PET imaging reveals over-activation induced metabolic changes in circuits involved in reward processing and interoception. Ketamine treatment ameliorates the blunted anticipatory arousal and reverses associated metabolic changes. These results demonstrate a causal role for primate sgACC/25 over-activity in selective aspects of impaired reward processing translationally relevant to anhedonia, and ketamine's modulation of an affective network to exert its action.
Collapse
|
188
|
Wei C, Han X, Weng D, Feng Q, Qi X, Li J, Luo M. Response dynamics of midbrain dopamine neurons and serotonin neurons to heroin, nicotine, cocaine, and MDMA. Cell Discov 2018; 4:60. [PMID: 30416749 PMCID: PMC6218454 DOI: 10.1038/s41421-018-0060-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/21/2018] [Accepted: 08/26/2018] [Indexed: 12/13/2022] Open
Abstract
Heroin, nicotine, cocaine, and MDMA are abused by billions of people. They are believed to target midbrain dopamine neurons and/or serotonin neurons, but their effects on the dynamic neuronal activity remain unclear in behaving states. By combining cell-type-specific fiber photometry of Ca2+ signals and intravenous drug infusion, here we show that these four drugs of abuse profoundly modulate the activity of mouse midbrain dopamine neurons and serotonin neurons with distinct potency and kinetics. Heroin strongly activates dopamine neurons, and only excites serotonin neurons at higher doses. Nicotine activates dopamine neurons in merely a few seconds, but produces minimal effects on serotonin neurons. Cocaine and MDMA cause long-lasting suppression of both dopamine neurons and serotonin neurons, although MDMA inhibits serotonin neurons more profoundly. Moreover, these inhibitory effects are mediated through the activity of dopamine and serotonin autoreceptors. These results suggest that the activity of dopamine neurons and that of serotonin neurons are more closely associated with the drug's reinforcing property and the drug's euphorigenic property, respectively. This study also shows that our methodology may facilitate further in-vivo interrogation of neural dynamics using animal models of drug addiction.
Collapse
Affiliation(s)
- Chao Wei
- 1School of Life Sciences, Peking University, Beijing, 100871 China.,2Peking University-Tsinghua University-NIBS Graduate Program, Peking University, Beijing, 100081 China.,3National Institute of Biological Sciences (NIBS), Beijing, 102206 China
| | - Xiao Han
- 4Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850 China
| | - Danwei Weng
- 3National Institute of Biological Sciences (NIBS), Beijing, 102206 China
| | - Qiru Feng
- 3National Institute of Biological Sciences (NIBS), Beijing, 102206 China
| | - Xiangbing Qi
- 3National Institute of Biological Sciences (NIBS), Beijing, 102206 China
| | - Jin Li
- 4Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850 China
| | - Minmin Luo
- 3National Institute of Biological Sciences (NIBS), Beijing, 102206 China.,5School of Life Sciences, Tsinghua University, Beijing, 100084 China
| |
Collapse
|
189
|
Ren J, Friedmann D, Xiong J, Liu CD, Ferguson BR, Weerakkody T, DeLoach KE, Ran C, Pun A, Sun Y, Weissbourd B, Neve RL, Huguenard J, Horowitz MA, Luo L. Anatomically Defined and Functionally Distinct Dorsal Raphe Serotonin Sub-systems. Cell 2018; 175:472-487.e20. [PMID: 30146164 PMCID: PMC6173627 DOI: 10.1016/j.cell.2018.07.043] [Citation(s) in RCA: 272] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 07/01/2018] [Accepted: 07/25/2018] [Indexed: 01/21/2023]
Abstract
The dorsal raphe (DR) constitutes a major serotonergic input to the forebrain and modulates diverse functions and brain states, including mood, anxiety, and sensory and motor functions. Most functional studies to date have treated DR serotonin neurons as a single population. Using viral-genetic methods, we found that subcortical- and cortical-projecting serotonin neurons have distinct cell-body distributions within the DR and differentially co-express a vesicular glutamate transporter. Further, amygdala- and frontal-cortex-projecting DR serotonin neurons have largely complementary whole-brain collateralization patterns, receive biased inputs from presynaptic partners, and exhibit opposite responses to aversive stimuli. Gain- and loss-of-function experiments suggest that amygdala-projecting DR serotonin neurons promote anxiety-like behavior, whereas frontal-cortex-projecting neurons promote active coping in the face of challenge. These results provide compelling evidence that the DR serotonin system contains parallel sub-systems that differ in input and output connectivity, physiological response properties, and behavioral functions.
