1
|
Kazmierska-Grebowska P, Żakowski W, Myślińska D, Sahu R, Jankowski MM. Revisiting serotonin's role in spatial memory: A call for sensitive analytical approaches. Int J Biochem Cell Biol 2024; 176:106663. [PMID: 39321568 DOI: 10.1016/j.biocel.2024.106663] [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: 03/14/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
The serotonergic system is involved in various psychiatric and neurological conditions, with serotonergic drugs often used in treatment. These conditions frequently affect spatial memory, which can serve as a model of declarative memory due to well-known cellular components and advanced methods that track neural activity and behavior with high temporal resolution. However, most findings on serotonin's effects on spatial learning and memory come from studies lacking refined analytical techniques and modern approaches needed to uncover the underlying neuronal mechanisms. This In Focus review critically investigates available studies to identify areas for further exploration. It finds that well-established behavioral models could yield more insights with modern tracking and data analysis approaches, while the cellular aspects of spatial memory remain underexplored. The review highlights the complex role of serotonin in spatial memory, which holds the potential for better understanding and treating memory-related disorders.
Collapse
Affiliation(s)
| | - Witold Żakowski
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Dorota Myślińska
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Ravindra Sahu
- BioTechMed Center, Multimedia Systems Department, Faculty of Electronics, Telecommunications and Informatics, Gdansk University of Technology, Gdansk, Poland
| | - Maciej M Jankowski
- BioTechMed Center, Multimedia Systems Department, Faculty of Electronics, Telecommunications and Informatics, Gdansk University of Technology, Gdansk, Poland.
| |
Collapse
|
2
|
Li J, Liu Y, Yin C, Zeng Y, Mei Y. Structural and functional remodeling of neural networks in β-amyloid driven hippocampal hyperactivity. Ageing Res Rev 2024; 101:102468. [PMID: 39218080 DOI: 10.1016/j.arr.2024.102468] [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: 06/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Early detection of Alzheimer's disease (AD) is essential for improving the patients outcomes and advancing our understanding of disease, allowing for timely intervention and treatment. However, accurate biomarkers are still lacking. Recent evidence indicates that hippocampal hyperexcitability precedes the diagnosis of AD decades ago, can predict cognitive decline. Thus, could hippocampal hyperactivity be a robust biomarker for early-AD, and what drives hippocampal hyperactivity in early-AD? these critical questions remain to be answered. Increasing clinical and experimental studies suggest that early hippocampal activation is closely associated with longitudinal β-amyloid (Aβ) accumulation, Aβ aggregates, in turn, enhances hippocampal activity. Therefore, in this narrative review, we discuss the role of Aβ-induced altered intrinsic neuronal properties as well as structural and functional remodeling of glutamatergic, GABAergic, cholinergic, noradrenergic, serotonergic circuits in hippocampal hyperactivity. In addition, we analyze the available therapies and trials that can potentially be used clinically to attenuate hippocampal hyperexcitability in AD. Overall, the present review sheds lights on the mechanism behind Aβ-induced hippocampal hyperactivity, and highlights that hippocampal hyperactivity could be a robust biomarker and therapeutic target in prodromal AD.
Collapse
Affiliation(s)
- Jinquan Li
- 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
| | - Yanjun Liu
- 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
| | - Chuhui Yin
- 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
| | - Yan Zeng
- 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.
| | - 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
|
3
|
Özçete ÖD, Banerjee A, Kaeser PS. Mechanisms of neuromodulatory volume transmission. Mol Psychiatry 2024; 29:3680-3693. [PMID: 38789677 PMCID: PMC11540752 DOI: 10.1038/s41380-024-02608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024]
Abstract
A wealth of neuromodulatory transmitters regulate synaptic circuits in the brain. Their mode of signaling, often called volume transmission, differs from classical synaptic transmission in important ways. In synaptic transmission, vesicles rapidly fuse in response to action potentials and release their transmitter content. The transmitters are then sensed by nearby receptors on select target cells with minimal delay. Signal transmission is restricted to synaptic contacts and typically occurs within ~1 ms. Volume transmission doesn't rely on synaptic contact sites and is the main mode of monoamines and neuropeptides, important neuromodulators in the brain. It is less precise than synaptic transmission, and the underlying molecular mechanisms and spatiotemporal scales are often not well understood. Here, we review literature on mechanisms of volume transmission and raise scientific questions that should be addressed in the years ahead. We define five domains by which volume transmission systems can differ from synaptic transmission and from one another. These domains are (1) innervation patterns and firing properties, (2) transmitter synthesis and loading into different types of vesicles, (3) architecture and distribution of release sites, (4) transmitter diffusion, degradation, and reuptake, and (5) receptor types and their positioning on target cells. We discuss these five domains for dopamine, a well-studied monoamine, and then compare the literature on dopamine with that on norepinephrine and serotonin. We include assessments of neuropeptide signaling and of central acetylcholine transmission. Through this review, we provide a molecular and cellular framework for volume transmission. This mechanistic knowledge is essential to define how neuromodulatory systems control behavior in health and disease and to understand how they are modulated by medical treatments and by drugs of abuse.
Collapse
Affiliation(s)
- Özge D Özçete
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Aditi Banerjee
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Yu LCH. Gastrointestinal pathophysiology in long COVID: Exploring roles of microbiota dysbiosis and serotonin dysregulation in post-infectious bowel symptoms. Life Sci 2024; 358:123153. [PMID: 39454992 DOI: 10.1016/j.lfs.2024.123153] [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: 08/27/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered an unprecedented public health crisis known as the coronavirus disease 2019 (COVID-19) pandemic. Gastrointestinal (GI) symptoms develop in patients during acute infection and persist after recovery from airway distress in a chronic form of the disease (long COVID). A high incidence of irritable bowel syndrome (IBS) manifested by severe abdominal pain and defecation pattern changes is reported in COVID patients. Although COVID is primarily considered a respiratory disease, fecal shedding of SARS-CoV-2 antigens positively correlates with bowel symptoms. Active viral infection in the GI tract was identified by human intestinal organoid studies showing SARS-CoV-2 replication in gut epithelial cells. In this review, we highlight the key findings in post-COVID bowel symptoms and explore possible mechanisms underlying the pathophysiology of the illness. These mechanisms include mucosal inflammation, gut barrier dysfunction, and microbiota dysbiosis during viral infection. Viral shedding through the GI route may be the primary factor causing the alteration of the microbiome ecosystem, particularly the virome. Recent evidence in experimental models suggested that microbiome dysbiosis could be further aggravated by epithelial barrier damage and immune activation. Moreover, altered microbiota composition has been associated with dysregulated serotonin pathways, resulting in intestinal nerve hypersensitivity. These mechanisms may explain the development of post-infectious IBS-like symptoms in long COVID. Understanding how coronavirus infection affects gut pathophysiology, including microbiome changes, would benefit the therapeutic advancement for managing post-infectious bowel symptoms.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
5
|
Fang K, Cheng W, Yu B. Effects of Electroacupuncture at Varied Frequencies on Analgesia and Mechanisms in Sciatic Nerve Cuffing-Induced Neuropathic Pain Mice. J Mol Neurosci 2024; 74:98. [PMID: 39414746 PMCID: PMC11485069 DOI: 10.1007/s12031-024-02276-6] [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: 08/19/2024] [Accepted: 10/03/2024] [Indexed: 10/18/2024]
Abstract
Addressing the intricate challenge of chronic neuropathic pain has significant implications for the physical and psychological well-being of patients, given its enduring nature. In contrast to opioids, electroacupuncture (EA) may potentially provide a safer and more efficacious therapeutic alternative. Our objective is to investigate the distinct analgesic effects and potential mechanisms of EA at frequencies of 2 Hz, 100 Hz, and 18 kHz in order to establish more precise frequency selection criteria for clinical interventions. Analgesic efficacy was evaluated through the measurement of mice's mechanical and thermal pain thresholds. Spinal cord inflammatory cytokines and neuropeptides were quantified via Quantitative Real-time PCR (qRT-PCR), Western blot, and immunofluorescence. Additionally, RNA sequencing (RNA-Seq) was conducted on the spinal cord from mice in the 18 kHz EA group for comprehensive transcriptomic analysis. The analgesic effect of EA on neuropathic pain in mice was frequency-dependent. Stimulation at 18 kHz provided superior and prolonged relief compared to 2 Hz and 100 Hz. Our research suggests that EA at frequencies of 2 Hz, 100 Hz, and 18 kHz significantly reduce the release of inflammatory cytokines. The analgesic effects of 2 Hz and 100 Hz stimulation are due to frequency-dependent regulation of opioid release in the spinal cord. Furthermore, 18 kHz stimulation has been shown to reduce spinal neuronal excitability by modulating the serotonergic pathway and downstream receptors in the spinal cord to alleviate neuropathic pain.
Collapse
Affiliation(s)
- Kexin Fang
- Tongji University School of Medicine, Shanghai, China
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, 2209 Guangxing Road, Songjiang District, Shanghai, China
| | - Wen Cheng
- Tongji University School of Medicine, Shanghai, China
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, 2209 Guangxing Road, Songjiang District, Shanghai, China
| | - Bin Yu
- Tongji University School of Medicine, Shanghai, China.
- Department of Anesthesiology, Yangzhi Rehabilitation Hospital Affiliated to Tongji University, School of Medicine, Tongji University, 2209 Guangxing Road, Songjiang District, Shanghai, China.
| |
Collapse
|
6
|
Stinson HE, Ninan I. Median raphe glutamatergic neuron-mediated enhancement of GABAergic transmission and suppression of long-term potentiation in the hippocampus. Heliyon 2024; 10:e38192. [PMID: 39386853 PMCID: PMC11462361 DOI: 10.1016/j.heliyon.2024.e38192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
The ascending neuromodulatory pathway from the median raphe nucleus (MRN) extends widely throughout midline/para-midline regions and robustly innervates the hippocampus. This neuromodulatory pathway is believed to be critical for regulating emotional and affective behaviors. Although the MRN primarily contains serotoninergic (5-HTergic), GABAergic, and glutamatergic neurons, glutamatergic neurons expressing vesicular glutamate transporter 3 (VGLUT3) form the primary MRN input to the hippocampus. Despite the earlier demonstration of the robust MRN VGLUT3 innervation of the hippocampus, little is known about how this MRN glutamatergic input modulates synaptic transmission and plasticity in the hippocampus. Our studies show that MRN VGLUT3 neurons activate serotonin 3a receptor (5-HT3aR)-expressing GABAergic neurons, including VGLUT3-expressing neurons, at the stratum radiatum (SR)/stratum lacunosum moleculare (SLM) border. This MRN VGLUT3 neuron-mediated glutamatergic transmission onto SR/SLM 5-HT3aR neurons is negatively regulated by 5-HT through 5-HT1B receptors. In agreement with the MRN VGLUT3 neuron-mediated activation of the 5-HT3aR GABAergic neurons, activation of MRN VGLUT3 projections induces a long-lasting increase in GABAergic transmission but not glutamatergic transmission in CA1 pyramidal neurons from male but not female mice. Consistent with the MRN VGLUT3 neuron-mediated enhancement of GABAergic transmission in male mice, activation of MRN VGLUT3 projections suppresses Schaffer collateral (SC)-CA1 long-term potentiation (LTP) in male but not female mice. Thus, our results show that MRN VGLUT3 neurons modulate the dorsal hippocampus by augmenting synaptic inhibition of CA1 pyramidal neurons and by suppressing SC-CA1 LTP in a sex-specific manner.
