1
|
Ohmura Y, Nagayasu K. Functional Diversity of Serotonin Neurons in the Dorsal and Median Raphe Nuclei in Emotional Responses. Neuropsychopharmacol Rep 2025; 45:e70015. [PMID: 40254954 PMCID: PMC12010045 DOI: 10.1002/npr2.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/22/2025] Open
Abstract
Of serotonergic nuclei in the central nervous system, mainly the dorsal raphe nucleus (DRN) and median raphe nucleus (MRN) project to the forebrain and midbrain; therefore, these nuclei are involved in emotional/cognitive functions and psychiatric disorders. Researchers have often generalized findings from the DRN to represent the functions of the entire serotonergic system, primarily due to the fact that the DRN is the largest serotonergic nucleus and due to the assumption that the serotonergic system operates as a single, cohesive unit. However, recent evidence is challenging this perspective and necessitating a reevaluation. In this brief review, we summarize recent studies demonstrating the functional diversity of the DRN alongside the functional unity of the MRN. These findings suggest that different subpopulations within the serotonergic system may exert opposing effects on emotional functions. Furthermore, this diversity-aware approach will help settle ongoing debates regarding the serotonin hypothesis of depression, which stems from the difficulty in the application of this approach in humans. We advocate for increased efforts to identify factors associated with these functional subgroups, which could lead to more targeted and effective interventions.
Collapse
Affiliation(s)
- Yu Ohmura
- Chinese Institute for Brain Research, Beijing (CIBR)BeijingChina
| | - Kazuki Nagayasu
- Laboratory of Molecular NeuropharmacologyGraduate School of Pharmaceutical Sciences, Osaka UniversitySuitaJapan
- Project for Neural NetworksGraduate School of Pharmaceutical Sciences, Osaka UniversitySuitaJapan
| |
Collapse
|
2
|
Rios SM, Mootz JRK, Phillips TJ, Ingram SL. Absence of TAAR1 function increases methamphetamine-induced excitability of dorsal raphe serotonin neurons and drives binge-level methamphetamine intake. Neuropsychopharmacology 2025; 50:1136-1144. [PMID: 39934409 PMCID: PMC12089393 DOI: 10.1038/s41386-025-02063-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
Methamphetamine (MA) is a potent psychostimulant capable of exerting both rewarding and aversive effects, the balance of which likely drives variation in voluntary MA intake. Understanding the genetic factors underlying sensitivity to these effects of MA is critical for developing effective treatments. The activity of dorsal raphe serotonin neurons is linked to reward processing. Here, we performed whole-cell patch-clamp electrophysiology in dorsal raphe serotonin neurons from mice with high or low MA intake corresponding with high or low MA reward sensitivity. The MA drinking (MADR) mice consist of the MA reward sensitive MA high drinking (MAHDR) and the MA reward insensitive MA low drinking (MALDR) lines. MA is a trace amine-associated receptor 1 (TAAR1) agonist, and MAHDR mice are homozygous for a mutation in the Taar1 gene, Taar1m1J, that encodes non-functional TAAR1, whereas MALDR mice possess at least one copy of the reference Taar1+ allele that encodes functional TAAR1. Our previous research using CRISPR-Cas9-generated MAHDR-Taar1+/+ knock-in mice in which Taar1m1J was replaced with Taar1+, and non-edited MAHDR-Taar1m1J/m1J controls demonstrated that lack of TAAR1 function is critical for heightened MA consumption and MA reward sensitivity. Here, electrophysiological recordings in the MADR lines demonstrate a MA-induced decrease in dorsal raphe serotonin neuron activity from MALDR, but not MAHDR mice. However, in the presence of serotonin autoreceptor antagonists, MA potentiates dorsal raphe serotonin neuron activity of MAHDR, but not MALDR mice. Importantly, potentiation in the presence of the antagonists is abolished in knock-in mice expressing functional TAAR1. The knock-in mice did not display binge-level MA intake, consistent with the loss of MA-reward sensitivity previously reported in mice with functional TAAR1. Finally, because MA is a substrate of the serotonin transporter, we evaluated whether the serotonin transporter is necessary for MA-induced potentiation of dorsal raphe serotonin neuron activity in mice with non-functional TAAR1. The serotonin transporter antagonist fluoxetine blocks MA-induced potentiation for both MAHDR and MAHDR-Taar1m1J/m1J mice. Thus, TAAR1 function directly impacts MA reward sensitivity and MA intake and serves as a critical regulator of MA-induced activity of dorsal raphe serotonin neurons through its interaction with the serotonin transporter.
Collapse
Affiliation(s)
- Samantha M Rios
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - John R K Mootz
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
- Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Susan L Ingram
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
3
|
Robke R, Sansi F, Arbab T, Tunez A, Moore M, Bartsch D, Schönig K, Willuhn I. Optogenetic Stimulation of Novel Tph2-Cre Rats Advances Insight into Serotonin's Role in Locomotion, Reinforcement, and Compulsivity. J Neurosci 2025; 45:e1424242025. [PMID: 40204433 PMCID: PMC12096035 DOI: 10.1523/jneurosci.1424-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Serotonin critically modulates the activity of many brain networks, including circuits that control motivation and responses to rewarding and aversive stimuli. Furthermore, the serotonin system is targeted by first-line pharmacological treatments for several psychiatric disorders, including obsessive-compulsive disorder. However, understanding the behavioral function of serotonin is hampered by methodological limitations: the (brainstem) location of serotonergic neuron cell-bodies is difficult to access, their innervation of the brain is diffuse, and they release serotonin in relatively low concentrations. Here, we advance this effort by developing novel Tph2-Cre rats, which we utilized to study serotonin in the context of motor, compulsive, and reinforced behaviors using optogenetics in both male and female rats. Specificity and sensitivity of Cre recombinase expression and Cre-dependent processes were validated immunohistochemically, and optogenetic induction of in vivo serotonin release was validated with fast-scan cyclic voltammetry. Optogenetic stimulation of serotonin neurons in the dorsal raphe nucleus did not initiate locomotion or alter aversion-induced locomotion, nor did it elicit (real-time) place preference, and it had no measurable effect on compulsive behavior in the schedule-induced polydipsia task. In contrast, this optogenetic stimulation moderately sustained ongoing spontaneous locomotion and robustly reinforced operant lever pressing for self-stimulation of serotonin neurons, which was exacerbated by food restriction. Together, this work both introduces a novel rat Cre line to study serotonin and advances our understanding of serotonin's behavioral functions. Complementing previous findings, we find that brainwide serotonin release has an overall relatively mild effect on behavior, which manifested only in the absence of natural reinforcers and was modulated by physiological state.
Collapse
Affiliation(s)
- Rhiannon Robke
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Francesca Sansi
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
| | - Tara Arbab
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Adria Tunez
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| | - Miranda Moore
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
| | - Dusan Bartsch
- Department of Molecular Biology, Central Institute of Mental Health, Mannheim 68159, Germany
| | - Kai Schönig
- Department of Molecular Biology, Central Institute of Mental Health, Mannheim 68159, Germany
| | - Ingo Willuhn
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105BA, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam 1105AZ, The Netherlands
| |
Collapse
|
4
|
Xu L, Cao Y, Zhang S, Du L, Wang W, Liu J, Wang D, Zhao D, Cui M, Jiang S, Qin G, Meng F, Zhang M, Li C. Sirtuin 1 underlies depression-related behaviors by modulating the serotonin system in the dorsal raphe nucleus in female mice. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111400. [PMID: 40374143 DOI: 10.1016/j.pnpbp.2025.111400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/08/2025] [Accepted: 05/10/2025] [Indexed: 05/17/2025]
Abstract
Major depressive disorder (MDD) is a primary driver of disability and greatly escalates the worldwide disease burden. Sirtuin 1 (Sirt1), a key regulator of cellular metabolism, is associated with genetic variations in MDD. We investigated how Sirt1 in serotonin (5-HT) neurons within the dorsal raphe nucleus (DRN) in mice affected behaviors associated with depression and susceptibility to stress. Our findings revealed that Sirt1 expression in the DRN was decreased when chronic unpredictable stress was induced in depressed female mice. Additionally, Sirt1 was co-localized with 5-HT neurons within the DRN, and its selective ablation in these neurons have induced depressive phenotypes in female mice but not in males. Adeno-associated virus-mediated knockdown of Sirt1 in adult female mice induced depressive behaviors, whereas Sirt1 overexpression eliminated these behaviors. Moreover, fiber-optic recordings showed a decrease in the neural excitability of 5-HT neurons and 5-HT levels in the DRN after Sirt1 knockdown. Furthermore, we observed that Sirt1 knockdown reduced the expression of tryptophan hydroxylase-2 (Tph2) and phosphorylation levels of extracellular signal-regulated kinase (ERK) and CAMP response element binding protein (CREB). Finally, variable molecular targets regarding immune responses and cytokine productions after Sirt1 knockdown were analyzed via high-throughput RNA-seq analysis of specimens from the DRN. The findings of this study emphasize the importance of Sirt1 for regulating depression-related behaviors in female mice by influencing the activity of 5-HT neurons in the DRN.
Collapse
Affiliation(s)
- Lihong Xu
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Yifan Cao
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Shasha Zhang
- Health Examination Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Lin Du
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Wentao Wang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Jing Liu
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Dan Wang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Di Zhao
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Shandong 264003, China
| | - Gaofeng Qin
- Department of Traditional Chinese Medicine, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China
| | - Fantao Meng
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| | - Mengdi Zhang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| | - Chen Li
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong 256603, China.
| |
Collapse
|
5
|
Ye X, Pang S, Ren X, Wang H, Chen M. Neurotransmitter modulation of sleep-wake States: From molecular mechanisms to therapeutic potential. Sleep Med 2025; 132:106547. [PMID: 40359849 DOI: 10.1016/j.sleep.2025.106547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/14/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
Sleep is one of the most fundamental physiological activities in humans and animals, and a normal sleep cycle is crucial for maintaining overall health. However, sleep disorders are increasingly becoming a major mental health issue affecting individuals and society, as well as a contributing factor to the onset of other diseases. Consequently, the development of novel therapeutic strategies for sleep disorders has emerged as a significant scientific challenge garnering widespread attention. Based on current research findings, focusing on neurotransmitters remains a promising approach for developing effective treatments. Neurotransmitters play a central role in regulating the sleep-wake cycle by precisely modulating the activity states of different brain regions. This review aims to elucidate the neural mechanisms underlying sleep initiation and function, thereby providing a comprehensive understanding of the complex nature of sleep as a physiological process. Furthermore, it seeks to uncover the potential pathological mechanisms of sleep disorders, offering a theoretical foundation and novel insights for precision medicine and drug development, ultimately reducing the negative impact of sleep disorders on individuals and society.
Collapse
Affiliation(s)
- Xinyi Ye
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Shuchao Pang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China.
| | - Xiaoliang Ren
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Hui Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Meiling Chen
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
6
|
McElroy BD, Li C, McCloskey NS, Alberici AR, Kirby LG. Exploring the effects of adolescent social isolation stress on the serotonin system and ethanol-motivated behaviors. Psychopharmacology (Berl) 2025; 242:763-781. [PMID: 39903245 PMCID: PMC11890253 DOI: 10.1007/s00213-025-06749-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
RATIONALE Alcohol is one of the most frequently used drugs of abuse and has a major impact on human health worldwide. People assigned female at birth and those with adverse childhood experiences are stress-vulnerable and more likely to report drinking as a means of "self-medication." Prior studies in our laboratory showed that adolescent social isolation stress (SIS) increases vulnerability to ethanol (EtOH) intake and consumption despite negative consequences in female rats. OBJECTIVES Here, we explored modulation of the dorsal raphe nucleus (DRN)-serotonin (5-HT) system, a sexually dimorphic neurotransmitter system involved in stress-reward interactions, to determine its contribution to EtOH-motivated behaviors in rats that have undergone SIS. RESULTS We employed electrophysiological and functional neuroanatomy strategies to show that both SIS and EtOH exposure induce persistent hypofunction of the DRN 5-HT system, particularly in females. Chemogenetic activation of DRN 5-HT neurons attenuated reward value for both EtOH and sucrose and elevated punished responding for EtOH in a stress-dependent manner. CONCLUSIONS Our results highlight an inverse relationship between EtOH consumption and the 5-HT system, the sex- and stress-dependent nature of this relationship, and a connection between DRN 5-HT signaling and acute responding to rewards and punishment. These data support the DRN 5-HT system as a potential target to treat aberrant alcohol consumption and drinking despite negative consequences in stress-vulnerable populations.
