51
|
Pierson SR, Kolling LJ, James TD, Pushpavathi SG, Marcinkiewcz CA. Serotonergic dysfunction may mediate the relationship between alcohol consumption and Alzheimer's disease. Pharmacol Res 2024; 203:107171. [PMID: 38599469 PMCID: PMC11088857 DOI: 10.1016/j.phrs.2024.107171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
The impact of Alzheimer's disease (AD) and its related dementias is rapidly expanding, and its mitigation remains an urgent social and technical challenge. To date there are no effective treatments or interventions for AD, but recent studies suggest that alcohol consumption is correlated with the risk of developing dementia. In this review, we synthesize data from preclinical, clinical, and epidemiological models to evaluate the combined role of alcohol consumption and serotonergic dysfunction in AD, underscoring the need for further research on this topic. We first discuss the limitations inherent to current data-collection methods, and how neuropsychiatric symptoms common among AD, alcohol use disorder, and serotonergic dysfunction may mask their co-occurrence. We additionally describe how excess alcohol consumption may accelerate the development of AD via direct effects on serotonergic function, and we explore the roles of neuroinflammation and proteostasis in mediating the relationship between serotonin, alcohol consumption, and AD. Lastly, we argue for a shift in current research to disentangle the pathogenic effects of alcohol on early-affected brainstem structures in AD.
Collapse
Affiliation(s)
- Samantha R Pierson
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | - Louis J Kolling
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | - Thomas D James
- Department of Neuroscience and Pharmacology, University of Iowa, United States
| | | | | |
Collapse
|
52
|
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
|
53
|
Gao H, Wang J, Zhang R, Luo T. Recent advances in neural mechanism of general anesthesia induced unconsciousness: insights from optogenetics and chemogenetics. Front Pharmacol 2024; 15:1360864. [PMID: 38655183 PMCID: PMC11035785 DOI: 10.3389/fphar.2024.1360864] [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: 12/24/2023] [Accepted: 03/20/2024] [Indexed: 04/26/2024] Open
Abstract
For over 170 years, general anesthesia has played a crucial role in clinical practice, yet a comprehensive understanding of the neural mechanisms underlying the induction of unconsciousness by general anesthetics remains elusive. Ongoing research into these mechanisms primarily centers around the brain nuclei and neural circuits associated with sleep-wake. In this context, two sophisticated methodologies, optogenetics and chemogenetics, have emerged as vital tools for recording and modulating the activity of specific neuronal populations or circuits within distinct brain regions. Recent advancements have successfully employed these techniques to investigate the impact of general anesthesia on various brain nuclei and neural pathways. This paper provides an in-depth examination of the use of optogenetic and chemogenetic methodologies in studying the effects of general anesthesia on specific brain nuclei and pathways. Additionally, it discusses in depth the advantages and limitations of these two methodologies, as well as the issues that must be considered for scientific research applications. By shedding light on these facets, this paper serves as a valuable reference for furthering the accurate exploration of the neural mechanisms underlying general anesthesia. It aids researchers and clinicians in effectively evaluating the applicability of these techniques in advancing scientific research and clinical practice.
Collapse
Affiliation(s)
- Hui Gao
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jingyi Wang
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Rui Zhang
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Tao Luo
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
54
|
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
|
55
|
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
|
56
|
O'Connell CJ, Reeder EL, Hymore JA, Brown RS, Notorgiacomo GA, Collins SM, Gudelsky GA, Robson MJ. Transcriptomic dynamics governing serotonergic dysregulation in the dorsal raphe nucleus following mild traumatic brain injury. Exp Neurol 2024; 374:114695. [PMID: 38246304 DOI: 10.1016/j.expneurol.2024.114695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/15/2023] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
Mild traumatic brain injury (mTBI) is a leading cause of disability in the United States, with neuropsychiatric disturbances such as depression, anxiety, PTSD, and social disturbances being common comorbidities following injury. The molecular mechanisms driving neuropsychiatric complications following neurotrauma are not well understood and current FDA-approved pharmacotherapies employed to ameliorate these comorbidities lack desired efficacy. Concerted efforts to understand the molecular mechanisms of and identify novel drug candidates for treating neurotrauma-elicited neuropsychiatric sequelae are clearly needed. Serotonin (5-HT) is linked to the etiology of neuropsychiatric disorders, however our understanding of how various forms of TBI directly affect 5-HT neurotransmission is limited. 5-HT neurons originate in the raphe nucleus (RN) of the midbrain and project throughout the brain to regulate diverse behavioral phenotypes. We hypothesize that the characterization of the dynamics governing 5-HT neurotransmission after injury will drive the discovery of novel drug targets and lead to a greater understanding of the mechanisms associated with neuropsychiatric disturbances following mild TBI (mTBI). Herein, we provide evidence that closed-head mTBI alters total DRN 5-HT levels, with RNA sequencing of the DRN revealing injury-derived alterations in transcripts required for the development, identity, and functional stability of 5-HT neurons. Further, using gene ontology analyses combined with immunohistological analyses, we have identified a novel mechanism of transcriptomic control within 5-HT neurons that may directly influence 5-HT neuron identity/function post-injury. These studies provide molecular evidence of injury-elicited 5-HT neuron dysregulation, data which may expedite the identification of novel therapeutic targets to attenuate TBI-elicited neuropsychiatric sequelae.