Collapse
Affiliation(s)
- Hannah E. Stinson
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ipe Ninan
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
7
|
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 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
|
8
|
Wang Z, Li D, Chen M, Yu X, Chen C, Chen Y, Zhang L, Shu Y. A comprehensive study on the regulation of Compound Zaoren Granules on cAMP/CREB signaling pathway and metabolic disorder in CUMS-PCPA induced insomnia rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 332:118401. [PMID: 38815875 DOI: 10.1016/j.jep.2024.118401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/07/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Compound Zaoren Granules (CZG), an optimized herbal formulation based on the traditional Chinese medicine prescription Suanzaoren decoction, are designed specifically for insomnia treatment. However, the mechanisms underlying its efficacy in treating insomnia are not yet fully understood. AIM OF THE STUDY The research investigated the mechanisms of CZG's improvement in insomnia by regulating cAMP/CREB signaling pathway and metabolic profiles. METHODS The main components of CZG were characterized by liquid chromatography-mass spectrometry (LC-MS). Subsequently, these validated components were applied to network pharmacological analysis to predict signaling pathways associated with insomnia. We evaluated the effect of CZG on BV-2 cells in vitro. We also evaluated the behavioral indexes of CUMS combined with PCPA induced insomnia in rats. HE staining and Nissl staining were used to observe the pathological damage of hippocampus. ELISA was used to detect the levels of various neurotransmitters, orexins, HPA axis, and inflammatory factors in insomnia rats. Then we detected the expression of cAMP/CREB signaling pathway through ELISA, WB, and IHC. Finally, the metabolomics was further analyzed by using UHPLC-QTOF-MS/MS to investigate the changes in the hippocampus of insomnia rats and the possible metabolic pathways were also speculated. RESULTS The results of CZG in vitro experiments showed that CZG has protective and anti-inflammatory effects on LPS induced BV-2 cells. A total of 161 chemical components were identified in CZG. After conducting network pharmacology analysis through these confirmed components, we select the cAMP/CREB signaling pathway for further investigate. The behavioral research results on insomnia rats showed that CZG significantly prolonged sleep time, mitigated brain tissue pathological damage, and exhibited liver protective properties. CZG treats insomnia by regulating the content of various neurotransmitters, reducing levels of orexin, HPA axis, and inflammatory factors. It can also treat insomnia by upregulating the expression of the cAMP/CREB signaling pathway. Hippocampus metabolomics analysis identified 69 differential metabolites associated with insomnia. The metabolic pathways related to these differential metabolites have also been predicted. CONCLUSION These results indicate that CZG can significantly prolong sleep time. CZG is used to treat insomnia by regulating various neurotransmitters, HPA axis, inflammatory factors, cAMP/CREB signaling pathways, and metabolic disorders.
Collapse
Affiliation(s)
- Zekun Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Danting Li
- Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China; Key laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Min Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Xiaocong Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Chen Chen
- Nanjing Women and Children's Healthcare Hospital, 210029, China
| | - Yajun Chen
- Nanjing Women and Children's Healthcare Hospital, 210029, China
| | - Lingfeng Zhang
- School of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, 211198, China
| | - Yachun Shu
- Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China; Jiangsu Province Seaside Rehabilitation Hospital, Lianyungang, 222042, China.
| |
Collapse
|
9
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
10
|
Fenstermacher SJ, Vonasek A, Gattuso H, Chaimowitz C, Dymecki SM, Jessell TM, Dasen JS. Potentiation of active locomotor state by spinal-projecting serotonergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615260. [PMID: 39386605 PMCID: PMC11463418 DOI: 10.1101/2024.09.26.615260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Animals produce diverse motor actions that enable expression of context-appropriate behaviors. Neuromodulators facilitate behavioral flexibility by altering the temporal dynamics and output of neural circuits. Discrete populations of serotonergic (5-HT) neurons target circuits in the brainstem and spinal cord, but their role in the control of motor behavior is unclear. Here we define the pre- and post-synaptic organization of the spinal-projecting serotonergic system and define a role in locomotor control. We show that while forebrain-targeting 5-HT neurons decrease their activity during locomotion, subpopulations of spinal projecting neurons increase their activity in a context-dependent manner. Optogenetic activation of ventrally projecting 5-HT neurons does not trigger initiation of movement, but rather enhances the speed and duration of ongoing locomotion over extended time scales. These findings indicate that the descending serotonergic system potentiates locomotor output and demonstrate a role for serotonergic neurons in modulating the temporal dynamics of motor circuits.
Collapse
Affiliation(s)
- Sara J. Fenstermacher
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine
| | - Ann Vonasek
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine
| | - Hannah Gattuso
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine
| | - Corryn Chaimowitz
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine
| | | | | | - Jeremy S. Dasen
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine
| |
Collapse
|
11
|
Li C, McCloskey NS, Inan S, Kirby LG. Role of serotonin neurons in the dorsal raphe nucleus in heroin self-administration and punishment. Neuropsychopharmacology 2024:10.1038/s41386-024-01993-1. [PMID: 39300273 DOI: 10.1038/s41386-024-01993-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
One hallmark of substance use disorder is continued drug use despite negative consequences. When drug-taking behavior is punished with aversive stimuli, i.e. footshock, rats can also be categorized into punishment-resistant or compulsive vs. punishment-sensitive or non-compulsive phenotypes. The serotonin (5-hydroxytryptamine, 5-HT) system modulates responses to both reward and punishment. The goal of the current study was to examine punishment phenotypes in heroin self-administration and to determine the role of dorsal raphe nucleus (DRN) 5-HT neurons in both basal and punished heroin self-administration. First, rats were exposed to punished heroin self-administration and neuronal excitability of DRN 5-HT neurons was compared between punishment-resistant and punishment-sensitive phenotypes using ex vivo electrophysiology. Second, DRN 5-HT neuronal activity was manipulated in vivo during basal and punished heroin self-administration using chemogenetic tools in a Tph2-iCre rat line. While rats separated into punishment-resistant and punishment-sensitive phenotypes for punished heroin self-administration, DRN 5-HT neuronal excitability did not differ between the phenotypes. While chemogenetic inhibition of DRN 5-HT neurons was without effect, chemogenetic activation of DRN 5-HT neurons increased both basal and punished heroin self-administration selectively in punishment-resistant animals. Additionally, the responsiveness to chemogenetic activation of DRN 5-HT neurons in basal self-administration and motivation for heroin in progressive ratio each predicted resistance to punishment. Therefore, our data support the role for the DRN 5-HT system in compulsive heroin self-administration.
Collapse
Affiliation(s)
- Chen Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Nicholas S McCloskey
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Lynn G Kirby
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, USA.
| |
Collapse
|
12
|
Xie X, Li W, Xiong Z, Xu J, Liao T, Sun L, Xu H, Zhang M, Zhou J, Xiong W, Fu Z, Li Z, Han Q, Cui D, Anthony DC. Metformin reprograms tryptophan metabolism via gut microbiome-derived bile acid metabolites to ameliorate depression-Like behaviors in mice. Brain Behav Immun 2024; 123:442-455. [PMID: 39303815 DOI: 10.1016/j.bbi.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
As an adjunct therapy, metformin enhances the efficacy of conventional antidepressant medications. However, its mode of action remains unclear. Here, metformin was found to ameliorate depression-like behaviors in mice exposed to chronic restraint stress (CRS) by normalizing the dysbiotic gut microbiome. Fecal transplants from metformin-treated mice ameliorated depressive behaviors in stressed mice. Microbiome profiling revealed that Akkermansia muciniphila (A. muciniphila), in particular, was markedly increased in the gut by metformin and that oral administration of this species alone was sufficient to reverse CRS-induced depressive behaviors and normalize aberrant stress-induced 5-hydroxytryptamine (5-HT) metabolism in the brain and gut. Untargeted metabolomic profiling further identified the bile acid metabolites taurocholate and deoxycholic acid as direct A. muciniphila-derived molecules that are, individually, sufficient to rescue the CRS-induced impaired 5-HT metabolism and depression-like behaviors. Thus, we report metformin reprograms 5-HT metabolism via microbiome-brain interactions to mitigate depressive syndromes, providing novel insights into gut microbiota-derived bile acids as potential therapeutic candidates for depressive mood disorders from bench to bedside.
Collapse
Affiliation(s)
- Xiaoxian Xie
- Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 201109, PR China; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Wenwen Li
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Ze Xiong
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Junyu Xu
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, PR China
| | - Tailin Liao
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, PR China
| | - Lei Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Haoshen Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Mengya Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Jiafeng Zhou
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Wenzheng Xiong
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Zezhi Li
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, PR China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, PR China.
| | - Qi Han
- Center for Brain Science Shanghai Children s Medical Center, Department of Anatomy and Physiology, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China; Shanghai Center for Brain Science and Brain-inspired Technology, Shanghai 200031, PR China.
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 201109, PR China.
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK
| |
Collapse
|
13
|
Gonye EC, Shi Y, Li K, Clements RT, Xu W, Bayliss DA. Intrinsic Molecular Proton Sensitivity Underlies GPR4 Effects on Retrotrapezoid Nucleus Neuronal Activation and CO 2-Stimulated Breathing. J Neurosci 2024; 44:e0799242024. [PMID: 39107057 PMCID: PMC11376338 DOI: 10.1523/jneurosci.0799-24.2024] [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: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024] Open
Abstract
An interoceptive homeostatic reflex monitors levels of CO2/H+ to maintain blood gas homeostasis and rapidly regulate tissue acid-base balance by driving lung ventilation and CO2 excretion-this CO2-evoked increase in respiration is the hypercapnic ventilatory reflex (HCVR). Retrotrapezoid nucleus (RTN) neurons provide crucial excitatory drive to downstream respiratory rhythm/pattern-generating circuits, and their activity is directly modulated by changes in CO2/H+ RTN neurons express GPR4 and TASK-2, global deletion of which abrogates CO2/H+ activation of RTN neurons and the HCVR. It has not been determined if the intrinsic pH sensitivity of these proton detectors is required for these effects. We used CRISPR/Cas9 genome editing to generate mice with mutations in either of two pH-sensing histidine residues in GPR4 to determine effects on RTN neuronal CO2/H+ sensitivity and the HCVR. In global GPR4(H81F) and GPR4(H167F) mice, CO2-stimulated breathing and CO2-induced RTN neuronal activation were strongly blunted, with no effect on hypoxia-stimulated breathing. In brainstem slices from GPR4(H81F) mice, peak firing of RTN neurons during bath acidification was significantly reduced compared with GPR4 wild-type mice, and a subpopulation of RTN neurons was rendered pH-insensitive, phenocopying previous results from GPR4-deleted mice. These effects were independent of changes in RTN number/distribution, neuronal excitability or transcript levels for GPR4 and TASK-2. CO2-stimulated breathing was reduced to a similar extent in GPR4(H81F) and TASK-2-deleted mice, with combined mutation yielding no additional deficit in the HCVR. Together, these data demonstrate that the intrinsic pH sensitivity of GPR4 is necessary for full elaboration of the HCVR.