Collapse
Affiliation(s)
- Bryan D McElroy
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St, MERB Room 857, Philadelphia, PA, 19140, USA
| | - Chen Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St, MERB Room 857, Philadelphia, PA, 19140, USA
| | - Nicholas S McCloskey
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St, MERB Room 857, Philadelphia, PA, 19140, USA
| | - Amber R Alberici
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St, MERB Room 857, Philadelphia, PA, 19140, USA
| | - Lynn G Kirby
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, 3500 N. Broad St, MERB Room 857, Philadelphia, PA, 19140, USA.
| |
Collapse
|
7
|
Singer HS, Pellicciotti J. The Role of CBGTC Synaptic Neurotransmission in the Pathophysiology of Tics. Psychiatr Clin North Am 2025; 48:203-216. [PMID: 39880513 DOI: 10.1016/j.psc.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
The pathophysiology of tic disorders involves an alteration in the transmission of messages through the cortico-basal ganglia-thalamo-cortical circuit. A major requirement for the passage of a message through this circuit is an intact chemically mediated synaptic neurotransmitter system (ie, neurotransmitters and second messengers). This article reviews the scientific evidence supporting the involvement of a variety of neurotransmitters (ie, dopamine, glutamate, gamma-aminobutyric acid, serotonin, acetylcholine, and the opioid system). Although there are favored neurotransmitter abnormalities, their complex interactions suggest the likelihood that several are involved in the production of tics.
Collapse
Affiliation(s)
- Harvey S Singer
- Department of Neurology, Johns Hopkins University School of Medicine, Kennedy Krieger Institute, Baltimore, MD, USA.
| | - Justin Pellicciotti
- Department of Neurology, Johns Hopkins University School of Medicine, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
8
|
Yang S, Xue J, Li Z, Zhang S, Zhang Z, Huang Z, Yung KKL, Lai KWC. Deep Learning-Based Ion Channel Kinetics Analysis for Automated Patch Clamp Recording. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404166. [PMID: 39737527 PMCID: PMC12083860 DOI: 10.1002/advs.202404166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/07/2024] [Indexed: 01/01/2025]
Abstract
The patch clamp technique is a fundamental tool for investigating ion channel dynamics and electrophysiological properties. This study proposes the first artificial intelligence framework for characterizing multiple ion channel kinetics of whole-cell recordings. The framework integrates machine learning for anomaly detection and deep learning for multi-class classification. The anomaly detection excludes recordings that are incompatible with ion channel behavior. The multi-class classification combined a 1D convolutional neural network, bidirectional long short-term memory, and an attention mechanism to capture the spatiotemporal patterns of the recordings. The framework achieves an accuracy of 97.58% in classifying 124 test datasets into six categories based on ion channel kinetics. The utility of the novel framework is demonstrated in two applications: Alzheimer's disease drug screening and nanomatrix-induced neuronal differentiation. In drug screening, the framework illustrates the inhibitory effects of memantine on endogenous channels, and antagonistic interactions among potassium, magnesium, and calcium ion channels. For nanomatrix-induced differentiation, the classifier indicates the effects of differentiation conditions on sodium and potassium channels associated with action potentials, validating the functional properties of differentiated neurons for Parkinson's disease treatment. The proposed framework is promising for enhancing the efficiency and accuracy of ion channel kinetics analysis in electrophysiological research.
Collapse
Affiliation(s)
- Shengjie Yang
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Jiaqi Xue
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Ziqi Li
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| | - Shiqing Zhang
- JNU‐HKUST Joint Laboratory for Neuroscience and Innovative Drug ResearchCollege of PharmacyJinan University601 West Huangpu Road, TianheGuangzhou510632China
| | - Zhang Zhang
- School of Public HealthGuangzhou Medical UniversityXinzao, PanyuGuangzhou511436China
| | - Zhifeng Huang
- Department of ChemistryChinese University of Hong KongShatinNew TerritoriesHong Kong SARChina
| | - Ken Kin Lam Yung
- Department of Science and Environmental StudiesEducation University of Hong Kong10 Lo Ping RoadTai PoNew TerritoriesHong Kong SARChina
| | - King Wai Chiu Lai
- Department of Biomedical EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SARChina
| |
Collapse
|
9
|
Zhang Y, Zhang P, Shin M, Chang Y, Abbott SBG, Venton BJ, Zhu JJ. Coding principles and mechanisms of serotonergic transmission modes. Mol Psychiatry 2025:10.1038/s41380-025-02930-4. [PMID: 39987232 DOI: 10.1038/s41380-025-02930-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 01/13/2025] [Accepted: 02/10/2025] [Indexed: 02/24/2025]
Abstract
Serotonin-mediated intercellular communication has been implicated in myriad human behaviors and diseases, yet how serotonin communicates and how the communication is regulated remain unclear due to limitations of available monitoring tools. Here, we report a method multiplexing genetically encoded sensor-based imaging and fast-scan cyclic voltammetry, enabling simultaneous recordings of synaptic, perisynaptic, proximate and distal extrasynaptic serotonergic transmission. Employing this method alongside a genetically encoded sensor-based image analysis program (GESIAP), we discovered that heterogeneous firing patterns of serotonergic neurons create various transmission modes in the mouse raphe nucleus and amygdala, encoding information of firing pulse frequency, number, and synchrony using neurotransmitter quantity, releasing synapse count, and synaptic and/or volume transmission. During tonic and low-frequency phasic activities, serotonin is confined within synaptic clefts due to efficient retrieval by perisynaptic transporters, mediating synaptic transmission modes. Conversely, during high-frequency, especially synchronized phasic activities, or when transporter inhibition, serotonin may surpass transporter capacity, and escape synaptic clefts through 1‒3 outlet channels, leading to volume transmission modes. Our results elucidate a mechanism of how channeled synaptic enclosures, synaptic properties, and transporters collaborate to define the coding principles of activity pattern-dependent serotonergic transmission modes.
Collapse
Affiliation(s)
- Yajun Zhang
- Departments of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Peng Zhang
- Departments of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Mimi Shin
- Departments of Chemistry, University of Virginia, Charlottesville, VA, 22904, USA
| | - Yuanyu Chang
- Departments of Chemistry, University of Virginia, Charlottesville, VA, 22904, USA
| | - Stephen B G Abbott
- Departments of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - B Jill Venton
- Departments of Chemistry, University of Virginia, Charlottesville, VA, 22904, USA
| | - J Julius Zhu
- Departments of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA.
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
10
|
Zhao R, Wang J, Chung SK, Xu B. New insights into anti-depression effects of bioactive phytochemicals. Pharmacol Res 2025; 212:107566. [PMID: 39746497 DOI: 10.1016/j.phrs.2024.107566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Depression is one of the most common psychological disorders, and due to its high prevalence and mortality rates, it imposes a significant disease burden. Contemporary treatments for depression involve various synthetic drugs, which have limitations such as side effects, single targets, and slow onset of action. Unlike synthetic medications, phytochemicals offer the benefits of a multi-target and multi-pathway mode of treatment for depression. In this literature review, we describe the pharmacological actions, experimental models, and clinical trials of the antidepressant effects of various phytochemicals. Additionally, we summarize the potential mechanisms by which these phytochemicals prevent depression, including regulating neurotransmitters and their receptors, the HPA axis, inflammatory responses, managing oxidative stress, neuroplasticity, and the gut microbiome. Phytochemicals exert therapeutic effects through multiple pathways and targets, making traditional Chinese medicine (TCM) a promising adjunctive antidepressant for the prevention, alleviation, and treatment of depression. Therefore, this review aims to provide robust evidence for subsequent research into developing phytochemical resources as effective antidepressant agents.
Collapse
Affiliation(s)
- Ruohan Zhao
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China
| | - Jingwen Wang
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China
| | - Sookja Kim Chung
- Faculty of Medicine, Macau University of Science and Technology, Macau, China.
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
11
|
Li C, McCloskey NS, Inan S, Kirby LG. Role of serotonin neurons in the dorsal raphe nucleus in heroin self-administration and punishment. Neuropsychopharmacology 2025; 50:596-604. [PMID: 39300273 PMCID: PMC11735851 DOI: 10.1038/s41386-024-01993-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
One hallmark of substance use disorder is continued drug use despite negative consequences. When drug-taking behavior is punished with aversive stimuli, i.e. footshock, rats can also be categorized into punishment-resistant or compulsive vs. punishment-sensitive or non-compulsive phenotypes. The serotonin (5-hydroxytryptamine, 5-HT) system modulates responses to both reward and punishment. The goal of the current study was to examine punishment phenotypes in heroin self-administration and to determine the role of dorsal raphe nucleus (DRN) 5-HT neurons in both basal and punished heroin self-administration. First, rats were exposed to punished heroin self-administration and neuronal excitability of DRN 5-HT neurons was compared between punishment-resistant and punishment-sensitive phenotypes using ex vivo electrophysiology. Second, DRN 5-HT neuronal activity was manipulated in vivo during basal and punished heroin self-administration using chemogenetic tools in a Tph2-iCre rat line. While rats separated into punishment-resistant and punishment-sensitive phenotypes for punished heroin self-administration, DRN 5-HT neuronal excitability did not differ between the phenotypes. While chemogenetic inhibition of DRN 5-HT neurons was without effect, chemogenetic activation of DRN 5-HT neurons increased both basal and punished heroin self-administration selectively in punishment-resistant animals. Additionally, the responsiveness to chemogenetic activation of DRN 5-HT neurons in basal self-administration and motivation for heroin in progressive ratio each predicted resistance to punishment. Therefore, our data support the role for the DRN 5-HT system in compulsive heroin self-administration.
Collapse
Affiliation(s)
- Chen Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Nicholas S McCloskey
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Lynn G Kirby
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA.
| |
Collapse
|
12
|
Zhu L, Yao D. Research advances in children's sleep and vitamin D levels. Ann Pediatr Endocrinol Metab 2025; 30:3-10. [PMID: 40049669 PMCID: PMC11917401 DOI: 10.6065/apem.2448076.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/05/2024] [Indexed: 03/20/2025] Open
Abstract
In recent years, studies have revealed that vitamin D, a steroid hormone essential for calcium and phosphorus metabolism, also plays a role in sleep. Adequate levels of vitamin D have been linked to improved sleep quality in children and effective prevention of sleep problems. This report is a review and summary of research on the significance of sleep, the stages of children's sleep, and the impact of vitamin D levels on sleep problems. Additionally, this report explores the mechanisms through which vitamin D improves sleep.
Collapse
Affiliation(s)
- Liuyan Zhu
- Department of Pediatric Health Care, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| | - Dan Yao
- Department of Pediatric Health Care, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
13
|
Koda M, Kawai H, Shirakawa H, Kaneko S, Nagayasu K. Effect of antidepressants and social defeat stress on the activity of dorsal raphe serotonin neurons in free-moving animals. J Pharmacol Sci 2025; 157:113-123. [PMID: 39828391 DOI: 10.1016/j.jphs.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/28/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025] Open
Abstract
Major depressive disorder (MDD) is among the most common mental disorders worldwide and is characterized by dysregulated reward processing associated with anhedonia. Selective serotonin reuptake inhibitors (SSRIs) are the first-line treatment for MDD; however, their onset of action is delayed. Recent reports have shown that serotonin neurons in the dorsal raphe nucleus (DRN) are activated by rewards and play a vital role in reward processing. However, whether antidepressant treatment affects the DRN serotonin neuronal response to rewards in awake animals remains unknown. In this study, we measured the activity of DRN serotonin neurons in awake mice and determined the effects of antidepressants and chronic stress on DRN serotonin neuronal activity. We found that acute treatment with citalopram, an SSRI, significantly decreased sucrose-induced activation of DRN serotonin neurons. The decrease in response to acute citalopram treatment was attenuated by chronic citalopram treatment. Acute treatment with (S)-WAY100135, a 5-HT1A receptor antagonist, dose-dependently inhibited the response to acute citalopram treatment. These results indicate that autoinhibition by activating 5-HT1A receptors via acute SSRI treatment may blunt the reward response, which can be recovered after chronic SSRI treatment.
