1
|
Lynn MB, Geddes SD, Chahrour M, Maillé S, Caya-Bissonnette L, Harkin E, Harvey-Girard É, Haj-Dahmane S, Naud R, Béïque JC. Nonlinear recurrent inhibition through facilitating serotonin release in the raphe. Nat Neurosci 2025; 28:1024-1037. [PMID: 40175691 DOI: 10.1038/s41593-025-01912-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 02/06/2025] [Indexed: 04/04/2025]
Abstract
Serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) receive a constellation of long-range inputs, yet guiding principles of local circuit organization and underlying computations in this nucleus are largely unknown. Using inputs from the lateral habenula to interrogate the processing features of the mouse DRN, we uncovered 5-HT1A receptor-mediated recurrent connections between 5-HT neurons, refuting classical theories of autoinhibition. Cellular electrophysiology and imaging of a genetically encoded 5-HT sensor revealed that these recurrent inhibitory connections spanned the raphe, were slow, stochastic, strongly facilitating and gated spike output. These features collectively conveyed highly nonlinear dynamics to this network, generating excitation-driven inhibition and winner-take-all computations. In vivo optogenetic activation of lateral habenula inputs to DRN, at frequencies where these computations are predicted to ignite, transiently disrupted expression of a reward-conditioned response in an auditory conditioning task. Together, these data identify a core computation supported by an unsuspected slow serotonergic recurrent inhibitory network.
Collapse
Affiliation(s)
- Michael B Lynn
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Sean D Geddes
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mohamad Chahrour
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Sébastien Maillé
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Léa Caya-Bissonnette
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Emerson Harkin
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Érik Harvey-Girard
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Richard Naud
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Neural Dynamics and AI, University of Ottawa, Ottawa, Ontario, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Department of Physics, STEM Complex, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean-Claude Béïque
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Centre for Neural Dynamics and AI, University of Ottawa, Ottawa, Ontario, Canada.
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
2
|
Oubraim S, Hausknecht K, Micov V, Shen RY, Haj-Dahmane S. Chemogenetic inhibition of prefrontal cortex inputs to dorsal raphe reduces anxiety behaviors in male rat model of fetal alcohol spectrum disorder. Sci Rep 2025; 15:14397. [PMID: 40275074 PMCID: PMC12022358 DOI: 10.1038/s41598-025-99181-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Prenatal ethanol exposure (PE) causes Fetal Alcohol Spectrum Disorders (FASD), characterized by cognitive, behavioral, and emotional deficits, including anxiety and depression. PE-induced alteration in the function of dorsal raphe nucleus (DRN) serotonin (5-HT) neurons is thought to be major contributing factor for increased anxiety. However, the precise neuronal circuits involved are unknown. Using electrophysiology, optogenetics, chemogenetics, and behavioral approaches, we find that PE preferentially potentiates medial prefrontal cortex (mPFC) glutamatergic inputs, but not lateral habenula (LHb), to DRN 5-HT neurons projecting to mPFC. Additionally, PE also increases the strength of LHb but not mPFC excitatory inputs to DRN 5-HT neurons projecting to central amygdala (Ce). This input and target selective effect of PE was mediated by a circuit-specific increase in nitric oxide (NO) signaling. Importantly, chemogenetic inhibition of mPFC-DRN neuronal circuit blunted anxiety-like behaviors in PE rats. As such, our results unraveled the DRN neuronal circuitries affected by PE, which gate FASD-induced anxiety-like behaviors.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Kathryn Hausknecht
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Veronika Micov
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 1021 Main Street, Buffalo, NY, 14203, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA.
- University at Buffalo Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 1021 Main Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
3
|
Zhang W, Jin Y, Zhou FM. Chronic Fluoxetine Treatment Desensitizes Serotoninergic Inhibition of GABAergic Inputs and Intrinsic Excitability of Dorsal Raphe Serotonin Neurons. Brain Sci 2025; 15:384. [PMID: 40309832 PMCID: PMC12025439 DOI: 10.3390/brainsci15040384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/29/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Dorsal raphe serotonin (5-hydroxytryptamine, 5-HT) neurons are spontaneously active and release 5-HT that is critical for normal brain function and regulates mood and emotion. Serotonin reuptake inhibitors (SSRIs) increase the synaptic and extracellular 5-HT level and are effective in treating depression. Treatment of two weeks or longer is often required for SSRIs to produce clinical benefits. The cellular mechanism underlying this delay is not fully understood. Methods and Results: Using whole-cell patch clamp recording in brain slices, here we show that the GABAergic inputs inhibit the spike firing of raphe 5-HT neurons. This GABAergic regulation was reduced by 5-HT; additionally, this 5-HT effect was prevented by the G-protein-activated inwardly rectifying potassium (GirK) channel inhibitor tertiapin-Q, indicating a contribution of 5-HT activation of GirK channels in GABAergic presynaptic axon terminals. Equally important, after 14 days of treatment with fluoxetine, a widely used SSRI type antidepressant, the 5-HT inhibition of GABAergic inputs was downregulated. Furthermore, chronic fluoxetine treatment downregulated the 5-HT activation of the inhibitory GirK current in 5-HT neurons. Conclusions: Taken together, our results suggest that chronic fluoxetine treatment, by blocking 5-HT reuptake and hence increasing the extracellular 5-HT level, can downregulate the function of 5-HT1B receptors on the GABAergic afferent axon terminals synapsing onto 5-HT neurons, allowing extrinsic GABAergic neurons to more effectively influence 5-HT neurons; simultaneously, chronic fluoxetine treatment also downregulated somatic 5-HT autoreceptor-activated GirK channel-mediated hyperpolarization and decrease in input resistance, rendering 5-HT neurons resistant to autoinhibition and leading to increased 5-HT neuron activity. These neuroplastic changes in raphe 5-HT neurons and their GABAergic afferents may contribute to the behavioral effect of SSRIs.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China;
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN 38163, USA;
| | - Ying Jin
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN 38163, USA;
- Institute of Science and Technology, Fudan University, Shanghai 200433, China
| | - Fu-Ming Zhou
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN 38163, USA;
| |
Collapse
|
4
|
Yu W, Zhang R, Zhang A, Mei Y. Deciphering the Functions of Raphe-Hippocampal Serotonergic and Glutamatergic Circuits and Their Deficits in Alzheimer's Disease. Int J Mol Sci 2025; 26:1234. [PMID: 39941002 PMCID: PMC11818420 DOI: 10.3390/ijms26031234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Subcortical innervation of the hippocampus by the raphe nucleus is essential for emotional and cognitive control. The two major afferents from raphe to hippocampus originate from serotonergic and glutamatergic neurons, of which the serotonergic control of hippocampal inhibitory network, theta activity, and synaptic plasticity have been extensively explored in the growing body of literature, whereas those of glutamatergic circuits have received little attention. Notably, both serotonergic and glutamatergic circuits between raphe and hippocampus are disrupted in Alzheimer's disease (AD), which may contribute to initiation and progression of behavioral and psychological symptoms of dementia. Thus, deciphering the mechanism underlying abnormal raphe-hippocampal circuits in AD is crucial to prevent dementia-associated emotional and cognitive symptoms. In this review, we summarize the anatomical, neurochemical, and electrophysiological diversity of raphe nuclei as well as the architecture of raphe-hippocampal circuitry. We then elucidate subcortical control of hippocampal activity by raphe nuclei and their role in regulation of emotion and cognition. Additionally, we present an overview of disrupted raphe-hippocampal circuits in AD pathogenesis and analyze the available therapies that can potentially be used clinically to alleviate the neuropsychiatric symptoms and cognitive decline in AD course.
Collapse
Affiliation(s)
| | | | | | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer’s Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
5
|
Zhukovskaya A, Zimmerman CA, Willmore L, Pan-Vazquez A, Janarthanan SR, Lynch LA, Falkner AL, Witten IB. Heightened lateral habenula activity during stress produces brainwide and behavioral substrates of susceptibility. Neuron 2024; 112:3940-3956.e10. [PMID: 39393349 PMCID: PMC11624084 DOI: 10.1016/j.neuron.2024.09.009] [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: 11/01/2023] [Revised: 07/04/2024] [Accepted: 09/08/2024] [Indexed: 10/13/2024]
Abstract
Some individuals are susceptible to chronic stress, and others are more resilient. While many brain regions implicated in learning are dysregulated after stress, little is known about whether and how neural teaching signals during stress differ between susceptible and resilient individuals. Here, we seek to determine if activity in the lateral habenula (LHb), which encodes a negative teaching signal, differs between susceptible and resilient mice during stress to produce different outcomes. After (but not before) chronic social defeat stress, the LHb is active when susceptible mice are in proximity of the aggressor strain. During stress, activity is higher in susceptible mice during aggressor interactions, and activation biases mice toward susceptibility. This manipulation generates a persistent and widespread increase in the balance of subcortical vs. cortical activity in susceptible mice. Taken together, our results indicate that heightened activity in the LHb during stress produces lasting brainwide and behavioral substrates of susceptibility.
Collapse
Affiliation(s)
- Anna Zhukovskaya
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | - Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | | | - Laura A Lynch
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Annegret L Falkner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA; Howard Hughes Medical Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
6
|
Hussein MN. Labeling of the serotonergic neuronal circuits emerging from the raphe nuclei via some retrograde tracers. Microsc Res Tech 2024; 87:2894-2914. [PMID: 39041701 DOI: 10.1002/jemt.24662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/20/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a very important neurotransmitter emerging from the raphe nuclei to several brain regions. Serotonergic neuronal connectivity has multiple functions in the brain. In this study, several techniques were used to trace serotonergic neurons in the dorsal raphe (DR) and median raphe (MnR) that project toward the arcuate nucleus of the hypothalamus (Arc), dorsomedial hypothalamic nucleus (DM), lateral hypothalamic area (LH), paraventricular hypothalamic nucleus (PVH), ventromedial hypothalamic nucleus (VMH), fasciola cinereum (FC), and medial habenular nucleus (MHb). Cholera toxin subunit B (CTB), retro-adeno-associated virus (rAAV-CMV-mCherry), glycoprotein-deleted rabies virus (RV-ΔG), and simultaneous microinjection of rAAV2-retro-Cre-tagBFP with AAV-dio-mCherry in C57BL/6 mice were used in this study. In addition, rAAV2-retro-Cre-tagBFP was microinjected into Ai9 mice. Serotonin immunohistochemistry was used for the detection of retrogradely traced serotonergic neurons in the raphe nuclei. The results indicated that rAAV2-retro-Cre-tagBFP microinjection in Ai9 mice was the best method for tracing serotonergic neuron circuits. All of the previously listed nuclei exhibited serotonergic neuronal projections from the DR and MnR, with the exception of the FC, which had very few projections from the DR. The serotonergic neuronal projections were directed toward the Arc by the subpeduncular tegmental (SPTg) nuclei. Moreover, the RV-ΔG tracer revealed monosynaptic non-serotonergic neuronal projections from the DR that were directed toward the Arc. Furthermore, rAAV tracers revealed monosynaptic serotonergic neuronal connections from the raphe nuclei toward Arc. These findings validate the variations in neurotropism among several retrograde tracers. The continued discovery of several novel serotonergic neural circuits is crucial for the future discovery of the functions of these circuits. RESEARCH HIGHLIGHTS: Various kinds of retrograde tracers were microinjected into C57BL/6 and Ai9 mice. The optimum method for characterizing serotonergic neuronal circuits is rAAV2-retro-Cre-tagBFP microinjection in Ai9 mice. The DR, MnR, and SPTg nuclei send monosynaptic serotonergic neuronal projections toward the arcuate nucleus of the hypothalamus. Whole-brain quantification analysis of retrograde-labeled neurons in different brain nuclei following rAAV2-retro-Cre-tagBFP microinjection in the Arc, DM, LH, and VMH is shown. Differential quantitative analysis of median and dorsal raphe serotonergic neurons emerging toward the PVH, DM, LH, Arc, VMH, MHb, and FC is shown.