Collapse
Affiliation(s)
- Elizabeth C Gonye
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Yingtang Shi
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Keyong Li
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Rachel T Clements
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| | - Wenhao Xu
- Genetically Engineered Mouse Model Core, University of Virginia, Charlottesville, Virginia 22903
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22903
| |
Collapse
|
14
|
Jankovic T, Bogicevic M, Knezevic NN. The role of nitric oxide and hormone signaling in chronic stress, anxiety, depression and post-traumatic stress disorder. Mol Cell Endocrinol 2024; 590:112266. [PMID: 38718853 DOI: 10.1016/j.mce.2024.112266] [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: 12/27/2023] [Revised: 04/20/2024] [Accepted: 04/30/2024] [Indexed: 05/24/2024]
Abstract
This paper provides a summary of the role of nitric oxide (NO) and hormones in the development of chronic stress, anxiety, depression, and post-traumatic stress disorder (PTSD). These mental health conditions are prevalent globally and involve complex molecular interactions. Although there is a significant amount of research and therapeutic options available, the underlying mechanisms of these disorders are still not fully understood. The primary pathophysiologic processes involved in chronic stress, anxiety, depression, and PTSD include dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, the intracellular influence of neuronal nitric oxide synthase (nNOS) on transcription factors, an inflammatory response with the formation of nitrergic oxidative species, and reduced serotonergic transmission in the dorsal raphe nucleus. Despite the extensive literature on this topic, there is a great need for further research to clarify the complexities inherent in these pathways, with the primary aim of improving psychiatric care.
Collapse
Affiliation(s)
- Tamara Jankovic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Marko Bogicevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA; Midwestern University Chicago College of Osteopathic Medicine, Downers Grove, IL, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA; Department of Anesthesiology, University of Illinois, Chicago, IL, USA; Department of Surgery, University of Illinois, Chicago, IL, USA.
| |
Collapse
|
15
|
Lu S, Zhao Q, Guan Y, Sun Z, Li W, Guo S, Zhang A. The communication mechanism of the gut-brain axis and its effect on central nervous system diseases: A systematic review. Biomed Pharmacother 2024; 178:117207. [PMID: 39067168 DOI: 10.1016/j.biopha.2024.117207] [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: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
Gut microbiota is involved in intricate and active metabolic processes the host's brain function, especially its role in immune responses, secondary metabolism, and symbiotic connections with the host. Gut microbiota can promote the production of essential metabolites, neurotransmitters, and other neuroactive chemicals that affect the development and treatment of central nervous system diseases. This article introduces the relevant pathways and manners of the communication between the brain and gut, summarizes a comprehensive overview of the current research status of key gut microbiota metabolites that affect the functions of the nervous system, revealing those adverse factors that affect typical communication between the brain-gut axis, and outlining the efforts made by researchers to alleviate these neurological diseases through targeted microbial interventions. The relevant pathways and manners of communication between the brain and gut contribute to the experimental design of new treatment plans and drug development. The factors that may cause changes in gut microbiota and affect metabolites, as well as current intervention methods are summarized, which helps improve gut microbiota brain dialogue, prevent adverse triggering factors from interfering with the gut microbiota system, and minimize neuropathological changes.
Collapse
Affiliation(s)
- Shengwen Lu
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Qiqi Zhao
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Yu Guan
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Zhiwen Sun
- Department of Gastroenterology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Wenhao Li
- School of Basic Medical Science of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Sifan Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China
| | - Aihua Zhang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China; INTI International University, Nilai 71800, Malaysia.
| |
Collapse
|
16
|
Wang J, Zhang Y, Yang H, Tian E, Guo Z, Chen J, Qiao C, Jiang H, Guo J, Zhou Z, Luo Q, Shi S, Yao H, Lu Y, Zhang S. Advanced progress of vestibular compensation in vestibular neural networks. CNS Neurosci Ther 2024; 30:e70037. [PMID: 39268632 PMCID: PMC11393560 DOI: 10.1111/cns.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024] Open
Abstract
Vestibular compensation is the natural process of recovery that occurs with acute peripheral vestibular lesion. Here, we summarize the current understanding of the mechanisms underlying vestibular compensation, focusing on the role of the medial vestibular nucleus (MVN), the central hub of the vestibular system, and its associated neural networks. The disruption of neural activity balance between the bilateral MVNs underlies the vestibular symptoms after unilateral vestibular damage, and this balance disruption can be partially reversed by the mutual inhibitory projections between the bilateral MVNs, and their top-down regulation by other brain regions via different neurotransmitters. However, the detailed mechanism of how MVN is involved in vestibular compensation and regulated remains largely unknown. A deeper understanding of the vestibular neural network and the neurotransmitter systems involved in vestibular compensation holds promise for improving treatment outcomes and developing more effective interventions for vestibular disorders.
Collapse
Affiliation(s)
- Jun Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- National Clinical Research Center for Otolaryngologic Diseases, Jiangxi Branch Center, Nanchang, China
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuejin Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Huajing Yang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - E Tian
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoqi Guo
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caijuan Qiao
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Hongqun Jiang
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- National Clinical Research Center for Otolaryngologic Diseases, Jiangxi Branch Center, Nanchang, China
| | - Jiaqi Guo
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhanghong Zhou
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Luo
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- National Clinical Research Center for Otolaryngologic Diseases, Jiangxi Branch Center, Nanchang, China
| | - Shiyu Shi
- Department of Rehabilitation, Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyi Yao
- Department of Rehabilitation, Liyuan Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yisheng Lu
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Sulin Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Park I, Choi M, Kim J, Jang S, Kim D, Kim J, Choe Y, Geum D, Yu SW, Choi JW, Moon C, Choe HK, Son GH, Kim K. Role of the circadian nuclear receptor REV-ERBα in dorsal raphe serotonin synthesis in mood regulation. Commun Biol 2024; 7:998. [PMID: 39147805 PMCID: PMC11327353 DOI: 10.1038/s42003-024-06647-y] [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: 11/06/2023] [Accepted: 07/29/2024] [Indexed: 08/17/2024] Open
Abstract
Affective disorders are frequently associated with disrupted circadian rhythms. The existence of rhythmic secretion of central serotonin (5-hydroxytryptamine, 5-HT) pattern has been reported; however, the functional mechanism underlying the circadian control of 5-HTergic mood regulation remains largely unknown. Here, we investigate the role of the circadian nuclear receptor REV-ERBα in regulating tryptophan hydroxylase 2 (Tph2), the rate-limiting enzyme of 5-HT synthesis. We demonstrate that the REV-ERBα expressed in dorsal raphe (DR) 5-HTergic neurons functionally competes with PET-1-a nuclear activator crucial for 5-HTergic neuron development. In mice, genetic ablation of DR 5-HTergic REV-ERBα increases Tph2 expression, leading to elevated DR 5-HT levels and reduced depression-like behaviors at dusk. Further, pharmacological manipulation of the mice DR REV-ERBα activity increases DR 5-HT levels and affects despair-related behaviors. Our findings provide valuable insights into the molecular and cellular link between the circadian rhythm and the mood-controlling DR 5-HTergic systems.
Collapse
Affiliation(s)
- Inah Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Mijung Choi
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Jeongah Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Sangwon Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Doyeon Kim
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Jihoon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Dongho Geum
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Seong-Woon Yu
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Ji-Woong Choi
- Department of Electrical Engineering and Computer Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Cheil Moon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Han Kyoung Choe
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Gi Hoon Son
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Legal Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Kyungjin Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| |
Collapse
|
18
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [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
|
19
|
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
|
20
|
Bao L, Chen M, Dai B, Lei Y, Qin D, Cheng M, Song W, He W, Chen B, Shen H. Nanoengineered therapeutic strategies targeting SNHG1 for mitigating microglial ischemia-reperfusion injury implications for hypoxic-ischemic encephalopathy. SLAS Technol 2024; 29:100167. [PMID: 39043303 DOI: 10.1016/j.slast.2024.100167] [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: 05/08/2024] [Revised: 06/05/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024]
Abstract
The purpose of this work is to investigate the function of SNHG1, a long non-coding RNA implicated in disease progression, apoptosis, and proliferation, in order to solve the problem of hypoxic-ischemic encephalopathy (HIE) in newborn care. We investigated the impact of overexpressing SNHG1 on hypoxia-induced apoptosis and studied its expression in BV2 microglial cells under hypoxic circumstances. As a result of modifying YY1 expression, SNHG1's overexpression prevents apoptosis, as our data demonstrate that it is considerably downregulated under hypoxia. We demonstrate that SNHG1 might potentially reduce microglial ischemia-reperfusion damage by using sophisticated nanoengineering drug delivery technologies to target it. This provides encouraging information for the therapy of ischemic epilepsy.
Collapse
Affiliation(s)
- Li Bao
- Department of Neonatology,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Mingzhi Chen
- Department of Thoracic and Cardiovascular Surgery,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Biao Dai
- Department of Science and Education,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Yong Lei
- Department of Neonatology,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Dani Qin
- Department of Pediatrics,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Mengke Cheng
- Department of Neonatology,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Wei Song
- Department of Neonatology,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Wenxia He
- Department of Neonatology,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Bingyu Chen
- Department of Pediatrics,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China
| | - Huiping Shen
- Department of Pediatrics,Yixing Hospital Affiliated to Jiangsu University,Yixing 214200, Jiangsu Province, China.
| |
Collapse
|
21
|
Kreshchenko ND, Ermakov AM. Morphometric analysis and functional insights into the serotonergic system of Girardia tigrina (Tricladida, Platyhelminthes). J Morphol 2024; 285:e21756. [PMID: 39086183 DOI: 10.1002/jmor.21756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Using immunocytochemistry, serotonergic nerve elements were documented in the nervous system of the planarian Girardia tigrina. Serotonin-immunopositive components were observed in the brain, ventral, dorsal and longitudinal nerve cords, transverse nerve commissures connecting the nerve cords, and in the nerve plexus. Whole-mount preparations of G. tigrina were analyzed by fluorescent and confocal laser scanning microscopy. An essential quantitative morphometric measurement of serotonin-immunopositive structures was conducted in three body regions (anterior, middle, and posterior) of the planarian. The number of serotonin neurons was maximal in the head region. The ventral nerve cords gradually decreased in thickness from anterior to posterior body ends. Physiological action of exogenously applied serotonin was studied in G. tigrina for the first time. It was found that serotonin (0.1 and 1 µmol L-1) accelerated eye regeneration. The transcriptome sequencing performed for the first time for the planarian G. tigrina revealed the transcripts of the tryptophan hydroxylase (trph), amino acid decarboxylase (aadc) and serotonin transporter (sert) genes. The data obtained indicate the presence of the components of serotonin pathway in G. tigrina. The identified transcripts can take part in serotonin turnover and participate in the realization of biological effects of serotonin in planarians, associated with eyes regeneration and differentiation.