Collapse
Affiliation(s)
- Jing Ren
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Drew Friedmann
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Jing Xiong
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Cindy D Liu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Brielle R Ferguson
- Department of Neurology and Neurological Sciences, Stanford, CA 94305, USA
| | - Tanya Weerakkody
- Department of Neurology and Neurological Sciences, Stanford, CA 94305, USA
| | - Katherine E DeLoach
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Chen Ran
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Albert Pun
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Yanwen Sun
- Department of Physics, Stanford University, Stanford, CA 94305, USA
| | - Brandon Weissbourd
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Rachael L Neve
- Delivery Technology Core, Massachusetts General Hospital, Cambridge, MA 02139, USA
| | - John Huguenard
- Department of Neurology and Neurological Sciences, Stanford, CA 94305, USA
| | - Mark A Horowitz
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
190
|
Garcia-Garcia AL, Canetta S, Stujenske JM, Burghardt NS, Ansorge MS, Dranovsky A, Leonardo ED. Serotonin inputs to the dorsal BNST modulate anxiety in a 5-HT 1A receptor-dependent manner. Mol Psychiatry 2018; 23:1990-1997. [PMID: 28761080 PMCID: PMC5794659 DOI: 10.1038/mp.2017.165] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/31/2017] [Accepted: 06/20/2017] [Indexed: 11/24/2022]
Abstract
Serotonin (5-HT) neurons project from the raphe nuclei throughout the brain where they act to maintain homeostasis. Here, we study 5-HT inputs into the bed nucleus of the stria terminalis (BNST), a major subdivision of the extended amygdala that has been proposed to regulate responses to anxiogenic environments in humans and rodents. While the dorsal part of the BNST (dBNST) receives dense 5-HT innervation, whether and how 5-HT in the dBNST normally modulates anxiety remains unclear. Using optogenetics, we demonstrate that activation of 5-HT terminals in the dBNST reduces anxiety in a highly anxiogenic environment. Further analysis revealed that optogenetic inhibition of 5-HT inputs into the dBNST increases anxiety in a less anxiogenic environment. We found that 5-HT predominantly hyperpolarizes dBNST neurons, reducing their activity in a manner that can be blocked by a 5-HT1A antagonist. Finally, we demonstrate that activation of 5-HT1A receptors in the dBNST is necessary for the anxiolytic effect observed following optogenetic stimulation of 5-HT inputs into the dBNST. These data reveal that 5-HT release in the dBNST modulates anxiety-like behavior via 5-HT1A receptors under naturalistic conditions.