Collapse
Affiliation(s)
- Mona N Hussein
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Histology and Cytology Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| |
Collapse
|
7
|
Zhou L, Zhang C, Xie Z, Yu Q, Wang J, Gong Y, Zhao J, Bai S, Yang L, Deng D, Zhang R, Shi Y. Neural Circuit Mechanisms of Sinisan formula for the Treatment of adolescent Depression: prefrontal cortex to dorsal raphe nucleus. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118529. [PMID: 38972528 DOI: 10.1016/j.jep.2024.118529] [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: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sinisan formula (SNSF), documented in the classic books Shanghan Lun, is known for its ability to regulate liver-qi and treat depression. However, its underlying mechanism, particularly its effects on dynamic real-time neuron activity and circuits remains to be fully elucidated. AIM OF THE STUDY This study aimed to investigate the antidepressant effect of SNSF and its central nervous system mechanism on depression-like behaviors, focusing on the prefrontal cortex (PFC) to dorsal raphe nucleus (DRN) neural circuit in a stress-induced adolescent animal model. MATERIALS AND METHODS SNSF comprised four herbs, the root of Bupleurum chinense DC., the root of Paeonia lactiflora Pall., the fruit of Citrus aurantium L., the rhizome of Glycyrrhiza uralensis Fisch., in equal propotions. The adolescent depression animal model was induced by maternal separation (MS) and chronic restraint stress (CRS). In-vivo multichannel physiological electrodes were implanted into the PFC on PND 28 and animals were recorded 5 times during PND 35-46. From PND 47, the behavioral tests were performed to evaluate the antidepressant efficacy of SNSF. Subsequently, brain tissue was collected for Western blot and immunofluorescence staining analysis. Retro virus was injected into the DRN to explore sources of projections received by serotonergic (5-HTergic) neurons. And the PFC-to-DRN circuit was activated or inhibited through chemogenetic techniques to investigate the effects of SNSF on depression-like behaviors. RESULTS Administration of SNSF for 18 days effectively alleviated depression-like behaviors in MS&CRS adolescent mice. The PFC emerged as the primary glutamatergic projection source of the DRN5-HT neurons. Following SNSF administration for 13/15/18 days, there was an increase in the firing rate of excitatory neurons and excitatory/inhibitory (E/I) ratio in the PFC. MS&CRS stress let to a reduction in the density of 5-HT+ and CaMKII + neurons in the DRN, accompanied by an increase in the density of GAD + neurons in the DRN, while SNSF administration reversed the alterations. Chemogenetic activation of the PFC-to-DRN circuit rescued the depression-like behaviors induced by MS&CRS, whereas suppression of this circuit attenuated the antidepressant effect of SNSF. CONCLUSIONS SNSF significantly mitigated depression-like behaviors in MS&CRS mice. SNSF exerts its antidepressant effects by increasing the E/I ratio in the PFC and enhancing glutamatergic projections from the PFC to the DRN.
Collapse
Affiliation(s)
- Liuchang Zhou
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Caixia Zhang
- Outpatient Department, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zedan Xie
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Qingying Yu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Junjie Wang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuwen Gong
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jinlan Zhao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Shasha Bai
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lei Yang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Di Deng
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Rong Zhang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
8
|
Yu CC, Wang XF, Wang J, Li C, Xiao J, Wang XS, Han R, Wang SQ, Lin YF, Kong LH, Du YJ. Electroacupuncture Alleviates Memory Deficits in APP/PS1 Mice by Targeting Serotonergic Neurons in Dorsal Raphe Nucleus. Curr Med Sci 2024; 44:987-1000. [PMID: 38990450 DOI: 10.1007/s11596-024-2908-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE Alzheimer's disease (AD) has become a significant global concern, but effective drugs able to slow down AD progression is still lacked. Electroacupuncture (EA) has been demonstrated to ameliorate cognitive impairment in individuals with AD. However, the underlying mechanisms remains poorly understood. This study aimed at examining the neuroprotective properties of EA and its potential mechanism of action against AD. METHODS APP/PS1 transgenic mice were employed to evaluate the protective effects of EA on Shenshu (BL 23) and Baihui (GV 20). Chemogenetic manipulation was used to activate or inhibit serotonergic neurons within the dorsal raphe nucleus (DRN). Learning and memory abilities were assessed by the novel object recognition and Morris water maze tests. Golgi staining, western blot, and immunostaining were utilized to determine EA-induced neuroprotection. RESULTS EA at Shenshu (BL 23) and Baihui (GV 20) effectively ameliorated learning and memory impairments in APP/PS1 mice. EA attenuated dendritic spine loss, increased the expression levels of PSD95, synaptophysin, and brain-derived neurotrophic factor in hippocampus. Activation of serotonergic neurons within the DRN can ameliorate cognitive deficits in AD by activating glutamatergic neurons mediated by 5-HT1B. Chemogenetic inhibition of serotonergic neurons in the DRN reversed the effects of EA on synaptic plasticity and memory. CONCLUSION EA can alleviate cognitive dysfunction in APP/PS1 mice by activating serotonergic neurons in the DRN. Further study is necessary to better understand how the serotonergic neurons-related neural circuits involves in EA-induced memory improvement in AD.
Collapse
Affiliation(s)
- Chao-Chao Yu
- Department of Rehabilitation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Xiao-Fei Wang
- Department of Rehabilitation, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430074, China
| | - Jia Wang
- Department of Acupuncture and Moxibustion, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, 430030, China
| | - Chu Li
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Juan Xiao
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xue-Song Wang
- College of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, 050299, China
| | - Rui Han
- Department of Child Rehabilitation Medicine, Qujing Hospital of Maternity and Childcare, Qujing, 655002, China
| | - Shu-Qin Wang
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Yuan-Fang Lin
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Li-Hong Kong
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
9
|
Cao Z, Yung WH, Ke Y. Distinct populations of lateral preoptic nucleus neurons jointly contribute to depressive-like behaviors through divergent projections in male mice. Neurobiol Stress 2024; 32:100667. [PMID: 39233784 PMCID: PMC11372801 DOI: 10.1016/j.ynstr.2024.100667] [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: 05/13/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
The lateral preoptic area (LPO) is a component of the hypothalamus involved in various physiological functions including sleep-wakefulness transition, thermoregulation, and water-salt balance. In this study, we discovered that distinct LPO excitatory neurons project separately to the aversive processing center lateral habenula (LHb) and the reward processing hub ventral tegmental area (VTA). Following chronic restraint stress (CRS), the LHb-projecting and VTA-projecting LPO neurons exhibited increased and decreased neuronal activities, respectively. Optogenetic activation of LHb-projecting LPO excitatory neurons and LPO excitatory neuronal terminals within LHb evoked aversion and avoidance behaviors, while activation of VTA-projecting LPO excitatory neurons and LPO excitatory neuronal terminals within VTA produced preference and exploratory behaviors in mice. Furthermore, either optogenetic inhibition of LHb-projecting LPO excitatory neurons or activation of VTA-projecting LPO excitatory neurons during CRS effectively prevented the development of depressive-like behaviors. Our study unveils, for the first-time, divergent pathways originating from LPO that regulate opposite affective states in mice and implicates that an imbalance of their activities could lead to depressive-like behaviors. These circuitries represent promising therapeutic targets to relieve emotional dysfunctions in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zhiping Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, 999077, Hong Kong, China
| | - Wing-Ho Yung
- Department of Neuroscience, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, 999077, Hong Kong, China
| |
Collapse
|
10
|
Liu X, Li H, Ma R, Tong X, Wu J, Huang X, So K, Tao Q, Huang L, Lin S, Ren C. Burst firing in Output-Defined Parallel Habenula Circuit Underlies the Antidepressant Effects of Bright Light Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401059. [PMID: 38863324 PMCID: PMC11321664 DOI: 10.1002/advs.202401059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/11/2024] [Indexed: 06/13/2024]
Abstract
Research highlights the significance of increased bursting in lateral habenula (LHb) neurons in depression and as a focal point for bright light treatment (BLT). However, the precise spike patterns of LHb neurons projecting to different brain regions during depression, their roles in depression development, and BLT's therapeutic action remain elusive. Here, LHb neurons are found projecting to the dorsal raphe nucleus (DRN), ventral tegmental area (VTA), and median raphe nucleus (MnR) exhibit increased bursting following aversive stimuli exposure, correlating with distinct depressive symptoms. Enhanced bursting in DRN-projecting LHb neurons is pivotal for anhedonia and anxiety, while concurrent bursting in LHb neurons projecting to the DRN, VTA, and MnR is essential for despair. Remarkably, reducing bursting in distinct LHb neuron subpopulations underlies the therapeutic effects of BLT on specific depressive behaviors. These findings provide valuable insights into the mechanisms of depression and the antidepressant action of BLT.
Collapse
Affiliation(s)
- Xianwei Liu
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Han Li
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Ruijia Ma
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Xiaohan Tong
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Jijin Wu
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Xiaodan Huang
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Kwok‐Fai So
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdao266113China
| | - Qian Tao
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdao266113China
- Department of Rehabilitation MedicineFirst Affiliated Hospital of Jinan UniversityPsychology DepartmentSchool of MedicineJinan UniversityGuangzhou510632China
| | - Lu Huang
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
| | - Song Lin
- Physiology DepartmentKey Laboratory of Viral Pathogenesis & Infection Prevention and Control, School of MedicineJinan UniversityGuangzhou510632China
| | - Chaoran Ren
- Department of Neurology and Stroke CenterFirst Affiliated Hospital of Jinan UniversityKey Laboratory of CNS Regeneration (Ministry of Education)Guangdong Key Laboratory of Non‐human Primate ResearchGHM Institute of CNS RegenerationJinan UniversityGuangzhou510632China
- Co‐innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdao266113China
| |
Collapse
|
11
|
Zhukovskaya A, Christopher Z, Willmore L, Pan Vazquez A, Janarthanan S, Falkner A, Witten I. Heightened lateral habenula activity during stress produces brainwide and behavioral substrates of susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.06.565681. [PMID: 39005438 PMCID: PMC11244933 DOI: 10.1101/2023.11.06.565681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Some individuals are susceptible to the experience of chronic stress and others are more resilient. While many brain regions implicated in learning are dysregulated after stress, little is known about whether and how neural teaching signals during stress differ between susceptible and resilient individuals. Here, we seek to determine if activity in the lateral habenula (LHb), which encodes a negative teaching signal, differs between susceptible and resilient mice during stress to produce different outcomes. After, but not before, chronic social defeat stress (CSDS), the LHb is active when susceptible mice are in the proximity of the aggressor strain. During stress itself, LHb activity is higher in susceptible mice during aggressor proximity, and activation of the LHb during stress biases mice towards susceptibility. This manipulation generates a persistent and widespread increase in the balance of subcortical versus cortical activity in susceptible mice. Taken together, our results indicate that heightened activity in the LHb during stress produces lasting brainwide and behavioral substrates of susceptibility.