Collapse
Affiliation(s)
- Natalia D Kreshchenko
- Laboratory of the Mechanisms of Cell Redox-Ragulation, Institute of Cell Biophysics of the Russian Academy of Sciences, Moscow, Russia
| | - Artem M Ermakov
- Laboratory of Genome Researches, Institute of Theoretical and Experimental Biophysics of the Russian Academy of Science, Moscow, Russia
| |
Collapse
|
22
|
Geng X, Wang X, Liu K, Xing Y, Xu J, Li Z, Zhang H, Hu M, Gao P, Chen D, Liu W, Li K, Wei S. ShuYu capsule alleviates emotional and physical symptoms of premenstrual dysphoric disorder: Impact on ALLO decline and GABA A receptor δ subunit in the PAG area. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155549. [PMID: 38810551 DOI: 10.1016/j.phymed.2024.155549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/05/2024] [Accepted: 03/18/2024] [Indexed: 05/31/2024]
Abstract
Premenstrual dysphoric disorder (PMDD) is a severe subtype of premenstrual syndrome in women of reproductive age, with its pathogenesis linked to the heightened sensitivity of type A γ -aminobutyric acid receptors (GABAAR) to neuroactive steroid hormone changes, particularly allopregnanolone (ALLO). While a low dose of fluoxetine, a classic selective serotonin reuptake inhibitor, is commonly used as a first-line drug to alleviate emotional disorders in PMDD in clinical settings, its mechanism of action is related to ALLO-GABAA receptor function. However, treating PMDD requires attention to both emotional and physical symptoms, such as pain sensitivity. This study aims to investigate the efficacy of ShuYu capsules, a traditional Chinese medicine, in simultaneously treating emotional and physical symptoms in a rat model of PMDD. Specifically, our focus centres on the midbrain periaqueductal grey (PAG), a region associated with emotion regulation and susceptibility to hyperalgesia. Considering the underlying mechanisms of ALLO-GABAA receptor function in the PAG region, we conducted a series of experiments to evaluate and define the effects of ShuYu capsules and uncover the relationship between the drug's efficacy and ALLO concentration fluctuations on GABAA receptor function in the PAG region. Our findings demonstrate that ShuYu capsules significantly improved oestrous cycle-dependant depression-like behaviour and reduced stress-induced hyperalgesia in rats with PMDD. Similar to the low dose of fluoxetine, ShuYu capsules targeted and mitigated the sharp decline in ALLO, rescued the upregulation of GABAAR subunit function, and activated PAG neurons in PMDD rats. The observed effects of ShuYu capsules suggest a central mechanism underlying PMDD symptoms, involving ALLO_GABAA receptor function in the PAG region. This study highlights the potential of traditional Chinese medicine in addressing both emotional and physical symptoms associated with PMDD, shedding light on novel therapeutic approaches for this condition.
Collapse
Affiliation(s)
- Xiwen Geng
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Xinyu Wang
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Kun Liu
- High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Ying Xing
- High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Jialing Xu
- High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Zifa Li
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Hao Zhang
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Minghui Hu
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Peng Gao
- Institute of Pharmaceutical Research, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Dan Chen
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China
| | - Wei Liu
- Department of Encephalopathy, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Ji'nan 250001, China.
| | - Kejian Li
- High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China.
| | - Sheng Wei
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; High Level Key Disciplines of Traditional Chinese Medicine, Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan 250355, China.
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Li YM, Shen CY, Jiang JG. Sedative and hypnotic effects of the saponins from a traditional edible plant Liriope spicata Lour. in PCPA-induced insomnia mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118049. [PMID: 38484954 DOI: 10.1016/j.jep.2024.118049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/07/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liriope spicata Lour., a species listed in the catalogue of 'Medicinal and Edible Homologous Species', is traditionally used for the treatment of fatigue, restlessness, insomnia and constipation. AIM OF THE STUDY This study is aimed to evaluate the sedative and hypnotic effect of the saponins from a natural plant L. spicata Lour. in vivo. MATERIALS AND METHODS The total saponin (LSTS) and purified saponin (LSPS) were extracted from L. spicata, followed by a thorough analysis of their major components using the HPLC-MS. Subsequently, the therapeutic efficacy of LSTS and LSPS was evaluated by the improvement of anxiety and depression behaviors of the PCPA-induced mice. RESULTS LSTS and LSPS exhibited similar saponin compositions but differ in their composition ratios, with liriopesides-type saponins accounting for a larger proportion in LSTS. Studies demonstrated that both LSTS and LSPS can extend sleep duration and immobility time, while reducing sleep latency in PCPA-induced mice. However, there was no significant difference in weight change among the various mice groups. Elisa results indicated that the LSTS and LSPS could decrease levels of NE, DA, IL-6, and elevate the levels of 5-HT, NO, PGD2 and TNF-α in mice plasma. LSTS enhanced the expression of neurotransmitter receptors, while LSPS exhibited a more pronounced effect in regulating the expression of inflammatory factors. In conclusion, the saponins derived from L. spicata might hold promise as ingredients for developing health foods with sedative and hypnotic effects, potentially related to the modulation of serotonergic and GABAAergic neuron expression, as well as immunomodulatory process.
Collapse
Affiliation(s)
- Yi-Meng Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Chun-Yan Shen
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China; Southern Medical University, School of Traditional Chinese Medicine, Guangzhou, 510515, China
| | - Jian-Guo Jiang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
26
|
Rogers JF, Vandendoren M, Prather JF, Landen JG, Bedford NL, Nelson AC. Neural cell-types and circuits linking thermoregulation and social behavior. Neurosci Biobehav Rev 2024; 161:105667. [PMID: 38599356 PMCID: PMC11163828 DOI: 10.1016/j.neubiorev.2024.105667] [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: 01/03/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Understanding how social and affective behavioral states are controlled by neural circuits is a fundamental challenge in neurobiology. Despite increasing understanding of central circuits governing prosocial and agonistic interactions, how bodily autonomic processes regulate these behaviors is less resolved. Thermoregulation is vital for maintaining homeostasis, but also associated with cognitive, physical, affective, and behavioral states. Here, we posit that adjusting body temperature may be integral to the appropriate expression of social behavior and argue that understanding neural links between behavior and thermoregulation is timely. First, changes in behavioral states-including social interaction-often accompany changes in body temperature. Second, recent work has uncovered neural populations controlling both thermoregulatory and social behavioral pathways. We identify additional neural populations that, in separate studies, control social behavior and thermoregulation, and highlight their relevance to human and animal studies. Third, dysregulation of body temperature is linked to human neuropsychiatric disorders. Although body temperature is a "hidden state" in many neurobiological studies, it likely plays an underappreciated role in regulating social and affective states.
Collapse
Affiliation(s)
- Joseph F Rogers
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Morgane Vandendoren
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Jonathan F Prather
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Jason G Landen
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Nicole L Bedford
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Adam C Nelson
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA.
| |
Collapse
|
27
|
Miquel-Rio L, Sarriés-Serrano U, Sancho-Alonso M, Florensa-Zanuy E, Paz V, Ruiz-Bronchal E, Manashirov S, Campa L, Pilar-Cuéllar F, Bortolozzi A. ER stress in mouse serotonin neurons triggers a depressive phenotype alleviated by ketamine targeting eIF2α signaling. iScience 2024; 27:109787. [PMID: 38711453 PMCID: PMC11070602 DOI: 10.1016/j.isci.2024.109787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/19/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Depression is a devastating mood disorder that causes significant disability worldwide. Current knowledge of its pathophysiology remains modest and clear biological markers are lacking. Emerging evidence from human and animal models reveals persistent alterations in endoplasmic reticulum (ER) homeostasis, suggesting that ER stress-related signaling pathways may be targets for prevention and treatment. However, the neurobiological basis linking the pathways involved in depression-related ER stress remains unknown. Here, we report that an induced model of ER stress in mouse serotonin (5-HT) neurons is associated with reduced Egr1-dependent 5-HT cellular activity and 5-HT neurotransmission, resulting in neuroplasticity deficits in forebrain regions and a depressive-like phenotype. Ketamine administration engages downstream eIF2α signaling to trigger rapid neuroplasticity events that rescue the depressive-like effects. Collectively, these data identify ER stress in 5-HT neurons as a cellular pathway involved in the pathophysiology of depression and show that eIF2α is critical in eliciting ketamine's fast antidepressant effects.
Collapse
Affiliation(s)
- Lluis Miquel-Rio
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- University of Barcelona (UB), 08036 Barcelona, Spain
| | - Unai Sarriés-Serrano
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- University of the Basque Country UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - María Sancho-Alonso
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Eva Florensa-Zanuy
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Department of Molecular and Cellular Signaling, Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), University of Cantabria-CSIC, 39011 Santander, Spain
| | - Verónica Paz
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Esther Ruiz-Bronchal
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Sharon Manashirov
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- miCure Therapeutics LTD., Tel-Aviv 6423902, Israel
| | - Leticia Campa
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Fuencisla Pilar-Cuéllar
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Department of Molecular and Cellular Signaling, Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), University of Cantabria-CSIC, 39011 Santander, Spain
| | - Analia Bortolozzi
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
- Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
28
|
McManus E, Muhlert N, Duncan NW. InSpectro-Gadget: A Tool for Estimating Neurotransmitter and Neuromodulator Receptor Distributions for MRS Voxels. Neuroinformatics 2024; 22:135-145. [PMID: 38386228 DOI: 10.1007/s12021-024-09654-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] [Accepted: 12/21/2023] [Indexed: 02/23/2024]
Abstract
Magnetic resonance spectroscopy (MRS) is widely used to estimate concentrations of glutamate and γ -aminobutyric acid (GABA) in specific regions of the living human brain. As cytoarchitectural properties differ across the brain, interpreting these measurements can be assisted by having knowledge of such properties for the MRS region(s) studied. In particular, some knowledge of likely local neurotransmitter receptor patterns can potentially give insights into the mechanistic environment GABA- and glutamatergic neurons are functioning in. This may be of particular utility when comparing two or more regions, given that the receptor populations may differ substantially across them. At the same time, when studying MRS data from multiple participants or timepoints, the homogeneity of the sample becomes relevant, as measurements taken from areas with different cytoarchitecture may be difficult to compare. To provide insights into the likely cytoarchitectural environment of user-defined regions-of-interest, we produced an easy to use tool - InSpectro-Gadget - that interfaces with receptor mRNA expression information from the Allen Human Brain Atlas. This Python tool allows users to input masks and automatically obtain a graphical overview of the receptor population likely to be found within. This includes comparison between multiple masks or participants where relevant. The receptors and receptor subunit genes featured include GABA- and glutamatergic classes, along with a wide range of neuromodulators. The functionality of the tool is explained here and its use is demonstrated through a set of example analyses. The tool is available at https://github.com/lizmcmanus/Inspectro-Gadget .
