1
|
O’Connell CJ, Robson MJ. Apples to oranges: environmentally derived, dynamic regulation of serotonin neuron subpopulations in adulthood? Neural Regen Res 2025; 20:2596-2597. [PMID: 39503429 PMCID: PMC11801279 DOI: 10.4103/nrr.nrr-d-24-00507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 02/08/2025] Open
Affiliation(s)
| | - Matthew J. Robson
- James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
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 2025:10.1038/s41386-025-02063-w. [PMID: 39934409 DOI: 10.1038/s41386-025-02063-w] [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: 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
|
Balasubramanian N, Wang R, Ismail S, Hartman B, Aboushaar Z, Marcinkiewcz CA. A New Insight into the Role of CART Peptide in Serotonergic Function and Anxiety. J Neurosci 2025; 45:e0467242024. [PMID: 39909575 PMCID: PMC11800755 DOI: 10.1523/jneurosci.0467-24.2024] [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: 03/07/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 02/07/2025] Open
Abstract
Cocaine- and amphetamine-regulated transcript (CART) peptide has been implicated in stress-related behaviors that are regulated by central serotonergic (5-HT) systems in the dorsal raphe nucleus (DRN). Here, we aimed to investigate the interaction between CART and DRN 5-HTergic systems after initially observing CART axonal terminals in the DRN. We found that microinfusion of CART peptide (55-102) into the DRN-induced anxiogenic effects in male C57BL/6J mice, while central administration of CART reduced c-Fos in 5-HTDRN neurons. This inhibitory effect of exogenous CART on 5-HTDRN activity and local 5-HT release was also demonstrated via in vivo fiber photometry coupled with calcium and 5-HT biosensors. CART inputs to the DRN were observed in various subcortical nuclei, but only those in the centrally projecting Edinger-Westphal nucleus (EWcp) were highly responsive to stress. Chemogenetic activation of these DRN-projecting CARTEWcp neurons recapitulated the effects of intra-DRN CART infusion on anxiety-like behavior in males, but not in females, suggesting a sex-specific role for this pathway. Interestingly, CARTEWcp projections to the DRN made direct synaptic contact primarily with non-5-HT neurons, which were also found to express putative CART receptors. Furthermore, chemogenetic stimulation of this CARTEWcp→DRN pathway inhibited 5-HT neurons while increasing activity in local GABAergic neurons. In summary, this study establishes for the first time a neuromodulatory role for CARTEWcp neurons in 5-HTDRN neurotransmission and suggests that CART may drive anxiety-like behavior by promoting feedforward inhibition of 5-HT neurons.
Collapse
Affiliation(s)
- Nagalakshmi Balasubramanian
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242
| | - Ruixiang Wang
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242
| | - Shafa Ismail
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242
| | - Benjamin Hartman
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242
| | - Zeid Aboushaar
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242
| | - Catherine A Marcinkiewcz
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
4
|
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) 2025:10.1007/s00213-025-06749-3. [PMID: 39903245 DOI: 10.1007/s00213-025-06749-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
5
|
Ruge O, Hoppe JPM, Dalle Molle R, Silveira PP. Early environmental influences on the orbito-frontal cortex function and its effects on behavior. Neurosci Biobehav Rev 2025; 169:106013. [PMID: 39814119 DOI: 10.1016/j.neubiorev.2025.106013] [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: 10/17/2024] [Revised: 01/07/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Early-life adversity during pre- and early post-natal phases can impact brain development and lead to maladaptive changes in executive function related behaviors. This increases the risk for a range of psychopathologies and physical diseases. Importantly, exposure to adversities during these periods is also linked to alterations in the orbito-frontal cortex (OFC) which is a key player in these executive functions. The OFC thus appears to be a central node in this association between early life stress and disease risk. Gaining a clear, and detailed understanding of the association between early life stress, OFC function, and executive function, as well as the underlying mechanisms mediating this association is relevant to inform potential therapeutic interventions. In this paper, we begin by reviewing evidence linking early life adversities to 1) alterations in behaviors regulated by the OFC and 2) changes in OFC anatomy and function. We then present insights into the underlying mechanisms for these changes, stemming from early life adversity models, and highlight important future directions for this line of research.
Collapse
Affiliation(s)
- Olivia Ruge
- Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - João Paulo Maires Hoppe
- Douglas Research Centre, McGill University, Montreal, QC, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | | | - Patricia Pelufo Silveira
- Douglas Research Centre, McGill University, Montreal, QC, Canada; Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Faraji N, Payami B, Ebadpour N, Gorji A. Vagus nerve stimulation and gut microbiota interactions: A novel therapeutic avenue for neuropsychiatric disorders. Neurosci Biobehav Rev 2025; 169:105990. [PMID: 39716559 DOI: 10.1016/j.neubiorev.2024.105990] [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: 11/18/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
The rising prevalence of treatment-resistant neuropsychiatric disorders underscores the need for innovative and effective treatment strategies. The gut microbiota (GM) plays a pivotal role in the progression of these diseases, influencing the brain and mental health through the gut-brain axis (GBA). The vagus nerve plays a significant role in the GBA, making it a key area of focus for potential novel therapeutic interventions. Vagus nerve stimulation (VNS) was introduced and approved as a treatment for refractory forms of some neuropsychological disorders, such as depression and epilepsy. Considering its impact on several brain regions that play a vital part in mood, motivation, affection, and cognitive function, the VNS has shown significant therapeutic potential for treating a variety of neuropsychiatric disorders. Using VNS to target the bidirectional communication pathways linking the GM and the VN could present an exciting and novel approach to treating neuropsychological disorders. Imbalances in the GM, such as dysbiosis, can impair the communication pathways between the gut and the brain, contributing to the development of neuropsychological disorders. VNS shows potential for modulating these interconnected systems, helping to restore balance. Interestingly, the composition of the GM may also influence the effectiveness of VNS, as it has the potential to modify the brain's response to this therapeutic approach. This study provides a comprehensive analysis of a relatively unexplored but noteworthy interaction between VNS and GM in the treatment of neuropsychiatric disorders. In addition, we discussed the mechanisms, therapeutic potential, and clinical implications of VNS on the GBA across neuropsychiatric disorders.
Collapse
Affiliation(s)
- Navid Faraji
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Payami
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Epilepsy Research Center, Department of Neurosurgery, Münster University, Germany; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
7
|
Pierson SR, Fiock KL, Wang R, Balasubramanian N, Reinhardt J, Khan KM, James TD, Hunter ML, Cooper BJ, Williamsen HR, Betters R, Deniz K, Lee G, Aldridge G, Hefti MM, Marcinkiewcz CA. Tau pathology in the dorsal raphe may be a prodromal indicator of Alzheimer's disease. Mol Psychiatry 2025; 30:532-546. [PMID: 39143322 DOI: 10.1038/s41380-024-02664-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 06/22/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
Protein aggregation in brainstem nuclei is thought to occur in the early stages of Alzheimer's disease (AD), but its specific role in driving prodromal symptoms and disease progression is largely unknown. The dorsal raphe nucleus (DRN) contains a large population of serotonin (5-hydroxytryptamine; 5-HT) neurons that regulate mood, reward-related behavior, and sleep, which are all disrupted in AD. We report here that tau pathology is present in the DRN of individuals 25-80 years old without a known history of dementia, and its prevalence was comparable to the locus coeruleus (LC). By comparison, fewer cases were positive for other pathological proteins including α-synuclein, β-amyloid, and TDP-43. To evaluate how early tau pathology impacts behavior, we overexpressed human P301L-tau in the DRN of mice and observed depressive-like behaviors and hyperactivity without deficits in spatial memory. Tau pathology was predominantly found in neurons relative to glia and colocalized with a significant proportion of Tph2-expressing neurons in the DRN. 5-HT neurons were also hyperexcitable in P301L-tauDRN mice, and there was an increase in the amplitude of excitatory post-synaptic currents (EPSCs). Moreover, astrocytic density was elevated in the DRN and accompanied by an increase in IL-1α and Frk expression, which suggests increased inflammatory signaling. Additionally, tau pathology was detected in axonal processes in the thalamus, hypothalamus, amygdala, and caudate putamen. A significant proportion of this tau pathology colocalized with the serotonin reuptake transporter (SERT), suggesting that tau may spread in an anterograde manner to regions outside the DRN. Together these results indicate that tau pathology accumulates in the DRN in a subset of individuals over 50 years and may lead to behavioral dysregulation, 5-HT neuronal dysfunction, and activation of local astrocytes which may be prodromal indicators of AD.