Collapse
|
12
|
Cao K, Zhong J, Wang S, Shi Y, Bai S, Zhao J, Yang L, Liang Q, Deng D, Zhang R. SiNiSan exerts antidepressant effects by modulating serotonergic/GABAergic neuron activity in the dorsal raphe nucleus region through NMDA receptor in the adolescent depression mouse model. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118040. [PMID: 38479542 DOI: 10.1016/j.jep.2024.118040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/29/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Kerun Cao
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jialong Zhong
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shanshan Wang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Bai
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinlan Zhao
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Yang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Liang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital Group, Shenzhen, 518000, China.
| | - Di Deng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
13
|
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
|
14
|
Chen X, Liu X, Luan S, Wang X, Zhang Y, Hao Y, Zhang Q, Zhang J, Zhao H. Optogenetic activation of the lateral habenula D1R-ventral tegmental area circuit induces depression-like behavior in mice. Eur Arch Psychiatry Clin Neurosci 2024; 274:867-878. [PMID: 38236282 DOI: 10.1007/s00406-023-01743-2] [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: 02/19/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
A number of different receptors are distributed in glutamatergic neurons of the lateral habenula (LHb). These glutamatergic neurons are involved in different neural pathways, which may identify how the LHb regulates various physiological functions. However, the role of dopamine D1 receptor (D1R)-expressing habenular neurons projecting to the ventral tegmental area (VTA) (LHbD1R-VTA) remains not well understood. In the current study, to determine the activity of D1R-expressing neurons in LHb, D1R-Cre mice were used to establish the chronic restraint stress (CRS) depression model. Adeno-associated virus was injected into bilateral LHb in D1R-Cre mice to examine whether optogenetic activation of the LHb D1R-expressing neurons and their projections could induce depression-like behavior. Optical fibers were implanted in the LHb and VTA, respectively. To investigate whether optogenetic inhibition of the LHbD1R-VTA circuit could produce antidepressant-like effects, the adeno-associated virus was injected into the bilateral LHb in the D1R-Cre CRS model, and optical fibers were implanted in the bilateral VTA. The D1R-expressing neuronal activity in the LHb was increased in the CRS depression model. Optogenetic activation of the D1R-expressing neurons in LHb induced behavioral despair and anhedonia, which could also be induced by activation of the LHbD1R-VTA axons. Conversely, optogenetic inhibition of the LHbD1R-VTA circuit improved behavioral despair and anhedonia in the CRS depression model. D1R-expressing glutamatergic neurons in the LHb and their projections to the VTA are involved in the occurrence and regulation of depressive-like behavior.
Collapse
Affiliation(s)
- Xiaowei Chen
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China
- Department of Rehabilitation Medicine, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Xiaofeng Liu
- Neuroscience Research Center, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Shuxin Luan
- Department of Mental Health, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Xuxin Wang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China
| | - Ying Zhang
- Department of Neurology, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Yulei Hao
- Neuroscience Research Center, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Qiang Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China
| | - Jiaming Zhang
- Department of Rehabilitation Medicine, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Hua Zhao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China.
- Neuroscience Research Center, First Hospital of Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
15
|
Zhang W, Jin Y, Zhou FM. Chronic fluoxetine treatment desensitizes serotoninergic inhibition of GABA inputs and the intrinsic excitability of dorsal raphe serotonin neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.592963. [PMID: 38766100 PMCID: PMC11100661 DOI: 10.1101/2024.05.07.592963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Dorsal raphe serotonin (5-hydroxytryptamine, 5-HT) neurons are spontaneously active and release 5-HT that is critical to normal brain function such mood and emotion. Serotonin reuptake inhibitors (SSRIs) increase the synaptic and extracellular 5-HT level and are effective in treating depression. Treatment of two weeks or longer is often required for SSRIs to exert clinical benefits. The cellular mechanism underlying this delay was not fully understood. Here we show that the GABAergic inputs inhibit the spike firing of raphe 5-HT neurons; this GABAergic regulation was reduced by 5-HT, which was prevented by G-protein-activated inwardly rectifying potassium (Girk) channel inhibitor tertiapin-Q, indicating a contribution of 5-HT activation of Girk channels in GABAergic presynaptic axon terminals. Equally important, after 14 days of treatment of fluoxetine, a widely used SSRI type antidepressant, this 5-HT inhibition of GABAergic inputs was substantially downregulated. Furthermore, the chronic fluoxetine treatment substantially downregulated the 5-HT activation of the inhibitory Girk current in 5-HT neurons. Taken together, our results suggest that chronic fluoxetine administration, by blocking 5-HT reuptake and hence increasing the extracellular 5-HT level, can downregulate the function of 5-HT1B receptors on the GABAergic afferent axon terminals synapsing onto 5-HT neurons, allowing extrinsic, behaviorally important GABA neurons to more effectively influence 5-HT neurons; simultaneously, chronic fluoxetine treatment also downregulate somatic 5-HT autoreceptor-activated Girk channel-mediated hyperpolarization and decrease in input resistance and intrinsic excitability, rendering 5-HT neurons resistant to autoinhibition and leading to increased 5-HT neuron activity, potentially contributing to the antidepressant effect of SSRIs.
Collapse
|
16
|
Castle ME, Flanigan ME. The role of brain serotonin signaling in excessive alcohol consumption and withdrawal: A call for more research in females. Neurobiol Stress 2024; 30:100618. [PMID: 38433994 PMCID: PMC10907856 DOI: 10.1016/j.ynstr.2024.100618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/05/2024] Open
Abstract
Alcohol Use Disorder (AUD) is a leading cause of death and disability worldwide, but current treatments are insufficient in fully addressing the symptoms that often lead to relapses in alcohol consumption. The brain's serotonin system has been implicated in AUD for decades and is a major regulator of stress-related behaviors associated with increased alcohol consumption. This review will discuss the current literature on the association between neurobiological adaptations in serotonin systems and AUD in humans as well as the effectiveness of serotonin receptor manipulations on alcohol-related behaviors like consumption and withdrawal. We will further discuss how these findings in humans relate to findings in animal models, including a comparison of systemic pharmacological manipulations modulating alcohol consumption. We next provide a detailed overview of brain region-specific roles for serotonin and serotonin receptor signaling in alcohol-related behaviors in preclinical animal models, highlighting the complexity of forming a cohesive model of serotonin function in AUD and providing possible avenues for more effective therapeutic intervention. Throughout the review, we discuss what is known about sex differences in the sequelae of AUD and the role of serotonin in these sequelae. We stress a critical need for additional studies in women and female animals so that we may build a clearer path to elucidating sex-specific serotonergic mechanisms and develop better treatments.
Collapse
Affiliation(s)
- Megan E. Castle
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Meghan E. Flanigan
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
17
|
Groos D, Helmchen F. The lateral habenula: A hub for value-guided behavior. Cell Rep 2024; 43:113968. [PMID: 38522071 DOI: 10.1016/j.celrep.2024.113968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/20/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
The habenula is an evolutionarily highly conserved diencephalic brain region divided into two major parts, medial and lateral. Over the past two decades, studies of the lateral habenula (LHb), in particular, have identified key functions in value-guided behavior in health and disease. In this review, we focus on recent insights into LHb connectivity and its functional relevance for different types of aversive and appetitive value-guided behavior. First, we give an overview of the anatomical organization of the LHb and its main cellular composition. Next, we elaborate on how distinct LHb neuronal subpopulations encode aversive and appetitive stimuli and on their involvement in more complex decision-making processes. Finally, we scrutinize the afferent and efferent connections of the LHb and discuss their functional implications for LHb-dependent behavior. A deepened understanding of distinct LHb circuit components will substantially contribute to our knowledge of value-guided behavior.
Collapse
Affiliation(s)
- Dominik Groos
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland.
| | - Fritjof Helmchen
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich, Zurich, Switzerland; University Research Priority Program (URPP), Adaptive Brain Circuits in Development and Learning, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Mitsui K, Takahashi A. Aggression modulator: Understanding the multifaceted role of the dorsal raphe nucleus. Bioessays 2024; 46:e2300213. [PMID: 38314963 DOI: 10.1002/bies.202300213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/07/2024]
Abstract
Aggressive behavior is instinctively driven behavior that helps animals to survive and reproduce and is closely related to multiple behavioral and physiological processes. The dorsal raphe nucleus (DRN) is an evolutionarily conserved midbrain structure that regulates aggressive behavior by integrating diverse brain inputs. The DRN consists predominantly of serotonergic (5-HT:5-hydroxytryptamine) neurons and decreased 5-HT activity was classically thought to increase aggression. However, recent studies challenge this 5-HT deficiency model, revealing a more complex role for the DRN 5-HT system in aggression. Furthermore, emerging evidence has shown that non-5-HT populations in the DRN and specific neural circuits contribute to the escalation of aggressive behavior. This review argues that the DRN serves as a multifaceted modulator of aggression, acting not only via 5-HT but also via other neurotransmitters and neural pathways, as well as different subsets of 5-HT neurons. In addition, we discuss the contribution of DRN neurons in the behavioral and physiological aspects implicated in aggressive behavior, such as arousal, reward, and impulsivity, to further our understanding of DRN-mediated aggression modulation.
Collapse
Affiliation(s)
- Koshiro Mitsui
- Laboratory of Behavioral Neurobiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Aki Takahashi
- Laboratory of Behavioral Neurobiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Institute of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
19
|
Wu Z, Shen Z, Xu Y, Chen S, Xiao S, Ye J, Zhang H, Ma X, Zhu Y, Zhu X, Jiang Y, Fang J, Liu B, He X, Gao S, Shao X, Liu J, Fang J. A neural circuit associated with anxiety-like behaviors induced by chronic inflammatory pain and the anxiolytic effects of electroacupuncture. CNS Neurosci Ther 2024; 30:e14520. [PMID: 38018559 PMCID: PMC11017463 DOI: 10.1111/cns.14520] [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: 07/21/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/30/2023] Open
Abstract
AIMS Negative emotions induced by chronic pain are a serious clinical problem. Electroacupuncture (EA) is a clinically proven safe and effective method to manage pain-related negative emotions. However, the circuit mechanisms underlying the effect of EA treatment on negative emotions remain unclear. METHODS Plantar injection of complete Freund's adjuvant (CFA) was performed to establish a rat model of chronic inflammatory pain-induced anxiety-like behaviors. Adeno-associated virus (AAV) tracing was used to identify excitatory synaptic transmission from the rostral anterior cingulate cortex (rACC) to the dorsal raphe nucleus (DRN). Employing chemogenetic approaches, we examined the role of the rACC-DRN circuit in chronic pain-induced anxiety-like behaviors and investigated whether EA could reverse chronic pain-induced dysfunctions of the rACC-DRN circuit and anxiety-like behaviors. RESULTS We found that chemogenetic activation of the rACC-DRN circuit alleviated CFA-induced anxiety-like behaviors, while chemogenetic inhibition of the rACC-DRN circuit resulted in short-term CFA-induced anxiety-like behaviors. Further research revealed that the development of CFA-induced anxiety-like behaviors was attributed to the dysfunction of rACC CaMKII neurons projecting to DRN serotonergic neurons (rACCCaMKII-DRN5-HT neurons) but not rACC CaMKII neurons projecting to DRN GABAergic neurons (rACCCaMKII-DRNGABA neurons). This is supported by the findings that chemogenetic activation of the rACCCaMKII-DRN5-HT circuit alleviates anxiety-like behaviors in rats with chronic pain, whereas neither chemogenetic inhibition nor chemogenetic activation of the rACCCaMKII-DRNGABA circuit altered CFA chronic pain-evoked anxiety-like behaviors in rats. More importantly, we found that EA could reverse chronic pain-induced changes in the activity of rACC CaMKII neurons and DRN 5-HTergic neurons and that chemogenetic inhibition of the rACCCaMKII-DRN5-HT circuit blocked the therapeutic effects of EA on chronic pain-induced anxiety-like behaviors. CONCLUSIONS Our data suggest that the reversal of rACCCaMKII-DRN5-HT circuit dysfunction may be a mechanism underlying the therapeutic effect of EA on chronic pain-induced anxiety-like behaviors.