Collapse
Affiliation(s)
| | - Nils Muhlert
- School of Health Sciences, University of Manchester, Manchester, UK
| | - Niall W Duncan
- Graduate Institute of Mind, Brain and Consciousness, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
29
|
Wang H, Sun Y, Wang W, Wang X, Zhang J, Bai Y, Wang K, Luan L, Yan J, Qin L. Mapping the 5-HTergic neural pathways in perimenopausal mice and elucidating the role of oestrogen receptors in 5-HT neurotransmission. Heliyon 2024; 10:e27976. [PMID: 38510058 PMCID: PMC10951590 DOI: 10.1016/j.heliyon.2024.e27976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Perimenopausal syndrome (PMS) encompasses neuropsychiatric symptoms, such as hot flashes and depression, which are associated with alterations in the 5-HTergic neural pathway in the brain. However, the specific changes and mechanisms underlying these alterations remain unclear. In this study, ovariectomized mice were used to successfully establish a perimenopause model, and the changes in the expression of 5-HT and its receptors (5-HT1AR and 5-HT2AR) across 72 brain regions in these ovariectomized mice were assessed by immunohistochemistry. Although both 5-HT and 5-HT1AR were widely expressed throughout the brain, only a limited number of regions expressed 5-HT2AR. Notably, decreased expression of 5-HT was observed across almost all brain regions in the ovariectomy (OVX) group compared with the Sham group. Altered expression of both receptors was found within areas related to hot flashes (the preoptic area) or mood disorders (the amygdala). Additionally, reduced oestrogen receptor (ER)α/β expression was detected in cells in the raphe nucleus (RN), an area known to regulate body temperature. Results showed that ERα/β positively regulate the transcriptional activity of the enzymes TPH2/MAOA, which are involved in serotonin metabolism during perimenopause. This study revealed the changes in 5-HT neuropathways (5-HT, 5-HT1AR and 5-HT2AR) in perimenopausal mice, mainly in brain regions related to regulation of the body temperature, mood, sleep and memory. This study clarified that the expression of oestrogen receptor decreased in perimenopause, which regulated the transcription levels of TPH2 and MAOA, and ultimately led to the reduction of 5-HT content, providing a new target for clinical diagnosis and treatment of perimenopausal diseases.
Collapse
Affiliation(s)
- Hanfei Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yanrong Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Wenjuan Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiangqiu Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jinglin Zhang
- Department of Dental Medicine, School of Dental Medicine, Yuncheng Vocational Nursing College, Yuncheng, 044000, China
| | - Yu Bai
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ke Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Liju Luan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Junhao Yan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lihua Qin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| |
Collapse
|
30
|
Hou G, Hao M, Duan J, Han MH. The Formation and Function of the VTA Dopamine System. Int J Mol Sci 2024; 25:3875. [PMID: 38612683 PMCID: PMC11011984 DOI: 10.3390/ijms25073875] [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: 10/20/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The midbrain dopamine system is a sophisticated hub that integrates diverse inputs to control multiple physiological functions, including locomotion, motivation, cognition, reward, as well as maternal and reproductive behaviors. Dopamine is a neurotransmitter that binds to G-protein-coupled receptors. Dopamine also works together with other neurotransmitters and various neuropeptides to maintain the balance of synaptic functions. The dysfunction of the dopamine system leads to several conditions, including Parkinson's disease, Huntington's disease, major depression, schizophrenia, and drug addiction. The ventral tegmental area (VTA) has been identified as an important relay nucleus that modulates homeostatic plasticity in the midbrain dopamine system. Due to the complexity of synaptic transmissions and input-output connections in the VTA, the structure and function of this crucial brain region are still not fully understood. In this review article, we mainly focus on the cell types, neurotransmitters, neuropeptides, ion channels, receptors, and neural circuits of the VTA dopamine system, with the hope of obtaining new insight into the formation and function of this vital brain region.
Collapse
Affiliation(s)
- Guoqiang Hou
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mei Hao
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiawen Duan
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ming-Hu Han
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
31
|
Hou X, Jing J, Jiang Y, Huang X, Xian Q, Lei T, Zhu J, Wong KF, Zhao X, Su M, Li D, Liu L, Qiu Z, Sun L. Nanobubble-actuated ultrasound neuromodulation for selectively shaping behavior in mice. Nat Commun 2024; 15:2253. [PMID: 38480733 PMCID: PMC10937988 DOI: 10.1038/s41467-024-46461-y] [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: 02/24/2023] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
Ultrasound is an acoustic wave which can noninvasively penetrate the skull to deep brain regions, enabling neuromodulation. However, conventional ultrasound's spatial resolution is diffraction-limited and low-precision. Here, we report acoustic nanobubble-mediated ultrasound stimulation capable of localizing ultrasound's effects to only the desired brain region in male mice. By varying the delivery site of nanobubbles, ultrasound could activate specific regions of the mouse motor cortex, evoking EMG signaling and limb movement, and could also, separately, activate one of two nearby deep brain regions to elicit distinct behaviors (freezing or rotation). Sonicated neurons displayed reversible, low-latency calcium responses and increased c-Fos expression in the sub-millimeter-scale region with nanobubbles present. Ultrasound stimulation of the relevant region also modified depression-like behavior in a mouse model. We also provide evidence of a role for mechanosensitive ion channels. Altogether, our treatment scheme allows spatially-targetable, repeatable and temporally-precise activation of deep brain circuits for neuromodulation without needing genetic modification.
Collapse
Affiliation(s)
- Xuandi Hou
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Jianing Jing
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Yizhou Jiang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Xiaohui Huang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Quanxiang Xian
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Ting Lei
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Jiejun Zhu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, Guangdong, China
| | - Kin Fung Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Xinyi Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Min Su
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Danni Li
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Langzhou Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China
| | - Zhihai Qiu
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, 519031, Guangdong, China
| | - Lei Sun
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, 999077, Hong Kong SAR, PR China.
| |
Collapse
|
32
|
Yao H, Wang X, Chi J, Chen H, Liu Y, Yang J, Yu J, Ruan Y, Xiang X, Pi J, Xu JF. Exploring Novel Antidepressants Targeting G Protein-Coupled Receptors and Key Membrane Receptors Based on Molecular Structures. Molecules 2024; 29:964. [PMID: 38474476 DOI: 10.3390/molecules29050964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Major Depressive Disorder (MDD) is a complex mental disorder that involves alterations in signal transmission across multiple scales and structural abnormalities. The development of effective antidepressants (ADs) has been hindered by the dominance of monoamine hypothesis, resulting in slow progress. Traditional ADs have undesirable traits like delayed onset of action, limited efficacy, and severe side effects. Recently, two categories of fast-acting antidepressant compounds have surfaced, dissociative anesthetics S-ketamine and its metabolites, as well as psychedelics such as lysergic acid diethylamide (LSD). This has led to structural research and drug development of the receptors that they target. This review provides breakthroughs and achievements in the structure of depression-related receptors and novel ADs based on these. Cryo-electron microscopy (cryo-EM) has enabled researchers to identify the structures of membrane receptors, including the N-methyl-D-aspartate receptor (NMDAR) and the 5-hydroxytryptamine 2A (5-HT2A) receptor. These high-resolution structures can be used for the development of novel ADs using virtual drug screening (VDS). Moreover, the unique antidepressant effects of 5-HT1A receptors in various brain regions, and the pivotal roles of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) and tyrosine kinase receptor 2 (TrkB) in regulating synaptic plasticity, emphasize their potential as therapeutic targets. Using structural information, a series of highly selective ADs were designed based on the different role of receptors in MDD. These molecules have the favorable characteristics of rapid onset and low adverse drug reactions. This review offers researchers guidance and a methodological framework for the structure-based design of ADs.
Collapse
Affiliation(s)
- Hanbo Yao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Xiaodong Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiaxin Chi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Haorong Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yilin Liu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiayi Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiaqi Yu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Xufu Xiang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
33
|
Wang X, Fu SQ, Yuan X, Yu F, Ji Q, Tang HW, Li RK, Huang S, Huang PQ, Qin WT, Zuo H, Du C, Yao LL, Li H, Li J, Li DX, Yang Y, Xiao SY, Tulamaiti A, Wang XF, Dai CH, Zhang X, Jiang SH, Hu LP, Zhang XL, Zhang ZG. A GAPDH serotonylation system couples CD8 + T cell glycolytic metabolism to antitumor immunity. Mol Cell 2024; 84:760-775.e7. [PMID: 38215751 DOI: 10.1016/j.molcel.2023.12.015] [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: 03/06/2023] [Revised: 10/04/2023] [Accepted: 12/12/2023] [Indexed: 01/14/2024]
Abstract
Apart from the canonical serotonin (5-hydroxytryptamine [5-HT])-receptor signaling transduction pattern, 5-HT-involved post-translational serotonylation has recently been noted. Here, we report a glyceraldehyde-3-phosphate dehydrogenase (GAPDH) serotonylation system that promotes the glycolytic metabolism and antitumor immune activity of CD8+ T cells. Tissue transglutaminase 2 (TGM2) transfers 5-HT to GAPDH glutamine 262 and catalyzes the serotonylation reaction. Serotonylation supports the cytoplasmic localization of GAPDH, which induces a glycolytic metabolic shift in CD8+ T cells and contributes to antitumor immunity. CD8+ T cells accumulate intracellular 5-HT for serotonylation through both synthesis by tryptophan hydroxylase 1 (TPH1) and uptake from the extracellular compartment via serotonin transporter (SERT). Monoamine oxidase A (MAOA) degrades 5-HT and acts as an intrinsic negative regulator of CD8+ T cells. The adoptive transfer of 5-HT-producing TPH1-overexpressing chimeric antigen receptor T (CAR-T) cells induced a robust antitumor response. Our findings expand the known range of neuroimmune interaction patterns by providing evidence of receptor-independent serotonylation post-translational modification.
Collapse
Affiliation(s)
- Xu Wang
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China.
| | - Sheng-Qiao Fu
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Xiao Yuan
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Feng Yu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, P.R. China
| | - Qian Ji
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Hao-Wen Tang
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Rong-Kun Li
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Shan Huang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Pei-Qi Huang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Wei-Ting Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Hao Zuo
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, P.R. China
| | - Chang Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Lin-Li Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Hui Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Jun Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Dong-Xue Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Yan Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Shu-Yu Xiao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Aziguli Tulamaiti
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Xue-Feng Wang
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Chun-Hua Dai
- Department of Radiation Oncology, Cancer Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P.R. China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, P.R. China.