Collapse
Affiliation(s)
- Samantha R Pierson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kimberly L Fiock
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Ruixiang Wang
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Jessica Reinhardt
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kanza M Khan
- Psychological Sciences Department, Daemen University, Amherst, NY, 14226, USA
| | - Thomas D James
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Mikayla L Hunter
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Benjamin J Cooper
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | | | - Ryan Betters
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kaancan Deniz
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
| | - Gloria Lee
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Georgina Aldridge
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Marco M Hefti
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA
| | - Catherine A Marcinkiewcz
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Alexander C, Jeon J, Nickerson K, Hassler S, Vasefi M. CBD and the 5-HT1A receptor: A medicinal and pharmacological review. Biochem Pharmacol 2025; 233:116742. [PMID: 39778776 DOI: 10.1016/j.bcp.2025.116742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/25/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Cannabidiol (CBD), a phytocannabinoid, has emerged as a promising candidate for addressing a wide array of symptoms. It has the ability to bind to multiple proteins and receptors, including 5-HT1AR, transient receptor potential vanilloid 1 (TRPV1), and cannabinoid receptors. However, CBD's pharmacodynamic interaction with 5-HT1AR and its medicinal outcomes are still debated. This review explores recent literature to elucidate these questions, highlighting the neurotherapeutic outcomes of this pharmacodynamic interaction and proposing a signaling pathway underlying the mechanism by which CBD desensitizes 5-HT1AR signaling. A comprehensive survey of the literature underscores CBD's multifaceted neurotherapeutic effects, which include antidepressant, anxiolytic, neuroprotective, antipsychotic, antiemetic, anti-allodynic, anti-epileptic, anti-degenerative, and addiction-treating properties, attributable in part to its interactions with 5-HT1AR. Furthermore, evidence suggests that the pharmacodynamic interaction between CBD and 5-HT1AR is contingent upon dosage. Moreover, we propose that CBD can induce desensitization of 5-HT1AR via both homologous and heterologous mechanisms. Homologous desensitization involves the recruitment of G protein-coupled receptor kinase 2 (GRK2) and β-arrestin, leading to receptor endocytosis. In contrast, heterologous desensitization is mediated by an elevated intracellular calcium level or activation of protein kinases, such as c-Jun N-terminal kinase (JNK), through the activity of other receptors.
Collapse
Affiliation(s)
- Claire Alexander
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA
| | - Jiyoon Jeon
- Department of Biology, Lamar University, Beaumont, TX, 77710, USA
| | - Kyle Nickerson
- Department of Biology, Baylor University, Waco, TX, 76706, USA
| | - Shayne Hassler
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houton, Houston, TX, 77204, USA
| | - Maryam Vasefi
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houton, Houston, TX, 77204, USA.
| |
Collapse
|
12
|
Kolling LJ, Chimenti MS, Marcinkiewcz CA. Spatial differences in gene expression across the dorsal raphe nucleus in a model of early Alzheimer's disease. J Alzheimers Dis 2025; 103:133-148. [PMID: 39584353 DOI: 10.1177/13872877241299119] [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: 11/26/2024]
Abstract
BACKGROUND Persons with Alzheimer's disease (AD) present with changes in mood, sleep, and arousal that may precede the clinical manifestation of cognitive decline. These early symptoms can be driven by changes in the serotonergic (5-HT) nuclei of the brainstem, particularly the dorsal raphe nucleus (DRN). It is unclear why all 5-HT neurons do not simultaneously develop AD pathology that progresses at the same rate. OBJECTIVE We sought to identify any underlying genetic components associated with susceptibility or resistance of 5-HT neurons to AD pathology. METHODS The Visium Spatial Gene Expression platform was used to identify transcriptomic changes across the DRN in a preclinical model of early AD, human tau-overexpressing mice (htau mice). We further used RNAscope and immunohistochemical assessment to validate findings of primary interest. RESULTS We find that the DRN of htau mice differentially expresses AD-related genes, including those related to kinase binding, ion channel activity, ligand-receptor interactions, and regulation of serine/threonine kinases. We further find that computational sub-clustering of the DRN is consistent with previous circuitry-driven characterizations, allowing for spatial bounding of distinct subregions within the DRN. Of these, we find the dorsolateral DRN is preferentially impacted by 5-HT neuron loss and development of tau pathology, which coincides with increased expression of the long noncoding RNA Map2k3os. CONCLUSIONS Map2k3os may serve regulatory roles relevant for tau phosphorylation and warrants further investigation to characterize its interactions. Overall, this report demonstrates the power of large-scale spatial transcriptomics technologies, while underscoring the need for convergent-data validation to overcome their limitations.
Collapse
Affiliation(s)
- Louis J Kolling
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Michael S Chimenti
- Iowa Institute of Human Genetics Bioinformatics Division, University of Iowa, IA City, IA, USA
| | | |
Collapse
|
13
|
Sun X, Liu F, Liu H, Guo L, Ma H, Zhu J, Qian Y. Molecular mechanisms and behavioral relevance underlying neural correlates of childhood neglect. J Affect Disord 2024; 367:795-805. [PMID: 39255872 DOI: 10.1016/j.jad.2024.09.020] [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: 05/08/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Childhood neglect is associated with brain changes, yet the molecular mechanisms and behavioral relevance underlying such associations remain elusive. METHODS We calculated fractional amplitude of low-frequency fluctuations (fALFF) using resting-state functional MRI and tested their correlation with childhood neglect across a large sample of 510 healthy young adults. Then, we investigated the spatial relationships of the identified neural correlates of childhood neglect with gene expression, neurotransmitter, and behavioral domain atlases. RESULTS We found that more severe childhood neglect was correlated with higher fALFF in the bilateral anterior cingulate cortex. Remarkably, the identified neural correlates of childhood neglect were spatially correlated with expression of gene categories primarily involving neuron, synapse, ion channel, cognitive and perceptual processes, and physiological response and regulation. Concurrently, there were significant associations between the neural correlates and specific neurotransmitter systems including serotonin and GABA. Finally, neural correlates of childhood neglect were associated with diverse behavioral domains implicating mental disorders, emotion, cognition, and sensory perception. LIMITATIONS The cross-sectional study design cannot unequivocally establish causality. CONCLUSIONS Our findings may not only add to the current knowledge regarding the relationship between childhood neglect and mental health, but also have clinical implications for developing preventive strategies for individuals exposed to childhood neglect who are at risk for mental disorders.
Collapse
Affiliation(s)
- Xuetian Sun
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Fujun Liu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Hu Liu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Lixin Guo
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Haining Ma
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China.
| | - Yinfeng Qian
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, China.