Collapse
Affiliation(s)
- Christopher J O'Connell
- University of Cincinnati, James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH, USA
| | - Evan L Reeder
- University of Cincinnati, James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH, USA
| | - Jacob A Hymore
- University of Cincinnati, James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH, USA
| | - Ryan S Brown
- University of Cincinnati, James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH, USA
| | | | - Sean M Collins
- University of Cincinnati, James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH, USA
| | - Gary A Gudelsky
- University of Cincinnati, James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthew J Robson
- University of Cincinnati, James L. Winkle College of Pharmacy, Division of Pharmaceutical Sciences, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
57
|
Wang H, Sun Y, Wang W, Wang X, Zhang J, Bai Y, Wang K, Luan L, Yan J, Qin L. Mapping the 5-HTergic neural pathways in perimenopausal mice and elucidating the role of oestrogen receptors in 5-HT neurotransmission. Heliyon 2024; 10:e27976. [PMID: 38510058 PMCID: PMC10951590 DOI: 10.1016/j.heliyon.2024.e27976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Perimenopausal syndrome (PMS) encompasses neuropsychiatric symptoms, such as hot flashes and depression, which are associated with alterations in the 5-HTergic neural pathway in the brain. However, the specific changes and mechanisms underlying these alterations remain unclear. In this study, ovariectomized mice were used to successfully establish a perimenopause model, and the changes in the expression of 5-HT and its receptors (5-HT1AR and 5-HT2AR) across 72 brain regions in these ovariectomized mice were assessed by immunohistochemistry. Although both 5-HT and 5-HT1AR were widely expressed throughout the brain, only a limited number of regions expressed 5-HT2AR. Notably, decreased expression of 5-HT was observed across almost all brain regions in the ovariectomy (OVX) group compared with the Sham group. Altered expression of both receptors was found within areas related to hot flashes (the preoptic area) or mood disorders (the amygdala). Additionally, reduced oestrogen receptor (ER)α/β expression was detected in cells in the raphe nucleus (RN), an area known to regulate body temperature. Results showed that ERα/β positively regulate the transcriptional activity of the enzymes TPH2/MAOA, which are involved in serotonin metabolism during perimenopause. This study revealed the changes in 5-HT neuropathways (5-HT, 5-HT1AR and 5-HT2AR) in perimenopausal mice, mainly in brain regions related to regulation of the body temperature, mood, sleep and memory. This study clarified that the expression of oestrogen receptor decreased in perimenopause, which regulated the transcription levels of TPH2 and MAOA, and ultimately led to the reduction of 5-HT content, providing a new target for clinical diagnosis and treatment of perimenopausal diseases.
Collapse
Affiliation(s)
- Hanfei Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yanrong Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Wenjuan Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiangqiu Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jinglin Zhang
- Department of Dental Medicine, School of Dental Medicine, Yuncheng Vocational Nursing College, Yuncheng, 044000, China
| | - Yu Bai
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ke Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Liju Luan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Junhao Yan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lihua Qin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| |
Collapse
|
58
|
Trujillo V, Camilo TA, Valentim-Lima E, Carbalan QSR, Dos-Santos RC, Felintro V, Reis LC, Lustrino D, Rorato R, Mecawi AS. Neonatal treatment with para-chlorophenylalanine (pCPA) induces adolescent hyperactivity associated with changes in the paraventricular nucleus Crh and Trh expressions. Behav Brain Res 2024; 462:114867. [PMID: 38246394 DOI: 10.1016/j.bbr.2024.114867] [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: 08/14/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024]
Abstract
Disruption of the brain serotoninergic (5-HT) system during development induces long-lasting changes in molecular profile, cytoarchitecture, and function of neurons, impacting behavioral regulation throughout life. In male and female rats, we investigate the effect of neonatal tryptophan hydroxylase (TPH) inhibition by using para-chlorophenylalanine (pCPA) on the expression of 5-HTergic system components and neuropeptides related to adolescent social play behavior regulation. We observed sex-dependent 5-HT levels decrease after pCPA-treatment in the dorsal raphe nucleus (DRN) at 17 and 35 days. Neonatal pCPA-treatment increased playing, social and locomotory behaviors assessed in adolescent rats of both sexes. The pCPA-treated rats demonstrated decreased Crh (17 days) and increased Trh (35 days) expression in the hypothalamic paraventricular nucleus (PVN). There was sex dimorphism in Htr2c (17 days) and VGF (35 days) in the prefrontal cortex, with the females expressing higher levels of it than males. Our results indicate that neonatal pCPA-treatment results in a long-lasting and sex-dependent DRN 5-HT synthesis changes, decreased Crh, and increased Trh expression in the PVN, resulting in a hyperactivity-like phenotype during adolescence. The present work demonstrates that the impairment of TPH function leads to neurobehavioral disorders related to hyperactivity and impulsivity, such as attention deficit hyperactivity disorder (ADHD).
Collapse
Affiliation(s)
- Verónica Trujillo
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil; Department of Physiology, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Tays Araújo Camilo
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Evandro Valentim-Lima
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Quézia S R Carbalan
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Brazil
| | - Raoni C Dos-Santos
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Brazil
| | - Viviane Felintro
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Brazil
| | - Luís C Reis
- Department of Physiological Sciences, Instituto de Ciências Biológicas e da Saúde, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica, Brazil
| | - Danilo Lustrino
- Laboratory of Basic and Behavioral Neuroendocrinology, Department of Physiology, Centro de Ciências Biológicas e da Saúde, Universidade Federal de Sergipe (UFS), São Cristóvão, Brazil
| | - Rodrigo Rorato
- Laboratory of Stress Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - André S Mecawi
- Laboratory of Molecular Neuroendocrinology, Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
59
|
Zhao Y, Huang CX, Gu Y, Zhao Y, Ren W, Wang Y, Chen J, Guan NN, Song J. Serotonergic modulation of vigilance states in zebrafish and mice. Nat Commun 2024; 15:2596. [PMID: 38519480 PMCID: PMC10959952 DOI: 10.1038/s41467-024-47021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
Vigilance refers to being alertly watchful or paying sustained attention to avoid potential threats. Animals in vigilance states reduce locomotion and have an enhanced sensitivity to aversive stimuli so as to react quickly to dangers. Here we report that an unconventional 5-HT driven mechanism operating at neural circuit level which shapes the internal state underlying vigilance behavior in zebrafish and male mice. The neural signature of internal vigilance state was characterized by persistent low-frequency high-amplitude neuronal synchrony in zebrafish dorsal pallium and mice prefrontal cortex. The neuronal synchronization underlying vigilance was dependent on intense release of 5-HT induced by persistent activation of either DRN 5-HT neuron or local 5-HT axon terminals in related brain regions via activation of 5-HTR7. Thus, we identify a mechanism of vigilance behavior across species that illustrates the interplay between neuromodulators and neural circuits necessary to shape behavior states.