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Li-Peng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Xue-Li Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| | - Zhi-Gang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, P.R. China.
| |
Collapse
|
34
|
De Filippo R, Schmitz D. Synthetic surprise as the foundation of the psychedelic experience. Neurosci Biobehav Rev 2024; 157:105538. [PMID: 38220035 PMCID: PMC10839673 DOI: 10.1016/j.neubiorev.2024.105538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/16/2024]
Abstract
Psychedelic agents, such as LSD and psilocybin, induce marked alterations in consciousness via activation of the 5-HT2A receptor (5-HT2ARs). We hypothesize that psychedelics enforce a state of synthetic surprise through the biased activation of the 5-HTRs system. This idea is informed by recent insights into the role of 5-HT in signaling surprise. The effects on consciousness, explained by the cognitive penetrability of perception, can be described within the predictive coding framework where surprise corresponds to prediction error, the mismatch between predictions and actual sensory input. Crucially, the precision afforded to the prediction error determines its effect on priors, enabling a dynamic interaction between top-down expectations and incoming sensory data. By integrating recent findings on predictive coding circuitry and 5-HT2ARs transcriptomic data, we propose a biological implementation with emphasis on the role of inhibitory interneurons. Implications arise for the clinical use of psychedelics, which may rely primarily on their inherent capacity to induce surprise in order to disrupt maladaptive patterns.
Collapse
Affiliation(s)
- Roberto De Filippo
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany.
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Einstein Center for Neuroscience, 10117 Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, 10117 Berlin, Germany; Humboldt-Universität zu Berlin, Bernstein Center for Computational Neuroscience, Philippstr. 13, 10115 Berlin, Germany
| |
Collapse
|
35
|
Li C, McElroy BD, Phillips J, McCloskey NS, Shi X, Unterwald EM, Kirby LG. Role of α1-GABA A receptors in the serotonergic dorsal raphe nucleus in models of opioid reward, anxiety, and depression. J Psychopharmacol 2024; 38:188-199. [PMID: 38293836 PMCID: PMC10921389 DOI: 10.1177/02698811241227672] [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] [Indexed: 02/01/2024]
Abstract
BACKGROUND The serotonin (5-hydroxytryptamine (5-HT))-mediated system plays an important role in stress-related psychiatric disorders and substance abuse. Our previous studies showed that stress and drug exposure can modulate the dorsal raphe nucleus (DRN)-5-HT system via γ-aminobutyric acid (GABA)A receptors. Moreover, GABAA receptor-mediated inhibition of serotonergic DRN neurons is required for stress-induced reinstatement of opioid seeking. AIM/METHODS To further test the role of GABAA receptors in the 5-HT system in stress and opioid-sensitive behaviors, our current study generated mice with conditional genetic deletions of the GABAA α1 subunit to manipulate GABAA receptors in either the DRN or the entire population of 5-HT neurons. The GABAA α1 subunit is a constituent of the most abundant GABAA subtype in the brain and the most highly expressed subunit in 5-HT DRN neurons. RESULTS Our results showed that mice with DRN-specific knockout of α1-GABAA receptors exhibited a normal phenotype in tests of anxiety- and depression-like behaviors as well as swim stress-induced reinstatement of morphine-conditioned place preference. By contrast, mice with 5-HT neuron-specific knockout of α1-GABAA receptors exhibited an anxiolytic phenotype at baseline and increased sensitivity to post-morphine withdrawal-induced anxiety. CONCLUSIONS Our data suggest that GABAA receptors on 5-HT neurons contribute to anxiety-like behaviors and sensitivity of those behaviors to opioid withdrawal.
Collapse
Affiliation(s)
- Chen Li
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Bryan D McElroy
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Jared Phillips
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville TN, USA
| | - Nicholas S McCloskey
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Xiangdang Shi
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Ellen M Unterwald
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| | - Lynn G Kirby
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
36
|
Weber LM, Divecha HR, Tran MN, Kwon SH, Spangler A, Montgomery KD, Tippani M, Bharadwaj R, Kleinman JE, Page SC, Hyde TM, Collado-Torres L, Maynard KR, Martinowich K, Hicks SC. The gene expression landscape of the human locus coeruleus revealed by single-nucleus and spatially-resolved transcriptomics. eLife 2024; 12:RP84628. [PMID: 38266073 PMCID: PMC10945708 DOI: 10.7554/elife.84628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
Norepinephrine (NE) neurons in the locus coeruleus (LC) make long-range projections throughout the central nervous system, playing critical roles in arousal and mood, as well as various components of cognition including attention, learning, and memory. The LC-NE system is also implicated in multiple neurological and neuropsychiatric disorders. Importantly, LC-NE neurons are highly sensitive to degeneration in both Alzheimer's and Parkinson's disease. Despite the clinical importance of the brain region and the prominent role of LC-NE neurons in a variety of brain and behavioral functions, a detailed molecular characterization of the LC is lacking. Here, we used a combination of spatially-resolved transcriptomics and single-nucleus RNA-sequencing to characterize the molecular landscape of the LC region and the transcriptomic profile of LC-NE neurons in the human brain. We provide a freely accessible resource of these data in web-accessible and downloadable formats.
Collapse
Affiliation(s)
- Lukas M Weber
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Matthew N Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Abby Spangler
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Kelsey D Montgomery
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Rahul Bharadwaj
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of MedicineBaltimoreUnited States
- Department of Neurology, Johns Hopkins School of MedicineBaltimoreUnited States
| | | | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of MedicineBaltimoreUnited States
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins School of MedicineBaltimoreUnited States
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of MedicineBaltimoreUnited States
- The Kavli Neuroscience Discovery Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
| |
Collapse
|
37
|
O’Connell CJ, Brown RS, Peach TM, Traubert OD, Schwierling HC, Notorgiacomo GA, Robson MJ. Strain in the Midbrain: Impact of Traumatic Brain Injury on the Central Serotonin System. Brain Sci 2024; 14:51. [PMID: 38248266 PMCID: PMC10813794 DOI: 10.3390/brainsci14010051] [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: 10/31/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Traumatic brain injury (TBI) is a pervasive public health crisis that severely impacts the quality of life of affected individuals. Like peripheral forms of trauma, TBI results from extraordinarily heterogeneous environmental forces being imparted on the cranial space, resulting in heterogeneous disease pathologies. This has made therapies for TBI notoriously difficult to develop, and currently, there are no FDA-approved pharmacotherapies specifically for the acute or chronic treatment of TBI. TBI is associated with changes in cognition and can precipitate the onset of debilitating psychiatric disorders like major depressive disorder (MDD), generalized anxiety disorder (GAD), and post-traumatic stress disorder (PTSD). Complicating these effects of TBI, FDA-approved pharmacotherapies utilized to treat these disorders often fail to reach the desired level of efficacy in the context of neurotrauma. Although a complicated association, decades of work have linked central serotonin (5-HT) neurotransmission as being involved in the etiology of a myriad of neuropsychiatric disorders, including MDD and GAD. 5-HT is a biogenic monoamine neurotransmitter that is highly conserved across scales of biology. Though the majority of 5-HT is isolated to peripheral sites such as the gastrointestinal (GI) tract, 5-HT neurotransmission within the CNS exerts exquisite control over diverse biological functions, including sleep, appetite and respiration, while simultaneously establishing normal mood, perception, and attention. Although several key studies have begun to elucidate how various forms of neurotrauma impact central 5-HT neurotransmission, a full determination of precisely how TBI disrupts the highly regulated dynamics of 5-HT neuron function and/or 5-HT neurotransmission has yet to be conceptually or experimentally resolved. The purpose of the current review is, therefore, to integrate the disparate bodies of 5-HT and TBI research and synthesize insight into how new combinatorial research regarding 5-HT neurotransmission and TBI may offer an informed perspective into the nature of TBI-induced neuropsychiatric complications.
Collapse
Affiliation(s)
- Christopher J. O’Connell
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | - Ryan S. Brown
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | - Taylor M. Peach
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | - Owen D. Traubert
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA;
| | - Hana C. Schwierling
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | | | - Matthew J. Robson
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
38
|
Harripaul R, Morini E, Salani M, Logan E, Kirchner E, Bolduc J, Chekuri A, Currall B, Yadav R, Erdin S, Talkowski ME, Gao D, Slaugenhaupt S. Transcriptome analysis in a humanized mouse model of familial dysautonomia reveals tissue-specific gene expression disruption in the peripheral nervous system. Sci Rep 2024; 14:570. [PMID: 38177237 PMCID: PMC10766950 DOI: 10.1038/s41598-023-51137-6] [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: 10/31/2023] [Accepted: 12/31/2023] [Indexed: 01/06/2024] Open
Abstract
Familial dysautonomia (FD) is a rare recessive neurodevelopmental disease caused by a splice mutation in the Elongator acetyltransferase complex subunit 1 (ELP1) gene. This mutation results in a tissue-specific reduction of ELP1 protein, with the lowest levels in the central and peripheral nervous systems (CNS and PNS, respectively). FD patients exhibit complex neurological phenotypes due to the loss of sensory and autonomic neurons. Disease symptoms include decreased pain and temperature perception, impaired or absent myotatic reflexes, proprioceptive ataxia, and progressive retinal degeneration. While the involvement of the PNS in FD pathogenesis has been clearly recognized, the underlying mechanisms responsible for the preferential neuronal loss remain unknown. In this study, we aimed to elucidate the molecular mechanisms underlying FD by conducting a comprehensive transcriptome analysis of neuronal tissues from the phenotypic mouse model TgFD9; Elp1Δ20/flox. This mouse recapitulates the same tissue-specific ELP1 mis-splicing observed in patients while modeling many of the disease manifestations. Comparison of FD and control transcriptomes from dorsal root ganglion (DRG), trigeminal ganglion (TG), medulla (MED), cortex, and spinal cord (SC) showed significantly more differentially expressed genes (DEGs) in the PNS than the CNS. We then identified genes that were tightly co-expressed and functionally dependent on the level of full-length ELP1 transcript. These genes, defined as ELP1 dose-responsive genes, were combined with the DEGs to generate tissue-specific dysregulated FD signature genes and networks. Within the PNS networks, we observed direct connections between Elp1 and genes involved in tRNA synthesis and genes related to amine metabolism and synaptic signaling. Importantly, transcriptomic dysregulation in PNS tissues exhibited enrichment for neuronal subtype markers associated with peptidergic nociceptors and myelinated sensory neurons, which are known to be affected in FD. In summary, this study has identified critical tissue-specific gene networks underlying the etiology of FD and provides new insights into the molecular basis of the disease.
Collapse
Affiliation(s)
- Ricardo Harripaul
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Elisabetta Morini
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Monica Salani
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Emily Logan
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Emily Kirchner
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Jessica Bolduc
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Anil Chekuri
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Benjamin Currall
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Rachita Yadav
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Serkan Erdin
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Michael E Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Dadi Gao
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Susan Slaugenhaupt
- Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Boston, MA, USA.