Collapse
Affiliation(s)
- Zemin Wu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Department of Acupuncture and Moxibustionthe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Zui Shen
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yingling Xu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
| | - Shaozong Chen
- Institution of Acupuncture and Moxibustion, Shandong University of Traditional Chinese MedicineJinanChina
| | - Siqi Xiao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jiayu Ye
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Haiyan Zhang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xinyi Ma
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yichen Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xixiao Zhu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Junfan Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Boyi Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Shuzhong Gao
- Institution of Acupuncture and Moxibustion, Shandong University of Traditional Chinese MedicineJinanChina
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jinggen Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- National Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture ResearchThe Third Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Department of Acupuncture and Moxibustionthe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
20
|
Kisner A, Polter AM. Maturation of glutamatergic transmission onto dorsal raphe serotonergic neurons. J Neurophysiol 2024; 131:626-637. [PMID: 38380827 PMCID: PMC11305679 DOI: 10.1152/jn.00037.2023] [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: 01/23/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 02/22/2024] Open
Abstract
Serotonergic neurons in the dorsal raphe nucleus (DRN) play important roles early in postnatal development in the maturation and modulation of higher-order emotional, sensory, and cognitive circuitry. The pivotal functions of these cells in brain development make them a critical substrate by which early experience can be wired into the brain. In this study, we investigated the maturation of synapses onto dorsal raphe serotonergic neurons in typically developing male and female mice using whole cell patch-clamp recordings in ex vivo brain slices. We show that while inhibition of these neurons is relatively stable across development, glutamatergic synapses greatly increase in strength between postnatal day 6 (P6) and P21-23. In contrast to forebrain regions, where the components making up glutamatergic synapses are dynamic across early life, we find that DRN excitatory synapses maintain a very high ratio of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) to N-methyl-d-aspartate (NMDA) receptors and a rectifying component of the AMPA response until adulthood. Overall, these findings reveal that the development of serotonergic neurons is marked by a significant refinement of glutamatergic synapses during the first three postnatal weeks. This suggests this time is a sensitive period of heightened plasticity for the integration of information from upstream brain areas. Genetic and environmental insults during this period could lead to alterations in serotonergic output, impacting both the development of forebrain circuits and lifelong neuromodulatory actions.NEW & NOTEWORTHY Serotonergic neurons are regulators of both the development of and ongoing activity in neuronal circuits controlling affective, cognitive, and sensory processing. Here, we characterize the maturation of extrinsic synaptic inputs onto these cells, showing that the first three postnatal weeks are a period of synaptic refinement and a potential window for experience-dependent plasticity in response to both enrichment and adversity.
Collapse
Affiliation(s)
- Alexandre Kisner
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Abigail M Polter
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
21
|
Gutierrez-Castellanos N, Sarra D, Godinho BS, Mainen ZF. Maturation of cortical input to dorsal raphe nucleus increases behavioral persistence in mice. eLife 2024; 13:e93485. [PMID: 38477558 PMCID: PMC10994666 DOI: 10.7554/elife.93485] [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: 10/12/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
The ability to persist toward a desired objective is a fundamental aspect of behavioral control whose impairment is implicated in several behavioral disorders. One of the prominent features of behavioral persistence is that its maturation occurs relatively late in development. This is presumed to echo the developmental time course of a corresponding circuit within late-maturing parts of the brain, such as the prefrontal cortex, but the specific identity of the responsible circuits is unknown. Here, we used a genetic approach to describe the maturation of the projection from layer 5 neurons of the neocortex to the dorsal raphe nucleus in mice. Using optogenetic-assisted circuit mapping, we show that this projection undergoes a dramatic increase in synaptic potency between postnatal weeks 3 and 8, corresponding to the transition from juvenile to adult. We then show that this period corresponds to an increase in the behavioral persistence that mice exhibit in a foraging task. Finally, we used a genetic targeting strategy that primarily affected neurons in the medial prefrontal cortex, to selectively ablate this pathway in adulthood and show that mice revert to a behavioral phenotype similar to juveniles. These results suggest that frontal cortical to dorsal raphe input is a critical anatomical and functional substrate of the development and manifestation of behavioral persistence.
Collapse
Affiliation(s)
| | - Dario Sarra
- Champalimaud Research, Champalimaud FoundationLisbonPortugal
- Nuffield Department of Clinical Neurosciences, University of OxfordOxfordUnited Kingdom
| | - Beatriz S Godinho
- Champalimaud Research, Champalimaud FoundationLisbonPortugal
- Nuffield Department of Clinical Neurosciences, University of OxfordOxfordUnited Kingdom
| | | |
Collapse
|
22
|
Martianova E, Sadretdinova R, Pageau A, Pausic N, Gentiletti TD, Leblanc D, Rivera AM, Labonté B, Proulx CD. Hypothalamic neuronal outputs transmit sensorimotor signals at the onset of locomotor initiation. iScience 2023; 26:108328. [PMID: 38026162 PMCID: PMC10665817 DOI: 10.1016/j.isci.2023.108328] [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: 07/11/2023] [Revised: 09/27/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
The lateral hypothalamus (LH) plays a critical role in sensory integration to organize behavior responses. However, how projection-defined LH neuronal outputs dynamically transmit sensorimotor signals to major downstream targets to organize behavior is unknown. Here, using multi-fiber photometry, we show that three major LH neuronal outputs projecting to the dorsal raphe nucleus (DRN), ventral tegmental area (VTA), and lateral habenula (LHb) exhibit significant coherent activity in mice engaging sensory-evoked or self-initiated motor responses. Increased activity at LH axon terminals precedes movement initiation during active coping responses and the activity of serotonin neurons and dopamine neurons. The optogenetic activation of LH axon terminals in either of the DRN, VTA, or LHb was sufficient to increase motor initiation but had different effects on passive avoidance and sucrose consumption. Our findings support the complementary role of three projection-defined LH neuronal outputs in the transmission of sensorimotor signals to major downstream regions at movement onset.
Collapse
Affiliation(s)
- Ekaterina Martianova
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Renata Sadretdinova
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Alicia Pageau
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Nikola Pausic
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Tommy Doucet Gentiletti
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Danahé Leblanc
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Arturo Marroquin Rivera
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Benoît Labonté
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| | - Christophe D. Proulx
- CERVO Brain Research Center, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, Canada
| |
Collapse
|
23
|
Shin Y, Kim S, Sohn JW. Serotonergic regulation of appetite and sodium appetite. J Neuroendocrinol 2023; 35:e13328. [PMID: 37525500 DOI: 10.1111/jne.13328] [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: 02/05/2023] [Revised: 06/27/2023] [Accepted: 07/15/2023] [Indexed: 08/02/2023]
Abstract
Serotonin is a neurotransmitter that is synthesized and released from the brainstem raphe nuclei to affect many brain functions. It is well known that the activity of raphe serotonergic neurons is changed in response to the changes in feeding status to regulate appetite via the serotonin receptors. Likewise, changes in volume status are known to alter the activity of raphe serotonergic neurons and drugs targeting serotonin receptors were shown to affect sodium appetite. Therefore, the central serotonin system appears to regulate ingestion of both food and salt, although neural mechanisms that induce appetite in response to hunger and sodium appetite in response to volume depletion are largely distinct from each other. In this review, we discuss our current knowledge regarding the regulation of ingestion - appetite and sodium appetite - by the central serotonin system.
Collapse
Affiliation(s)
- Yurim Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Seungjik Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| |
Collapse
|
24
|
Feng YY, Bromberg-Martin ES, Monosov IE. Dorsal raphe neurons signal integrated value during multi-attribute decision-making. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553745. [PMID: 37662243 PMCID: PMC10473596 DOI: 10.1101/2023.08.17.553745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
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 grappling with reward uncertainty. However, whether and how DRN neurons signal reward amount, reward delay, and reward uncertainty during multi-attribute value-based decision-making, where subjects consider all these attributes to make a choice, is unclear. 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. Remarkably, these neurons commonly integrated offer attributes in a manner that reflected monkeys' overall 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 DRN participates in integrated value computations, guiding theories of DRN in decision-making and psychiatric disease.
Collapse
Affiliation(s)
- Yang-Yang Feng
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | | | - Ilya E. Monosov
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Washington University Pain Center, Washington University, St. Louis, Missouri, USA
- Department of Neurosurgery, Washington University, St. Louis, Missouri, USA
- Department of Electrical Engineering, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
25
|
Nageeb Hasan SM, Clarke CL, McManamon Strand TP, Bambico FR. Putative pathological mechanisms of late-life depression and Alzheimer's Disease. Brain Res 2023:148423. [PMID: 37244602 DOI: 10.1016/j.brainres.2023.148423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by progressive impairment in cognition and memory. AD is accompanied by several neuropsychiatric symptoms, with depression being the most prominent. Although depression has long been known to be associated with AD, controversial findings from preclinical and clinical studies have obscured the precise nature of this association. However recent evidence suggests that depression could be a prodrome or harbinger of AD. Evidence indicates that the major central serotonergic nucleus-the dorsal raphe nucleus (DRN)-shows very early AD pathology: neurofibrillary tangles made of hyperphosphorylated tau protein and degenerated neurites. AD and depression share common pathophysiologies, including functional deficits of the serotonin (5-HT) system. 5-HT receptors have modulatory effects on the progression of AD pathology i.e., reduction in Aβ load, increased hyper-phosphorylation of tau, decreased oxidative stress etc. Moreover, preclinical models show a role for specific channelopathies that result in abnormal regional activational and neuroplasticity patterns. One of these concerns the pathological upregulation of the small conductance calcium-activated potassium (SK) channel in corticolimbic structure. This has also been observed in the DRN in both diseases. The SKC is a key regulator of cell excitability and long-term potentiation (LTP). SKC over-expression is positively correlated with aging and cognitive decline, and is evident in AD. Pharmacological blockade of SKCs has been reported to reverse symptoms of depression and AD. Thus, aberrant SKC functioning could be related to depression pathophysiology and diverts its late-life progression towards the development of AD. We summarize findings from preclinical and clinical studies suggesting a molecular linkage between depression and AD pathology. We also provide a rationale for considering SKCs as a novel pharmacological target for the treatment of AD-associated symptoms.