Collapse
Affiliation(s)
- Yang Zhao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Chun-Xiao Huang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Yiming Gu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Yacong Zhao
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Wenjie Ren
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Yutong Wang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Jinjin Chen
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China
| | - Na N Guan
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China.
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China.
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Jianren Song
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
- Clinical Center for Brain and Spinal Cord Research, Tongji University, 200092, Shanghai, China.
- Frontiers Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai, China.
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
60
|
Gullino LS, Fuller C, Dunn P, Collins HM, El Mestikawy S, Sharp T. Evidence for a Role of 5-HT-glutamate Co-releasing Neurons in Acute Stress Mechanisms. ACS Chem Neurosci 2024; 15:1185-1196. [PMID: 38377469 PMCID: PMC10958520 DOI: 10.1021/acschemneuro.3c00758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/22/2024] Open
Abstract
A major subpopulation of midbrain 5-hydroxytryptamine (5-HT) neurons expresses the vesicular glutamate transporter 3 (VGLUT3) and co-releases 5-HT and glutamate, but the function of this co-release is unclear. Given the strong links between 5-HT and uncontrollable stress, we used a combination of c-Fos immunohistochemistry and conditional gene knockout mice to test the hypothesis that glutamate co-releasing 5-HT neurons are activated by stress and involved in stress coping. Acute, uncontrollable swim stress increased c-Fos immunoreactivity in neurons co-expressing VGLUT3 and the 5-HT marker tryptophan hydroxylase 2 (TPH2) in the dorsal raphe nucleus (DRN). This effect was localized in the ventral DRN subregion and prevented by the antidepressant fluoxetine. In contrast, a more controllable stressor, acute social defeat, had no effect on c-Fos immunoreactivity in VGLUT3-TPH2 co-expressing neurons in the DRN. To test whether activation of glutamate co-releasing 5-HT neurons was causally linked to stress coping, mice with a specific deletion of VGLUT3 in 5-HT neurons were exposed to acute swim stress. Compared to wildtype controls, the mutant mice showed increased climbing behavior, a measure of active coping. Wildtype mice also showed increased climbing when administered fluoxetine, revealing an interesting parallel between the behavioral effects of genetic loss of VGLUT3 in 5-HT neurons and 5-HT reuptake inhibition. We conclude that 5-HT-glutamate co-releasing neurons are recruited by exposure to uncontrollable stress. Furthermore, natural variation in the balance of 5-HT and glutamate co-released at the 5-HT synapse may impact stress susceptibility.
Collapse
Affiliation(s)
- L. Sophie Gullino
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Cara Fuller
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Poppy Dunn
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Helen M. Collins
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| | - Salah El Mestikawy
- Douglas
Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC H4H
1R3, Canada
- Sorbonne
Université, INSERM, CNRS, Neuroscience Paris Seine –
Institut de Biologie Paris Seine (NPS – IBPS), 75005 Paris, France
| | - Trevor Sharp
- University
Department of Pharmacology, University of
Oxford, Mansfield Road, Oxford OX1 3QT, U.K.
| |
Collapse
|
61
|
Li HQ, Jiang W, Ling L, Pratelli M, Chen C, Gupta V, Godavarthi SK, Spitzer NC. Generalized fear after acute stress is caused by change in neuronal cotransmitter identity. Science 2024; 383:1252-1259. [PMID: 38484078 PMCID: PMC11830151 DOI: 10.1126/science.adj5996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/22/2024] [Indexed: 03/19/2024]
Abstract
Overgeneralization of fear to harmless situations is a core feature of anxiety disorders resulting from acute stress, yet the mechanisms by which fear becomes generalized are poorly understood. In this study, we show that generalized fear in mice results from a transmitter switch from glutamate to γ-aminobutyric acid (GABA) in serotonergic neurons of the lateral wings of the dorsal raphe. Similar change in transmitter identity was found in the postmortem brains of individuals with posttraumatic stress disorder (PTSD). Overriding the transmitter switch in mice prevented the acquisition of generalized fear. Corticosterone release and activation of glucocorticoid receptors mediated the switch, and prompt antidepressant treatment blocked the cotransmitter switch and generalized fear. Our results provide important insight into the mechanisms involved in fear generalization.