Collapse
Affiliation(s)
- Alvaro L. Garcia-Garcia
- Dranovsky-Leonardo (ADL) lab, Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032,Address correspondence to AGG at , to AD at and to EDL at . Telephone: (001) (646) 774-7105. Fax: (001) (646) 774-7117
| | - Sarah Canetta
- Department of Psychiatry, Columbia University, New York, NY 10032
| | - Joseph M. Stujenske
- Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032
| | - Nesha S. Burghardt
- Department of Psychology, Hunter College, City University of New York, 695 Park Avenue, New York, NY 10065
| | - Mark S. Ansorge
- Department of Psychiatry, Columbia University, New York, NY 10032
| | - Alex Dranovsky
- Dranovsky-Leonardo (ADL) lab, Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032,Address correspondence to AGG at , to AD at and to EDL at . Telephone: (001) (646) 774-7105. Fax: (001) (646) 774-7117
| | - E. David Leonardo
- Dranovsky-Leonardo (ADL) lab, Department of Psychiatry, Division of Integrative Neuroscience, Columbia University and the New York State Psychiatric Institute, 1051 Riverside Dr. Box 87, New York, NY 10032,Address correspondence to AGG at , to AD at and to EDL at . Telephone: (001) (646) 774-7105. Fax: (001) (646) 774-7117
| |
Collapse
|
191
|
Höflich A, Michenthaler P, Kasper S, Lanzenberger R. Circuit Mechanisms of Reward, Anhedonia, and Depression. Int J Neuropsychopharmacol 2018; 22:105-118. [PMID: 30239748 PMCID: PMC6368373 DOI: 10.1093/ijnp/pyy081] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/12/2018] [Indexed: 12/23/2022] Open
Abstract
Pleasure and motivation are important factors for goal-directed behavior and well-being in both animals and humans. Intact hedonic capacity requires an undisturbed interplay between a number of different brain regions and transmitter systems. Concordantly, dysfunction of networks encoding for reward have been shown in depression and other psychiatric disorders. The development of technological possibilities to investigate connectivity on a functional level in humans and to directly influence networks in animals using optogenetics among other techniques has provided new important insights in this field of research.In this review, we aim to provide an overview on the neurobiological substrates of anhedonia on a network level. For this purpose, definition of anhedonia and the involved reward components are described first, then current data on reward networks in healthy individuals and in depressed patients are summarized, and the roles of different neurotransmitter systems involved in reward processing are specified. Based on this information, the impact of different therapeutic approaches on reward processing is described with a particular focus on deep brain stimulation (DBS) as a possibility for a direct modulation of human brain structures in vivo.Overall, results of current studies emphasize the importance of anhedonia in psychiatric disorders and the relevance of targeting this phenotype for a successful psychiatric treatment. However, more data incorporating these results for the refinement of methodological approaches are needed to be able to develop individually tailored therapeutic concepts based on both clinical and neurobiological profiles of patients.
Collapse
Affiliation(s)
- Anna Höflich
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Paul Michenthaler
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria,Correspondence: Rupert Lanzenberger, MD, PD, Department of Psychiatry and Psychotherapy, Medical University of Vienna, Waehringer Guertel 18–20, 1090 Vienna, Austria ()
| |
Collapse
|
192
|
Hamed A, Kursa MB. Inter-individual differences in serotonin and glutamate co-transmission reflect differentiation in context-induced conditioned 50-kHz USVs response after morphine withdrawal. Brain Struct Funct 2018; 223:3149-3167. [PMID: 29774428 PMCID: PMC6132671 DOI: 10.1007/s00429-018-1683-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/09/2018] [Indexed: 12/13/2022]
Abstract
A growing body of research provides compelling evidence that in rats 50-kHz USVs are a form of expression of positive emotions. Context-induced 50-kHz USVs emission is variable among rats, indicating individual differences in contextual response bound up with pharmacological reward. The aims of this study were to: extract the most important neurotransmitters related to context-induced conditioned 50-kHz USVs response; find biological basis of existing inter-individual differences in context-induced conditioned 50-kHz USVs response; create a model of all-to-all neurotransmitters correlations. The data collected here confirms that re-exposure to the context of morphine administration after the withdrawal period increases the level of 50-kHz USVs and this contextual response is associated with elevated serotonin concentrations in amygdala, hippocampus and mPFC and with increased Glu/Gln ratio in nucleus accumbens. The concentration of serotonin increases simultaneously in amygdala, nucleus accumbens and hippocampus. Moreover, 5-HT concentration in amygdala is bound up with glutamate level in this structure as well as in hippocampus. Furthermore, Glu/Gln ratio in nucleus accumbens has strong associations with Glu/Gln ratio simultaneously in VTA, amygdala, striatum and hippocampus. All-to-all-analysis indicate that concentration of glutamate in hippocampus is proportional to glutamate in VTA and GABA concentration in the hippocampus. We have also demonstrated that Glu/GABA ratio in VTA and amygdala was elevated after post withdrawal re-exposure to the pharmacological reward paired context. Presented analysis indicates a strong correlation between serotonergic and glutamatergic systems in context-induced conditioned response. The strength of this co-transmission correlates with the number of 50-kHz USVs emitted in response to morphine-paired context.