Collapse
Affiliation(s)
- S M Nageeb Hasan
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada.
| | - Courtney Leigh Clarke
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada
| | | | - Francis Rodriguez Bambico
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada; Behavioural Neurobiology Laboratory, Centre for Addiction and Mental Health, Toronto, ON, M5T1R8, Canada
| |
Collapse
|
26
|
Neural mechanism underlying depressive-like state associated with social status loss. Cell 2023; 186:560-576.e17. [PMID: 36693374 DOI: 10.1016/j.cell.2022.12.033] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/13/2022] [Accepted: 12/20/2022] [Indexed: 01/25/2023]
Abstract
Downward social mobility is a well-known mental risk factor for depression, but its neural mechanism remains elusive. Here, by forcing mice to lose against their subordinates in a non-violent social contest, we lower their social ranks stably and induce depressive-like behaviors. These rank-decline-associated depressive-like behaviors can be reversed by regaining social status. In vivo fiber photometry and single-unit electrophysiological recording show that forced loss, but not natural loss, generates negative reward prediction error (RPE). Through the lateral hypothalamus, the RPE strongly activates the brain's anti-reward center, the lateral habenula (LHb). LHb activation inhibits the medial prefrontal cortex (mPFC) that controls social competitiveness and reinforces retreats in contests. These results reveal the core neural mechanisms mutually promoting social status loss and depressive behaviors. The intertwined neuronal signaling controlling mPFC and LHb activities provides a mechanistic foundation for the crosstalk between social mobility and psychological disorder, unveiling a promising target for intervention.
Collapse
|
27
|
Behera CK, Joshi A, Wang DH, Sharp T, Wong-Lin K. Degeneracy and stability in neural circuits of dopamine and serotonin neuromodulators: A theoretical consideration. Front Comput Neurosci 2023; 16:950489. [PMID: 36761394 PMCID: PMC9905743 DOI: 10.3389/fncom.2022.950489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 12/30/2022] [Indexed: 01/26/2023] Open
Abstract
Degenerate neural circuits perform the same function despite being structurally different. However, it is unclear whether neural circuits with interacting neuromodulator sources can themselves degenerate while maintaining the same neuromodulatory function. Here, we address this by computationally modeling the neural circuits of neuromodulators serotonin and dopamine, local glutamatergic and GABAergic interneurons, and their possible interactions, under reward/punishment-based conditioning tasks. The neural modeling is constrained by relevant experimental studies of the VTA or DRN system using, e.g., electrophysiology, optogenetics, and voltammetry. We first show that a single parsimonious, sparsely connected neural circuit model can recapitulate several separate experimental findings that indicated diverse, heterogeneous, distributed, and mixed DRNVTA neuronal signaling in reward and punishment tasks. The inability of this model to recapitulate all observed neuronal signaling suggests potentially multiple circuits acting in parallel. Then using computational simulations and dynamical systems analysis, we demonstrate that several different stable circuit architectures can produce the same observed network activity profile, hence demonstrating degeneracy. Due to the extensive D2-mediated connections in the investigated circuits, we simulate the D2 receptor agonist by increasing the connection strengths emanating from the VTA DA neurons. We found that the simulated D2 agonist can distinguish among sub-groups of the degenerate neural circuits based on substantial deviations in specific neural populations' activities in reward and punishment conditions. This forms a testable model prediction using pharmacological means. Overall, this theoretical work suggests the plausibility of degeneracy within neuromodulator circuitry and has important implications for the stable and robust maintenance of neuromodulatory functions.
Collapse
Affiliation(s)
- Chandan K. Behera
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Derry∼Londonderry, United Kingdom,*Correspondence: Chandan K. Behera,
| | - Alok Joshi
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Derry∼Londonderry, United Kingdom
| | - Da-Hui Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China,School of Systems Science, Beijing Normal University, Beijing, China
| | - Trevor Sharp
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - KongFatt Wong-Lin
- Intelligent Systems Research Centre, School of Computing, Engineering and Intelligent Systems, Ulster University, Derry∼Londonderry, United Kingdom,KongFatt Wong-Lin,
| |
Collapse
|
28
|
Kisner A, Polter AM. Maturation of glutamatergic transmission onto dorsal raphe serotonergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.19.524776. [PMID: 36711665 PMCID: PMC9882295 DOI: 10.1101/2023.01.19.524776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Serotonergic neurons in the dorsal raphe nucleus (DRN) play important roles early in postnatal development in the maturation and modulation of higher order emotional, sensory, and cognitive circuitry. This unique position makes these cells a substrate by which early experience can be wired into brain. In this study, we have investigated the maturation of synapses onto dorsal raphe serotonergic neurons in typically developing male and female mice using whole-cell patch-clamp recordings in ex vivo brain slices. We show that while inhibition of these neurons is relatively stable across development, glutamatergic synapses greatly increase in strength between P6 and P21-23. In contrast to forebrain regions, where the components making up glutamatergic synapses are dynamic across early life, we find that the makeup of these synapses onto DRN serotonergic neurons is largely stable after P15. DRN excitatory synapses maintain a very high ratio of AMPA to NMDA receptors and a rectifying component of the AMPA response throughout the lifespan. Overall, these findings reveal that the development of serotonergic neurons is marked by a significant refinement of glutamatergic synapses during the first 3 postnatal weeks. This suggests this time as a sensitive period of heightened plasticity for integration of information from upstream brain areas and that genetic and environmental insults during this period could lead to alterations in serotonergic output, impacting both the development of forebrain circuits and lifelong neuromodulatory actions.
Collapse
Affiliation(s)
- Alexandre Kisner
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
- Current address: Department of Neuroscience, American University, Washington DC 20016
| | - Abigail M. Polter
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| |
Collapse
|
29
|
Harkin EF, Lynn MB, Payeur A, Boucher JF, Caya-Bissonnette L, Cyr D, Stewart C, Longtin A, Naud R, Béïque JC. Temporal derivative computation in the dorsal raphe network revealed by an experimentally driven augmented integrate-and-fire modeling framework. eLife 2023; 12:72951. [PMID: 36655738 PMCID: PMC9977298 DOI: 10.7554/elife.72951] [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: 08/10/2021] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
By means of an expansive innervation, the serotonin (5-HT) neurons of the dorsal raphe nucleus (DRN) are positioned to enact coordinated modulation of circuits distributed across the entire brain in order to adaptively regulate behavior. Yet the network computations that emerge from the excitability and connectivity features of the DRN are still poorly understood. To gain insight into these computations, we began by carrying out a detailed electrophysiological characterization of genetically identified mouse 5-HT and somatostatin (SOM) neurons. We next developed a single-neuron modeling framework that combines the realism of Hodgkin-Huxley models with the simplicity and predictive power of generalized integrate-and-fire models. We found that feedforward inhibition of 5-HT neurons by heterogeneous SOM neurons implemented divisive inhibition, while endocannabinoid-mediated modulation of excitatory drive to the DRN increased the gain of 5-HT output. Our most striking finding was that the output of the DRN encodes a mixture of the intensity and temporal derivative of its input, and that the temporal derivative component dominates this mixture precisely when the input is increasing rapidly. This network computation primarily emerged from prominent adaptation mechanisms found in 5-HT neurons, including a previously undescribed dynamic threshold. By applying a bottom-up neural network modeling approach, our results suggest that the DRN is particularly apt to encode input changes over short timescales, reflecting one of the salient emerging computations that dominate its output to regulate behavior.
Collapse
Affiliation(s)
- Emerson F Harkin
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Michael B Lynn
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Alexandre Payeur
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
- Department of Physics, University of OttawaOttawaCanada
| | - Jean-François Boucher
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Léa Caya-Bissonnette
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Dominic Cyr
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Chloe Stewart
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - André Longtin
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
- Department of Physics, University of OttawaOttawaCanada
| | - Richard Naud
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
- Department of Physics, University of OttawaOttawaCanada
| | - Jean-Claude Béïque
- Brain and Mind Research Institute, Centre for Neural Dynamics, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| |
Collapse
|
30
|
Bąk J, Bobula B, Hess G. Restraint Stress and Repeated Corticosterone Administration Differentially Affect Neuronal Excitability, Synaptic Transmission and 5-HT 7 Receptor Reactivity in the Dorsal Raphe Nucleus of Young Adult Male Rats. Int J Mol Sci 2022; 23:ijms232214303. [PMID: 36430779 PMCID: PMC9698125 DOI: 10.3390/ijms232214303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Exogenous corticosterone administration reduces GABAergic transmission and impairs its 5-HT7 receptor-dependent modulation in the rat dorsal raphe nucleus (DRN), but it is largely unknown how neuronal functions of the DRN are affected by repeated physical and psychological stress. This study compared the effects of repeated restraint stress and corticosterone injections on DRN neuronal excitability, spontaneous synaptic transmission, and its 5-HT7 receptor-dependent modulation. Male Wistar rats received corticosterone injections for 7 or 14 days or were restrained for 10 min twice daily for 3 days. Repeated restraint stress and repeated corticosterone administration evoked similar changes in performance in the forced swim test. They increased the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) recorded from DRN neurons. In contrast to the treatment with corticosterone, restraint stress-induced changes in sEPSC kinetics and decreased intrinsic excitability of DRN neurons did not modify inhibitory transmission. Repeated injections of the 5-HT7 receptor antagonist SB 269970 ameliorated the effects of restraint on excitability and sEPSC frequency but did not restore the altered kinetics of sEPSCs. Thus, repeated restraint stress and repeated corticosterone administration differ in consequences for the intrinsic excitability of DRN projection neurons and their excitatory and inhibitory synaptic inputs. Effects of repeated restraint stress on DRN neurons can be partially abrogated by blocking the 5-HT7 receptor.
Collapse
|
31
|
Nentwig TB, Vaughan DT, Braunscheidel KM, Browning BD, Woodward JJ, Chandler LJ. The lateral habenula is not required for ethanol dependence-induced escalation of drinking. Neuropsychopharmacology 2022; 47:2123-2131. [PMID: 35717465 PMCID: PMC9556754 DOI: 10.1038/s41386-022-01357-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/06/2022] [Accepted: 05/31/2022] [Indexed: 12/30/2022]
Abstract
The lateral habenula (LHb) is an epithalamic nuclei that has been shown to signal the aversive properties of ethanol. The present study tested the hypothesis that activity of the LHb is required for the acquisition and/or expression of dependence-induced escalation of ethanol drinking and somatic withdrawal symptoms. Male Sprague-Dawley rats completed 4 weeks of baseline drinking under a standard intermittent access two-bottle choice (2BC) paradigm before undergoing 2 weeks of daily chronic intermittent ethanol (CIE) via vapor inhalation. Following this CIE exposure period, rats resumed 2BC drinking to assess dependence-induced changes in voluntary ethanol consumption. CIE exposed rats exhibited a significant increase in ethanol drinking that was associated with high levels of blood alcohol and a reduction in somatic symptoms of ethanol withdrawal. However, despite robust cFos activation in the LHb during ethanol withdrawal, chemogenetic inhibition of the LHb did not alter either ethanol consumption or somatic signs of ethanol withdrawal. Consistent with this observation, ablating LHb outputs via electrolytic lesions of the fasciculus retroflexus (FR) did not alter the acquisition of somatic withdrawal symptoms or escalation of ethanol drinking in CIE-exposed rats. The LHb controls activity of the rostromedial tegmental nucleus (RMTg), a midbrain nucleus activated by aversive experiences including ethanol withdrawal. During ethanol withdrawal, both FR lesioned and sham control rats exhibited similar cFos activation in the RMTg, suggesting that RMTg activation during ethanol withdrawal does not require LHb input. These data suggest that, at least in male rats, the LHb is not necessary for the acquisition or expression of escalation of ethanol consumption or expression of somatic symptoms of ethanol withdrawal. Overall, our findings provide evidence that the LHb is dispensable for some of the negative consequences of ethanol withdrawal.