Collapse
Affiliation(s)
- Hui-quan Li
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Wuji Jiang
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Li Ling
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Marta Pratelli
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Cong Chen
- Department of Cellular and Molecular Medicine, University of California San Diego; La Jolla, California 92093
| | - Vaidehi Gupta
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Swetha K. Godavarthi
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| | - Nicholas C. Spitzer
- Neurobiology Department, School of Biological Sciences and Center for Neural Circuits and Behavior, University of California San Diego; La Jolla, California 92093
- Kavli Institute for Brain and Mind, University of California San Diego; La Jolla, California 92093
| |
Collapse
|
62
|
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
|
63
|
Zhu X, Huang JY, Dong WY, Tang HD, Xu S, Wu Q, Zhang H, Cheng PK, Jin Y, Zhu MY, Zhao W, Mao Y, Wang H, Zhang Y, Wang H, Tao W, Tian Y, Bai L, Zhang Z. Somatosensory cortex and central amygdala regulate neuropathic pain-mediated peripheral immune response via vagal projections to the spleen. Nat Neurosci 2024; 27:471-483. [PMID: 38291284 DOI: 10.1038/s41593-023-01561-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/13/2023] [Indexed: 02/01/2024]
Abstract
Pain involves neuroimmune crosstalk, but the mechanisms of this remain unclear. Here we showed that the splenic T helper 2 (TH2) immune cell response is differentially regulated in male mice with acute versus chronic neuropathic pain and that acetylcholinergic neurons in the dorsal motor nucleus of the vagus (AChDMV) directly innervate the spleen. Combined in vivo recording and immune cell profiling revealed the following two distinct circuits involved in pain-mediated peripheral TH2 immune response: glutamatergic neurons in the primary somatosensory cortex (GluS1HL)→AChDMV→spleen circuit and GABAergic neurons in the central nucleus of the amygdala (GABACeA)→AChDMV→spleen circuit. The acute pain condition elicits increased excitation from GluS1HL neurons to spleen-projecting AChDMV neurons and increased the proportion of splenic TH2 immune cells. The chronic pain condition increased inhibition from GABACeA neurons to spleen-projecting AChDMV neurons and decreased splenic TH2 immune cells. Our study thus demonstrates how the brain encodes pain-state-specific immune responses in the spleen.
Collapse
Affiliation(s)
- Xia Zhu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Ji-Ye Huang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Wan-Ying Dong
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Hao-Di Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Si Xu
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei, P. R. China
| | - Qielan Wu
- Department of Oncology, The First Affiliated Hospital of USTC, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Huimin Zhang
- Department of Oncology, The First Affiliated Hospital of USTC, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Ping-Kai Cheng
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Yuxin Jin
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Meng-Yu Zhu
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, P. R. China
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, P. R. China
| | - Wan Zhao
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of University of Science and Technique of China, Hefei, P. R. China
| | - Yu Mao
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
- Department of Anesthesiology and Pain Management, The First Affiliated Hospital of Anhui Medical University, Hefei, P. R. China
| | - Haitao Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, P. R. China
| | - Yan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Hao Wang
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, P. R. China
| | - Wenjuan Tao
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, P. R. China.
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, P. R. China.
| | - Yanghua Tian
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei, P. R. China.
| | - Li Bai
- Department of Oncology, The First Affiliated Hospital of USTC, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China.
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China.
- Department of Biophysics and Neurobiology, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, P. R. China.
- The Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China.
| |
Collapse
|
64
|
Bonaldo B, Casile A, Ostuni MT, Bettarelli M, Nasini S, Marraudino M, Panzica G, Gotti S. Perinatal exposure to bisphenol A or S: Effects on anxiety-related behaviors and serotonergic system. CHEMOSPHERE 2024; 349:140827. [PMID: 38042429 DOI: 10.1016/j.chemosphere.2023.140827] [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: 06/22/2023] [Revised: 11/06/2023] [Accepted: 11/26/2023] [Indexed: 12/04/2023]
Abstract
Bisphenols, synthetic organic compounds used in the production of plastics, are an extremely abundant class of Endocrine Disrupting Chemicals, i.e., exogenous chemicals or mixtures of chemicals that can interfere with any aspect of hormone action. Exposure to BPs can lead to a wide range of effects, and it is especially dangerous if it occurs during specific critical periods of life. Focusing on the perinatal exposure to BPA or its largely used substitute BPS, we investigated the effects on anxiety-related behaviors and the serotonergic system, which is highly involved in controlling these behaviors, in adult mice. We treated C57BL/6J dams orally with a dose of 4 μg/kg body weight/day (i.e., EFSA TDI) of BPA or BPS dissolved in corn oil or with vehicle alone, at the onset of mating and continued treatment until the offspring were weaned. Adult offspring of both sexes performed the elevated plus maze and the open field tests. Then, we analyzed the serotonergic system in dorsal (DR) and median (MnR) raphe nuclei by immunohistochemical techniques. Behavioral tests highlighted alterations in BPA- and BPS-treated mice, suggesting different effects of the bisphenols exposure on anxiety-related behavior in males (anxiolytic) and females (anxiogenic). The analysis of the serotonergic system highlighted a sex dimorphism in the DR only, with control females showing higher values of serotonin immunoreactivity (5-HT-ir) than control males. BPA-treated males displayed a significant increase of 5-HT-ir in all analyzed nuclei, whereas BPS-treated males showed an increase in ventral DR only. In females, both bisphenols-treated groups showed a significant increase of 5-HT-ir in dorsal DR compared to the controls, and BPA-treated females also showed a significant increase in MnR.These results provide evidence that exposure during the early phases of life to BPA or BPS alters anxiety and the raphe serotonergic neurons in a sex-dependent manner.