Collapse
Affiliation(s)
- Masashi Koda
- 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; Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-cho, Abeno-ku, Osaka, 545-8585, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shuji Kaneko
- 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; Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Japan; Project for Neural Networks, Drug Innovation Center, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, 565-0871, Japan.
| |
Collapse
|
14
|
Yu W, Zhang R, Zhang A, Mei Y. Deciphering the Functions of Raphe-Hippocampal Serotonergic and Glutamatergic Circuits and Their Deficits in Alzheimer's Disease. Int J Mol Sci 2025; 26:1234. [PMID: 39941002 PMCID: PMC11818420 DOI: 10.3390/ijms26031234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Subcortical innervation of the hippocampus by the raphe nucleus is essential for emotional and cognitive control. The two major afferents from raphe to hippocampus originate from serotonergic and glutamatergic neurons, of which the serotonergic control of hippocampal inhibitory network, theta activity, and synaptic plasticity have been extensively explored in the growing body of literature, whereas those of glutamatergic circuits have received little attention. Notably, both serotonergic and glutamatergic circuits between raphe and hippocampus are disrupted in Alzheimer's disease (AD), which may contribute to initiation and progression of behavioral and psychological symptoms of dementia. Thus, deciphering the mechanism underlying abnormal raphe-hippocampal circuits in AD is crucial to prevent dementia-associated emotional and cognitive symptoms. In this review, we summarize the anatomical, neurochemical, and electrophysiological diversity of raphe nuclei as well as the architecture of raphe-hippocampal circuitry. We then elucidate subcortical control of hippocampal activity by raphe nuclei and their role in regulation of emotion and cognition. Additionally, we present an overview of disrupted raphe-hippocampal circuits in AD pathogenesis and analyze the available therapies that can potentially be used clinically to alleviate the neuropsychiatric symptoms and cognitive decline in AD course.
Collapse
Affiliation(s)
| | | | | | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer’s Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
15
|
Lan T, Li Y, Chen X, Wang W, Wang C, Lou H, Chen S, Yu S. Exercise-Activated mPFC Tri-Synaptic Pathway Ameliorates Depression-Like Behaviors in Mouse. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408618. [PMID: 39574315 PMCID: PMC11744721 DOI: 10.1002/advs.202408618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/03/2024] [Indexed: 01/21/2025]
Abstract
Exercise is considered as playing a pivotal role in the modulation of emotional responses. However, a precise circuit that mediates the effects of exercise on depression have yet to be elucidated. Here, a molecularly defined tri-synaptic pathway circuit is identified that correlates motor inputs with antidepressant effects. With this pathway, initial inputs from neurons within the dorsal root ganglia (DRG) project to excitatory neurons in the gracile nucleus (GR), which in turn connect with 5-HTergic neurons in the dorsal raphe nucleus (DRN), eventually coursing to excitatory pyramidal neurons within the medial prefrontal cortex (mPFC). Exercise activates this pathway, with the result that depressive- and anxiety-like behaviors in mice are significantly reduced. In addition, it is found that exercise may exert antidepressant effects through regulating synaptic plasticity within this tri-synaptic pathway. These findings reveal a hindbrain-to-forebrain neuronal circuit that specifically modulates depression and provides a potential mechanism for the antidepressant effects of exercise.
Collapse
Affiliation(s)
- Tian Lan
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Ye Li
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Xiao Chen
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Wenjing Wang
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Changmin Wang
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Haiyan Lou
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PharmacologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Shihong Chen
- Department of Endocrinology and MetabolismThe Second Hospital of Shandong UniversityJinanShandong250033China
| | - Shuyan Yu
- Shandong Key Laboratory of Mental Disorders and Intelligent ControlThe Second Hospital of Shandong UniversitySchool of Basic Medical SciencesShandong UniversityJinanShandong250012China
- Department of PhysiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
- Department of Medical Psychology and EthicsSchool of Basic Medical sciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| |
Collapse
|
16
|
Ontiveros-Araiza LF. The Neurobehavioral State hypothesis. Biosystems 2025; 247:105361. [PMID: 39521269 DOI: 10.1016/j.biosystems.2024.105361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/02/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Since the early attempts to understand the brain made by Greek philosophers more than 2000 years ago, one of the main questions in neuroscience has been how the brain perceives all the stimuli in the environment and uses this information to implement a response. Recent hypotheses of the neural code rely on the existence of an ideal observer, whether on specific areas of the cerebral cortex or distributed network composed of cortical and subcortical elements. The Neurobehavioral State hypothesis stipulates that neurons are in a quasi-stable state due to the dynamic interaction of their molecular components. This increases their computational capabilities and electrophysiological behavior further than a binary active/inactive state. Together, neuronal populations across the brain learn to identify and associate internal and external stimuli with actions and emotions. Furthermore, such associations can be stored through the regulation of neuronal components as new quasi-stable states. Using this framework, behavior arises as the result of the dynamic interaction between internal and external stimuli together with previously established quasi-stable states that delineate the behavioral response. Finally, the Neurobehavioral State hypothesis is firmly grounded on present evidence of the complex dynamics within the brain, from the molecular to the network level, and avoids the need for a central observer by proposing the brain configures itself through experience-driven associations.
Collapse
Affiliation(s)
- Luis Fernando Ontiveros-Araiza
- Department of Cognitive Neuroscience, Division of Neuroscience, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
17
|
Kawai H. [Role of Median Raphe Serotonergic Neurons in Positive and Negative Information Processing]. YAKUGAKU ZASSHI 2025; 145:79-84. [PMID: 39894484 DOI: 10.1248/yakushi.24-00150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Serotonergic neurons play a critical role in processing reward and aversive information. Rewarding stimuli activate serotonergic neurons in the dorsal raphe nucleus (DRN), whereas optogenetic activation of DRN serotonergic neurons induces reward-like effects. However, the pharmacological enhancement of serotonin neurotransmission does not induce rewarding or aversive effects. These findings suggest the presence of another serotonergic neuron that plays a role opposite to that of the DRN in processing reward and aversion information. Previous reports suggested that the median raphe nucleus (MRN) processes negative emotional stimuli. To elucidate the function of MRN serotonergic neurons in these processes, we recorded the changes in serotonergic activity in mice in response to rewarding and aversive stimuli. We also used optogenetic manipulation to determine whether these changes could induce rewarding and aversive behaviors. The activity of MRN serotonergic neurons decreased in response to rewarding stimuli and increased after aversive stimuli. Optogenetic inhibition of MRN serotonergic neurons induced reward-related behavior, while optogenetic stimulation induced aversion-related behavior. Furthermore, we found that the projection pathway from MRN serotonergic neurons to the interpeduncular nucleus is crucial for these processes. These results indicate that MRN serotonergic neurons play a pivotal role in processing reward and aversive information, functioning oppositely to DRN neurons.
Collapse
Affiliation(s)
- Hiroyuki Kawai
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka Metropolitan University
| |
Collapse
|
18
|
Niitani K, Nishida R, Futami Y, Nishitani N, Deyama S, Kaneda K. Activation of ventral pallidum-projecting neurons in the nucleus accumbens via 5-HT 2C receptor stimulation regulates motivation for wheel running in male mice. Neuropharmacology 2024; 261:110181. [PMID: 39393590 DOI: 10.1016/j.neuropharm.2024.110181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Rodents have a strong motivation for wheel running; however, the neural mechanisms that regulate their motivation remain unknown. We investigated the possible involvement of serotonin (5-HT) systems in regulating motivation for wheel running in male mice. Systemic administration of a 5-HT1A receptor antagonist (WAY100635) increased the number of wheel rotations, whereas administration of a 5-HT2A or 5-HT2C receptor antagonist (volinanserin or SB242084, respectively) decreased it. In the open field test, neither WAY100635 nor volinanserin affected locomotor activity, whereas SB242084 increased locomotor activity. To identify the brain regions on which these antagonists act, we locally injected these into the motivational circuitry, including the nucleus accumbens (NAc), dorsomedial striatum (DM-Str), and medial prefrontal cortex (mPFC). Injection of SB242084 into the NAc, but not the DM-Str or mPFC, reduced the number of wheel rotations without altering locomotor activity. The local administration of WAY100635 or volinanserin to these brain regions did not affect the number of wheel rotations. Immunohistochemical analyses revealed that wheel running increased the number of c-Fos-positive cells in the NAc medial shell (NAc-MS), which was reduced by systemic SB242084 administration. In vitro slice whole-cell recordings showed that bath application of the 5-HT2C receptor agonist lorcaserin increased the frequency of spontaneous excitatory and inhibitory postsynaptic currents in the ventral tegmental area (VTA)-projecting neurons, whereas it only increased the frequency of spontaneous excitatory postsynaptic currents in ventral pallidum (VP)-projecting neurons in the NAc-MS. These findings suggest that the activation of VP-projecting NAc-MS neurons via 5-HT2C receptor stimulation regulates motivation for wheel running.
Collapse
Affiliation(s)
- Kazuhei Niitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ryoma Nishida
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yusaku Futami
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
19
|
Hutchens SED, Khurram I, Hurley LM. Solitude and serotonin: juvenile isolation alters the covariation between social behavior and cFos expression by serotonergic neurons. Front Neurosci 2024; 18:1446866. [PMID: 39502712 PMCID: PMC11535725 DOI: 10.3389/fnins.2024.1446866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/19/2024] [Indexed: 11/08/2024] Open
Abstract
Variation in the mutual responsiveness of social partners to each other can be reflected in behavioral suites that covary with neural activity in ways that track the salience or valence of interactions. Juvenile social isolation alters social behavior and neural activity during social interaction, but whether and how it alters the covariation between behavior and neural activity has not been as well explored. To address this issue, four classes of experimental subjects: isolated males, socially housed males, isolated females, and socially housed females, were paired with an opposite-sex social partner that had been socially housed. Social behaviors and c-Fos expression in the serotonergic dorsal raphe nucleus (DRN) were then measured in subjects following the social interactions. Relative to social housing, postweaning isolation led to a decrease in the density of neurons double-labeled for tryptophan hydroxylase and c-Fos in the dorsomedial subdivision of the DRN, regardless of sex. Vocal and non-vocal behaviors were also affected by isolation. In interactions with isolated males, both ultrasonic vocalization (USVs) and broadband vocalizations (squeaks) increased in conjunction with greater male investigation of females. Neural and behavioral measures also correlated with each other. In the isolated male group, the density of double-labeled neurons in the dorsomedial DRN was negatively correlated with USV production and positively correlated with a principal component of non-vocal behavior corresponding to greater defensive kicking by females and less investigation and mounting behavior. This correlation was reversed in direction for socially housed males, and for isolated males versus isolated females. These findings confirm that the dynamics of social interactions are reflected in c-Fos activation in the dorsomedial DRN, and suggest an altered responsiveness of serotonergic neurons to social interaction following social isolation in males, in parallel with an altered male response to female cues.