Collapse
Affiliation(s)
- Adam Hamed
- Laboratory of Spatial Memory, Department of Cellular and Molecular Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Miron Bartosz Kursa
- Interdisciplinary Centre for Mathematical and Computational Modelling, University of Warsaw, Pawinskiego 5A, 02-106, Warsaw, Poland
| |
Collapse
|
193
|
Keesom SM, Morningstar MD, Sandlain R, Wise BM, Hurley LM. Social isolation reduces serotonergic fiber density in the inferior colliculus of female, but not male, mice. Brain Res 2018; 1694:94-103. [DOI: 10.1016/j.brainres.2018.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/15/2018] [Accepted: 05/11/2018] [Indexed: 12/26/2022]
|
194
|
5-HT release in nucleus accumbens rescues social deficits in mouse autism model. Nature 2018; 560:589-594. [PMID: 30089910 DOI: 10.1038/s41586-018-0416-4] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 06/27/2018] [Indexed: 01/07/2023]
Abstract
Dysfunction in prosocial interactions is a core symptom of autism spectrum disorder. However, the neural mechanisms that underlie sociability are poorly understood, limiting the rational development of therapies to treat social deficits. Here we show in mice that bidirectional modulation of the release of serotonin (5-HT) from dorsal raphe neurons in the nucleus accumbens bidirectionally modifies sociability. In a mouse model of a common genetic cause of autism spectrum disorder-a copy number variation on chromosome 16p11.2-genetic deletion of the syntenic region from 5-HT neurons induces deficits in social behaviour and decreases dorsal raphe 5-HT neuronal activity. These sociability deficits can be rescued by optogenetic activation of dorsal raphe 5-HT neurons, an effect requiring and mimicked by activation of 5-HT1b receptors in the nucleus accumbens. These results demonstrate an unexpected role for 5-HT action in the nucleus accumbens in social behaviours, and suggest that targeting this mechanism may prove therapeutically beneficial.
Collapse
|
195
|
Expression map of 78 brain-expressed mouse orphan GPCRs provides a translational resource for neuropsychiatric research. Commun Biol 2018; 1:102. [PMID: 30271982 PMCID: PMC6123746 DOI: 10.1038/s42003-018-0106-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/06/2018] [Indexed: 12/26/2022] Open
Abstract
Orphan G-protein-coupled receptors (oGPCRs) possess untapped potential for drug discovery. In the brain, oGPCRs are generally expressed at low abundance and their function is understudied. Expression profiling is an essential step to position oGPCRs in brain function and disease, however public databases provide only partial information. Here, we fine-map expression of 78 brain-oGPCRs in the mouse, using customized probes in both standard and supersensitive in situ hybridization. Images are available at http://ogpcr-neuromap.douglas.qc.ca. This searchable database contains over 8000 coronal brain sections across 1350 slides, providing the first public mapping resource dedicated to oGPCRs. Analysis with public mouse (60 oGPCRs) and human (56 oGPCRs) genome-wide datasets identifies 25 oGPCRs with potential to address emotional and/or cognitive dimensions of psychiatric conditions. We probe their expression in postmortem human brains using nanoString, and included data in the resource. Correlating human with mouse datasets reveals excellent suitability of mouse models for oGPCRs in neuropsychiatric research. Aliza Ehrlich et al. report the fine-mapping of orphan GPCR (oGPCR) transcripts in the mouse brain using in situ hybridization and provide a public resource for data mining. The authors also mapped 25 selected oGPCRs in human brains, identifying oGPCRs with high correlation between species and potential roles in neuropsychiatric disorders.