Collapse
Affiliation(s)
- Brigitta Bonaldo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043 Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy; Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy.
| | - Antonino Casile
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043 Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy; School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri, 9, Camerino, 62032, Italy
| | - Marialaura Teresa Ostuni
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043 Orbassano, Turin, Italy
| | - Martina Bettarelli
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043 Orbassano, Turin, Italy
| | - Sofia Nasini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Meneghetti 2, 35131, Padua, PD, Italy
| | - Marilena Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043 Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy
| | - GianCarlo Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043 Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy
| | - Stefano Gotti
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Regione Gonzole, 10-10043 Orbassano, Turin, Italy; Department of Neuroscience "Rita Levi-Montalcini", University of Turin, Via Cherasco 15, 10126, Turin, Italy
| |
Collapse
|
65
|
Zhang Y, Pan YD, Zheng WY, Li HY, Zhu MZ, Ou Yang WJ, Qian Y, Turecki G, Mechawar N, Zhu XH. Enhancing HIF-1α-P2X2 signaling in dorsal raphe serotonergic neurons promotes psychological resilience. Redox Biol 2024; 69:103005. [PMID: 38150991 PMCID: PMC10788260 DOI: 10.1016/j.redox.2023.103005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 12/29/2023] Open
Abstract
Major depressive disorder (MDD) is a devastating condition. Although progress has been made in the past seven decades, patients with MDD continue to receive an inadequate treatment, primarily due to the late onset of first-line antidepressant drugs and to their acute withdrawal symptoms. Resilience is the ability to rebound from adversity in a healthy manner and many people have psychological resilience. Revealing the mechanisms and identifying methods promoting resilience will hopefully lead to more effective prevention strategies and treatments for depression. In this study, we found that intermittent hypobaric hypoxia training (IHHT), a method for training pilots and mountaineers, enhanced psychological resilience in adult mice. IHHT produced a sustained antidepressant-like effect in mouse models of depression by inducing long-term (up to 3 months after this treatment) overexpression of hypoxia-inducible factor (HIF)-1α in the dorsal raphe nucleus (DRN) of adult mice. Moreover, DRN-infusion of cobalt chloride, which mimics hypoxia increasing HIF-1α expression, triggered a rapid and long-lasting antidepressant-like effect. Down-regulation of HIF-1α in the DRN serotonergic (DRN5-HT) neurons attenuated the effects of IHHT. HIF-1α translationally regulated the expression of P2X2, and conditionally knocking out P2rx2 (encodes P2X2 receptors) in DRN5-HT neurons, in turn, attenuated the sustained antidepressant-like effect of IHHT, but not its acute effect. In line with these results, a single sub-anesthetic dose of ketamine enhanced HIF-1α-P2X2 signaling, which is essential for its rapid and long-lasting antidepressant-like effect. Notably, we found that P2X2 protein levels were significantly lower in the DRN of patients with MDD than that of control subjects. Together, these findings elucidate the molecular mechanism underlying IHHT promoting psychological resilience and highlight enhancing HIF-1α-P2X2 signaling in DRN5-HT neurons as a potential avenue for screening novel therapeutic treatments for MDD.
Collapse
Affiliation(s)
- Yuan Zhang
- School of Psychology, Shenzhen University, Shenzhen, China; Research Center for Brain Health, Pazhou Lab, Guangzhou, China
| | - Yi-da Pan
- Research Center for Brain Health, Pazhou Lab, Guangzhou, China
| | - Wen-Ying Zheng
- School of Psychology, Shenzhen University, Shenzhen, China; Research Center for Brain Health, Pazhou Lab, Guangzhou, China
| | - Huan-Yu Li
- Research Center for Brain Health, Pazhou Lab, Guangzhou, China
| | - Min-Zhen Zhu
- Research Center for Brain Health, Pazhou Lab, Guangzhou, China
| | - Wen-Jie Ou Yang
- School of Psychology, Shenzhen University, Shenzhen, China; Research Center for Brain Health, Pazhou Lab, Guangzhou, China
| | - Yu Qian
- Research Center for Brain Health, Pazhou Lab, Guangzhou, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Gustavo Turecki
- Department of Psychiatry, McGill University, McGill Group for Suicide Studies, Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, (Québec), Canada
| | - Naguib Mechawar
- Department of Psychiatry, McGill University, McGill Group for Suicide Studies, Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, (Québec), Canada
| | - Xin-Hong Zhu
- School of Psychology, Shenzhen University, Shenzhen, China; Research Center for Brain Health, Pazhou Lab, Guangzhou, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.
| |
Collapse
|
66
|
Yang H, You L, Wang Z, Yang L, Wang X, Wu W, Zhi H, Rong G, Sheng Y, Liu X, Liu L. Bile duct ligation elevates 5-HT levels in cerebral cortex of rats partly due to impairment of brain UGT1A6 expression and activity via ammonia accumulation. Redox Biol 2024; 69:103019. [PMID: 38163420 PMCID: PMC10794929 DOI: 10.1016/j.redox.2023.103019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024] Open
Abstract
Hepatic encephalopathy (HE) is often associated with endogenous serotonin (5-HT) disorders. However, the reason for elevated brain 5-HT levels due to liver failure remains unclear. This study aimed to investigate the mechanism by which liver failure increases brain 5-HT levels and the role in behavioral abnormalities in HE. Using bile duct ligation (BDL) rats as a HE model, we verified the elevated 5-HT levels in the cortex but not in the hippocampus and striatum, and found that this cortical 5-HT overload may be caused by BDL-mediated inhibition of UDP-glucuronosyltransferase 1A6 (UGT1A6) expression and activity in the cortex. The intraventricular injection of the UGT1A6 inhibitor diclofenac into rats demonstrated that the inhibition of brain UGT1A6 activity significantly increased cerebral 5-HT levels and induced HE-like behaviors. Co-immunofluorescence experiments demonstrated that UGT1A6 is primarily expressed in astrocytes. In vitro studies confirmed that NH4Cl activates the ROS-ERK pathway to downregulate UGT1A6 activity and expression in U251 cells, which can be reversed by the oxidative stress antagonist N-acetyl-l-cysteine and the ERK inhibitor U0126. Silencing Hepatocyte Nuclear Factor 4α (HNF4α) suppressed UGT1A6 expression whilst overexpressing HNF4α increased Ugt1a6 promotor activity. Meanwhile, both NH4Cl and the ERK activator TBHQ downregulated HNF4α and UGT1A6 expression. In the cortex of hyperammonemic rats, we also found activation of the ROS-ERK pathway, decreases in HNF4α and UGT1A6 expression, and increases in brain 5-HT content. These results prove that the ammonia-mediated ROS-ERK pathway activation inhibits HNF4α expression to downregulate UGT1A6 expression and activity, thereby increasing cerebral 5-HT content and inducing manic-like HE symptoms. This is the first study to reveal the mechanism of elevated cortical 5-HT concentration in a state of liver failure and elucidate its association with manic-like behaviors in HE.