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Luppi AI, Girn M, Rosas FE, Timmermann C, Roseman L, Erritzoe D, Nutt DJ, Stamatakis EA, Spreng RN, Xing L, Huttner WB, Carhart-Harris RL. A role for the serotonin 2A receptor in the expansion and functioning of human transmodal cortex. Brain 2024; 147:56-80. [PMID: 37703310 DOI: 10.1093/brain/awad311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
Integrating independent but converging lines of research on brain function and neurodevelopment across scales, this article proposes that serotonin 2A receptor (5-HT2AR) signalling is an evolutionary and developmental driver and potent modulator of the macroscale functional organization of the human cerebral cortex. A wealth of evidence indicates that the anatomical and functional organization of the cortex follows a unimodal-to-transmodal gradient. Situated at the apex of this processing hierarchy-where it plays a central role in the integrative processes underpinning complex, human-defining cognition-the transmodal cortex has disproportionately expanded across human development and evolution. Notably, the adult human transmodal cortex is especially rich in 5-HT2AR expression and recent evidence suggests that, during early brain development, 5-HT2AR signalling on neural progenitor cells stimulates their proliferation-a critical process for evolutionarily-relevant cortical expansion. Drawing on multimodal neuroimaging and cross-species investigations, we argue that, by contributing to the expansion of the human cortex and being prevalent at the apex of its hierarchy in the adult brain, 5-HT2AR signalling plays a major role in both human cortical expansion and functioning. Owing to its unique excitatory and downstream cellular effects, neuronal 5-HT2AR agonism promotes neuroplasticity, learning and cognitive and psychological flexibility in a context-(hyper)sensitive manner with therapeutic potential. Overall, we delineate a dual role of 5-HT2ARs in enabling both the expansion and modulation of the human transmodal cortex.
Collapse
Affiliation(s)
- Andrea I Luppi
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
- Leverhulme Centre for the Future of Intelligence, University of Cambridge, Cambridge, CB2 1SB, UK
- The Alan Turing Institute, London, NW1 2DB, UK
| | - Manesh Girn
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
| | - Fernando E Rosas
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
- Data Science Institute, Imperial College London, London, SW7 2AZ, UK
- Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Christopher Timmermann
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Leor Roseman
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David Erritzoe
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David J Nutt
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Emmanuel A Stamatakis
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - R Nathan Spreng
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Robin L Carhart-Harris
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
40
|
Ligneul R, Mainen ZF. Serotonin. Curr Biol 2023; 33:R1216-R1221. [PMID: 38052167 DOI: 10.1016/j.cub.2023.09.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Serotonin, also known as 5-hydroxytryptamine or 5-HT, is a neuromodulator widely recognized for its role in various psychoactive drugs. These drugs can exhibit antidepressant, antipsychotic, anxiolytic, empathogenic, or psychedelic effects, depending on their specific interactions with the serotonin system as well as other neuromodulators such as noradrenaline, dopamine, and oxytocin. This has led to a widespread belief that the neurochemical processes taking place deep inside our brains affect our subjective experiences and mental health. However, a scientific understanding of how neuromodulators' functions relate to drug effects remains elusive.
Collapse
Affiliation(s)
- Romain Ligneul
- Champalimaud Foundation, Lisbon, Portugal; INSERM, Centre de Recherche en Neurosciences de Lyon, Bron, France
| | | |
Collapse
|
41
|
Cheng H, Lou Q, Lai N, Chen L, Zhang S, Fei F, Gao C, Wu S, Han F, Liu J, Guo Y, Chen Z, Xu C, Wang Y. Projection-defined median raphe Pet + subpopulations are diversely implicated in seizure. Neurobiol Dis 2023; 189:106358. [PMID: 37977434 DOI: 10.1016/j.nbd.2023.106358] [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: 08/24/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
The raphe nuclei, the primary resource of forebrain 5-HT, play an important but heterogeneous role in regulating subcortical excitabilities. Fundamental circuit organizations of different median raphe (MR) subsystems are far from completely understood. In the present study, using cell-specific viral tracing, Ca2+ fiber photometry and epilepsy model, we map out the forebrain efferent and afferent of different MR Pet+ subpopulations and their divergent roles in epilepsy. We found that PetMR neurons send both collateral and parallel innervations to different downstream regions through different subpopulations. Notably, CA3-projecting PetMR subpopulations are largely distinct from habenula (Hb)-projecting PetMR subpopulations in anatomical distribution and topological organization, while majority of the CA3-projecting PetMR subpopulations are overlapped with the medial septum (MS)-projecting PetMR subpopulations. Further, using Ca2+ fiber photometry, we monitor activities of PetMR neurons in hippocampal-kindling seizure, a classical epilepsy model with pathological mechanisms caused by excitation-inhibition imbalance. We found that soma activities of PetMR neurons are heterogeneous during different periods of generalized seizures. These divergent activities are contributed by different projection-defined PetMR subpopulations, manifesting as increased activities in CA3 but decreased activity in Hb resulting from their upstream differences. Together, our findings provide a novel framework of MR subsystems showing that projection-defined MR Pet+ subpopulations are topologically heterogenous with divergent circuit connections and are diversely implicated in seizures. This may help in the understanding of heterogeneous nature of MR 5-HTergic subsystems and the paradox roles of 5-HTergic systems in epilepsy.
Collapse
Affiliation(s)
- Heming Cheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qiuwen Lou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Nanxi Lai
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Liying Chen
- Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Shuo Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310003, China
| | - Fan Fei
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chenshu Gao
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shuangshuang Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jinggen Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi Guo
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310061, China.
| |
Collapse
|
42
|
Ma H, Gu L, Wang Y, Xu Q, Zhang Y, Shao W, Yu Q, Lian X, Liu L, Gu J, Ji N, Liu X, Nagayasu K, Zhang H. The States of Different 5-HT Receptors Located in the Dorsal Raphe Nucleus Are Crucial for Regulating the Awakening During General Anesthesia. Mol Neurobiol 2023; 60:6931-6948. [PMID: 37516665 DOI: 10.1007/s12035-023-03519-0] [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: 03/13/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2023]
Abstract
General anesthesia is widely used in various clinical practices due to its ability to cause loss of consciousness. However, the exact mechanism of anesthesia-induced unconsciousness remains unclear. It is generally thought that arousal-related brain nuclei are involved. 5-Hydroxytryptamine (5-HT) is closely associated with sleep arousal. Here, we explore the role of the 5-HT system in anesthetic awakening through pharmacological interventions and optogenetic techniques. Our data showed that exogenous administration of 5-hydroxytryptophan (5-HTP) and optogenetic activation of 5-HT neurons in the dorsal raphe nucleus (DR) could significantly shorten the emergence time of sevoflurane anesthesia in mice, suggesting that regulation of the 5-HT system using both endogenous and exogenous approaches could mediate delayed emergence. In addition, we first discovered that the different 5-HT receptors located in the DR, known as 5-HT autoreceptors, are essential for the regulation of general anesthetic awakening, with 5-HT1A and 5-HT2A/C receptors playing a regulatory role. These results can provide a reliable theoretical basis as well as potential targets for clinical intervention to prevent delayed emergence and some postoperative risks.
Collapse
Affiliation(s)
- HaiXiang Ma
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
- Medical College of Jining Medical University, Ningji, 272067, Shandong, China
| | - LeYuan Gu
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - YuLing Wang
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Qing Xu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yuanli Zhang
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - WeiHui Shao
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Qian Yu
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - XiTing Lian
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lu Liu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - JiaXuan Gu
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Na Ji
- Department of Anesthesia, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - XiaoLing Liu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - HongHai Zhang
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China.
- Medical College of Jining Medical University, Ningji, 272067, Shandong, China.
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310006, China.
| |
Collapse
|
43
|
Huang J, Huang W, Yi J, Deng Y, Li R, Chen J, Shi J, Qiu Y, Wang T, Chen X, Zhang X, Xiang AP. Mesenchymal stromal cells alleviate depressive and anxiety-like behaviors via a lung vagal-to-brain axis in male mice. Nat Commun 2023; 14:7406. [PMID: 37973914 PMCID: PMC10654509 DOI: 10.1038/s41467-023-43150-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Major depressive disorder (MDD) is one of the most common and disabling mental disorders, and current strategies remain inadequate. Although mesenchymal stromal cells (MSCs) have shown beneficial effects in experimental models of depression, underlying mechanisms remain elusive. Here, using murine depression models, we demonstrated that MSCs could alleviate depressive and anxiety-like behaviors not due to a reduction in proinflammatory cytokines, but rather activation of dorsal raphe nucleus (DRN) 5-hydroxytryptamine (5-HT) neurons. Mechanistically, peripheral delivery of MSCs activated pulmonary innervating vagal sensory neurons, which projected to the nucleus tractus solitarius, inducing the release of 5-HT in DRN. Furthermore, MSC-secreted brain-derived neurotrophic factor activated lung sensory neurons through tropomyosin receptor kinase B (TrkB), and inhalation of a TrkB agonist also achieved significant therapeutic effects in male mice. This study reveals a role of peripheral MSCs in regulating central nervous system function and demonstrates a potential "lung vagal-to-brain axis" strategy for MDD.
Collapse
Affiliation(s)
- Jing Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Junzhe Yi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yiwen Deng
- Key Laboratory of Medical Transformation of Jiujiang, Jiujiang University, Jiujiang, Jiangxi, 332005, China
| | - Ruijie Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jieying Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jiahao Shi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yuan Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
44
|
Collins SA, Stinson HE, Himes A, Nestor-Kalinoski A, Ninan I. Sex-specific modulation of the medial prefrontal cortex by glutamatergic median raphe neurons. SCIENCE ADVANCES 2023; 9:eadg4800. [PMID: 37948526 PMCID: PMC10637752 DOI: 10.1126/sciadv.adg4800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023]
Abstract
A substantial proportion of raphe neurons are glutamatergic. However, little is known about how these glutamatergic neurons modulate the forebrain. We investigated how glutamatergic median raphe nucleus (MRN) input modulates the medial prefrontal cortex (mPFC), a critical component of fear circuitry. We show that vesicular glutamate transporter 3 (VGLUT3)-expressing MRN neurons activate VGLUT3- and somatostatin-expressing neurons in the mPFC. Consistent with this modulation of mPFC GABAergic neurons, activation of MRN (VGLUT3) neurons enhances GABAergic transmission in mPFC pyramidal neurons and attenuates fear memory in female but not male mice. Serotonin plays a key role in MRN (VGLUT3) neuron-mediated GABAergic plasticity in the mPFC. In agreement with these female-specific effects, we observed sex differences in glutamatergic transmission onto MRN (VGLUT3) neurons and in mPFC (VGLUT3) neuron-mediated dual release of glutamate and GABA. Our results demonstrate a cell type-specific modulation of the mPFC by MRN (VGLUT3) neurons and reveal a sex-specific role of this neuromodulation in mPFC synaptic plasticity.