Collapse
|
196
|
van Galen KA, Ter Horst KW, Booij J, la Fleur SE, Serlie MJ. The role of central dopamine and serotonin in human obesity: lessons learned from molecular neuroimaging studies. Metabolism 2018; 85:325-339. [PMID: 28970033 DOI: 10.1016/j.metabol.2017.09.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Obesity results from an imbalance between energy intake and expenditure, and many studies have aimed to determine why obese individuals continue to (over)consume food under conditions of caloric excess. The two major "neurotransmitter hypotheses" of obesity state that increased food intake is partially driven by decreased dopamine-mediated reward and decreased serotonin-mediated homeostatic feedback in response to food intake. Using molecular neuroimaging studies to visualize and quantify aspects of the central dopamine and serotonin systems in vivo, recent PET and SPECT studies have also implicated alterations in these systems in human obesity. The interpretation of these data, however, is more complex than it may appear. Here, we discuss important characteristics and limitations of current radiotracer methods and use this framework to comprehensively review the available human data on central dopamine and serotonin in obesity. On the basis of the available evidence, we conclude that obesity is associated with decreased central dopaminergic and serotonergic signaling and that future research, especially in long-term follow-up and interventional settings, is needed to advance our understanding of the neuronal pathophysiology of obesity in humans.
Collapse
Affiliation(s)
- Katy A van Galen
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands
| | - Kasper W Ter Horst
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands
| | - Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands; Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands.
| |
Collapse
|
197
|
Reward probability and timing uncertainty alter the effect of dorsal raphe serotonin neurons on patience. Nat Commun 2018; 9:2048. [PMID: 29858574 PMCID: PMC5984631 DOI: 10.1038/s41467-018-04496-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 05/03/2018] [Indexed: 12/28/2022] Open
Abstract
Recent experiments have shown that optogenetic activation of serotonin neurons in the dorsal raphe nucleus (DRN) in mice enhances patience in waiting for future rewards. Here, we show that serotonin effect in promoting waiting is maximized by both high probability and high timing uncertainty of reward. Optogenetic activation of serotonergic neurons prolongs waiting time in no-reward trials in a task with 75% food reward probability, but not with 50 or 25% reward probabilities. Serotonin effect in promoting waiting increases when the timing of reward presentation becomes unpredictable. To coherently explain the experimental data, we propose a Bayesian decision model of waiting that assumes that serotonin neuron activation increases the prior probability or subjective confidence of reward delivery. The present data and modeling point to the possibility of a generalized role of serotonin in resolving trade-offs, not only between immediate and delayed rewards, but also between sensory evidence and subjective confidence.