Collapse
Affiliation(s)
- Hanyu Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Linjun You
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, 210009, Nanjing, China
| | - Zhongyan Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lu Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xun Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Wenhan Wu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hao Zhi
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guangmei Rong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yun Sheng
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaodong Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Li Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China; Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
67
|
O’Connell CJ, Brown RS, Peach TM, Traubert OD, Schwierling HC, Notorgiacomo GA, Robson MJ. Strain in the Midbrain: Impact of Traumatic Brain Injury on the Central Serotonin System. Brain Sci 2024; 14:51. [PMID: 38248266 PMCID: PMC10813794 DOI: 10.3390/brainsci14010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/23/2024] Open
Abstract
Traumatic brain injury (TBI) is a pervasive public health crisis that severely impacts the quality of life of affected individuals. Like peripheral forms of trauma, TBI results from extraordinarily heterogeneous environmental forces being imparted on the cranial space, resulting in heterogeneous disease pathologies. This has made therapies for TBI notoriously difficult to develop, and currently, there are no FDA-approved pharmacotherapies specifically for the acute or chronic treatment of TBI. TBI is associated with changes in cognition and can precipitate the onset of debilitating psychiatric disorders like major depressive disorder (MDD), generalized anxiety disorder (GAD), and post-traumatic stress disorder (PTSD). Complicating these effects of TBI, FDA-approved pharmacotherapies utilized to treat these disorders often fail to reach the desired level of efficacy in the context of neurotrauma. Although a complicated association, decades of work have linked central serotonin (5-HT) neurotransmission as being involved in the etiology of a myriad of neuropsychiatric disorders, including MDD and GAD. 5-HT is a biogenic monoamine neurotransmitter that is highly conserved across scales of biology. Though the majority of 5-HT is isolated to peripheral sites such as the gastrointestinal (GI) tract, 5-HT neurotransmission within the CNS exerts exquisite control over diverse biological functions, including sleep, appetite and respiration, while simultaneously establishing normal mood, perception, and attention. Although several key studies have begun to elucidate how various forms of neurotrauma impact central 5-HT neurotransmission, a full determination of precisely how TBI disrupts the highly regulated dynamics of 5-HT neuron function and/or 5-HT neurotransmission has yet to be conceptually or experimentally resolved. The purpose of the current review is, therefore, to integrate the disparate bodies of 5-HT and TBI research and synthesize insight into how new combinatorial research regarding 5-HT neurotransmission and TBI may offer an informed perspective into the nature of TBI-induced neuropsychiatric complications.
Collapse
Affiliation(s)
- Christopher J. O’Connell
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | - Ryan S. Brown
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | - Taylor M. Peach
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | - Owen D. Traubert
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA;
| | - Hana C. Schwierling
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
| | | | - Matthew J. Robson
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA; (C.J.O.); (R.S.B.); (T.M.P.)
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
68
|
Jeong M, Choi JH, Jang H, Sohn DH, Wang Q, Lee J, Yao L, Lee EJ, Fan J, Pratelli M, Wang EH, Snyder CN, Wang XY, Shin S, Gittis AH, Sung TC, Spitzer NC, Lim BK. Viral vector-mediated transgene delivery with novel recombinase systems for targeting neuronal populations defined by multiple features. Neuron 2024; 112:56-72.e4. [PMID: 37909037 PMCID: PMC10916502 DOI: 10.1016/j.neuron.2023.09.038] [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: 07/18/2022] [Revised: 05/21/2023] [Accepted: 09/26/2023] [Indexed: 11/02/2023]
Abstract
A comprehensive understanding of neuronal diversity and connectivity is essential for understanding the anatomical and cellular mechanisms that underlie functional contributions. With the advent of single-cell analysis, growing information regarding molecular profiles leads to the identification of more heterogeneous cell types. Therefore, the need for additional orthogonal recombinase systems is increasingly apparent, as heterogeneous tissues can be further partitioned into increasing numbers of specific cell types defined by multiple features. Critically, new recombinase systems should work together with pre-existing systems without cross-reactivity in vivo. Here, we introduce novel site-specific recombinase systems based on ΦC31 bacteriophage recombinase for labeling multiple cell types simultaneously and a novel viral strategy for versatile and robust intersectional expression of any transgene. Together, our system will help researchers specifically target different cell types with multiple features in the same animal.
Collapse
Affiliation(s)
- Minju Jeong
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jun-Hyeok Choi
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hyeonseok Jang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Qingdi Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joann Lee
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Li Yao
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eun Ji Lee
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jiachen Fan
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marta Pratelli
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric H Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christen N Snyder
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Xiao-Yun Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sora Shin
- Center for Neurobiology Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA, USA; Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Aryn H Gittis
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Tsung-Chang Sung
- Transgenic Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nicholas C Spitzer
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Byung Kook Lim
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
69
|
Kang SJ, Kim JH, Kim DI, Roberts BZ, Han S. A pontomesencephalic PACAPergic pathway underlying panic-like behavioral and somatic symptoms in mice. Nat Neurosci 2024; 27:90-101. [PMID: 38177337 PMCID: PMC11195305 DOI: 10.1038/s41593-023-01504-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/19/2023] [Indexed: 01/06/2024]
Abstract
Panic disorder is characterized by uncontrollable fear accompanied by somatic symptoms that distinguish it from other anxiety disorders. Neural mechanisms underlying these unique symptoms are not completely understood. Here, we report that the pituitary adenylate cyclase-activating polypeptide (PACAP)-expressing neurons in the lateral parabrachial nucleus projecting to the dorsal raphe are crucial for panic-like behavioral and physiological alterations. These neurons are activated by panicogenic stimuli but inhibited in conditioned fear and anxiogenic conditions. Activating these neurons elicits strong defensive behaviors and rapid cardiorespiratory increase without creating aversive memory, whereas inhibiting them attenuates panic-associated symptoms. Chemogenetic or pharmacological inhibition of downstream PACAP receptor-expressing dorsal raphe neurons abolishes panic-like symptoms. The pontomesencephalic PACAPergic pathway is therefore a likely mediator of panicogenesis, and may be a promising therapeutic target for treating panic disorder.