Collapse
Affiliation(s)
- Sarah E. D. Hutchens
- Hurley Laboratory, Department of Biology, Indiana University, Bloomington, IN, United States
| | - Izza Khurram
- Hurley Laboratory, Department of Biology, Indiana University, Bloomington, IN, United States
| | - Laura M. Hurley
- Hurley Laboratory, Department of Biology, Indiana University, Bloomington, IN, United States
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, United States
| |
Collapse
|
20
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
21
|
Zhang S, Xie X, Xu Y, Mi J, Li Z, Guo Z, Xu G. Effects of transcranial magneto-acoustic stimulation on cognitive function and neural signal transmission in the hippocampal CA1 region of mice. Neuroscience 2024; 556:86-95. [PMID: 39047971 DOI: 10.1016/j.neuroscience.2024.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/16/2023] [Accepted: 01/29/2024] [Indexed: 07/27/2024]
Abstract
As a new means of brain neuroregulation and research, transcranial magneto-acoustic stimulation (TMAS) uses the coupling effect of ultrasound and a static magnetic field to regulate neural activity in the corresponding brain areas. Calcium ions can promote the secretion of neurotransmitters and play a key role in the transmission of neural signals in brain cognition. In this study, to explore the effects of TMAS on cognitive function and neural signaling in the CA1 region of the hippocampus, TMAS was applied to male 2-month-old C57 mice with a magnetic field strength of 0.3 T and ultrasound intensity of 2.6 W/cm2. First, the efficiency of neural signaling in the CA1 region of the mouse hippocampus was detected by fiber photometry. Second, the effects of TMAS on cognitive function in mice were investigated through multiple behavioral experiments, including spatial learning and memory ability, anxiety and desire for novelty. The experimental results showed that TMAS could improve cognitive function in mice, and the efficiency of neural signaling in the CA1 area of the hippocampus was significantly increased during stimulation and maintained for one week after stimulation. In addition, the neural signaling efficiency in the CA1 area of the hippocampus increased in the open field (OF) experiment and recovered after one week, the neural signaling efficiency in the new object exploration (NOE) experiment was significantly enhanced, and the intensity slowed after one week. In conclusion, TMAS enhances cognitive performance and promotes neural signaling in the CA1 region of the mouse hippocampus.
Collapse
Affiliation(s)
- Shuai Zhang
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China.
| | - Xiaofeng Xie
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Yihao Xu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Jinrui Mi
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Zichun Li
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Zhongsheng Guo
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| | - Guizhi Xu
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, Hebei University of Technology, Tianjin 300130, China; Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin 300130, China; Hebei Key Laboratory of Electromagnetic Field and Electrical Reliability, Hebei University of Technology, Tianjin 300130, China
| |
Collapse
|
22
|
Taira M, Miyazaki KW, Miyazaki K, Chen J, Okitsu-Sakurayama S, Chaudhary A, Nishio M, Miyake T, Yamanaka A, Tanaka KF, Doya K. The differential effect of optogenetic serotonergic manipulation on sustained motor actions and waiting for future rewards in mice. Front Neurosci 2024; 18:1433061. [PMID: 39385850 PMCID: PMC11461476 DOI: 10.3389/fnins.2024.1433061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Serotonin is an essential neuromodulator that affects behavioral and cognitive functions. Previous studies have shown that activation of serotonergic neurons in the dorsal raphe nucleus (DRN) promotes patience to wait for future rewards. However, it is still unclear whether serotonergic neurons also regulate persistence to act for future rewards. Here we used optogenetic activation and inhibition of DRN serotonergic neurons to examine their effects on sustained motor actions for future rewards. We trained mice to perform waiting and repeated lever-pressing tasks with variable reward delays and tested effects of optogenetic activation and inhibition of DRN serotonergic neurons on task performance. Interestingly, in the lever-pressing task, mice tolerated longer delays as they repeatedly pressed a lever than in the waiting task, suggesting that lever-pressing actions may not simply be costly, but may also be subjectively rewarding. Optogenetic activation of DRN serotonergic neurons prolonged waiting duration in the waiting task, consistent with previous studies. However, its effect on lever presses was nuanced, and was detected only by focusing on the period before premature reward check and by subtracting the trends within and across sessions using generalized linear model. While optogenetic inhibition decreased waiting, it did not affect lever pressing time or numbers. These results revealed that the necessity of motor actions may increase motivation for delayed rewards and that DRN serotonergic neurons more significantly promote waiting rather than persistent motor actions for future rewards.
Collapse
Affiliation(s)
- Masakazu Taira
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- Department of Psychology, University of Sydney, Camperdown, NSW, Australia
| | - Kayoko W. Miyazaki
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Katsuhiko Miyazaki
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Jianning Chen
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Shiho Okitsu-Sakurayama
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Anupama Chaudhary
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Mika Nishio
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- School of Medicine, Tohoku University, Sendai, Japan
| | - Tsukasa Miyake
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
- School of Medicine, University of the Ryukyus, Okinawa, Japan
| | | | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Doya
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
23
|
Zhang Y, Shen J, Xie F, Liu Z, Yin F, Cheng M, Wang L, Cai M, Herzog H, Wu P, Zhang Z, Zhan C, Liu T. Feedforward inhibition of stress by brainstem neuropeptide Y neurons. Nat Commun 2024; 15:7603. [PMID: 39217143 PMCID: PMC11365948 DOI: 10.1038/s41467-024-51956-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Resistance to stress is a key determinant for mammalian functioning. While many studies have revealed neural circuits and substrates responsible for initiating and mediating stress responses, little is known about how the brain resists to stress and prevents overreactions. Here, we identified a previously uncharacterized neuropeptide Y (NPY) neuronal population in the dorsal raphe nucleus and ventrolateral periaqueductal gray region (DRN/vlPAG) with anxiolytic effects in male mice. NPYDRN/vlPAG neurons are rapidly activated by various stressful stimuli. Inhibiting these neurons exacerbated hypophagic and anxiety responses during stress, while activation significantly ameliorates acute stress-induced hypophagia and anxiety levels and transmits positive valence. Furthermore, NPYDRN/vlPAG neurons exert differential but synergic anxiolytic effects via inhibitory projections to the paraventricular thalamic nucleus (PVT) and the lateral hypothalamic area (LH). Together, our findings reveal a feedforward inhibition neural mechanism underlying stress resistance and suggest NPYDRN/vlPAG neurons as a potential therapeutic target for stress-related disorders.
Collapse
Grants
- the National Key R&D Program of China (2019YFA0801900, 2018YFA0800300), the National Natural Science Foundation of China (9235730017, 92249302, 32150610475, 31971074), Innovation Team and Talents Cultivation Program of National Administration of Traditional Chinese Medicine (ZYYCXTD-D-202001), Faculty Resources Project of College of Life Sciences, Inner Mongolia University (2022-102)
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, the National Natural Science Foundation of China (32171144) and Shanghai Pujiang Program (22PJD007).
- the STI2030-Major Projects (2021ZD0203900),the National Natural Science Foundation of China (32271063, 31822026, 31500860), Research Funds of Center for Advanced Interdisciplinary Science and Biomedicine of IHM (QYPY20220018)
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- Hefei National Research center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Jiayi Shen
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Famin Xie
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Zhiwei Liu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fangfang Yin
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Mingxiu Cheng
- National Institute of Biological Sciences, Beijing, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Liang Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Meiting Cai
- Hefei National Research center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Herbert Herzog
- St Vincent's Centre for Applied Medical Research, Faculty of Medicine, UNSW, Sydney, NSW, Australia
- Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Ping Wu
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Zhi Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| | - Cheng Zhan
- Hefei National Research center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
- Human Phenome Institute, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, China.
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Shanghai, China.
- School of Life Sciences, Inner Mongolia University, Hohhot, China.
| |
Collapse
|
24
|
Jing S, Geng C, Liu P, Wang D, Li Q, Li A. Serotonergic input from the dorsal raphe nucleus shapes learning-associated odor responses in the olfactory bulb. Acta Physiol (Oxf) 2024; 240:e14198. [PMID: 38958443 DOI: 10.1111/apha.14198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/29/2024] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
AIM Neural activity in the olfactory bulb (OB) can represent odor information during different brain and behavioral states. For example, the odor responses of mitral/tufted (M/T) cells in the OB change during learning of odor-discrimination tasks and, at the network level, beta power increases and the high gamma (HG) power decreases during odor presentation in such tasks. However, the neural mechanisms underlying these observations remain poorly understood. Here, we investigate whether serotonergic modulation from the dorsal raphe nucleus (DRN) to the OB is involved in shaping activity during the learning process in a go/no-go task in mice. METHODS Fiber photometry was used to record the population activity of DRN serotonergic neurons during a go/no-go task. In vivo electrophysiology was used to record neural activity (single units and local field potentials) in the OB during the go/no-go task. Real-time place preference (RTPP) and intracranial light administration in a specific subarea (iClass) tests were used to assess the ability of mice to encoding reward information. RESULTS Odor-evoked population activity in serotonergic neurons in the DRN was shaped during the learning process in a go/no-go task. In the OB, neural activity from oscillations to single cells showed complex, learning-associated changes and ability to encode information during an odor discrimination task. However, these properties were not observed after ablation of DRN serotonergic neurons. CONCLUSION The activity of neural networks and single cells in the OB, and their ability to encode information about odor value, are shaped by serotonergic projections from the DRN.
Collapse
Affiliation(s)
- Siqi Jing
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Chi Geng
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Penglai Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Dejuan Wang
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Qun Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| | - Anan Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
25
|
Bremshey S, Groß J, Renken K, Masseck OA. The role of serotonin in depression-A historical roundup and future directions. J Neurochem 2024; 168:1751-1779. [PMID: 38477031 DOI: 10.1111/jnc.16097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Depression is one of the most common psychiatric disorders worldwide, affecting approximately 280 million people, with probably much higher unrecorded cases. Depression is associated with symptoms such as anhedonia, feelings of hopelessness, sleep disturbances, and even suicidal thoughts. Tragically, more than 700 000 people commit suicide each year. Although depression has been studied for many decades, the exact mechanisms that lead to depression are still unknown, and available treatments only help a fraction of patients. In the late 1960s, the serotonin hypothesis was published, suggesting that serotonin is the key player in depressive disorders. However, this hypothesis is being increasingly doubted as there is evidence for the influence of other neurotransmitters, such as noradrenaline, glutamate, and dopamine, as well as larger systemic causes such as altered activity in the limbic network or inflammatory processes. In this narrative review, we aim to contribute to the ongoing debate on the involvement of serotonin in depression. We will review the evolution of antidepressant treatments, systemic research on depression over the years, and future research applications that will help to bridge the gap between systemic research and neurotransmitter dynamics using biosensors. These new tools in combination with systemic applications, will in the future provide a deeper understanding of the serotonergic dynamics in depression.
Collapse
Affiliation(s)
- Svenja Bremshey
- Synthetic Biology, University of Bremen, Bremen, Germany
- Neuropharmacology, University of Bremen, Bremen, Germany
| | - Juliana Groß
- Synthetic Biology, University of Bremen, Bremen, Germany
| | - Kim Renken
- Synthetic Biology, University of Bremen, Bremen, Germany
| | | |
Collapse
|
26
|
Ramkumar R, Edge-Partington M, Terstege DJ, Adigun K, Ren Y, Khan NS, Rouhi N, Jamani NF, Tsutsui M, Epp JR, Sargin D. Long-Term Impact of Early-Life Stress on Serotonin Connectivity. Biol Psychiatry 2024; 96:287-299. [PMID: 38316332 DOI: 10.1016/j.biopsych.2024.01.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
BACKGROUND Chronic childhood stress is a prominent risk factor for developing affective disorders, yet mechanisms underlying this association remain unclear. Maintenance of optimal serotonin (5-HT) levels during early postnatal development is critical for the maturation of brain circuits. Understanding the long-lasting effects of early-life stress (ELS) on serotonin-modulated brain connectivity is crucial to develop treatments for affective disorders arising from childhood stress. METHODS Using a mouse model of chronic developmental stress, we determined the long-lasting consequences of ELS on 5-HT circuits and behavior in females and males. Using FosTRAP mice, we cross-correlated regional c-Fos density to determine brain-wide functional connectivity of the raphe nucleus. We next performed in vivo fiber photometry to establish ELS-induced deficits in 5-HT dynamics and optogenetics to stimulate 5-HT release to improve behavior. RESULTS Adult female and male mice exposed to ELS showed heightened anxiety-like behavior. ELS further enhanced susceptibility to acute stress by disrupting the brain-wide functional connectivity of the raphe nucleus and the activity of 5-HT neuron population, in conjunction with increased orbitofrontal cortex (OFC) activity and disrupted 5-HT release in medial OFC. Optogenetic stimulation of 5-HT terminals in the medial OFC elicited an anxiolytic effect in ELS mice in a sex-dependent manner. CONCLUSIONS These findings suggest a significant disruption in 5-HT-modulated brain connectivity in response to ELS, with implications for sex-dependent vulnerability. The anxiolytic effect of the raphe-medial OFC circuit stimulation has potential implications for developing targeted stimulation-based treatments for affective disorders that arise from early life adversities.