| |
Collapse
|
14
|
Hussein MN. Labeling of the serotonergic neuronal circuits emerging from the raphe nuclei via some retrograde tracers. Microsc Res Tech 2024; 87:2894-2914. [PMID: 39041701 DOI: 10.1002/jemt.24662] [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: 02/13/2024] [Revised: 06/20/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a very important neurotransmitter emerging from the raphe nuclei to several brain regions. Serotonergic neuronal connectivity has multiple functions in the brain. In this study, several techniques were used to trace serotonergic neurons in the dorsal raphe (DR) and median raphe (MnR) that project toward the arcuate nucleus of the hypothalamus (Arc), dorsomedial hypothalamic nucleus (DM), lateral hypothalamic area (LH), paraventricular hypothalamic nucleus (PVH), ventromedial hypothalamic nucleus (VMH), fasciola cinereum (FC), and medial habenular nucleus (MHb). Cholera toxin subunit B (CTB), retro-adeno-associated virus (rAAV-CMV-mCherry), glycoprotein-deleted rabies virus (RV-ΔG), and simultaneous microinjection of rAAV2-retro-Cre-tagBFP with AAV-dio-mCherry in C57BL/6 mice were used in this study. In addition, rAAV2-retro-Cre-tagBFP was microinjected into Ai9 mice. Serotonin immunohistochemistry was used for the detection of retrogradely traced serotonergic neurons in the raphe nuclei. The results indicated that rAAV2-retro-Cre-tagBFP microinjection in Ai9 mice was the best method for tracing serotonergic neuron circuits. All of the previously listed nuclei exhibited serotonergic neuronal projections from the DR and MnR, with the exception of the FC, which had very few projections from the DR. The serotonergic neuronal projections were directed toward the Arc by the subpeduncular tegmental (SPTg) nuclei. Moreover, the RV-ΔG tracer revealed monosynaptic non-serotonergic neuronal projections from the DR that were directed toward the Arc. Furthermore, rAAV tracers revealed monosynaptic serotonergic neuronal connections from the raphe nuclei toward Arc. These findings validate the variations in neurotropism among several retrograde tracers. The continued discovery of several novel serotonergic neural circuits is crucial for the future discovery of the functions of these circuits. RESEARCH HIGHLIGHTS: Various kinds of retrograde tracers were microinjected into C57BL/6 and Ai9 mice. The optimum method for characterizing serotonergic neuronal circuits is rAAV2-retro-Cre-tagBFP microinjection in Ai9 mice. The DR, MnR, and SPTg nuclei send monosynaptic serotonergic neuronal projections toward the arcuate nucleus of the hypothalamus. Whole-brain quantification analysis of retrograde-labeled neurons in different brain nuclei following rAAV2-retro-Cre-tagBFP microinjection in the Arc, DM, LH, and VMH is shown. Differential quantitative analysis of median and dorsal raphe serotonergic neurons emerging toward the PVH, DM, LH, Arc, VMH, MHb, and FC is shown.
Collapse
Affiliation(s)
- Mona N Hussein
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Histology and Cytology Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| |
Collapse
|
15
|
Camargo A, Nilsson A, Shariatgorji R, Appleton E, Branzell N, Doyon D, Giovenzana M, Zhang X, Dautan D, Andren PE, Svenningsson P. Enduring modulation of dorsal raphe nuclei regulates (R,S)-ketamine-mediated resilient stress-coping behavior. Mol Psychiatry 2024:10.1038/s41380-024-02853-6. [PMID: 39592824 DOI: 10.1038/s41380-024-02853-6] [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: 04/21/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
Ketamine may be a novel pharmacologic approach to enhance resilience and protect against stress-related disorders, but the molecular targets underlying this response remain to be fully characterized. The multifunctional protein p11 is crucial in the pathophysiology of depression and antidepressant responses. However, it is still unclear whether p11 plays a role in the pro-resilience effects induced by ketamine. Here, we demonstrated that prophylactic administration of ketamine buffers passive stress-induced maladaptive phenotypes induced by chronic stress exposure. Spatial neurotransmitter and metabolite analysis revealed that prophylactic ketamine was also effective in blunting stress-induced disturbances of tryptophan metabolism in dorsal raphe nuclei (DRN). Additionally, we demonstrated that ketamine prevented chronic restraint stress-induced p11 reduction in DRN, a highly p11-enriched region. Furthermore, we provide novel evidence indicating that p11 deficiency regulates susceptibility to stress-induced depression-related phenotypes, and these behavioral maladaptations are dependent, at least in part, on p11 function in serotonergic neurons. Spatial neurotransmitter and metabolite analysis also showed a reduction of tryptophan and dopamine metabolism in DRN of serotonergic p11-deficient mice. Viral-mediated downregulation of p11 within DRN induced a stress-susceptible phenotype. Finally, our results also unveiled that the ability of ketamine to elicit a pro-resilience response against stress-induced maladaptive phenotypes was occluded when p11 was selectively deleted in serotonergic neurons. Altogether, we showed a previously unexplored role of the DRN circuit in regulating stress susceptibility and resilience-enhancing actions of ketamine.
Collapse
Affiliation(s)
- Anderson Camargo
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Anna Nilsson
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Reza Shariatgorji
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ellen Appleton
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Niclas Branzell
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Doyon
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mattia Giovenzana
- Department of Medicine and Surgery, University of Milano Bicocca Monza, Monza, Italy
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Dautan
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Per E Andren
- Department of Pharmaceutical Biosciences, Spatial Mass Spectrometry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
16
|
Cardozo Pinto DF, Pomrenze MB, Guo MY, Touponse GC, Chen APF, Bentzley BS, Eshel N, Malenka RC. Opponent control of reinforcement by striatal dopamine and serotonin. Nature 2024:10.1038/s41586-024-08412-x. [PMID: 39586475 DOI: 10.1038/s41586-024-08412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
The neuromodulators dopamine (DA) and serotonin (5-hydroxytryptamine; 5HT) powerfully regulate associative learning1-8. Similarities in the activity and connectivity of these neuromodulatory systems have inspired competing models of how DA and 5HT interact to drive the formation of new associations9-14. However, these hypotheses have not been tested directly because it has not been possible to interrogate and manipulate multiple neuromodulatory systems in a single subject. Here we establish a mouse model that enables simultaneous genetic access to the brain's DA and 5HT neurons. Anterograde tracing revealed the nucleus accumbens (NAc) to be a putative hotspot for the integration of convergent DA and 5HT signals. Simultaneous recording of DA and 5HT axon activity, together with genetically encoded DA and 5HT sensor recordings, revealed that rewards increase DA signalling and decrease 5HT signalling in the NAc. Optogenetically dampening DA or 5HT reward responses individually produced modest behavioural deficits in an appetitive conditioning task, while blunting both signals together profoundly disrupted learning and reinforcement. Optogenetically reproducing DA and 5HT reward responses together was sufficient to drive the acquisition of new associations and supported reinforcement more potently than either manipulation did alone. Together, these results demonstrate that striatal DA and 5HT signals shape learning by exerting opponent control of reinforcement.
Collapse
Affiliation(s)
- Daniel F Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew B Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Michaela Y Guo
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Gavin C Touponse
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Allen P F Chen
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Neir Eshel
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
17
|
Ren L, Fan Y, Wu W, Qian Y, He M, Li X, Wang Y, Yang Y, Wen X, Zhang R, Li C, Chen X, Hu J. Anxiety disorders: Treatments, models, and circuitry mechanisms. Eur J Pharmacol 2024; 983:176994. [PMID: 39271040 DOI: 10.1016/j.ejphar.2024.176994] [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: 06/09/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
Anxiety disorders are one of the most prevalent mental health conditions worldwide, imposing a significant burden on individuals affected by them and society in general. Current research endeavors aim to enhance the effectiveness of existing anxiolytic drugs and reduce their side effects through optimization or the development of new treatments. Several anxiolytic novel drugs have been produced as a result of discovery-focused research. However, many drug candidates that show promise in preclinical rodent model studies fail to offer any substantive clinical benefits to patients. This review provides an overview of the diagnosis and classification of anxiety disorders together with a systematic review of anxiolytic drugs with a focus on their targets, therapeutic applications, and side effects. It also provides a concise overview of the constraints and disadvantages associated with frequently administered anxiolytic drugs. Additionally, the study comprehensively reviews animal models used in anxiety studies and their associated molecular mechanisms, while also summarizing the brain circuitry related to anxiety. In conclusion, this article provides a valuable foundation for future anxiolytic drug discovery efforts.