Collapse
Affiliation(s)
- Todd B Nentwig
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Dylan T Vaughan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin M Braunscheidel
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Neuroscience Mount Sinai, New York, NY, USA
| | - Brittney D Browning
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
32
|
Souza R, Bueno D, Lima LB, Muchon MJ, Gonçalves L, Donato J, Shammah-Lagnado SJ, Metzger M. Top-down projections of the prefrontal cortex to the ventral tegmental area, laterodorsal tegmental nucleus, and median raphe nucleus. Brain Struct Funct 2022; 227:2465-2487. [DOI: 10.1007/s00429-022-02538-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
|
33
|
Rahaman SM, Chowdhury S, Mukai Y, Ono D, Yamaguchi H, Yamanaka A. Functional Interaction Between GABAergic Neurons in the Ventral Tegmental Area and Serotonergic Neurons in the Dorsal Raphe Nucleus. Front Neurosci 2022; 16:877054. [PMID: 35663550 PMCID: PMC9160575 DOI: 10.3389/fnins.2022.877054] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/21/2022] [Indexed: 12/02/2022] Open
Abstract
GABAergic neurons in the ventral tegmental area (VTA) have brain-wide projections and are involved in multiple behavioral and physiological functions. Here, we revealed the responsiveness of Gad67+ neurons in VTA (VTAGad67+) to various neurotransmitters involved in the regulation of sleep/wakefulness by slice patch clamp recording. Among the substances tested, a cholinergic agonist activated, but serotonin, dopamine and histamine inhibited these neurons. Dense VTAGad67+ neuronal projections were observed in brain areas regulating sleep/wakefulness, including the central amygdala (CeA), dorsal raphe nucleus (DRN), and locus coeruleus (LC). Using a combination of electrophysiology and optogenetic studies, we showed that VTAGad67+ neurons inhibited all neurons recorded in the DRN, but did not inhibit randomly recorded neurons in the CeA and LC. Further examination revealed that the serotonergic neurons in the DRN (DRN5–HT) were monosynaptically innervated and inhibited by VTAGad67+ neurons. All recorded DRN5–HT neurons received inhibitory input from VTAGad67+ neurons, while only one quarter of them received inhibitory input from local GABAergic neurons. Gad67+ neurons in the DRN (DRNGad67+) also received monosynaptic inhibitory input from VTAGad67+ neurons. Taken together, we found that VTAGad67+ neurons were integrated in many inputs, and their output inhibits DRN5–HT neurons, which may regulate physiological functions including sleep/wakefulness.
Collapse
Affiliation(s)
- Sheikh Mizanur Rahaman
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Srikanta Chowdhury
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Ono
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Yamaguchi
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
- *Correspondence: Akihiro Yamanaka,
| |
Collapse
|
34
|
Grossman CD, Bari BA, Cohen JY. Serotonin neurons modulate learning rate through uncertainty. Curr Biol 2022; 32:586-599.e7. [PMID: 34936883 PMCID: PMC8825708 DOI: 10.1016/j.cub.2021.12.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 10/11/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
Abstract
Regulating how fast to learn is critical for flexible behavior. Learning about the consequences of actions should be slow in stable environments, but accelerate when that environment changes. Recognizing stability and detecting change are difficult in environments with noisy relationships between actions and outcomes. Under these conditions, theories propose that uncertainty can be used to modulate learning rates ("meta-learning"). We show that mice behaving in a dynamic foraging task exhibit choice behavior that varied as a function of two forms of uncertainty estimated from a meta-learning model. The activity of dorsal raphe serotonin neurons tracked both types of uncertainty in the foraging task as well as in a dynamic Pavlovian task. Reversible inhibition of serotonin neurons in the foraging task reproduced changes in learning predicted by a simulated lesion of meta-learning in the model. We thus provide a quantitative link between serotonin neuron activity, learning, and decision making.
Collapse
Affiliation(s)
- Cooper D Grossman
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Bilal A Bari
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Jeremiah Y Cohen
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
35
|
Zhang GM, Wu HY, Cui WQ, Peng W. Multi-level variations of lateral habenula in depression: A comprehensive review of current evidence. Front Psychiatry 2022; 13:1043846. [PMID: 36386995 PMCID: PMC9649931 DOI: 10.3389/fpsyt.2022.1043846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Despite extensive research in recent decades, knowledge of the pathophysiology of depression in neural circuits remains limited. Recently, the lateral habenula (LHb) has been extensively reported to undergo a series of adaptive changes at multiple levels during the depression state. As a crucial relay in brain networks associated with emotion regulation, LHb receives excitatory or inhibitory projections from upstream brain regions related to stress and cognition and interacts with brain regions involved in emotion regulation. A series of pathological alterations induced by aberrant inputs cause abnormal function of the LHb, resulting in dysregulation of mood and motivation, which present with depressive-like phenotypes in rodents. Herein, we systematically combed advances from rodents, summarized changes in the LHb and related neural circuits in depression, and attempted to analyze the intrinsic logical relationship among these pathological alterations. We expect that this summary will greatly enhance our understanding of the pathological processes of depression. This is advantageous for fostering the understanding and screening of potential antidepressant targets against LHb.
Collapse
Affiliation(s)
- Guang-Ming Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hong-Yun Wu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wen-Qiang Cui
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Peng
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
36
|
Cerebellin-2 regulates a serotonergic dorsal raphe circuit that controls compulsive behaviors. Mol Psychiatry 2021; 26:7509-7521. [PMID: 34158618 PMCID: PMC8692491 DOI: 10.1038/s41380-021-01187-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
Cerebellin-1 (Cbln1) and cerebellin-2 (Cbln2) are secreted glycoproteins that are expressed in distinct subsets of neurons throughout the brain. Cbln1 and Cbln2 simultaneously bind to presynaptic neurexins and postsynaptic GluD1 and GluD2, thereby forming trans-synaptic adhesion complexes. Genetic associations link cerebellins, neurexins and GluD's to neuropsychiatric disorders involving compulsive behaviors, such as Tourette syndrome, attention-deficit hyperactivity disorder (ADHD), and obsessive-compulsive disorder (OCD). Extensive evidence implicates dysfunction of serotonergic signaling in these neuropsychiatric disorders. Here, we report that constitutive Cbln2 KO mice, but not Cbln1 KO mice, display robust compulsive behaviors, including stereotypic pattern running, marble burying, explosive jumping, and excessive nest building, and exhibit decreased brain serotonin levels. Strikingly, treatment of Cbln2 KO mice with the serotonin precursor 5-hydroxytryptophan or the serotonin reuptake-inhibitor fluoxetine alleviated compulsive behaviors. Conditional deletion of Cbln2 both from dorsal raphe neurons and from presynaptic neurons synapsing onto dorsal raphe neurons reproduced the compulsive behaviors of Cbln2 KO mice. Finally, injection of recombinant Cbln2 protein into the dorsal raphe of Cbln2 KO mice largely reversed their compulsive behaviors. Taken together, our results show that Cbln2 controls compulsive behaviors by regulating serotonergic circuits in the dorsal raphe.
Collapse
|
37
|
Xu Z, Feng Z, Zhao M, Sun Q, Deng L, Jia X, Jiang T, Luo P, Chen W, Tudi A, Yuan J, Li X, Gong H, Luo Q, Li A. Whole-brain connectivity atlas of glutamatergic and GABAergic neurons in the mouse dorsal and median raphe nuclei. eLife 2021; 10:65502. [PMID: 34792021 PMCID: PMC8626088 DOI: 10.7554/elife.65502] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 11/17/2021] [Indexed: 11/25/2022] Open
Abstract
The dorsal raphe nucleus (DR) and median raphe nucleus (MR) contain populations of glutamatergic and GABAergic neurons that regulate diverse behavioral functions. However, their whole-brain input-output circuits remain incompletely elucidated. We used viral tracing combined with fluorescence micro-optical sectioning tomography to generate a comprehensive whole-brain atlas of inputs and outputs of glutamatergic and GABAergic neurons in the DR and MR. We found that these neurons received inputs from similar upstream brain regions. The glutamatergic and GABAergic neurons in the same raphe nucleus had divergent projection patterns with differences in critical brain regions. Specifically, MR glutamatergic neurons projected to the lateral habenula through multiple pathways. Correlation and cluster analysis revealed that glutamatergic and GABAergic neurons in the same raphe nucleus received heterogeneous inputs and sent different collateral projections. This connectivity atlas further elucidates the anatomical architecture of the raphe nuclei, which could facilitate better understanding of their behavioral functions.
Collapse
Affiliation(s)
- Zhengchao Xu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao Feng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Mengting Zhao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Qingtao Sun
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Lei Deng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Xueyan Jia
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Pan Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Wu Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Ayizuohere Tudi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Xiangning Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Science, Shanghai, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China.,School of Biomedical Engineering, Hainan University, Haikou, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.,HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Science, Shanghai, China
| |
Collapse
|
38
|
Della Vecchia A, Arone A, Piccinni A, Mucci F, Marazziti D. GABA System in Depression: Impact on Pathophysiology and Psychopharmacology. Curr Med Chem 2021; 29:5710-5730. [PMID: 34781862 DOI: 10.2174/0929867328666211115124149] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/21/2021] [Accepted: 09/30/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The pathophysiology of major depressive disorder (MDD), one of the major causes of worldwide disability, is still largely unclear, despite the increasing data reporting evidence of multiple alterations of different systems. Recently, there was a renewed interest in the signalling of gamma aminobutyric acid (GABA) - the main inhibitory neurotransmitter. OBJECTIVE The aim of this study was to review and comment on the available literature about the involvement of GABA in MDD, as well as on novel GABAergic compounds possibly useful as antidepressants. METHODS We carried out a narrative review through Pubmed, Google Scholar and Scopus, by using specific keywords. RESULTS The results, derived from various research tools, strongly support the presence of a deficiency of the GABA system in MDD, which appears to be restored by common antidepressant treatments. More recent publications would indicate the complex interactions between GABA and all the other processes involved in MDD, such as monoamine neurotransmission, hypothalamus-pituitary adrenal axis functioning, neurotrophism, and immune response. Taken together, all these findings seem to further support the complexity of the pathophysiology of MDD, possibly reflecting the heterogeneity of the clinical pictures. CONCLUSION Although further data are necessary to support the specificity of GABA deficiency in MDD, the available findings would suggest that novel GABAergic compounds might constitute innovative therapeutic strategies in MDD.