Collapse
Affiliation(s)
- Raksha Ramkumar
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Moriah Edge-Partington
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dylan J Terstege
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kabirat Adigun
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yi Ren
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nazmus S Khan
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nahid Rouhi
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Naila F Jamani
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mio Tsutsui
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan R Epp
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derya Sargin
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
27
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
28
|
Boillot M, ter Horst J, López JR, Di Fazio I, Steens ILM, Cohen MX, Homberg JR. Serotonin transporter knockout in rats reduces beta- and gamma-band functional connectivity between the orbitofrontal cortex and amygdala during auditory discrimination. Cereb Cortex 2024; 34:bhae334. [PMID: 39128940 PMCID: PMC11317204 DOI: 10.1093/cercor/bhae334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 06/27/2024] [Indexed: 08/13/2024] Open
Abstract
The orbitofrontal cortex and amygdala collaborate in outcome-guided decision-making through reciprocal projections. While serotonin transporter knockout (SERT-/-) rodents show changes in outcome-guided decision-making, and in orbitofrontal cortex and amygdala neuronal activity, it remains unclear whether SERT genotype modulates orbitofrontal cortex-amygdala synchronization. We trained SERT-/- and SERT+/+ male rats to execute a task requiring to discriminate between two auditory stimuli, one predictive of a reward (CS+) and the other not (CS-), by responding through nose pokes in opposite-side ports. Overall, task acquisition was not influenced by genotype. Next, we simultaneously recorded local field potentials in the orbitofrontal cortex and amygdala of both hemispheres while the rats performed the task. Behaviorally, SERT-/- rats showed a nonsignificant trend for more accurate responses to the CS-. Electrophysiologically, orbitofrontal cortex-amygdala synchronization in the beta and gamma frequency bands during response selection was significantly reduced and associated with decreased hubness and clustering coefficient in both regions in SERT-/- rats compared to SERT+/+ rats. Conversely, theta synchronization at the time of behavioral response in the port associated with reward was similar in both genotypes. Together, our findings reveal the modulation by SERT genotype of the orbitofrontal cortex-amygdala functional connectivity during an auditory discrimination task.
Collapse
Affiliation(s)
- Morgane Boillot
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, Netherlands
| | - Jordi ter Horst
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, Netherlands
| | - José Rey López
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, Netherlands
| | - Ilaria Di Fazio
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, Netherlands
| | - Indra L M Steens
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, Netherlands
| | - Michael X Cohen
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, Netherlands
| | - Judith R Homberg
- Department of Cognitive Neuroscience, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Kapittelweg 29, 6525 EN, Nijmegen, Netherlands
| |
Collapse
|
29
|
Maddaloni G, Chang YJ, Senft RA, Dymecki SM. Adaptation to photoperiod via dynamic neurotransmitter segregation. Nature 2024; 632:147-156. [PMID: 39020173 DOI: 10.1038/s41586-024-07692-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/07/2024] [Indexed: 07/19/2024]
Abstract
Changes in the amount of daylight (photoperiod) alter physiology and behaviour1,2. Adaptive responses to seasonal photoperiods are vital to all organisms-dysregulation associates with disease, including affective disorders3 and metabolic syndromes4. The circadian rhythm circuitry is implicated in such responses5,6, yet little is known about the precise cellular substrates that underlie phase synchronization to photoperiod change. Here we identify a brain circuit and system of axon branch-specific and reversible neurotransmitter deployment that are critical for behavioural and sleep adaptation to photoperiod. A type of neuron called mrEn1-Pet17 in the mouse brainstem median raphe nucleus segregates serotonin from VGLUT3 (also known as SLC17A8, a proxy for glutamate) to different axonal branches that innervate specific brain regions involved in circadian rhythm and sleep-wake timing8,9. This branch-specific neurotransmitter deployment did not distinguish between daylight and dark phase; however, it reorganized with change in photoperiod. Axonal boutons, but not cell soma, changed neurochemical phenotype upon a shift away from equinox light/dark conditions, and these changes were reversed upon return to equinox conditions. When we genetically disabled Vglut3 in mrEn1-Pet1 neurons, sleep-wake periods, voluntary activity and clock gene expression did not synchronize to the new photoperiod or were delayed. Combining intersectional rabies virus tracing and projection-specific neuronal silencing, we delineated a preoptic area-to-mrEn1Pet1 connection that was responsible for decoding the photoperiodic inputs, driving the neurotransmitter reorganization and promoting behavioural synchronization. Our results reveal a brain circuit and periodic, branch-specific neurotransmitter deployment that regulates organismal adaptation to photoperiod change.
Collapse
Affiliation(s)
- G Maddaloni
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Y J Chang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - R A Senft
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - S M Dymecki
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Henderson F, Dumas S, Gangarossa G, Bernard V, Pujol M, Poirel O, Pietrancosta N, El Mestikawy S, Daumas S, Fabre V. Regulation of stress-induced sleep perturbations by dorsal raphe VGLUT3 neurons in male mice. Cell Rep 2024; 43:114411. [PMID: 38944834 DOI: 10.1016/j.celrep.2024.114411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/07/2024] [Accepted: 06/12/2024] [Indexed: 07/02/2024] Open
Abstract
Exposure to stressors has profound effects on sleep that have been linked to serotonin (5-HT) neurons of the dorsal raphe nucleus (DR). However, the DR also comprises glutamatergic neurons expressing vesicular glutamate transporter type 3 (DRVGLUT3), leading us to examine their role. Cell-type-specific tracing revealed that DRVGLUT3 neurons project to brain areas regulating arousal and stress. We found that chemogenetic activation of DRVGLUT3 neurons mimics stress-induced sleep perturbations. Furthermore, deleting VGLUT3 in the DR attenuated stress-induced sleep perturbations, especially after social defeat stress. In the DR, VGLUT3 is found in subsets of 5-HT and non-5-HT neurons. We observed that both populations are activated by acute stress, including those projecting to the ventral tegmental area. However, deleting VGLUT3 in 5-HT neurons minimally affected sleep regulation. These findings suggest that VGLUT3 expression in the DR drives stress-induced sleep perturbations, possibly involving non-5-HT DRVGLUT3 neurons.
Collapse
Affiliation(s)
- Fiona Henderson
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France
| | | | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France; Institut Universitaire de France (IUF), Paris, France
| | - Véronique Bernard
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France
| | - Marine Pujol
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France
| | - Odile Poirel
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France
| | - Nicolas Pietrancosta
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France; Sorbonne Université, CNRS UMR 7203, Laboratoire des BioMolécules, 75005 Paris, France
| | - Salah El Mestikawy
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France; Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montréal, QC H4H 1R3, Canada
| | - Stéphanie Daumas
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France.
| | - Véronique Fabre
- Sorbonne Université, CNRS UMR 8246, INSERM U1130 - Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), 75005 Paris, France.
| |
Collapse
|
31
|
Fulton KA, Zimmerman D, Samuel A, Vogt K, Datta SR. Common principles for odour coding across vertebrates and invertebrates. Nat Rev Neurosci 2024; 25:453-472. [PMID: 38806946 DOI: 10.1038/s41583-024-00822-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
The olfactory system is an ideal and tractable system for exploring how the brain transforms sensory inputs into behaviour. The basic tasks of any olfactory system include odour detection, discrimination and categorization. The challenge for the olfactory system is to transform the high-dimensional space of olfactory stimuli into the much smaller space of perceived objects and valence that endows odours with meaning. Our current understanding of how neural circuits address this challenge has come primarily from observations of the mechanisms of the brain for processing other sensory modalities, such as vision and hearing, in which optimized deep hierarchical circuits are used to extract sensory features that vary along continuous physical dimensions. The olfactory system, by contrast, contends with an ill-defined, high-dimensional stimulus space and discrete stimuli using a circuit architecture that is shallow and parallelized. Here, we present recent observations in vertebrate and invertebrate systems that relate the statistical structure and state-dependent modulation of olfactory codes to mechanisms of perception and odour-guided behaviour.
Collapse
Affiliation(s)
- Kara A Fulton
- Department of Neuroscience, Harvard Medical School, Boston, MA, USA
| | - David Zimmerman
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Aravi Samuel
- Department of Physics, Harvard University, Cambridge, MA, USA
| | - Katrin Vogt
- Department of Physics, Harvard University, Cambridge, MA, USA.
- Department of Biology, University of Konstanz, Konstanz, Germany.
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, Konstanz, Germany.
| | | |
Collapse
|
32
|
Cheng J, Chen L, Zheng YN, Liu J, Zhang L, Zhang XM, Huang L, Yuan QL. Disfunction of dorsal raphe nucleus-hippocampus serotonergic-HTR3 transmission results in anxiety phenotype of Neuroplastin 65-deficient mice. Acta Pharmacol Sin 2024; 45:1393-1405. [PMID: 38528118 PMCID: PMC11192762 DOI: 10.1038/s41401-024-01252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/26/2024] [Indexed: 03/27/2024]
Abstract
Anxiety disorders are the most common psychiatric condition, but the etiology of anxiety disorders remains largely unclear. Our previous studies have shown that neuroplastin 65 deficiency (NP65-/-) mice exhibit abnormal social and mental behaviors and decreased expression of tryptophan hydroxylase 2 (TPH2) protein. However, whether a causal relationship between TPH2 reduction and anxiety disorders exists needs to be determined. In present study, we found that replenishment of TPH2 in dorsal raphe nucleus (DRN) enhanced 5-HT level in the hippocampus and alleviated anxiety-like behaviors. In addition, injection of AAV-NP65 in DRN significantly increased TPH2 expression in DRN and hippocampus, and reduced anxiety-like behaviors. Acute administration of exogenous 5-HT or HTR3 agonist SR57227A in hippocampus mitigated anxiety-like behaviors in NP65-/- mice. Moreover, replenishment of TPH2 in DRN partly repaired the impairment of long-term potentiation (LTP) maintenance in hippocampus of NP65-/- mice. Finally, we found that loss of NP65 lowered transcription factors Lmx1b expression in postnatal stage and replenishment of NP65 in DRN reversed the decrease in Lmx1b expression of NP65-/- mice. Together, our findings reveal that NP65 deficiency induces anxiety phenotype by downregulating DRN-hippocampus serotonergic-HTR3 transmission. These studies provide a novel and insightful view about NP65 function, suggesting an attractive potential target for treatment of anxiety disorders.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ling Chen
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ya-Ni Zheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Juan Liu
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Lei Zhang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xiao-Ming Zhang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Liang Huang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiong-Lan Yuan
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
33
|
Corradetti D, Bernardi A, Corradetti R. Deep learning models for atypical serotonergic cells recognition. J Neurosci Methods 2024; 407:110158. [PMID: 38703797 DOI: 10.1016/j.jneumeth.2024.110158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/15/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The serotonergic system modulates brain processes via functionally distinct subpopulations of neurons with heterogeneous properties, including their electrophysiological activity. In extracellular recordings, serotonergic neurons to be investigated for their functional properties are commonly identified on the basis of "typical" features of their activity, i.e. slow regular firing and relatively long duration of action potentials. Thus, due to the lack of equally robust criteria for discriminating serotonergic neurons with "atypical" features from non-serotonergic cells, the physiological relevance of the diversity of serotonergic neuron activities results largely understudied. NEW METHODS We propose deep learning models capable of discriminating typical and atypical serotonergic neurons from non-serotonergic cells with high accuracy. The research utilized electrophysiological in vitro recordings from serotonergic neurons identified by the expression of fluorescent proteins specific to the serotonergic system and non-serotonergic cells. These recordings formed the basis of the training, validation, and testing data for the deep learning models. The study employed convolutional neural networks (CNNs), known for their efficiency in pattern recognition, to classify neurons based on the specific characteristics of their action potentials. RESULTS The models were trained on a dataset comprising 27,108 original action potential samples, alongside an extensive set of 12 million synthetic action potential samples, designed to mitigate the risk of overfitting the background noise in the recordings, a potential source of bias. Results show that the models achieved high accuracy and were further validated on "non-homogeneous" data, i.e., data unknown to the model and collected on different days from those used for the training of the model, to confirm their robustness and reliability in real-world experimental conditions. COMPARISON WITH EXISTING METHODS Conventional methods for identifying serotonergic neurons allow recognition of serotonergic neurons defined as typical. Our model based on the analysis of the sole action potential reliably recognizes over 94% of serotonergic neurons including those with atypical features of spike and activity. CONCLUSION The model is ready for use in experiments conducted with the here described recording parameters. We release the codes and procedures allowing to adapt the model to different acquisition parameters or for identification of other classes of spontaneously active neurons.