Collapse
Affiliation(s)
- Li Ren
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China.
| | - Yue Fan
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Wenjian Wu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Yuanxin Qian
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Miao He
- College of Life Sciences and Medicine, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Xinlong Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Yizhu Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Yu Yang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Xuetong Wen
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Ruijia Zhang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Chenhang Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Xin Chen
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, 611137, China
| | - Jingqing Hu
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
18
|
Schmitz N, Hodzic S, Riedemann T. Common and contrasting effects of 5-HTergic signaling in pyramidal cells and SOM interneurons of the mouse cortex. Neuropsychopharmacology 2024:10.1038/s41386-024-02022-x. [PMID: 39511335 DOI: 10.1038/s41386-024-02022-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/03/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a powerful modulator of neuronal activity within the central nervous system and dysfunctions of the serotonergic system have been linked to several neuropsychiatric disorders such as major depressive disorders or schizophrenia. The anterior cingulate cortex (aCC) plays an important role in cognitive capture of stimuli and valence processing and it is densely innervated by serotonergic fibers from the nucleus raphe. In order to understand how pathophysiological 5-HT signalling can lead to neuropsychiatric diseases, it is important to understand the physiological actions of 5-HT on cortical circuits. Therefore, we combined electrophysiological recordings with pharmacology and immunocytochemistry to investigate the effects of 5-HT on Somatostatin-positive interneurons (SOM-INs) and compared these to supragranular pyramidal cells (PCs). This comparison allowed us to identify common and contrasting effects of 5-HT on SOM-INs and PCs of the aCC resulting in a specific modulation of the excitation-to-inhibition balance in PCs but not in SOM-INs.
Collapse
Affiliation(s)
- Nathalie Schmitz
- Department of Physiological Genomics, Institute of Physiology, Biomedical Center, Ludwig-Maximilians-Universität München, 82152, Planegg-Martinsried, Germany
| | - Sadat Hodzic
- Department of Physiological Genomics, Institute of Physiology, Biomedical Center, Ludwig-Maximilians-Universität München, 82152, Planegg-Martinsried, Germany
| | - Therese Riedemann
- Department of Physiological Genomics, Institute of Physiology, Biomedical Center, Ludwig-Maximilians-Universität München, 82152, Planegg-Martinsried, Germany.
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology, Paracelsus Medical University, Strubergasse 22, 5020, Salzburg, Austria.
| |
Collapse
|
19
|
Pomrenze MB, Vaillancourt S, Salgado JS, Raymond KB, Llorach P, Touponse GC, Cardozo Pinto DF, Rastegar Z, Casey AB, Eshel N, Malenka RC, Heifets BD. 5-HT 2C receptors in the nucleus accumbens constrain the rewarding effects of MDMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.20.619256. [PMID: 39484424 PMCID: PMC11527024 DOI: 10.1101/2024.10.20.619256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
MDMA is a promising adjunct to psychotherapy and has well-known abuse liability, although less than other amphetamine analogs. While the reinforcing dopamine (DA)-releasing properties of MDMA are on par with methamphetamine (METH), MDMA is a far more potent serotonin (5-HT) releaser, via the 5-HT transporter (SERT). MDMA-mediated 5-HT release in a major reward center, the nucleus accumbens (NAc), drives prosocial behaviors via 5-HT1BR activation. We hypothesized that this prosocial mechanism contributes to the reduced reinforcing properties of MDMA compared to METH and used a platform of assays to predict the balance of prosocial and abuse-linked effects of (R)-MDMA, a novel entactogen in clinical development. NAc DA release, measured by GRAB-DA photometry in vivo, increased in proportion to MDMA (7.5 and 15 mg/kg, i.p.) and METH (2 mg/kg i.p.)-conditioned place preference (CPP). Using conditional knockouts (cKOs) for DAT and SERT, microdialysis, and photometry, we found that MDMA-released 5-HT limited MDMA-released DA through actions in the NAc, rather than at ventral tegmental area DAergic cell bodies. SERT cKO reduced the MDMA dose required for CPP three-fold. This enhanced MDMA-CPP and increased DA release were replicated by intra-NAc infusion of either a 5-HT reuptake inhibitor (escitalopram) to prevent MDMA interaction with SERT, or a 5-HT2CR antagonist (SB242084), but not by the 5-HT1BR antagonist NAS-181. These data support separate mechanisms for the low abuse potential versus prosocial effect of MDMA. Using this platform of assays, (R)-MDMA is predicted to have prosocial effects and low abuse potential.
Collapse
Affiliation(s)
- Matthew B. Pomrenze
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Sam Vaillancourt
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Juliana S. Salgado
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Kendall B. Raymond
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Pierre Llorach
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Gavin C. Touponse
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Daniel F. Cardozo Pinto
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Zahra Rastegar
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Austen B. Casey
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Neir Eshel
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Robert C. Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
| | - Boris D. Heifets
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
20
|
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
|
21
|
Song SY. The cerebrospinal fluid (CSF)-contacting raphe nucleus (CsfR) in mice. Neurosci Lett 2024; 841:137969. [PMID: 39236800 DOI: 10.1016/j.neulet.2024.137969] [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: 08/07/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
A unique nucleus, the cerebrospinal fluid-contacting nucleus (CsfR), has been identified in the brain parenchyma. This nucleus features neurons with somas located within the parenchyma and processes extending into the cerebrospinal fluid (CSF). This anatomical configuration suggests that the CsfR may serve as a crucial interface between the nervous and body fluid regulatory systems, potentially playing a significant role in overall physiological modulation. Despite its importance, the precise biological significance of the CsfR remains to be fully elucidated. Previous research has characterized the CsfR, providing detailed information on its position, neighboring structures, neuron distribution, and 3D reconstruction in both rats and non-human primates, with stereotaxic coordinates specifically provided for the rat model. Given the relevance of mice as a model organism, especially the C57BL/6J strain, this study aims to explore the existence and morphology of the CsfR in mice. Our findings confirm the presence of the CsfR, consistently located in the ventral gray area of the lower part of the aqueduct and the upper part of the fourth ventricle floor. It is bilaterally symmetrical and heart-shaped in the coronal plane, which differs slightly from the Y-shape observed in coronal sections of rats. This study provides significant references for researchers investigating this specialized nucleus.
Collapse
Affiliation(s)
- Si-Yuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
22
|
Spring MG, Nautiyal KM. Striatal Serotonin Release Signals Reward Value. J Neurosci 2024; 44:e0602242024. [PMID: 39117457 PMCID: PMC11466065 DOI: 10.1523/jneurosci.0602-24.2024] [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: 03/25/2024] [Revised: 07/02/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Serotonin modulates diverse phenotypes and functions including depressive, aggressive, impulsive, and feeding behaviors, all of which have reward-related components. To date, research has focused on understanding these effects by measuring and manipulating dorsal raphe serotonin neurons and using single-receptor approaches. These studies have led to a better understanding of the heterogeneity of serotonin actions on behavior; however, they leave open many questions about the timing and location of serotonin's actions modulating the neural circuits that drive these behaviors. Recent advances in genetically encoded fluorescent biosensors, including the GPCR activation-based sensor for serotonin (GRAB-5-HT), enable the measurement of serotonin release in mice on a timescale compatible with a single rewarding event without corelease confounds. Given substantial evidence from slice electrophysiology experiments showing that serotonin influences neural activity of the striatal circuitry, and the known role of the dorsal medial striatal (DMS) in reward-directed behavior, we focused on understanding the parameters and timing that govern serotonin release in the DMS in the context of reward consumption, external reward value, internal state, and cued reward. Overall, we found that serotonin release is associated with each of these and encodes reward anticipation, value, approach, and consumption in the DMS.
Collapse
Affiliation(s)
- Mitchell G Spring
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Katherine M Nautiyal
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, New Hampshire 03755
| |
Collapse
|
23
|
Schoofs A, Miroschnikow A, Schlegel P, Zinke I, Schneider-Mizell CM, Cardona A, Pankratz MJ. Serotonergic modulation of swallowing in a complete fly vagus nerve connectome. Curr Biol 2024; 34:4495-4512.e6. [PMID: 39270641 PMCID: PMC7616834 DOI: 10.1016/j.cub.2024.08.025] [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: 02/06/2024] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
How the body interacts with the brain to perform vital life functions, such as feeding, is a fundamental issue in physiology and neuroscience. Here, we use a whole-animal scanning transmission electron microscopy volume of Drosophila to map the neuronal circuits that connect the entire enteric nervous system to the brain via the insect vagus nerve at synaptic resolution. We identify a gut-brain feedback loop in which Piezo-expressing mechanosensory neurons in the esophagus convey food passage information to a cluster of six serotonergic neurons in the brain. Together with information on food value, these central serotonergic neurons enhance the activity of serotonin receptor 7-expressing motor neurons that drive swallowing. This elemental circuit architecture includes an axo-axonic synaptic connection from the glutamatergic motor neurons innervating the esophageal muscles onto the mechanosensory neurons that signal to the serotonergic neurons. Our analysis elucidates a neuromodulatory sensory-motor system in which ongoing motor activity is strengthened through serotonin upon completion of a biologically meaningful action, and it may represent an ancient form of motor learning.