Collapse
Affiliation(s)
- Sukjae J Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jong-Hyun Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
| | - Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Benjamin Z Roberts
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Neuroscience Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
- Neuroscience Graduate Program, University of California San Diego, La Jolla, CA, USA.
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea.
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
70
|
Koelle S, Mastrovito D, Whitesell JD, Hirokawa KE, Zeng H, Meila M, Harris JA, Mihalas S. Modeling the cell-type-specific mesoscale murine connectome with anterograde tracing experiments. Netw Neurosci 2023; 7:1497-1512. [PMID: 38144695 PMCID: PMC10745083 DOI: 10.1162/netn_a_00337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 09/10/2023] [Indexed: 12/26/2023] Open
Abstract
The Allen Mouse Brain Connectivity Atlas consists of anterograde tracing experiments targeting diverse structures and classes of projecting neurons. Beyond regional anterograde tracing done in C57BL/6 wild-type mice, a large fraction of experiments are performed using transgenic Cre-lines. This allows access to cell-class-specific whole-brain connectivity information, with class defined by the transgenic lines. However, even though the number of experiments is large, it does not come close to covering all existing cell classes in every area where they exist. Here, we study how much we can fill in these gaps and estimate the cell-class-specific connectivity function given the simplifying assumptions that nearby voxels have smoothly varying projections, but that these projection tensors can change sharply depending on the region and class of the projecting cells. This paper describes the conversion of Cre-line tracer experiments into class-specific connectivity matrices representing the connection strengths between source and target structures. We introduce and validate a novel statistical model for creation of connectivity matrices. We extend the Nadaraya-Watson kernel learning method that we previously used to fill in spatial gaps to also fill in gaps in cell-class connectivity information. To do this, we construct a "cell-class space" based on class-specific averaged regionalized projections and combine smoothing in 3D space as well as in this abstract space to share information between similar neuron classes. Using this method, we construct a set of connectivity matrices using multiple levels of resolution at which discontinuities in connectivity are assumed. We show that the connectivities obtained from this model display expected cell-type- and structure-specific connectivities. We also show that the wild-type connectivity matrix can be factored using a sparse set of factors, and analyze the informativeness of this latent variable model.
Collapse
Affiliation(s)
- Samson Koelle
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Statistics, University of Washington, Seattle, WA, USA
| | | | | | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Marina Meila
- Department of Statistics, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
71
|
He J, Zhu Y, Wu C, Wu J, Chen Y, Yuan M, Cheng Z, Zeng L, Ji X. Transcranial ultrasound neuromodulation facilitates isoflurane-induced general anesthesia recovery and improves cognition in mice. ULTRASONICS 2023; 135:107132. [PMID: 37604030 DOI: 10.1016/j.ultras.2023.107132] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/13/2023] [Accepted: 08/05/2023] [Indexed: 08/23/2023]
Abstract
Delayed arousal and cognitive dysfunction are common, especially in older patients after general anesthesia (GA). Elevating central nervous system serotonin (5-HT) levels can promote recovery from GA and increase synaptic plasticity to improve cognition. Ultrasound neuromodulation has become a noninvasive physical intervention therapy with high spatial resolution and penetration depth, which can modulate neuronal excitability to treat psychiatric and neurodegenerative diseases. This study aims to use ultrasound to noninvasively modulate the brain 5-HT levels of mice to promote recovery from GA and improve cognition in mice. The dorsal raphe nucleus (DRN) of mice during GA was stimulated by the 1.1 MHz ultrasound with a negative pressure of 356 kPa, and the liquid chromatography coupled tandem mass spectrometry (LC-MS/MS) method was used to measure the DRN 5-HT concentrations. The mice's recovery time from GA was assessed, and the cognition was evaluated through spontaneous alternation Y-maze and novel object recognition (NOR) tests. After ultrasound stimulation, the mice's DRN 5-HT levels were significantly increased (control: 554.0 ± 103.2 ng/g, anesthesia + US: 664.2 ± 84.1 ng/g, *p = 0.0389); the GA recovery time (return of the righting reflex (RORR) emergence latency time) of mice was significantly reduced (anesthesia: 331.6 ± 70 s, anesthesia + US: 223.2 ± 67.7 s, *p = 0.0215); the spontaneous rotation behavior score of mice was significantly increased (anesthesia: 59.46 ± 5.26 %, anesthesia + US: 68.55 ± 5.24 %; *p = 0.0126); the recognition index was significantly increased (anesthesia: 55.02 ± 6.23 %, anesthesia + US: 78.52 ± 12.21 %; ***p = 0.0009). This study indicates that ultrasound stimulation of DRN increases serotonin levels, accelerates recovery from anesthesia, and improves cognition, which could be an important strategy for treating delayed arousal, postoperative delirium, or even lasting cognitive dysfunction after GA.