Collapse
Affiliation(s)
- Daniele Corradetti
- Grupo de Fisica Matematica, Instituto Superior Tecnico, Av. Rovisco Pais, Lisboa, 1049-001, Portugal; Departamento de Matematica, Universidade do Algarve, Campus de Gambelas, Faro, 8005-139, Faro, Portugal.
| | | | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale G. Pieraccini 6, Firenze, 50139, Toscana, Italy.
| |
Collapse
|
34
|
Feng YY, Bromberg-Martin ES, Monosov IE. Dorsal raphe neurons integrate the values of reward amount, delay, and uncertainty in multi-attribute decision-making. Cell Rep 2024; 43:114341. [PMID: 38878290 DOI: 10.1016/j.celrep.2024.114341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/27/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
The dorsal raphe nucleus (DRN) is implicated in psychiatric disorders that feature impaired sensitivity to reward amount, impulsivity when facing reward delays, and risk-seeking when confronting reward uncertainty. However, it has been unclear whether and how DRN neurons signal reward amount, reward delay, and reward uncertainty during multi-attribute value-based decision-making, where subjects consider these attributes to make a choice. We recorded DRN neurons as monkeys chose between offers whose attributes, namely expected reward amount, reward delay, and reward uncertainty, varied independently. Many DRN neurons signaled offer attributes, and this population tended to integrate the attributes in a manner that reflected monkeys' preferences for amount, delay, and uncertainty. After decision-making, in response to post-decision feedback, these same neurons signaled signed reward prediction errors, suggesting a broader role in tracking value across task epochs and behavioral contexts. Our data illustrate how the DRN participates in value computations, guiding theories about the role of the DRN in decision-making and psychiatric disease.
Collapse
Affiliation(s)
- Yang-Yang Feng
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | | | - Ilya E Monosov
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, USA; Washington University Pain Center, Washington University, St. Louis, MO, USA; Department of Neurosurgery, Washington University, St. Louis, MO, USA; Department of Electrical Engineering, Washington University, St. Louis, MO, USA.
| |
Collapse
|
35
|
Bahi A. Serotonin transporter knockdown relieves depression-like behavior and ethanol-induced CPP in mice after chronic social defeat stress. Behav Brain Res 2024; 466:114998. [PMID: 38614210 DOI: 10.1016/j.bbr.2024.114998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/15/2024]
Abstract
Patients with stress-triggered major depression disorders (MDD) can often seek comfort or temporary relief through alcohol consumption, as they may turn to it as a means of self-medication or coping with overwhelming emotions. The use of alcohol as a coping mechanism for stressful events can escalate, fostering a cycle where the temporary relief it provides from depression can deepen into alcohol dependence, exacerbating both conditions. Although, the specific mechanisms involved in stress-triggered alcohol dependence and MDD comorbidities are not well understood, a large body of literature suggests that the serotonin transporter (SERT) plays a critical role in these abnormalities. To further investigate this hypothesis, we used a lentiviral-mediated knockdown approach to examine the role of hippocampal SERT knockdown in social defeat stress-elicited depression like behavior and ethanol-induced place preference (CPP). The results showed that social defeat stress-pro depressant effects were reversed following SERT knockdown demonstrated by increased sucrose preference, shorter latency to feed in the novelty suppressed feeding test, and decreased immobility time in the tail suspension and forced swim tests. Moreover, and most importantly, social stress-induced ethanol-CPP acquisition and reinstatement were significantly reduced following hippocampal SERT knockdown using short hairpin RNA shRNA-expressing lentiviral vectors. Finally, we confirmed that SERT hippocampal mRNA expression correlated with measures of depression- and ethanol-related behaviors by Pearson's correlation analysis. Taken together, our data suggest that hippocampal serotoninergic system is involved in social stress-triggered mood disorders as well as in the acquisition and retrieval of ethanol contextual memory and that blockade of this transporter can decrease ethanol rewarding properties.
Collapse
Affiliation(s)
- Amine Bahi
- College of Medicine, Ajman University, Ajman, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Department of Anatomy, CMHS, UAE University, Al Ain, United Arab Emirates.
| |
Collapse
|
36
|
Hamada HT, Abe Y, Takata N, Taira M, Tanaka KF, Doya K. Optogenetic activation of dorsal raphe serotonin neurons induces brain-wide activation. Nat Commun 2024; 15:4152. [PMID: 38755120 PMCID: PMC11099070 DOI: 10.1038/s41467-024-48489-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Serotonin is a neuromodulator that affects multiple behavioral and cognitive functions. Nonetheless, how serotonin causes such a variety of effects via brain-wide projections and various receptors remains unclear. Here we measured brain-wide responses to optogenetic stimulation of serotonin neurons in the dorsal raphe nucleus (DRN) of the male mouse brain using functional MRI with an 11.7 T scanner and a cryoprobe. Transient activation of DRN serotonin neurons caused brain-wide activation, including the medial prefrontal cortex, the striatum, and the ventral tegmental area. The same stimulation under anesthesia with isoflurane decreased brain-wide activation, including the hippocampal complex. These brain-wide response patterns can be explained by DRN serotonergic projection topography and serotonin receptor expression profiles, with enhanced weights on 5-HT1 receptors. Together, these results provide insight into the DR serotonergic system, which is consistent with recent discoveries of its functions in adaptive behaviors.
Collapse
Affiliation(s)
- Hiro Taiyo Hamada
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
- Research & Development Department, Araya Inc, Tokyo, Japan.
| | - Yoshifumi Abe
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Norio Takata
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Masakazu Taira
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Doya
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
37
|
Castle ME, Flanigan ME. The role of brain serotonin signaling in excessive alcohol consumption and withdrawal: A call for more research in females. Neurobiol Stress 2024; 30:100618. [PMID: 38433994 PMCID: PMC10907856 DOI: 10.1016/j.ynstr.2024.100618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/05/2024] Open
Abstract
Alcohol Use Disorder (AUD) is a leading cause of death and disability worldwide, but current treatments are insufficient in fully addressing the symptoms that often lead to relapses in alcohol consumption. The brain's serotonin system has been implicated in AUD for decades and is a major regulator of stress-related behaviors associated with increased alcohol consumption. This review will discuss the current literature on the association between neurobiological adaptations in serotonin systems and AUD in humans as well as the effectiveness of serotonin receptor manipulations on alcohol-related behaviors like consumption and withdrawal. We will further discuss how these findings in humans relate to findings in animal models, including a comparison of systemic pharmacological manipulations modulating alcohol consumption. We next provide a detailed overview of brain region-specific roles for serotonin and serotonin receptor signaling in alcohol-related behaviors in preclinical animal models, highlighting the complexity of forming a cohesive model of serotonin function in AUD and providing possible avenues for more effective therapeutic intervention. Throughout the review, we discuss what is known about sex differences in the sequelae of AUD and the role of serotonin in these sequelae. We stress a critical need for additional studies in women and female animals so that we may build a clearer path to elucidating sex-specific serotonergic mechanisms and develop better treatments.
Collapse
Affiliation(s)
- Megan E. Castle
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Meghan E. Flanigan
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
38
|
Kohtz AS, Zhao J, Aston-Jones G. Serotonin Signaling in Hippocampus during Initial Cocaine Abstinence Drives Persistent Drug Seeking. J Neurosci 2024; 44:e1505212024. [PMID: 38514181 PMCID: PMC11044100 DOI: 10.1523/jneurosci.1505-21.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 03/23/2024] Open
Abstract
The initiation of abstinence after chronic drug self-administration is stressful. Cocaine-seeking behavior on the first day of the absence of the expected drug (Extinction Day 1, ED1) is reduced by blocking 5-HT signaling in dorsal hippocampal cornu ammonis 1 (CA1) in both male and female rats. We hypothesized that the experience of ED1 can substantially influence later relapse behavior and that dorsal raphe (DR) serotonin (5-HT) input to CA1 may be involved. We inhibited 5-HT1A/1B receptors (WAY-100635 plus GR-127935), or DR input (chemogenetics), in CA1 on ED1 to test the role of this pathway on cocaine-seeking persistence 2 weeks later. We also inhibited 5-HT1A or 5-HT1B receptors in CA1 during conditioned place preference (CPP) for cocaine, to examine mechanisms involved in the persistent effects of ED1 manipulations. Inhibition of DR inputs, or 5-HT1A/1B signaling, in CA1 decreased drug seeking on ED1 and decreased cocaine seeking 2 weeks later revealing that 5-HT signaling in CA1 during ED1 contributes to persistent drug seeking during abstinence. In addition, 5-HT1B antagonism alone transiently decreased drug-associated memory performance when given prior to a CPP test, whereas similar antagonism of 5-HT1A alone had no such effect but blocked CPP retrieval on a test 24 h later. These CPP findings are consistent with prior work showing that DR inputs to CA1 augment recall of the drug-associated context and drug seeking via 5-HT1B receptors and prevent consolidation of the updated nondrug context via 5-HT1A receptors. Thus, treatments that modulate 5-HT-dependent memory mechanisms in CA1 during initial abstinence may facilitate later maintenance of abstinence.
Collapse
Affiliation(s)
- Amy S Kohtz
- Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Joshua Zhao
- Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University, Piscataway, New Jersey 08854
| |
Collapse
|
39
|
Lee WL, Westergaard X, Hwu C, Hwu J, Fiala T, Lacefield C, Boltaev U, Mendieta AM, Lin L, Sonders MS, Brown KR, He K, Asher WB, Javitch JA, Sulzer D, Sames D. Molecular Design of SERTlight: A Fluorescent Serotonin Probe for Neuronal Labeling in the Brain. J Am Chem Soc 2024; 146:9564-9574. [PMID: 38557024 DOI: 10.1021/jacs.3c11617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The serotonergic transmitter system plays fundamental roles in the nervous system in neurotransmission, synaptic plasticity, pathological processes, and therapeutic effects of antidepressants and psychedelics, as well as in the gastrointestinal and circulatory systems. We introduce a novel small molecule fluorescent agent, termed SERTlight, that specifically labels serotonergic neuronal cell bodies, dendrites, and axonal projections as a serotonin transporter (SERT) fluorescent substrate. SERTlight was developed by an iterative molecular design process, based on an aminoethyl-quinolone system, to integrate structural elements that impart SERT substrate activity, sufficient fluorescent brightness, and a broad absence of pharmacological activity, including at serotonin (5-hydroxytryptamine, 5HT) receptors, other G protein-coupled receptors (GPCRs), ion channels, and monoamine transporters. The high labeling selectivity is not achieved by high affinity binding to SERT itself but rather by a sufficient rate of SERT-mediated transport of SERTlight, resulting in accumulation of these molecules in 5HT neurons and yielding a robust and selective optical signal in the mammalian brain. SERTlight provides a stable signal, as it is not released via exocytosis nor by reverse SERT transport induced by 5HT releasers such as MDMA. SERTlight is optically, pharmacologically, and operationally orthogonal to a wide range of genetically encoded sensors, enabling multiplexed imaging. SERTlight enables labeling of distal 5HT axonal projections and simultaneous imaging of the release of endogenous 5HT using the GRAB5HT sensor, providing a new versatile molecular tool for the study of the serotonergic system.