Collapse
Affiliation(s)
- Andreas Schoofs
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | - Anton Miroschnikow
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | - Philipp Schlegel
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 TN1, UK; MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK
| | - Ingo Zinke
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany
| | | | - Albert Cardona
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Trumpington, Cambridge CB2 0QH, UK; Janelia Research Campus, Howard Hughes Medical Institute, Helix Drive, Ashburn, VA 20147, USA; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Place, Cambridge CB2 3EL, UK
| | - Michael J Pankratz
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Straße, Bonn 53115, Germany.
| |
Collapse
|
24
|
Phalip A, Netser S, Wagner S. Understanding the neurobiology of social behavior through exploring brain-wide dynamics of neural activity. Neurosci Biobehav Rev 2024; 165:105856. [PMID: 39159735 DOI: 10.1016/j.neubiorev.2024.105856] [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: 05/10/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Social behavior is highly complex and adaptable. It can be divided into multiple temporal stages: detection, approach, and consummatory behavior. Each stage can be further divided into several cognitive and behavioral processes, such as perceiving social cues, evaluating the social and non-social contexts, and recognizing the internal/emotional state of others. Recent studies have identified numerous brain-wide circuits implicated in social behavior and suggested the existence of partially overlapping functional brain networks underlying various types of social and non-social behavior. However, understanding the brain-wide dynamics underlying social behavior remains challenging, and several brain-scale dynamics (macro-, meso-, and micro-scale levels) need to be integrated. Here, we suggest leveraging new tools and concepts to explore social brain networks and integrate those different levels. These include studying the expression of immediate-early genes throughout the entire brain to impartially define the structure of the neuronal networks involved in a given social behavior. Then, network dynamics could be investigated using electrode arrays or multi-channel fiber photometry. Finally, tools like high-density silicon probes and miniscopes can probe neural activity in specific areas and across neuronal populations at the single-cell level.
Collapse
Affiliation(s)
- Adèle Phalip
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| | - Shai Netser
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Shlomo Wagner
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
25
|
Yu CC, Wang XF, Wang J, Li C, Xiao J, Wang XS, Han R, Wang SQ, Lin YF, Kong LH, Du YJ. Electroacupuncture Alleviates Memory Deficits in APP/PS1 Mice by Targeting Serotonergic Neurons in Dorsal Raphe Nucleus. Curr Med Sci 2024; 44:987-1000. [PMID: 38990450 DOI: 10.1007/s11596-024-2908-9] [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: 05/01/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE Alzheimer's disease (AD) has become a significant global concern, but effective drugs able to slow down AD progression is still lacked. Electroacupuncture (EA) has been demonstrated to ameliorate cognitive impairment in individuals with AD. However, the underlying mechanisms remains poorly understood. This study aimed at examining the neuroprotective properties of EA and its potential mechanism of action against AD. METHODS APP/PS1 transgenic mice were employed to evaluate the protective effects of EA on Shenshu (BL 23) and Baihui (GV 20). Chemogenetic manipulation was used to activate or inhibit serotonergic neurons within the dorsal raphe nucleus (DRN). Learning and memory abilities were assessed by the novel object recognition and Morris water maze tests. Golgi staining, western blot, and immunostaining were utilized to determine EA-induced neuroprotection. RESULTS EA at Shenshu (BL 23) and Baihui (GV 20) effectively ameliorated learning and memory impairments in APP/PS1 mice. EA attenuated dendritic spine loss, increased the expression levels of PSD95, synaptophysin, and brain-derived neurotrophic factor in hippocampus. Activation of serotonergic neurons within the DRN can ameliorate cognitive deficits in AD by activating glutamatergic neurons mediated by 5-HT1B. Chemogenetic inhibition of serotonergic neurons in the DRN reversed the effects of EA on synaptic plasticity and memory. CONCLUSION EA can alleviate cognitive dysfunction in APP/PS1 mice by activating serotonergic neurons in the DRN. Further study is necessary to better understand how the serotonergic neurons-related neural circuits involves in EA-induced memory improvement in AD.
Collapse
Affiliation(s)
- Chao-Chao Yu
- Department of Rehabilitation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Xiao-Fei Wang
- Department of Rehabilitation, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430074, China
| | - Jia Wang
- Department of Acupuncture and Moxibustion, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, 430030, China
| | - Chu Li
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Juan Xiao
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xue-Song Wang
- College of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, 050299, China
| | - Rui Han
- Department of Child Rehabilitation Medicine, Qujing Hospital of Maternity and Childcare, Qujing, 655002, China
| | - Shu-Qin Wang
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Yuan-Fang Lin
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Li-Hong Kong
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
26
|
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
|
27
|
Zou J, Chen H, Chen X, Lin Z, Yang Q, Tie C, Wang H, Niu L, Guo Y, Zheng H. Noninvasive closed-loop acoustic brain-computer interface for seizure control. Theranostics 2024; 14:5965-5981. [PMID: 39346532 PMCID: PMC11426232 DOI: 10.7150/thno.99820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/03/2024] [Indexed: 10/01/2024] Open
Abstract
Rationale: The brain-computer interface (BCI) is core tasks in comprehensively understanding the brain, and is one of the most significant challenges in neuroscience. The development of novel non-invasive neuromodulation technique will drive major innovations and breakthroughs in the field of BCI. Methods: We develop a new noninvasive closed-loop acoustic brain-computer interface (aBCI) for decoding the seizure onset based on the electroencephalography and triggering ultrasound stimulation of the vagus nerve to terminate seizures. Firstly, we create the aBCI system and decode the onset of seizure via a multi-level threshold model based on the analysis of wireless-collected electroencephalogram (EEG) signals recorded from above the hippocampus. Then, the different acoustic parameters induced acoustic radiation force were used to stimulate the vagus nerve in a rat model of epilepsy-induced by pentylenetetrazole. Finally, the results of epileptic EEG signal triggering ultrasound stimulation of the vagus nerve to control seizures. In addition, the mechanism of aBCI control seizures were investigated by real-time quantitative polymerase chain reaction (RT-qPCR). Results: In a rat model of epilepsy, the aBCI system selectively actives mechanosensitive neurons in the nodose ganglion while suppressing neuronal excitability in the hippocampus and amygdala, and stops seizures rapidly upon ultrasound stimulation of the vagus nerve. Physical transection or chemical blockade of the vagus nerve pathway abolish the antiepileptic effects of aBCI. In addition, aBCI shows significant antiepileptic effects compared to conventional vagus nerve electrical stimulation in an acute experiment. Conclusions: Closed-loop aBCI provides a novel, safe and effective tool for on-demand stimulation to treat abnormal neuronal discharges, opening the door to next generation non-invasive BCI.