Collapse
Affiliation(s)
- Alessandra Della Vecchia
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa. Italy
| | - Alessandro Arone
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa. Italy
| | - Armando Piccinni
- Saint Camillus International University of Health and Medical Sciences, Rome. Italy
| | - Federico Mucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena. Italy
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa. Italy
| |
Collapse
|
39
|
Bhave VM, Nectow AR. The dorsal raphe nucleus in the control of energy balance. Trends Neurosci 2021; 44:946-960. [PMID: 34663507 DOI: 10.1016/j.tins.2021.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/04/2021] [Accepted: 09/23/2021] [Indexed: 01/07/2023]
Abstract
Energy balance is orchestrated by an extended network of highly interconnected nuclei across the central nervous system. While much is known about the hypothalamic circuits regulating energy homeostasis, the 'extra-hypothalamic' circuits involved are relatively poorly understood. In this review, we focus on the brainstem's dorsal raphe nucleus (DRN), integrating decades of research linking this structure to the physiologic and behavioral responses that maintain proper energy stores. DRN neurons sense and respond to interoceptive and exteroceptive cues related to energy imbalance and in turn induce appropriate alterations in energy intake and expenditure. The DRN is also molecularly differentiable, with different populations playing distinct and often opposing roles in controlling energy balance. These populations are integrated into the extended circuit known to regulate energy balance. Overall, this review summarizes the key evidence demonstrating an important role for the DRN in regulating energy balance.
Collapse
Affiliation(s)
- Varun M Bhave
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Alexander R Nectow
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
40
|
Alexander C, Vasefi M. Cannabidiol and the corticoraphe circuit in post-traumatic stress disorder. IBRO Neurosci Rep 2021; 11:88-102. [PMID: 34485973 PMCID: PMC8408530 DOI: 10.1016/j.ibneur.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 01/06/2023] Open
Abstract
Post-Traumatic Stress Disorder (PTSD), characterized by re-experiencing, avoidance, negative affect, and impaired memory processing, may develop after traumatic events. PTSD is complicated by impaired plasticity and medial prefrontal cortex (mPFC) activity, hyperactivity of the amygdala, and impaired fear extinction. Cannabidiol (CBD) is a promising candidate for treatment due to its multimodal action that enhances plasticity and calms hyperexcitability. CBD’s mechanism in the mPFC of PTSD patients has been explored extensively, but literature on the mechanism in the dorsal raphe nucleus (DRN) is lacking. Following the PRISMA guidelines, we examined current literature regarding CBD in PTSD and overlapping symptomologies to propose a mechanism by which CBD treats PTSD via corticoraphe circuit. Acute CBD inhibits excess 5-HT release from DRN to amygdala and releases anandamide (AEA) onto amygdala inputs. By first reducing amygdala and DRN hyperactivity, CBD begins to ameliorate activity disparity between mPFC and amygdala. Chronic CBD recruits the mPFC, creating harmonious corticoraphe signaling. DRN releases enough 5-HT to ameliorate mPFC hypoactivity, while the mPFC continuously excites DRN 5-HT neurons via glutamate. Meanwhile, AEA regulates corticoraphe activity to stabilize signaling. AEA prevents DRN GABAergic interneurons from inhibiting 5-HT release so the DRN can assist the mPFC in overcoming its hypoactivity. DRN-mediated restoration of mPFC activity underlies CBD’s mechanism on fear extinction and learning of stress coping. CBD reduces PTSD symptoms via the DRN and corticoraphe circuit. Acute effects of CBD reduce DRN-amygdala excitatory signaling to lessen the activity disparity between amygdala and mPFC. Chronic CBD officially resolves mPFC hypoactivity by facilitating 5-HT release from DRN to mPFC. CBD-facilitated endocannabinoid signaling stabilizes DRN activity and restores mPFC inhibitory control. Chronically administered CBD acts via the corticoraphe circuit to favor fear extinction over fear memory reconsolidation.
Collapse
Key Words
- 2-AG, 2-arachidonoylglycerol
- 5-HT, Serotonin
- 5-HT1AR, 5-HT Receptor Type 1A
- 5-HT2AR, 5-HT Receptor Type 2 A
- AEA, Anandamide
- CB1R, Cannabinoid Receptor Type 1
- CB2R, Cannabinoid Receptor Type 2
- CBD, Cannabidiol
- COVID-19, SARS-CoV-2
- Cannabidiol
- DRN, Dorsal Raphe Nucleus
- ERK1/2, Extracellular Signal-Related Kinases Type 1 or Type 2
- FAAH, Fatty Acid Amide Hydrolase
- GABA, Gamma-Aminobutyric Acid
- GPCRs, G-Protein Coupled Receptors
- NMDAR, N-Methyl-D-aspartate Receptors
- PET, Positron Emission Tomography
- PFC, DRN and Raphe
- PFC, Prefrontal Cortex
- PTSD
- PTSD, Post-Traumatic Stress Disorder
- SSNRI, Selective Norepinephrine Reuptake Inhibitor
- SSRI, Selective Serotonin Reuptake Inhibitor
- Serotonin
- TRPV1, Transient Receptor Potential Vanilloid 1 Channels
- Traumatic Stress
- fMRI, Functional Magnetic Resonance Imaging
- mPFC, Medial Prefrontal Cortex
Collapse
Affiliation(s)
- Claire Alexander
- Department of Biology, Lamar University, Beaumont, TX 77710, USA
| | - Maryam Vasefi
- Department of Biology, Lamar University, Beaumont, TX 77710, USA
| |
Collapse
|
41
|
Lyu S, Guo Y, Zhang L, Tang G, Li R, Yang J, Gao S, Li W, Liu J. Downregulation of astroglial glutamate transporter GLT-1 in the lateral habenula is associated with depressive-like behaviors in a rat model of Parkinson's disease. Neuropharmacology 2021; 196:108691. [PMID: 34197892 DOI: 10.1016/j.neuropharm.2021.108691] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/06/2021] [Accepted: 06/24/2021] [Indexed: 01/11/2023]
Abstract
Recent studies show that neuron-glial communication plays an important role in neurological diseases. Particularly, dysfunction of astroglial glutamate transporter GLT-1 has been involved in various neuropsychiatric disorders, including Parkinson's disease (PD) and depression. Our previous studies indicated hyperactivity of neurons in the lateral habenula (LHb) of hemiparkinsonian rats with depressive-like behaviors. Thus, we hypothesized that impaired expression or function of GLT-1 in the LHb might be a potential contributor to LHb hyperactivity, which consequently induces PD-related depression. In the study, unilateral lesions of the substantia nigra pars compacta (SNc) by 6-hydroxydopamine in rats induced depressive-like behaviors and resulted in neuronal hyperactivity as well as increased glutamate levels in the LHb compared to sham-lesioned rats. Intra-LHb injection of GLT-1 inhibitor WAY-213613 induced the depressive-like behaviors in both groups, but the dose producing behavioral effects in the lesioned rats was lower than that of sham-lesioned rats. In the two groups of rats, WAY-213613 increased the firing rate of LHb neurons and extracellular levels of glutamate, and these excitatory effects in the lesioned rats lasted longer than those in sham-lesioned rats. The functional changes of the GLT-1 which primarily expresses in astrocytes in the LHb may attribute to its downregulation after degeneration of the nigrostriatal pathway. Bioinformatics analysis showed that GLT-1 is correlated with various biomarkers of PD and depression risks. Collectively, our study suggests that astroglial GLT-1 in the LHb regulates the firing activity of the neurons, whereupon its downregulation and dysfunction are closely associated with PD-related depression.
Collapse
Affiliation(s)
- Shuxuan Lyu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Yuan Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Li Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Guoyi Tang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Ruotong Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jie Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Shasha Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Wenjuan Li
- Department of Rehabilitation Medicine, The Second Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jian Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| |
Collapse
|
42
|
Supramammillary neurons projecting to the septum regulate dopamine and motivation for environmental interaction in mice. Nat Commun 2021; 12:2811. [PMID: 33990558 PMCID: PMC8121914 DOI: 10.1038/s41467-021-23040-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 04/06/2021] [Indexed: 12/29/2022] Open
Abstract
The supramammillary region (SuM) is a posterior hypothalamic structure, known to regulate hippocampal theta oscillations and arousal. However, recent studies reported that the stimulation of SuM neurons with neuroactive chemicals, including substances of abuse, is reinforcing. We conducted experiments to elucidate how SuM neurons mediate such effects. Using optogenetics, we found that the excitation of SuM glutamatergic (GLU) neurons was reinforcing in mice; this effect was relayed by their projections to septal GLU neurons. SuM neurons were active during exploration and approach behavior and diminished activity during sucrose consumption. Consistently, inhibition of SuM neurons disrupted approach responses, but not sucrose consumption. Such functions are similar to those of mesolimbic dopamine neurons. Indeed, the stimulation of SuM-to-septum GLU neurons and septum-to-ventral tegmental area (VTA) GLU neurons activated mesolimbic dopamine neurons. We propose that the supramammillo-septo-VTA pathway regulates arousal that reinforces and energizes behavioral interaction with the environment.
Collapse
|
43
|
Liu PF, Wang Y, Zhang R, Xu L, Li JB, Mu D. Propofol modulates inhibitory inputs in paraventricular thalamic nucleus of mice. Neurosci Lett 2021; 756:135950. [PMID: 33979698 DOI: 10.1016/j.neulet.2021.135950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 05/07/2021] [Indexed: 11/19/2022]
Abstract
The mechanisms of general anaesthetics such as propofol have drawn substantial attention. The effects of propofol on inhibitory postsynaptic currents are not exactly the same in different brain nuclei. Recent studies revealed that the paraventricular thalamic nucleus (PVT) is a critical nucleus modulating wakefulness. However, the effects of propofol on PVT neurons and the mechanisms underlying such effects remain unknown. Here, we performed the whole-cell recording of the PVT neurons in acute brain slices and bath application of propofol. We found that propofol hyperpolarized the membrane potentials of the PVT neurons and suppressed the action potentials induced by step-current injection. Propofol did not affect the spontaneous inhibitory postsynaptic currents (sIPSCs) amplitude or frequency, but prolonged the sIPSCs half-width. Besides, propofol increased miniature inhibitory synaptic currents (mIPSCs) frequency and half-width. Furthermore, propofol could induce GABAA receptors-mediated tonic inhibitory currents dose-dependently. Thus, our results demonstrate that propofol hyperpolarizes PVT neurons by modulating inhibitory currents via GABAA receptors in mice.