Collapse
Affiliation(s)
- Wei-Li Lee
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Xavier Westergaard
- Department of Biological Sciences, Columbia University, New York, New York 10027, United States
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Christopher Hwu
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Jennifer Hwu
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Tomas Fiala
- Department of Chemistry, Columbia University, New York, New York 10027, United States
- Laboratory of Organic Chemistry, ETH Zürich, D-CHAB, Vladimir-Prelog-Weg 3, 8093 Zürich, Switzerland
| | - Clay Lacefield
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Systems Neuroscience, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Umed Boltaev
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Adriana M Mendieta
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Lisa Lin
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Mark S Sonders
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Keaon R Brown
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Keer He
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Wesley B Asher
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Neurology, Columbia University Irving Medical Center, New York, New York 10032, United States
- Department of Molecular Pharmacology and Therapeutics, Columbia University Irving Medical Center, New York, New York 10032, United States
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York 10032, United States
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, New York 10027, United States
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York 10027, United States
| |
Collapse
|
40
|
Mitsui K, Takahashi A. Aggression modulator: Understanding the multifaceted role of the dorsal raphe nucleus. Bioessays 2024; 46:e2300213. [PMID: 38314963 DOI: 10.1002/bies.202300213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/07/2024]
Abstract
Aggressive behavior is instinctively driven behavior that helps animals to survive and reproduce and is closely related to multiple behavioral and physiological processes. The dorsal raphe nucleus (DRN) is an evolutionarily conserved midbrain structure that regulates aggressive behavior by integrating diverse brain inputs. The DRN consists predominantly of serotonergic (5-HT:5-hydroxytryptamine) neurons and decreased 5-HT activity was classically thought to increase aggression. However, recent studies challenge this 5-HT deficiency model, revealing a more complex role for the DRN 5-HT system in aggression. Furthermore, emerging evidence has shown that non-5-HT populations in the DRN and specific neural circuits contribute to the escalation of aggressive behavior. This review argues that the DRN serves as a multifaceted modulator of aggression, acting not only via 5-HT but also via other neurotransmitters and neural pathways, as well as different subsets of 5-HT neurons. In addition, we discuss the contribution of DRN neurons in the behavioral and physiological aspects implicated in aggressive behavior, such as arousal, reward, and impulsivity, to further our understanding of DRN-mediated aggression modulation.
Collapse
Affiliation(s)
- Koshiro Mitsui
- Laboratory of Behavioral Neurobiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aki Takahashi
- Laboratory of Behavioral Neurobiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Institute of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
41
|
Gullino LS, Fuller C, Dunn P, Collins HM, El Mestikawy S, Sharp T. Evidence for a Role of 5-HT-glutamate Co-releasing Neurons in Acute Stress Mechanisms. ACS Chem Neurosci 2024; 15:1185-1196. [PMID: 38377469 PMCID: PMC10958520 DOI: 10.1021/acschemneuro.3c00758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/22/2024] Open
Abstract
A major subpopulation of midbrain 5-hydroxytryptamine (5-HT) neurons expresses the vesicular glutamate transporter 3 (VGLUT3) and co-releases 5-HT and glutamate, but the function of this co-release is unclear. Given the strong links between 5-HT and uncontrollable stress, we used a combination of c-Fos immunohistochemistry and conditional gene knockout mice to test the hypothesis that glutamate co-releasing 5-HT neurons are activated by stress and involved in stress coping. Acute, uncontrollable swim stress increased c-Fos immunoreactivity in neurons co-expressing VGLUT3 and the 5-HT marker tryptophan hydroxylase 2 (TPH2) in the dorsal raphe nucleus (DRN). This effect was localized in the ventral DRN subregion and prevented by the antidepressant fluoxetine. In contrast, a more controllable stressor, acute social defeat, had no effect on c-Fos immunoreactivity in VGLUT3-TPH2 co-expressing neurons in the DRN. To test whether activation of glutamate co-releasing 5-HT neurons was causally linked to stress coping, mice with a specific deletion of VGLUT3 in 5-HT neurons were exposed to acute swim stress. Compared to wildtype controls, the mutant mice showed increased climbing behavior, a measure of active coping. Wildtype mice also showed increased climbing when administered fluoxetine, revealing an interesting parallel between the behavioral effects of genetic loss of VGLUT3 in 5-HT neurons and 5-HT reuptake inhibition. We conclude that 5-HT-glutamate co-releasing neurons are recruited by exposure to uncontrollable stress. Furthermore, natural variation in the balance of 5-HT and glutamate co-released at the 5-HT synapse may impact stress susceptibility.
Collapse
Affiliation(s)
- L. Sophie Gullino
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Cara Fuller
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Poppy Dunn
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Helen M. Collins
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Salah El Mestikawy
- Douglas
Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC H4H
1R3, Canada
- Sorbonne
Université, INSERM, CNRS, Neuroscience Paris Seine –
Institut de Biologie Paris Seine (NPS – IBPS), 75005 Paris, France
| | - Trevor Sharp
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| |
Collapse
|
42
|
Song R, Soler-Cedeño O, Xi ZX. Optical Intracranial Self-Stimulation (oICSS): A New Behavioral Model for Studying Drug Reward and Aversion in Rodents. Int J Mol Sci 2024; 25:3455. [PMID: 38542425 PMCID: PMC10970671 DOI: 10.3390/ijms25063455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/10/2024] [Accepted: 03/17/2024] [Indexed: 11/03/2024] Open
Abstract
Brain-stimulation reward, also known as intracranial self-stimulation (ICSS), is a commonly used procedure for studying brain reward function and drug reward. In electrical ICSS (eICSS), an electrode is surgically implanted into the medial forebrain bundle (MFB) in the lateral hypothalamus or the ventral tegmental area (VTA) in the midbrain. Operant lever responding leads to the delivery of electrical pulse stimulation. The alteration in the stimulation frequency-lever response curve is used to evaluate the impact of pharmacological agents on brain reward function. If a test drug induces a leftward or upward shift in the eICSS response curve, it implies a reward-enhancing or abuse-like effect. Conversely, if a drug causes a rightward or downward shift in the functional response curve, it suggests a reward-attenuating or aversive effect. A significant drawback of eICSS is the lack of cellular selectivity in understanding the neural substrates underlying this behavior. Excitingly, recent advancements in optical ICSS (oICSS) have facilitated the development of at least three cell type-specific oICSS models-dopamine-, glutamate-, and GABA-dependent oICSS. In these new models, a comparable stimulation frequency-lever response curve has been established and employed to study the substrate-specific mechanisms underlying brain reward function and a drug's rewarding versus aversive effects. In this review article, we summarize recent progress in this exciting research area. The findings in oICSS have not only increased our understanding of the neural mechanisms underlying drug reward and addiction but have also introduced a novel behavioral model in preclinical medication development for treating substance use disorders.
Collapse
Affiliation(s)
- Rui Song
- Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology (BIPT), 27th Taiping Road, Beijing 100850, China
| | - Omar Soler-Cedeño
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse (NIDA), Intramural Research Program (IRP), Baltimore, MD 21224, USA;
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse (NIDA), Intramural Research Program (IRP), Baltimore, MD 21224, USA;
| |
Collapse
|
43
|
Ritchie JL, Qi S, Christian RJ, Greenwood MJ, Grenz HI, Swatzell SE, Krych PJ, Fuchs RA. Requisite role of dorsal raphé in contextual cocaine-memory reconsolidation. Neuropharmacology 2024; 246:109832. [PMID: 38176535 PMCID: PMC10901441 DOI: 10.1016/j.neuropharm.2023.109832] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Memory reconsolidation is a process by which labile drug memories are restabilized in long-term memory stores, permitting their enduring control over drug-seeking behaviors. In the present study, we investigated the involvement of the dorsal raphé nuclei (DRN) in cocaine-memory reconsolidation. Sprague-Dawley rats (male, female) were trained to self-administer cocaine in a distinct environmental context to establish contextual drug memories. They then received extinction training in a different context. Next, the rats were re-exposed to the cocaine-predictive context for 15 min to reactivate their cocaine memories or remained in their home cages (no-reactivation control). Memory reactivation was sufficient to increase c-Fos expression, an index of neuronal activation, in the DRN, but not in the median raphé nuclei, during reconsolidation, compared to no reactivation. To determine whether DRN neuronal activity was necessary for cocaine-memory reconsolidation, rats received intra-DRN baclofen plus muscimol (BM; GABAB/A agonists) or vehicle microinfusions immediately after or 6 h after a memory reactivation session conducted with or without lever access. The effects of DRN functional inactivation on long-term memory strength, as indicated by the magnitude of context-induced cocaine seeking, were assessed 72 h later. Intra-DRN BM treatment immediately after memory reactivation with or without lever access attenuated subsequent context-induced cocaine-seeking behavior, independent of sex. Conversely, BM treatment in the adjacent periaqueductal gray (PAG) immediately after memory reactivation, or BM treatment in the DRN 6 h after memory reactivation, did not alter responding. Together, these findings indicate that the DRN plays a requisite role in maintaining cocaine-memory strength during reconsolidation.
Collapse
Affiliation(s)
- J L Ritchie
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - S Qi
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - R J Christian
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - M J Greenwood
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - H I Grenz
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - S E Swatzell
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - P J Krych
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - R A Fuchs
- Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA, USA; Washington State University Alcohol and Drug Abuse Research Program, Pullman, WA, USA.
| |
Collapse
|
44
|
Izidoro Ribeiro R, Almeida Carvalho G, Almeida Chiareli R, Vieira de Assis Lima I, Quaglio Bellozi PM, Oliveira-Lima OC, Oliveira Giacomelli Á, Birbrair A, Santiago Gomez R, Pinheiro de Oliveira AC, Ulrich H, Cunha Xavier Pinto M. Glycine transporter-1 inhibition by NFPS promotes neuroprotection against striatal damage models. Neurosci Lett 2024; 826:137715. [PMID: 38460902 DOI: 10.1016/j.neulet.2024.137715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
The striatum, an essential component of the brain's motor and reward systems, plays a pivotal role in a wide array of cognitive processes. Its dysfunction is a hallmark of neurodegenerative diseases like Parkinson's disease (PD) and Huntington's disease (HD), leading to profound motor and cognitive deficits. These conditions are often related to excitotoxicity, primarily due to overactivation of NMDA receptors (NMDAR). In the synaptic cleft, glycine transporter type 1 (GlyT1) controls the glycine levels, a NMDAR co-agonist, which modulates NMDAR function. This research explored the neuroprotective potential of NFPS, a GlyT1 inhibitor, in murine models of striatal injury. Employing models of neurotoxicity induced by 6-hydroxydopamine (PD model) and quinolinic acid (HD model), we assessed the effectiveness of NFPS pre-treatment in maintaining the integrity of striatal neurons and averting neuronal degeneration. The results indicated that NFPS pre-treatment conferred significant neuroprotection, reducing neuronal degeneration, protecting dopaminergic neurons, and preserving dendritic spines within the striatum. Additionally, this pre-treatment notably mitigated motor impairments resulting from striatal damage. The study revealed that GlyT1 inhibition led to substantial changes in the ratios of NMDAR subunits GluN2A/GluN1 and GluN2B/GluN1, 24 h after NFPS treatment. These findings underscore the neuroprotective efficacy of GlyT1 inhibition, proposing it as a viable therapeutic strategy for striatum-related damage.