Collapse
Affiliation(s)
- Junjie Zou
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Houminji Chen
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoyan Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhengrong Lin
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qihang Yang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Changjun Tie
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hong Wang
- The Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lili Niu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yanwu Guo
- The National Key Clinic Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hairong Zheng
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Fu Y, Cheng HW. The Influence of Cecal Microbiota Transplantation on Chicken Injurious Behavior: Perspective in Human Neuropsychiatric Research. Biomolecules 2024; 14:1017. [PMID: 39199404 PMCID: PMC11352350 DOI: 10.3390/biom14081017] [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: 06/18/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Numerous studies have evidenced that neuropsychiatric disorders (mental illness and emotional disturbances) with aggression (or violence) pose a significant challenge to public health and contribute to a substantial economic burden worldwide. Especially, social disorganization (or social inequality) associated with childhood adversity has long-lasting effects on mental health, increasing the risk of developing neuropsychiatric disorders. Intestinal bacteria, functionally as an endocrine organ and a second brain, release various immunomodulators and bioactive compounds directly or indirectly regulating a host's physiological and behavioral homeostasis. Under various social challenges, stress-induced dysbiosis increases gut permeability causes serial reactions: releasing neurotoxic compounds, leading to neuroinflammation and neuronal injury, and eventually neuropsychiatric disorders associated with aggressive, violent, or impulsive behavior in humans and various animals via a complex bidirectional communication of the microbiota-gut-brain (MGB) axis. The dysregulation of the MGB axis has also been recognized as one of the reasons for the prevalence of social stress-induced injurious behaviors (feather pecking, aggression, and cannibalistic pecking) in chickens. However, existing knowledge of preventing and treating these disorders in both humans and chickens is not well understood. In previous studies, we developed a non-mammal model in an abnormal behavioral investigation by rationalizing the effects of gut microbiota on injurious behaviors in chickens. Based on our earlier success, the perspective article outlines the possibility of reducing stress-induced injurious behaviors in chickens through modifying gut microbiota via cecal microbiota transplantation, with the potential for providing a biotherapeutic rationale for preventing injurious behaviors among individuals with mental disorders via restoring gut microbiota diversity and function.
Collapse
Affiliation(s)
- Yuechi Fu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA;
| | - Heng-Wei Cheng
- Livestock Behavior Research Unit, USDA-ARS, West Lafayette, IN 47907, USA
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Yan C, Liu Z. The role of periaqueductal gray astrocytes in anxiety-like behavior induced by acute stress. Biochem Biophys Res Commun 2024; 720:150073. [PMID: 38754161 DOI: 10.1016/j.bbrc.2024.150073] [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: 12/10/2023] [Revised: 04/19/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
Astrocytes in the central nervous system play a vital role in modulating synaptic transmission and neuronal activation by releasing gliotransmitters. The 5-HTergic neurons in the ventrolateral periaqueductal gray (vlPAG) are important in anxiety processing. However, it remains uncertain whether the regulation of astrocytic activity on vlPAG 5-HTergic neurons is involved in anxiety processing. Here, through chemogenetic manipulation, we explored the impact of astrocytic activity in the PAG on the regulation of anxiety. To determine the role of astrocytes in the control of anxiety, we induced anxiety-like behaviors in mice through foot shock and investigated their effects on synaptic transmission and neuronal excitability in vlPAG 5-HTergic neurons. Foot shock caused anxiety-like behaviors, which were accompanied with the increase of the amplitude and frequency of miniature excitatory postsynaptic currents (mEPSCs), the area of slow inward currents (SICs), and the spike frequency of action potentials (AP) in vlPAG 5-HTergic neurons. The chemogenetic inhibition of vlPAG astrocytes was found to attenuate stress-induced anxiety-like behaviors and decrease the heightened synaptic transmission and neuronal excitability of vlPAG 5-HTergic neurons. Conversely, chemogenetic activation of vlPAG astrocytes triggered anxiety-like behaviors, enhanced synaptic transmission, and increased the excitability of vlPAG 5-HTergic neurons in unstressed mice. In summary, this study has provided initial insights into the pathway by which astrocytes influence behavior through the rapid regulation of associated neurons. This offers a new perspective for the investigation of the biological mechanisms underlying anxiety.
Collapse
Affiliation(s)
- Chuanting Yan
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, 199 Chang'an South Road, Xi'an, 710062, China; Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, 555 Qiangye Road, Shanghai, 201210, China
| | - Zhiqiang Liu
- MOE Key Laboratory of Modern Teaching Technology, Shaanxi Normal University, 199 Chang'an South Road, Xi'an, 710062, China.
| |
Collapse
|
34
|
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
|
35
|
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
|
36
|
Liu X, Li H, Ma R, Tong X, Wu J, Huang X, So K, Tao Q, Huang L, Lin S, Ren C. Burst firing in Output-Defined Parallel Habenula Circuit Underlies the Antidepressant Effects of Bright Light Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401059. [PMID: 38863324 PMCID: PMC11321664 DOI: 10.1002/advs.202401059] [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: 01/29/2024] [Revised: 05/11/2024] [Indexed: 06/13/2024]
Abstract
Research highlights the significance of increased bursting in lateral habenula (LHb) neurons in depression and as a focal point for bright light treatment (BLT). However, the precise spike patterns of LHb neurons projecting to different brain regions during depression, their roles in depression development, and BLT's therapeutic action remain elusive. Here, LHb neurons are found projecting to the dorsal raphe nucleus (DRN), ventral tegmental area (VTA), and median raphe nucleus (MnR) exhibit increased bursting following aversive stimuli exposure, correlating with distinct depressive symptoms. Enhanced bursting in DRN-projecting LHb neurons is pivotal for anhedonia and anxiety, while concurrent bursting in LHb neurons projecting to the DRN, VTA, and MnR is essential for despair. Remarkably, reducing bursting in distinct LHb neuron subpopulations underlies the therapeutic effects of BLT on specific depressive behaviors. These findings provide valuable insights into the mechanisms of depression and the antidepressant action of BLT.
Collapse
Affiliation(s)
- Xianwei Liu
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Han Li
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Ruijia Ma
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Xiaohan Tong
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Jijin Wu
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Xiaodan Huang
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Kwok‐Fai So
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdao266113China
| | - Qian Tao
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdao266113China
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Jinan UniversityPsychology DepartmentSchool of MedicineJinan UniversityGuangzhou510632China
| | - Lu Huang
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Song Lin
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Chaoran Ren
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdao266113China
| |
Collapse
|
37
|
Liu H, Qu N, Gonzalez NV, Palma MA, Chen H, Xiong J, Choubey A, Li Y, Li X, Yu M, Liu H, Tu L, Zhang N, Yin N, Conde KM, Wang M, Bean JC, Han J, Scarcelli NA, Yang Y, Saito K, Cui H, Tong Q, Sun Z, Wang C, Cai X, Lu L, He Y, Xu Y. A Light-Responsive Neural Circuit Suppresses Feeding. J Neurosci 2024; 44:e2192232024. [PMID: 38897723 PMCID: PMC11270527 DOI: 10.1523/jneurosci.2192-23.2024] [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: 11/23/2023] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Light plays an essential role in a variety of physiological processes, including vision, mood, and glucose homeostasis. However, the intricate relationship between light and an animal's feeding behavior has remained elusive. Here, we found that light exposure suppresses food intake, whereas darkness amplifies it in male mice. Interestingly, this phenomenon extends its reach to diurnal male Nile grass rats and healthy humans. We further show that lateral habenula (LHb) neurons in mice respond to light exposure, which in turn activates 5-HT neurons in the dorsal Raphe nucleus (DRN). Activation of the LHb→5-HTDRN circuit in mice blunts darkness-induced hyperphagia, while inhibition of the circuit prevents light-induced anorexia. Together, we discovered a light-responsive neural circuit that relays the environmental light signals to regulate feeding behavior in mice.
Collapse
Affiliation(s)
- Hailan Liu
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030 .
| | - Na Qu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Wuhan 430012, China .
- Wuhan Hospital for Psychotherapy, Wuhan 430012, China
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430012, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan 430012, China
- Affiliated Wuhan Mental Health Center, Jianghan University, Wuhan 430012, China
| | | | - Marco A Palma
- Human Behavior Laboratory, Texas A&M University, College Station, Texas 77843
| | - Huamin Chen
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Wuhan 430012, China
- Wuhan Hospital for Psychotherapy, Wuhan 430012, China
- Affiliated Wuhan Mental Health Center, Jianghan University, Wuhan 430012, China
| | - Jiani Xiong
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health Center, Wuhan 430012, China
- Wuhan Hospital for Psychotherapy, Wuhan 430012, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan 430012, China
| | - Abhinav Choubey
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, Texas 77030
| | - Yongxiang Li
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Xin Li
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, Texas 77030
| | - Meng Yu
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Hesong Liu
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Longlong Tu
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Nan Zhang
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Na Yin
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Kristine Marie Conde
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Mengjie Wang
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Jonathan Carter Bean
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Junying Han
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Nikolas Anthony Scarcelli
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Yongjie Yang
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Kenji Saito
- Department of Pharmacology and Neuroscience, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Huxing Cui
- Department of Pharmacology and Neuroscience, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
- F.O.E. Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - Zheng Sun
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, Texas 77030
| | - Chunmei Wang
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Xing Cai
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Li Lu
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Yang He
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030
| | - Yong Xu
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas 77030 .