Collapse
Affiliation(s)
- Stuart A. Collins
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Hannah E. Stinson
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Amanda Himes
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Andrea Nestor-Kalinoski
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ipe Ninan
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
45
|
Xu T, Cao L, Duan J, Li Y, Li Y, Hu Z, Li S, Zhang M, Wang G, Guo F, Lu J. Uncovering the role of FOXA2 in the Development of Human Serotonin Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303884. [PMID: 37679064 PMCID: PMC10646255 DOI: 10.1002/advs.202303884] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/08/2023] [Indexed: 09/09/2023]
Abstract
Directed differentiation of serotonin neurons (SNs) from human pluripotent stem cells (hPSCs) provides a valuable tool for uncovering the mechanism of human SN development and the associated neuropsychiatric disorders. Previous studies report that FOXA2 is expressed by serotonergic progenitors (SNPs) and functioned as a serotonergic fate determinant in mouse. However, in the routine differentiation experiments, it is accidentally found that less SNs and more non-neuronal cells are obtained from SNP stage with higher percentage of FOXA2-positive cells. This phenomenon prompted them to question the role of FOXA2 as an intrinsic fate determinant for human SN differentiation. Herein, by direct differentiation of engineered hPSCs into SNs, it is found that the SNs are not derived from FOXA2-lineage cells; FOXA2-knockout hPSCs can still differentiate into mature and functional SNs with typical serotonergic identity; FOXA2 overexpression suppresses the SN differentiation, indicating that FOXA2 is not intrinsically required for human SN differentiation. Furthermore, repressing FOXA2 expression by retinoic acid (RA) and dynamically modulating Sonic Hedgehog (SHH) signaling pathway promotes human SN differentiation. This study uncovers the role of FOXA2 in human SN development and improves the differentiation efficiency of hPSCs into SNs by repressing FOXA2 expression.
Collapse
Affiliation(s)
- Ting Xu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Jinjin Duan
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Yingqi Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - You Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Zhangsen Hu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Shuanqing Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Meihui Zhang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Guanhao Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Fei Guo
- Key Laboratory of Receptor ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center)Frontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
- Suzhou Institute of Tongji UniversitySuzhou215101China
| |
Collapse
|
46
|
McElroy BD, Li C, McCloskey NS, Kirby LG. Sex differences in ethanol consumption and drinking despite negative consequences following adolescent social isolation stress in male and female rats. Physiol Behav 2023; 271:114322. [PMID: 37573960 PMCID: PMC10592127 DOI: 10.1016/j.physbeh.2023.114322] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/19/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Alcohol use disorder (AUD) is a debilitating psychiatric disorder characterized by drinking despite negative social and biological consequences. AUDs make up 71% of substance use disorders, with relapse rates as high as 80%. Current treatments stem from data conducted largely in males and fail to target the psychological distress motivating drinking in stress-vulnerable and at-risk populations. Here we employed a rat model and hypothesized that early life stress would reveal sex differences in ethanol intake and drinking despite negative consequences in adulthood. Rats were group housed or isolated postweaning to evaluate sex and stress effects on ethanol consumption in homecage drinking, self-administration (SA), and punished SA (drinking despite negative consequences) in adulthood. Stressed rats showed elevated homecage ethanol intake, an effect more pronounced in females. During SA, males were more sensitive to stress-induced elevations of drinking over time, but females drank more overall. Stressed rats, regardless of sex, responded more for ethanol than their non-stressed counterparts. Stressed females showed greater resistance to punishment-suppressed SA than stressed males, indicating a more stress-resistant drinking phenotype. Results support our hypothesis that adolescent social isolation stress enhances adult ethanol intake in a sex- and model-dependent manner with females being especially sensitive to early life stress-induced elevations in ethanol intake and punished SA in adulthood. Our findings echo the clinical literature which indicates that stress-vulnerable populations are more likely to 'self-medicate' with substances. Elucidating a potential mechanism that underlies why vulnerable populations 'self-medicate' with alcohol can lead towards developing catered pharmacotherapeutics that could reduce punishment-resistant drinking and relapse.
Collapse
Affiliation(s)
- Bryan D McElroy
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University Philadelphia, PA, 19140, United States of America.
| | - Chen Li
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University Philadelphia, PA, 19140, United States of America
| | - Nicholas S McCloskey
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University Philadelphia, PA, 19140, United States of America
| | - Lynn G Kirby
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University Philadelphia, PA, 19140, United States of America
| |
Collapse
|
47
|
Harripaul R, Morini E, Salani M, Logan E, Kirchner E, Bolduc J, Chekuri A, Currall B, Yadav R, Erdin S, Talkowski ME, Gao D, Slaugenhaupt S. Transcriptome analysis in a humanized mouse model of familial dysautonomia reveals tissue-specific gene expression disruption in the peripheral nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559870. [PMID: 37808686 PMCID: PMC10557663 DOI: 10.1101/2023.09.28.559870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Familial dysautonomia (FD) is a rare recessive neurodevelopmental disease caused by a splice mutation in the Elongator acetyltransferase complex subunit 1 ( ELP1 ) gene. This mutation results in a tissue-specific reduction of ELP1 protein, with the lowest levels in the central and peripheral nervous systems (CNS and PNS, respectively). FD patients exhibit complex neurological phenotypes due to the loss of sensory and autonomic neurons. Disease symptoms include decreased pain and temperature perception, impaired or absent myotatic reflexes, proprioceptive ataxia, and progressive retinal degeneration. While the involvement of the PNS in FD pathogenesis has been clearly recognized, the underlying mechanisms responsible for the preferential neuronal loss remain unknown. In this study, we aimed to elucidate the molecular mechanisms underlying FD by conducting a comprehensive transcriptome analysis of neuronal tissues from the phenotypic mouse model TgFD9 ; Elp1 Δ 20/flox . This mouse recapitulates the same tissue-specific ELP1 mis-splicing observed in patients while modeling many of the disease manifestations. Comparison of FD and control transcriptomes from dorsal root ganglion (DRG), trigeminal ganglion (TG), medulla (MED), cortex, and spinal cord (SC) showed significantly more differentially expressed genes (DEGs) in the PNS than the CNS. We then identified genes that were tightly co-expressed and functionally dependent on the level of full-length ELP1 transcript. These genes, defined as ELP1 dose-responsive genes, were combined with the DEGs to generate tissue-specific dysregulated FD signature genes and networks. Within the PNS networks, we observed direct connections between Elp1 and genes involved in tRNA synthesis and genes related to amine metabolism and synaptic signaling. Importantly, transcriptomic dysregulation in PNS tissues exhibited enrichment for neuronal subtype markers associated with peptidergic nociceptors and myelinated sensory neurons, which are known to be affected in FD. In summary, this study has identified critical tissue-specific gene networks underlying the etiology of FD and provides new insights into the molecular basis of the disease.
Collapse
|
48
|
Collins SA, Stinson HE, Himes A, Ninan I. Sex-specific modulation of the medial prefrontal cortex by glutamatergic median raphe neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555555. [PMID: 37693545 PMCID: PMC10491205 DOI: 10.1101/2023.08.30.555555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The current understanding of the neuromodulatory role of the median raphe nucleus (MRN) is primarily based on its putative serotonergic output. However, a significant proportion of raphe neurons are glutamatergic. The present study investigated how glutamatergic MRN input modulates the medial prefrontal cortex (mPFC), a critical component of the fear circuitry. Our studies show that VGLUT3-expressing MRN neurons modulate VGLUT3- and somatostatin-expressing neurons in the mPFC. Consistent with this modulation of mPFC GABAergic neurons, activation of MRN (VGLUT3) neurons suppresses mPFC pyramidal neuron activity and attenuates fear memory in female but not male mice. In agreement with these female-specific effects, we observed sex differences in glutamatergic transmission onto MRN (VGLUT3) neurons and mPFC (VGLUT3) neuron-mediated dual release of glutamate and GABA. Thus, our results demonstrate a cell type-specific modulation of the mPFC by MRN (VGLUT3) neurons and reveal a sex-specific role of this neuromodulation in mPFC synaptic plasticity and fear memory.
Collapse
Affiliation(s)
| | | | - Amanda Himes
- Department of Neurosciences, University of Toledo, Toledo, Ohio
| | - Ipe Ninan
- Department of Neurosciences, University of Toledo, Toledo, Ohio
| |
Collapse
|
49
|
Mays KC, Haiman JH, Janušonis S. An experimental platform for stochastic analyses of single serotonergic fibers in the mouse brain. Front Neurosci 2023; 17:1241919. [PMID: 37869509 PMCID: PMC10587471 DOI: 10.3389/fnins.2023.1241919] [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: 06/17/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
The self-organization of the serotonergic matrix, a massive axon meshwork in all vertebrate brains, is driven by the structural and dynamical properties of its constitutive elements. Each of these elements, a single serotonergic axon (fiber), has a unique trajectory and can be supported by a soma that executes one of the many available transcriptional programs. This "individuality" of serotonergic neurons necessitates the development of specialized methods for single-fiber analyses, both at the experimental and theoretical levels. We developed an integrated platform that facilitates experimental isolation of single serotonergic fibers in brain tissue, including regions with high fiber densities, and demonstrated the potential of their quantitative analyses based on stochastic modeling. Single fibers were visualized using two transgenic mouse models, one of which is the first implementation of the Brainbow toolbox in this system. The trajectories of serotonergic fibers were automatically traced in the three spatial dimensions with a novel algorithm, and their properties were captured with a single parameter associated with the directional von Mises-Fisher probability distribution. The system represents an end-to-end workflow that can be imported into various studies, including those investigating serotonergic dysfunction in brain disorders. It also supports new research directions inspired by single-fiber analyses in the serotonergic matrix, including supercomputing simulations and modeling in physics.
Collapse
Affiliation(s)
| | | | - Skirmantas Janušonis
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
50
|
Hou Y, Liang H, Fan C, Feng Y. 5-Hydroxytryptamine and postoperative nausea and vomiting after microvascular decompression surgery. J Clin Neurosci 2023; 116:27-31. [PMID: 37597331 DOI: 10.1016/j.jocn.2023.08.010] [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: 06/05/2023] [Revised: 08/05/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND The incidence of postoperative nausea and vomiting (PONV) after microvascular decompression (MVD) surgery is high; however, its underlying mechanisms remain unknown. Serum 5-hydroxytryptamine (5-HT) levels are elevated in patients with PONV. However, the relationship between 5-HT and patients experiencing PONV after MVD surgery is still unknown. Therefore, we hypothesized that 5-HT levels are associated with PONV after MVD surgery. METHODS This prospective study included 85 patients with hemifacial spasm who received MVD surgery. Blood samples were collected preoperatively, postoperatively, and on postoperative day 1, and cerebrospinal fluid samples were collected intraoperatively. 5-HT levels were detected by enzyme-linked immunosorbent assay (ELISA). The incidence and severity of PONV were evaluated at 2, 6, and 24 h after MVD surgery. RESULTS In the multivariate regression analysis, PONV within 24 h after MVD surgery was associated with elevated cerebrospinal fluid 5-HT levels [odds ratio (OR) = 1.21, 95% confidence interval (CI): 1.01-1.45, p = 0.044], and reduction of intraocular pressure [OR = 11.54, 95% CI: 1.43-92.84, p = 0.022]. Receiver operating characteristic curve analysis revealed an area under the curve of 0.873 (95% CI: 0.77-0.98, p < 0.001). CONCLUSION Our study found that the cerebrospinal fluid 5-HT levels is an independent risk factor for PONV within 24 h after MVD surgery.
Collapse
Affiliation(s)
- Yuantao Hou
- Department of Anesthesiology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China.
| | - Hansheng Liang
- Department of Anesthesiology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China.
| | - Cungang Fan
- Department of Neurosurgery, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China.
| | - Yi Feng
- Department of Anesthesiology, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, China.
| |
Collapse
|