Collapse
Affiliation(s)
- Raul Izidoro Ribeiro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Gustavo Almeida Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Raphaela Almeida Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil
| | - Isabel Vieira de Assis Lima
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | - Paula Maria Quaglio Bellozi
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | | | - Alexander Birbrair
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | - Renato Santiago Gomez
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo-SP, Brazil
| | - Mauro Cunha Xavier Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia-GO, Brazil.
| |
Collapse
|
45
|
Nardone S, De Luca R, Zito A, Klymko N, Nicoloutsopoulos D, Amsalem O, Brannigan C, Resch JM, Jacobs CL, Pant D, Veregge M, Srinivasan H, Grippo RM, Yang Z, Zeidel ML, Andermann ML, Harris KD, Tsai LT, Arrigoni E, Verstegen AMJ, Saper CB, Lowell BB. A spatially-resolved transcriptional atlas of the murine dorsal pons at single-cell resolution. Nat Commun 2024; 15:1966. [PMID: 38438345 PMCID: PMC10912765 DOI: 10.1038/s41467-024-45907-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024] Open
Abstract
The "dorsal pons", or "dorsal pontine tegmentum" (dPnTg), is part of the brainstem. It is a complex, densely packed region whose nuclei are involved in regulating many vital functions. Notable among them are the parabrachial nucleus, the Kölliker Fuse, the Barrington nucleus, the locus coeruleus, and the dorsal, laterodorsal, and ventral tegmental nuclei. In this study, we applied single-nucleus RNA-seq (snRNA-seq) to resolve neuronal subtypes based on their unique transcriptional profiles and then used multiplexed error robust fluorescence in situ hybridization (MERFISH) to map them spatially. We sampled ~1 million cells across the dPnTg and defined the spatial distribution of over 120 neuronal subtypes. Our analysis identified an unpredicted high transcriptional diversity in this region and pinpointed the unique marker genes of many neuronal subtypes. We also demonstrated that many neuronal subtypes are transcriptionally similar between humans and mice, enhancing this study's translational value. Finally, we developed a freely accessible, GPU and CPU-powered dashboard ( http://harvard.heavy.ai:6273/ ) that combines interactive visual analytics and hardware-accelerated SQL into a data science framework to allow the scientific community to query and gain insights into the data.
Collapse
Affiliation(s)
- Stefano Nardone
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Roberto De Luca
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Antonino Zito
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Nataliya Klymko
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA
| | | | - Oren Amsalem
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Cory Brannigan
- HEAVY.AI, 100 Montgomery St Fl 5, San Francisco, California, 94104, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center. University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Christopher L Jacobs
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Deepti Pant
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Molly Veregge
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Harini Srinivasan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan M Grippo
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Zongfang Yang
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Mark L Zeidel
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA
| | - Mark L Andermann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Kenneth D Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Linus T Tsai
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elda Arrigoni
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Anne M J Verstegen
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA.
| | - Clifford B Saper
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA.
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Lindeman S, Fu X, Reinert JK, Fukunaga I. Value-related learning in the olfactory bulb occurs through pathway-dependent perisomatic inhibition of mitral cells. PLoS Biol 2024; 22:e3002536. [PMID: 38427708 PMCID: PMC10936853 DOI: 10.1371/journal.pbio.3002536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 03/13/2024] [Accepted: 02/05/2024] [Indexed: 03/03/2024] Open
Abstract
Associating values to environmental cues is a critical aspect of learning from experiences, allowing animals to predict and maximise future rewards. Value-related signals in the brain were once considered a property of higher sensory regions, but their wide distribution across many brain regions is increasingly recognised. Here, we investigate how reward-related signals begin to be incorporated, mechanistically, at the earliest stage of olfactory processing, namely, in the olfactory bulb. In head-fixed mice performing Go/No-Go discrimination of closely related olfactory mixtures, rewarded odours evoke widespread inhibition in one class of output neurons, that is, in mitral cells but not tufted cells. The temporal characteristics of this reward-related inhibition suggest it is odour-driven, but it is also context-dependent since it is absent during pseudo-conditioning and pharmacological silencing of the piriform cortex. Further, the reward-related modulation is present in the somata but not in the apical dendritic tuft of mitral cells, suggesting an involvement of circuit components located deep in the olfactory bulb. Depth-resolved imaging from granule cell dendritic gemmules suggests that granule cells that target mitral cells receive a reward-related extrinsic drive. Thus, our study supports the notion that value-related modulation of olfactory signals is a characteristic of olfactory processing in the primary olfactory area and narrows down the possible underlying mechanisms to deeper circuit components that contact mitral cells perisomatically.
Collapse
Affiliation(s)
- Sander Lindeman
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Xiaochen Fu
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Janine Kristin Reinert
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Izumi Fukunaga
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
47
|
Asim M, Wang H, Chen X, He J. Potentiated GABAergic neuronal activities in the basolateral amygdala alleviate stress-induced depressive behaviors. CNS Neurosci Ther 2024; 30:e14422. [PMID: 37715582 PMCID: PMC10915993 DOI: 10.1111/cns.14422] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/22/2023] [Accepted: 08/14/2023] [Indexed: 09/17/2023] Open
Abstract
AIMS Major depressive disorder is a severe psychiatric disorder that afflicts ~17% of the world population. Neuroimaging investigations of depressed patients have consistently reported the dysfunction of the basolateral amygdala in the pathophysiology of depression. However, how the BLA and related circuits are implicated in the pathogenesis of depression is poorly understood. METHODS Here, we combined fiber photometry, immediate early gene expression (c-fos), optogenetics, chemogenetics, behavioral analysis, and viral tracing techniques to provide multiple lines of evidence of how the BLA neurons mediate depressive-like behavior. RESULTS We demonstrated that the aversive stimuli elevated the neuronal activity of the excitatory BLA neurons (BLACAMKII neurons). Optogenetic activation of CAMKII neurons facilitates the induction of depressive-like behavior while inhibition of these neurons alleviates the depressive-like behavior. Next, we found that the chemogenetic inhibition of GABAergic neurons in the BLA (BLAGABA ) increased the firing frequency of CAMKII neurons and mediates the depressive-like phenotypes. Finally, through fiber photometry recording and chemogenetic manipulation, we proved that the activation of BLAGABA neurons inhibits BLACAMKII neuronal activity and alleviates depressive-like behavior in the mice. CONCLUSION Thus, through evaluating BLAGABA and BLACAMKII neurons by distinct interaction, the BLA regulates depressive-like behavior.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- Department of Biomedical ScienceCity University of Hong KongKowloon TongPeople's Republic of China
| | - Huajie Wang
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- Department of Biomedical ScienceCity University of Hong KongKowloon TongPeople's Republic of China
| | - Xi Chen
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- City University of Hong Kong Shenzhen Research InstituteShenzhenPeople's Republic of China
| | - Jufang He
- Department of NeuroscienceCity University of Hong KongKowloon TongPeople's Republic of China
- City University of Hong Kong Shenzhen Research InstituteShenzhenPeople's Republic of China
| |
Collapse
|
48
|
Ogelman R, Gomez Wulschner LE, Hoelscher VM, Hwang IW, Chang VN, Oh WC. Serotonin modulates excitatory synapse maturation in the developing prefrontal cortex. Nat Commun 2024; 15:1368. [PMID: 38365905 PMCID: PMC10873381 DOI: 10.1038/s41467-024-45734-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Serotonin (5-HT) imbalances in the developing prefrontal cortex (PFC) are linked to long-term behavioral deficits. However, the synaptic mechanisms underlying 5-HT-mediated PFC development are unknown. We found that chemogenetic suppression and enhancement of 5-HT release in the PFC during the first two postnatal weeks decreased and increased the density and strength of excitatory spine synapses, respectively, on prefrontal layer 2/3 pyramidal neurons in mice. 5-HT release on single spines induced structural and functional long-term potentiation (LTP), requiring both 5-HT2A and 5-HT7 receptor signals, in a glutamatergic activity-independent manner. Notably, LTP-inducing 5-HT stimuli increased the long-term survival of newly formed spines ( ≥ 6 h) via 5-HT7 Gαs activation. Chronic treatment of mice with fluoxetine, a selective serotonin-reuptake inhibitor, during the first two weeks, but not the third week of postnatal development, increased the density and strength of excitatory synapses. The effect of fluoxetine on PFC synaptic alterations in vivo was abolished by 5-HT2A and 5-HT7 receptor antagonists. Our data describe a molecular basis of 5-HT-dependent excitatory synaptic plasticity at the level of single spines in the PFC during early postnatal development.
Collapse
Affiliation(s)
- Roberto Ogelman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Luis E Gomez Wulschner
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria M Hoelscher
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - In-Wook Hwang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria N Chang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Won Chan Oh
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
49
|
Pan YD, Zhang Y, Zheng WY, Zhu MZ, Li HY, Ouyang WJ, Wen QQ, Zhu XH. Intermittent Hypobaric Hypoxia Ameliorates Autistic-Like Phenotypes in Mice. J Neurosci 2024; 44:e1665232023. [PMID: 38124211 PMCID: PMC10869151 DOI: 10.1523/jneurosci.1665-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/27/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by persistent deficits in social communication and stereotyped behaviors. Although major advances in basic research on autism have been achieved in the past decade, and behavioral interventions can mitigate the difficulties that individuals with autism experience, little is known about the many fundamental issues of the interventions, and no specific medication has demonstrated efficiency for the core symptoms of ASD. Intermittent hypobaric hypoxia (IHH) is characterized by repeated exposure to lowered atmospheric pressure and oxygen levels, which triggers multiple physiological adaptations in the body. Here, using two mouse models of ASD, male Shank3B -/- and Fmr1 -/y mice, we found that IHH training at an altitude of 5,000 m for 4 h per day, for 14 consecutive days, ameliorated autistic-like behaviors. Moreover, IHH training enhanced hypoxia inducible factor (HIF) 1α in the dorsal raphe nucleus (DRN) and activated the DRN serotonergic neurons. Infusion of cobalt chloride into the DRN, to mimic IHH in increasing HIF1α expression or genetically knockdown PHD2 to upregulate HIF1α expression in the DRN serotonergic neurons, alleviated autistic-like behaviors in Shank3B -/- mice. In contrast, downregulation of HIF1α in DRN serotonergic neurons induced compulsive behaviors. Furthermore, upregulating HIF1α in DRN serotonergic neurons increased the firing rates of these neurons, whereas downregulation of HIF1α in DRN serotonergic neurons decreased their firing rates. These findings suggest that IHH activated DRN serotonergic neurons via upregulation of HIF1α, and thus ameliorated autistic-like phenotypes, providing a novel therapeutic option for ASD.
Collapse
Affiliation(s)
- Yi-da Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Yuan Zhang
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Wen-Ying Zheng
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Min-Zhen Zhu
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Huan-Yu Li
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Wen-Jie Ouyang
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Qin-Qing Wen
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xin-Hong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
50
|
Zhu R, Lan Y, Qian X, Zhao J, Wang G, Tian P, Chen W. Streptococcus salivarius subsp. thermophilus CCFM1312 enhanced mice resilience to activity-based anorexia. Food Funct 2024; 15:1431-1442. [PMID: 38224462 DOI: 10.1039/d3fo04663j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Probiotic intervention, already showing promise in the treatment of various psychiatric disorders like depression, emerges as a potential therapy for anorexia nervosa (AN) with minimal side effects. In this study, we established an activity-based anorexia (ABA) model to probe the pathogenesis of AN and assess the impact of probiotics on ABA mice. ABA resulted in a compensatory increase in duodenal ghrelin levels, impairing the regulation of feeding and the brain reward system. Intervention with Streptococcus salivarius subsp. thermophilus CCFM1312 ameliorated these ABA-induced effects, and the activation of neurons in the nucleus tractus solitarius (NTS) was observed following probiotic administration, revealing the advantageous role of probiotics in AN through the vagus nerve. Furthermore, our metabolomics analysis of cecal contents unveiled that S. salivarius subsp. thermophilus CCFM1312 modulated gut microbiota metabolism and thereby regulated intestinal ghrelin levels.
Collapse
Affiliation(s)
- Ran Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yuming Lan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|