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
38
|
Chen M, Wang C, Lin Y, Chen Y, Xie W, Huang X, Zhang F, Fu C, Zhuang K, Zou T, Can D, Li H, Wu S, Luo C, Zhang J. Dorsal raphe nucleus-hippocampus serotonergic circuit underlies the depressive and cognitive impairments in 5×FAD male mice. Transl Neurodegener 2024; 13:34. [PMID: 39044270 PMCID: PMC11267773 DOI: 10.1186/s40035-024-00425-w] [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: 12/04/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Depressive symptoms often occur in patients with Alzheimer's disease (AD) and exacerbate the pathogenesis of AD. However, the neural circuit mechanisms underlying the AD-associated depression remain unclear. The serotonergic system plays crucial roles in both AD and depression. METHODS We used a combination of in vivo trans-synaptic circuit-dissecting anatomical approaches, chemogenetic manipulations, optogenetic manipulations, pharmacological methods, behavioral testing, and electrophysiological recording to investigate dorsal raphe nucleus serotonergic circuit in AD-associated depression in AD mouse model. RESULTS We found that the activity of dorsal raphe nucleus serotonin neurons (DRN5-HT) and their projections to the dorsal hippocampal CA1 (dCA1) terminals (DRN5-HT-dCA1CaMKII) both decreased in brains of early 5×FAD mice. Chemogenetic or optogenetic activation of the DRN5-HT-dCA1CaMKII neural circuit attenuated the depressive symptoms and cognitive impairments in 5×FAD mice through serotonin receptor 1B (5-HT1BR) and 4 (5-HT4R). Pharmacological activation of 5-HT1BR or 5-HT4R attenuated the depressive symptoms and cognitive impairments in 5×FAD mice by regulating the DRN5-HT-dCA1CaMKII neural circuit to improve synaptic plasticity. CONCLUSIONS These findings provide a new mechanistic connection between depression and AD and provide potential pharmaceutical prevention targets for AD.
Collapse
Affiliation(s)
- Meiqin Chen
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Chenlu Wang
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, 361000, China
| | - Yinan Lin
- Department of Anesthesiology, First Affiliated Hospital of Xiamen University, Xiamen, 361000, China
| | - Yanbing Chen
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wenting Xie
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xiaoting Huang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Fan Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Congrui Fu
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China
| | - Kai Zhuang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Tingting Zou
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Dan Can
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Huifang Li
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jie Zhang
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, College of Basic Medicine, Hebei Medical University, Shijiazhuang, 050017, China.
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, 361102, China.
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Institute of Neuroscience, Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
39
|
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
|
40
|
Xie L, Zhao J, Li Y, Bai J. PET brain imaging in neurological disorders. Phys Life Rev 2024; 49:100-111. [PMID: 38574584 DOI: 10.1016/j.plrev.2024.03.007] [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: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024]
Abstract
Brain disorders are a series of conditions with damage or loss of neurons, such as Parkinson's disease (PD), Alzheimer's disease (AD), or drug dependence. These individuals have gradual deterioration of cognitive, motor, and other central nervous system functions affected. This degenerative trajectory is intricately associated with dysregulations in neurotransmitter systems. Positron Emission Tomography (PET) imaging, employing radiopharmaceuticals and molecular imaging techniques, emerges as a crucial tool for detecting brain biomarkers. It offers invaluable insights for early diagnosis and distinguishing brain disorders. This article comprehensively reviews the application and progress of conventional and novel PET imaging agents in diagnosing brain disorders. Furthermore, it conducts a thorough analysis on merits and limitations. The article also provides a forward-looking perspective in the future development directions of PET imaging agents for diagnosing brain disorders and proposes potential innovative strategies. It aims to furnish clinicians and researchers with an all-encompassing overview of the latest advancements and forthcoming trends in the utilization of PET imaging for diagnosing brain disorders.
Collapse
Affiliation(s)
- Lijun Xie
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China; Laboratory of Molecular Neurobiology, Medical school, Kunming University of Science and Technology, Kunming 650500, PR China; Department of Nuclear Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, PR China
| | - Jihua Zhao
- Department of Nuclear Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, PR China
| | - Ye Li
- Laboratory of Molecular Neurobiology, Medical school, Kunming University of Science and Technology, Kunming 650500, PR China.
| | - Jie Bai
- Laboratory of Molecular Neurobiology, Medical school, Kunming University of Science and Technology, Kunming 650500, PR China.
| |
Collapse
|
41
|
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] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/26/2024] [Indexed: 03/27/2024] Open
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
|
42
|
Ertürk A. Deep 3D histology powered by tissue clearing, omics and AI. Nat Methods 2024; 21:1153-1165. [PMID: 38997593 DOI: 10.1038/s41592-024-02327-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/28/2024] [Indexed: 07/14/2024]
Abstract
To comprehensively understand tissue and organism physiology and pathophysiology, it is essential to create complete three-dimensional (3D) cellular maps. These maps require structural data, such as the 3D configuration and positioning of tissues and cells, and molecular data on the constitution of each cell, spanning from the DNA sequence to protein expression. While single-cell transcriptomics is illuminating the cellular and molecular diversity across species and tissues, the 3D spatial context of these molecular data is often overlooked. Here, I discuss emerging 3D tissue histology techniques that add the missing third spatial dimension to biomedical research. Through innovations in tissue-clearing chemistry, labeling and volumetric imaging that enhance 3D reconstructions and their synergy with molecular techniques, these technologies will provide detailed blueprints of entire organs or organisms at the cellular level. Machine learning, especially deep learning, will be essential for extracting meaningful insights from the vast data. Further development of integrated structural, molecular and computational methods will unlock the full potential of next-generation 3D histology.
Collapse
Affiliation(s)
- Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany.
- School of Medicine, Koç University, İstanbul, Turkey.
- Deep Piction GmbH, Munich, Germany.
| |
Collapse
|
43
|
Liu H, He Y, Liu H, Brouwers B, Yin N, Lawler K, Keogh JM, Henning E, Lee DK, Yu M, Tu L, Zhang N, Conde KM, Han J, Yan Z, Scarcelli NA, Liao L, Xu J, Tong Q, Zheng H, Sun Z, Yang Y, Wang C, He Y, Farooqi IS, Xu Y. Neural circuits expressing the serotonin 2C receptor regulate memory in mice and humans. SCIENCE ADVANCES 2024; 10:eadl2675. [PMID: 38941473 PMCID: PMC11212768 DOI: 10.1126/sciadv.adl2675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/22/2024] [Indexed: 06/30/2024]
Abstract
Declined memory is a hallmark of Alzheimer's disease (AD). Experiments in rodents and human postmortem studies suggest that serotonin (5-hydroxytryptamine, 5-HT) plays a role in memory, but the underlying mechanisms are unknown. Here, we investigate the role of 5-HT 2C receptor (5-HT2CR) in regulating memory. Transgenic mice expressing a humanized HTR2C mutation exhibit impaired plasticity of hippocampal ventral CA1 (vCA1) neurons and reduced memory. Further, 5-HT neurons project to and synapse onto vCA1 neurons. Disruption of 5-HT synthesis in vCA1-projecting neurons or deletion of 5-HT2CRs in the vCA1 impairs neural plasticity and memory. We show that a selective 5-HT2CR agonist, lorcaserin, improves synaptic plasticity and memory in an AD mouse model. Cumulatively, we demonstrate that hippocampal 5-HT2CR signaling regulates memory, which may inform the use of 5-HT2CR agonists in the treatment of dementia.