Collapse
|
198
|
Henke A, Kovalyova Y, Dunn M, Dreier D, Gubernator NG, Dincheva I, Hwu C, Šebej P, Ansorge MS, Sulzer D, Sames D. Toward Serotonin Fluorescent False Neurotransmitters: Development of Fluorescent Dual Serotonin and Vesicular Monoamine Transporter Substrates for Visualizing Serotonin Neurons. ACS Chem Neurosci 2018; 9:925-934. [PMID: 29281252 PMCID: PMC6342556 DOI: 10.1021/acschemneuro.7b00320] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Ongoing efforts in our laboratories focus on design of optical reporters known as fluorescent false neurotransmitters (FFNs) that enable the visualization of uptake into, packaging within, and release from individual monoaminergic neurons and presynaptic sites in the brain. Here, we introduce the molecular probe FFN246 as an expansion of the FFN platform to the serotonergic system. Combining the acridone fluorophore with the ethylamine recognition element of serotonin, we identified FFN54 and FFN246 as substrates for both the serotonin transporter and the vesicular monoamine transporter 2 (VMAT2). A systematic structure-activity study revealed the basic structural chemotype of aminoalkyl acridones required for serotonin transporter (SERT) activity and enabled lowering the background labeling of these probes while maintaining SERT activity, which proved essential for obtaining sufficient signal in the brain tissue (FFN246). We demonstrate the utility of FFN246 for direct examination of SERT activity and SERT inhibitors in 96-well cell culture assays, as well as specific labeling of serotonergic neurons of the dorsal raphe nucleus in the living tissue of acute mouse brain slices. While we found only minor FFN246 accumulation in serotonergic axons in murine brain tissue, FFN246 effectively traces serotonin uptake and packaging in the soma of serotonergic neurons with improved photophysical properties and loading parameters compared to known serotonin-based fluorescent tracers.
Collapse
Affiliation(s)
- Adam Henke
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| | - Yekaterina Kovalyova
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| | - Matthew Dunn
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| | - Dominik Dreier
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| | - Niko G Gubernator
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| | - Iva Dincheva
- Department of Psychiatry , Columbia University , New York , New York 10032 , United States
| | - Christopher Hwu
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| | - Peter Šebej
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| | - Mark S Ansorge
- Department of Psychiatry , Columbia University , New York , New York 10032 , United States
| | - David Sulzer
- Department of Psychiatry , Columbia University , New York , New York 10032 , United States
- Department of Neurology , Columbia University , New York , New York 10032 , United States
- Department of Pharmacology , Columbia University , New York , New York 10032 , United States
| | - Dalibor Sames
- Department of Chemistry and Neuro Technology Center at Columbia University , New York , New York 10027 , United States
| |
Collapse
|
199
|
Distinct Anatomical Connectivity Patterns Differentiate Subdivisions of the Nonlemniscal Auditory Thalamus in Mice. Cereb Cortex 2018; 29:2437-2454. [DOI: 10.1093/cercor/bhy115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/04/2018] [Indexed: 11/14/2022] Open
|
200
|
Jiang H, Kim HF. Anatomical Inputs From the Sensory and Value Structures to the Tail of the Rat Striatum. Front Neuroanat 2018; 12:30. [PMID: 29773980 PMCID: PMC5943565 DOI: 10.3389/fnana.2018.00030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/05/2018] [Indexed: 11/17/2022] Open
Abstract
The caudal region of the rodent striatum, called the tail of the striatum (TS), is a relatively small area but might have a distinct function from other striatal subregions. Recent primate studies showed that this part of the striatum has a unique function in encoding long-term value memory of visual objects for habitual behavior. This function might be due to its specific connectivity. We identified inputs to the rat TS and compared those with inputs to the dorsomedial striatum (DMS) in the same animals. The TS directly received anatomical inputs from both sensory structures and value-coding regions, but the DMS did not. First, inputs from the sensory cortex and sensory thalamus to the TS were found; visual, auditory, somatosensory and gustatory cortex and thalamus projected to the TS but not to the DMS. Second, two value systems innervated the TS; dopamine and serotonin neurons in the lateral part of the substantia nigra pars compacta (SNc) and dorsal raphe nucleus projected to the TS, respectively. The DMS received inputs from the separate group of dopamine neurons in the medial part of the SNc. In addition, learning-related regions of the limbic system innervated the TS; the temporal areas and the basolateral amygdala selectively innervated the TS, but not the DMS. Our data showed that both sensory and value-processing structures innervated the TS, suggesting its plausible role in value-guided sensory-motor association for habitual behavior.
Collapse
Affiliation(s)
- Haiyan Jiang
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, South Korea
| | - Hyoung F Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, South Korea.,Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, South Korea
| |
Collapse
|