Collapse
Affiliation(s)
- Peng-Fei Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Xu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Bao Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Di Mu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
44
|
Kim E, Bari BA, Cohen JY. Subthreshold basis for reward-predictive persistent activity in mouse prefrontal cortex. Cell Rep 2021; 35:109082. [PMID: 33951442 PMCID: PMC8167820 DOI: 10.1016/j.celrep.2021.109082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/30/2020] [Accepted: 04/13/2021] [Indexed: 11/30/2022] Open
Abstract
Nervous systems maintain information internally using persistent activity changes. The mechanisms by which this activity arises are incompletely understood. We study prefrontal cortex (PFC) in mice performing behaviors in which stimuli predicted rewards at different delays with different probabilities. We measure membrane potential (Vm) from pyramidal neurons across layers. Reward-predictive persistent firing increases arise due to sustained increases in mean and variance of Vm and are terminated by reward or via centrally generated mechanisms based on reward expectation. Other neurons show persistent decreases in firing rates, maintained by persistent hyperpolarization that is robust to intracellular perturbation. Persistent activity is layer (L)- and cell-type-specific. Neurons with persistent depolarization are primarily located in upper L5, whereas those with persistent hyperpolarization are mostly found in lower L5. L2/3 neurons do not show persistent activity. Thus, reward-predictive persistent activity in PFC is spatially organized and conveys information about internal state via synaptic mechanisms. Kim et al. show sustained changes in membrane potential and firing rates in mouse frontal cortex leading up to an expected reward. These dynamics rely on underlying changes in mean and variance, directly testing prior theoretical studies. Neurons showing increased and decreased activity changes are located in different cortical layers.
Collapse
Affiliation(s)
- Eunyoung Kim
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bilal A Bari
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeremiah Y Cohen
- The Solomon H. Snyder Department of Neuroscience, Brain Science Institute, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
45
|
Vahid-Ansari F, Albert PR. Rewiring of the Serotonin System in Major Depression. Front Psychiatry 2021; 12:802581. [PMID: 34975594 PMCID: PMC8716791 DOI: 10.3389/fpsyt.2021.802581] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin is a key neurotransmitter that is implicated in a wide variety of behavioral and cognitive phenotypes. Originating in the raphe nuclei, 5-HT neurons project widely to innervate many brain regions implicated in the functions. During the development of the brain, as serotonin axons project and innervate brain regions, there is evidence that 5-HT plays key roles in wiring the developing brain, both by modulating 5-HT innervation and by influencing synaptic organization within corticolimbic structures. These actions are mediated by 14 different 5-HT receptors, with region- and cell-specific patterns of expression. More recently, the role of the 5-HT system in synaptic re-organization during adulthood has been suggested. The 5-HT neurons have the unusual capacity to regrow and reinnervate brain regions following insults such as brain injury, chronic stress, or altered development that result in disconnection of the 5-HT system and often cause depression, anxiety, and cognitive impairment. Chronic treatment with antidepressants that amplify 5-HT action, such as selective serotonin reuptake inhibitors (SSRIs), appears to accelerate the rewiring of the 5-HT system by mechanisms that may be critical to the behavioral and cognitive improvements induced in these models. In this review, we survey the possible 5-HT receptor mechanisms that could mediate 5-HT rewiring and assess the evidence that 5-HT-mediated brain rewiring is impacting recovery from mental illness. By amplifying 5-HT-induced rewiring processes using SSRIs and selective 5-HT agonists, more rapid and effective treatments for injury-induced mental illness or cognitive impairment may be achieved.
Collapse
Affiliation(s)
- Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
46
|
Ndyabawe K, Cipriano M, Zhao W, Haidekker M, Yao K, Mao L, Kisaalita WS. Brain-on-a-Chip Device for Modeling Multiregional Networks. ACS Biomater Sci Eng 2020; 7:350-359. [PMID: 33320530 DOI: 10.1021/acsbiomaterials.0c00895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Animal models are frequently used in drug discovery because they represent a mammalian in vivo model system, they are the closest approximation to the human brain, and experimentation in humans is not ethical. Working with postmortem human brain samples is challenging and developing human in vitro systems, which mimic the in vivo human brain, has been challenging. However, the use of animal models in drug discovery for human neurological diseases is currently under scrutiny because data from animal models has come with variations due to genetic differences. Evidence from the literature suggests that techniques to reconstruct multiple neurotransmission projections, which characterize neurological disease circuits in humans, in vitro, have not been demonstrated. This paper presents a multicompartment microdevice for patterning neurospheres and specification of neural stem cell fate toward networks of multiple neuronal phenotypes. We validated our design by specification of human neural stem cells to dopaminergic and GABAergic neurons in different compartments of the device, simultaneously. The neurospheres formed unrestricted robust neuronal circuits between arrays of neurospheres in all compartments of the device. Such a device design may provide a basis for formation of multineurotransmission circuits to model functional connectivity between specific human brain regions, in vitro, using human-derived neural stem cells. This work finds relevance in neurological disease modeling and drug screening using human cell-based assays and may provide the impetus for shifting from animal-based models.
Collapse
|
47
|
Azizi SA. Monoamines: Dopamine, Norepinephrine, and Serotonin, Beyond Modulation, "Switches" That Alter the State of Target Networks. Neuroscientist 2020; 28:121-143. [PMID: 33292070 DOI: 10.1177/1073858420974336] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
How do monoamines influence the perceptual and behavioral aspects of brain function? A library of information regarding the genetic, molecular, cellular, and function of monoamines in the nervous system and other organs has accumulated. We briefly review monoamines' anatomy and physiology and discuss their effects on the target neurons and circuits. Monoaminergic cells in the brain stem receive inputs from sensory, limbic, and prefrontal areas and project extensively to the forebrain and hindbrain. We review selected studies on molecular, cellular, and electrophysiological effects of monoamines on the brain's target areas. The idea is that monoamines, by reversibly modulating the "primary" information processing circuits, regulate and switch the functions of brain networks and can reversibly alter the "brain states," such as consciousness, emotions, and movements. Monoamines, as the drivers of normal motor and sensory brain operations, including housekeeping, play essential roles in pathogenesis of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Sayed Ausim Azizi
- Department of Neurology, Global Neuroscience Institute, Chester, PA, USA
| |
Collapse
|
48
|
Lyu S, Guo Y, Zhang L, Wang Y, Tang G, Li R, Yang J, Gao S, Ma B, Liu J. Blockade of GABA transporter-1 and GABA transporter-3 in the lateral habenula improves depressive-like behaviors in a rat model of Parkinson's disease. Neuropharmacology 2020; 181:108369. [DOI: 10.1016/j.neuropharm.2020.108369] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 12/23/2022]
|
49
|
The Emerging Role of LHb CaMKII in the Comorbidity of Depressive and Alcohol Use Disorders. Int J Mol Sci 2020; 21:ijms21218123. [PMID: 33143210 PMCID: PMC7663385 DOI: 10.3390/ijms21218123] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 01/05/2023] Open
Abstract
Depressive disorders and alcohol use disorders are widespread among the general population and are significant public health and economic burdens. Alcohol use disorders often co-occur with other psychiatric conditions and this dual diagnosis is called comorbidity. Depressive disorders invariably contribute to the development and worsening of alcohol use disorders, and vice versa. The mechanisms underlying these disorders and their comorbidities remain unclear. Recently, interest in the lateral habenula, a small epithalamic brain structure, has increased because it becomes hyperactive in depression and alcohol use disorders, and can inhibit dopamine and serotonin neurons in the midbrain reward center, the hypofunction of which is believed to be a critical contributor to the etiology of depressive disorders and alcohol use disorders as well as their comorbidities. Additionally, calcium/calmodulin-dependent protein kinase II (CaMKII) in the lateral habenula has emerged as a critical player in the etiology of these comorbidities. This review analyzes the interplay of CaMKII signaling in the lateral habenula associated with depressive disorders and alcohol use disorders, in addition to the often-comorbid nature of these disorders. Although most of the CaMKII signaling pathway's core components have been discovered, much remains to be learned about the biochemical events that propagate and link between depression and alcohol abuse. As the field rapidly advances, it is expected that further understanding of the pathology involved will allow for targeted treatments.
Collapse
|
50
|
Ge R, Dai Y. Three-Week Treadmill Exercise Enhances Persistent Inward Currents, Facilitates Dendritic Plasticity, and Upregulates the Excitability of Dorsal Raphe Serotonin Neurons in ePet-EYFP Mice. Front Cell Neurosci 2020; 14:575626. [PMID: 33177992 PMCID: PMC7595958 DOI: 10.3389/fncel.2020.575626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022] Open
Abstract
Exercise plays a key role in preventing or treating mental or motor disorders caused by dysfunction of the serotonergic system. However, the electrophysiological and ionic channel mechanisms underlying these effects remain unclear. In this study, we investigated the effects of 3-week treadmill exercise on the electrophysiological and channel properties of dorsal raphe nucleus (DRN). Serotonin (5-HT) neurons in ePet-EYFP mice, using whole-cell patch clamp recording. Treadmill exercise was induced in ePet-EYFP mice of P21–24 for 3 weeks, and whole-cell patch clamp recording was performed on EYFP-positive 5-HT neurons from DRN slices of P42–45 mice. Experiment data showed that 5-HT neurons in the DRN were a heterogeneous population with multiple firing patterns (single firing, phasic firing, and tonic firing). Persistent inward currents (PICs) with multiple patterns were expressed in 5-HT neurons and composed of Cav1.3 (Ca-PIC) and sodium (Na-PIC) components. Exercise hyperpolarized the voltage threshold for action potential (AP) by 3.1 ± 1.0 mV (control: n = 14, exercise: n = 18, p = 0.005) and increased the AP amplitude by 6.7 ± 3.0 mV (p = 0.031) and firing frequency by more than 22% especially within a range of current stimulation stronger than 70 pA. A 3-week treadmill exercise was sufficient to hyperpolarize PIC onset by 2.6 ± 1.3 mV (control: −53.4 ± 4.7 mV, n = 28; exercise: −56.0 ± 4.7 mV, n = 25, p = 0.050) and increase the PIC amplitude by 28% (control: 193.6 ± 81.8 pA; exercise: 248.5 ± 105.4 pA, p = 0.038). Furthermore, exercise hyperpolarized Na-PIC onset by 3.8 ± 1.8 mV (control: n = 8, exercise: n = 9, p = 0.049) and increased the Ca-PIC amplitude by 23% (p = 0.013). The exercise-induced enhancement of the PIC amplitude was mainly mediated by Ca-PIC and hyperpolarization of PIC onset by Na-PIC. Moreover, exercise facilitated dendritic plasticity, which was shown as the increased number of branch points by 1.5 ± 0.5 (p = 0.009) and dendritic branches by 2.1 ± 0.6 (n = 20, p = 0.001) and length by 732.0 ± 100.1 μm (p < 0.001) especially within the range of 50–200 μm from the soma. Functional analysis suggested that treadmill exercise enhanced Na-PIC for facilitation of spike initiation and Ca-PIC for regulation of repetitive firing. We concluded that PICs broadly existed in DRN 5-HT neurons and could influence serotonergic neurotransmission in juvenile mice and that 3-week treadmill exercise induced synaptic adaptations, enhanced PICs, and thus upregulated the excitability of the 5-HT neurons.
Collapse
Affiliation(s)
- Renkai Ge
- Shanghai Key Laboratory of Multidimensional Information Processing, School of Communication and Electronic Engineering, East China Normal University, Shanghai, China.,School of Physical Education and Health Care, East China Jiaotong University, Nanchang, China
| | - Yue Dai
- Shanghai Key Laboratory of Multidimensional Information Processing, School of Communication and Electronic Engineering, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education and Health Care, East China Normal University, Shanghai, China
| |
Collapse
|