Collapse
Affiliation(s)
- Hesong Liu
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang He
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hailan Liu
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bas Brouwers
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Na Yin
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Julia M. Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Dong-Kee Lee
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng Yu
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Longlong Tu
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nan Zhang
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kristine M. Conde
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Junying Han
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zili Yan
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nikolas A. Scarcelli
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zheng Sun
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yongjie Yang
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chunmei Wang
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Brain Glycemic and Metabolism Control Department, Louisiana State University, Baton Rouge, LA 70808, USA
| | - I. Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Yong Xu
- USDA/ARS, Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
44
|
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
|
45
|
Zhang D, Wei Y. Distinct Neural Mechanisms Between Anesthesia Induction and Emergence: A Narrative Review. Anesth Analg 2024:00000539-990000000-00840. [PMID: 38861419 DOI: 10.1213/ane.0000000000007114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Anesthesia induction and emergence are critical periods for perioperative safety in the clinic. Traditionally, the emergence from general anesthesia has been recognized as a simple inverse process of induction resulting from the elimination of general anesthetics from the central nervous system. However, accumulated evidence has indicated that anesthesia induction and emergence are not mirror-image processes because of the occurrence of hysteresis/neural inertia in both animals and humans. An increasing number of studies have highlighted the critical role of orexinergic neurons and their involved circuits in the selective regulation of emergence but not the induction of general anesthesia. Moreover, additional brain regions have also been implicated in distinct neural mechanisms for anesthesia induction and emergence, which extends the concept that anesthetic induction and emergence are not antiparallel processes. Here, we reviewed the current literature and summarized the evidence regarding the differential mechanism of neural modulation in anesthesia induction and emergence, which will facilitate the understanding of the underlying neural mechanism for emergence from general anesthesia.
Collapse
Affiliation(s)
- Donghang Zhang
- From the Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Anesthesiology, Weill Cornell Medicine, New York, New York
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| |
Collapse
|
46
|
Kikuchi S, Iwasaki Y, Yoshioka M, Hino K, Morita SY, Tada R, Uchimura Y, Kubo Y, Kobayashi T, Kinoshita Y, Hayashi M, Furusho Y, Tamiaki H, Ishiyama H, Kuroda M, Udagawa J. Solitary and Synergistic Effects of Different Hydrophilic and Hydrophobic Phospholipid Moieties on Rat Behaviors. Pharmaceutics 2024; 16:762. [PMID: 38931883 PMCID: PMC11207216 DOI: 10.3390/pharmaceutics16060762] [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: 04/10/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Glycerophospholipids have hydrophobic and hydrophilic moieties. Previous studies suggest that phospholipids with different moieties have different effects on rodent behavior; however, the relationship between chemical structures and behavioral effects remains unclear. To clarify the functions of phospholipid moieties, we injected male rats with phospholipids with different moieties and conducted behavioral tests. Exploratory activity was reduced by phosphatidylethanolamine (PE)(18:0/22:6) but not PE(18:0/18:0) or PE(18:0/20:4). Conversely, exploratory activity was increased by plasmanyl PE(16:0/22:6), which harbors an alkyl-ether linkage, but not by phosphatidylcholine (PC)(16:0/22:6) or plasmanyl PC(16:0/22:6). Docosahexaenoic acid (DHA)(22:6) and an alkyl-ether linkage in PE were thus postulated to be involved in exploratory activity. Anxiety-like behavior was reduced by plasmenyl PC(18:0/20:4), which harbors a vinyl-ether linkage, but not by PC(18:0/20:4) or plasmanyl PC(18:0/20:4), suggesting the anxiolytic effects of vinyl-ether linkage. The activation of social interaction was suppressed by PE(18:0/18:0), PE(18:0/22:6), PC(16:0/22:6), plasmanyl PE(16:0/22:6), and plasmanyl PC(16:0/22:6) but not by PE(18:0/20:4), plasmenyl PE(18:0/20:4), or plasmanyl PC(18:0/22:6). DHA may suppress social interaction, whereas arachidonic acid(20:4) or a combination of alkyl-ether linkage and stearic acid(18:0) may restore social deficits. Our findings indicate the characteristic effects of different phospholipid moieties on rat behavior, and may help to elucidate patterns between chemical structures and their effects.
Collapse
Affiliation(s)
- Shuhei Kikuchi
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Yugo Iwasaki
- College of Bioscience and Biotechnology, Chubu University, Kasugai 487-8501, Aichi, Japan;
| | - Mina Yoshioka
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Kodai Hino
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Shin-ya Morita
- Department of Pharmacotherapeutics, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| | - Ryu Tada
- Molecular Engineering Institute, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| | - Yasuhiro Uchimura
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Yoshinori Kubo
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Tomoya Kobayashi
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Yusuke Kinoshita
- Graduate School of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (Y.K.); (H.T.)
| | - Masahiro Hayashi
- Department of Marine Biology and Environmental Science, Faculty of Agriculture, University of Miyazaki, Miyazaki 889-2192, Miyazaki, Japan;
| | - Yoshio Furusho
- Department of Chemistry, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan
| | - Hitoshi Tamiaki
- Graduate School of Life Sciences, Ritsumeikan University, Kusatsu 525-8577, Shiga, Japan; (Y.K.); (H.T.)
| | - Hiroaki Ishiyama
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Minoru Kuroda
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| | - Jun Udagawa
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan; (S.K.); (Y.U.); (M.K.)
| |
Collapse
|
47
|
Meng J, Niu J, Zhang N, Hao Y, Gao T, Wei M. Modified conjunctival sac plasty and personalized eye model implantation in the treatment of patients with conjunctival sac stenosis and sunken eye socket. Minerva Med 2024; 115:384-386. [PMID: 37962896 DOI: 10.23736/s0026-4806.23.08844-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Affiliation(s)
- Jikai Meng
- Department of Corneal, Orbital and Ocular Plastic Surgery, Handan City Eye Hospital (The Third Hospital of Handan), Handan, China
| | - Jie Niu
- Department of Internal Medicine of Fundus Diseases, Handan City Eye Hospital (The Third Hospital of Handan), Handan, China
| | - Ning Zhang
- Department of Corneal, Orbital and Ocular Plastic Surgery, Handan City Eye Hospital (The Third Hospital of Handan), Handan, China
| | - Yongna Hao
- Department of Corneal, Orbital and Ocular Plastic Surgery, Handan City Eye Hospital (The Third Hospital of Handan), Handan, China
| | - Tieying Gao
- Department of Corneal, Orbital and Ocular Plastic Surgery, Handan City Eye Hospital (The Third Hospital of Handan), Handan, China
| | - Min Wei
- Department of Corneal, Orbital and Ocular Plastic Surgery, Handan City Eye Hospital (The Third Hospital of Handan), Handan, China -
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Liang YF, Chen XQ, Zhang MT, Tang HY, Shen GM. Research Progress of Central and Peripheral Corticotropin-Releasing Hormone in Irritable Bowel Syndrome with Comorbid Dysthymic Disorders. Gut Liver 2024; 18:391-403. [PMID: 37551453 PMCID: PMC11096901 DOI: 10.5009/gnl220346] [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: 08/05/2022] [Revised: 04/26/2023] [Accepted: 05/22/2023] [Indexed: 08/09/2023] Open
Abstract
Irritable bowel syndrome (IBS) is considered a stress disorder characterized by psychological and gastrointestinal dysfunction. IBS patients not only suffer from intestinal symptoms such as abdominal pain, diarrhea, or constipation but also, experience dysthymic disorders such as anxiety and depression. Studies have found that corticotropin-releasing hormone plays a key role in IBS with comorbid dysthymic disorders. Next, we will summarize the effects of corticotropin-releasing hormone from the central nervous system and periphery on IBS with comorbid dysthymic disorders and relevant treatments based on published literatures in recent years.
Collapse
Affiliation(s)
- Yi Feng Liang
- College of Acupuncture and Massage, Anhui University of Chinese Medicine, Hefei, China
| | - Xiao Qi Chen
- College of Acupuncture and Massage, Anhui University of Chinese Medicine, Hefei, China
| | - Meng Ting Zhang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - He Yong Tang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Guo Ming Shen
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|