Collapse
Affiliation(s)
- Jiaru He
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Yiyue Zhu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Canwen Wu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Junwei Wu
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Yan Chen
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Maodan Yuan
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhongwen Cheng
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Lvming Zeng
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China
| | - Xuanrong Ji
- State Key Laboratory of Precision Electronic Manufacturing Technology and Equipment, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
72
|
Suzuki M, Pennartz CMA, Aru J. How deep is the brain? The shallow brain hypothesis. Nat Rev Neurosci 2023; 24:778-791. [PMID: 37891398 DOI: 10.1038/s41583-023-00756-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/29/2023]
Abstract
Deep learning and predictive coding architectures commonly assume that inference in neural networks is hierarchical. However, largely neglected in deep learning and predictive coding architectures is the neurobiological evidence that all hierarchical cortical areas, higher or lower, project to and receive signals directly from subcortical areas. Given these neuroanatomical facts, today's dominance of cortico-centric, hierarchical architectures in deep learning and predictive coding networks is highly questionable; such architectures are likely to be missing essential computational principles the brain uses. In this Perspective, we present the shallow brain hypothesis: hierarchical cortical processing is integrated with a massively parallel process to which subcortical areas substantially contribute. This shallow architecture exploits the computational capacity of cortical microcircuits and thalamo-cortical loops that are not included in typical hierarchical deep learning and predictive coding networks. We argue that the shallow brain architecture provides several critical benefits over deep hierarchical structures and a more complete depiction of how mammalian brains achieve fast and flexible computational capabilities.
Collapse
Affiliation(s)
- Mototaka Suzuki
- Department of Cognitive and Systems Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| | - Cyriel M A Pennartz
- Department of Cognitive and Systems Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaan Aru
- Institute of Computer Science, University of Tartu, Tartu, Estonia.
| |
Collapse
|
73
|
Ma H, Gu L, Wang Y, Xu Q, Zhang Y, Shao W, Yu Q, Lian X, Liu L, Gu J, Ji N, Liu X, Nagayasu K, Zhang H. The States of Different 5-HT Receptors Located in the Dorsal Raphe Nucleus Are Crucial for Regulating the Awakening During General Anesthesia. Mol Neurobiol 2023; 60:6931-6948. [PMID: 37516665 DOI: 10.1007/s12035-023-03519-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2023]
Abstract
General anesthesia is widely used in various clinical practices due to its ability to cause loss of consciousness. However, the exact mechanism of anesthesia-induced unconsciousness remains unclear. It is generally thought that arousal-related brain nuclei are involved. 5-Hydroxytryptamine (5-HT) is closely associated with sleep arousal. Here, we explore the role of the 5-HT system in anesthetic awakening through pharmacological interventions and optogenetic techniques. Our data showed that exogenous administration of 5-hydroxytryptophan (5-HTP) and optogenetic activation of 5-HT neurons in the dorsal raphe nucleus (DR) could significantly shorten the emergence time of sevoflurane anesthesia in mice, suggesting that regulation of the 5-HT system using both endogenous and exogenous approaches could mediate delayed emergence. In addition, we first discovered that the different 5-HT receptors located in the DR, known as 5-HT autoreceptors, are essential for the regulation of general anesthetic awakening, with 5-HT1A and 5-HT2A/C receptors playing a regulatory role. These results can provide a reliable theoretical basis as well as potential targets for clinical intervention to prevent delayed emergence and some postoperative risks.
Collapse
Affiliation(s)
- HaiXiang Ma
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
- Medical College of Jining Medical University, Ningji, 272067, Shandong, China
| | - LeYuan Gu
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - YuLing Wang
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Qing Xu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yuanli Zhang
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - WeiHui Shao
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Qian Yu
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - XiTing Lian
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Lu Liu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - JiaXuan Gu
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Na Ji
- Department of Anesthesia, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - XiaoLing Liu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - HongHai Zhang
- Department of Anesthesiology, The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310006, China.
- Medical College of Jining Medical University, Ningji, 272067, Shandong, China.
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310006, China.
| |
Collapse
|
74
|
Bang D, Luo Y, Barbosa LS, Batten SR, Hadj-Amar B, Twomey T, Melville N, White JP, Torres A, Celaya X, Ramaiah P, McClure SM, Brewer GA, Bina RW, Lohrenz T, Casas B, Chiu PH, Vannucci M, Kishida KT, Witcher MR, Montague PR. Noradrenaline tracks emotional modulation of attention in human amygdala. Curr Biol 2023; 33:5003-5010.e6. [PMID: 37875110 PMCID: PMC10957395 DOI: 10.1016/j.cub.2023.09.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 09/01/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023]
Abstract
The noradrenaline (NA) system is one of the brain's major neuromodulatory systems; it originates in a small midbrain nucleus, the locus coeruleus (LC), and projects widely throughout the brain.1,2 The LC-NA system is believed to regulate arousal and attention3,4 and is a pharmacological target in multiple clinical conditions.5,6,7 Yet our understanding of its role in health and disease has been impeded by a lack of direct recordings in humans. Here, we address this problem by showing that electrochemical estimates of sub-second NA dynamics can be obtained using clinical depth electrodes implanted for epilepsy monitoring. We made these recordings in the amygdala, an evolutionarily ancient structure that supports emotional processing8,9 and receives dense LC-NA projections,10 while patients (n = 3) performed a visual affective oddball task. The task was designed to induce different cognitive states, with the oddball stimuli involving emotionally evocative images,11 which varied in terms of arousal (low versus high) and valence (negative versus positive). Consistent with theory, the NA estimates tracked the emotional modulation of attention, with a stronger oddball response in a high-arousal state. Parallel estimates of pupil dilation, a common behavioral proxy for LC-NA activity,12 supported a hypothesis that pupil-NA coupling changes with cognitive state,13,14 with the pupil and NA estimates being positively correlated for oddball stimuli in a high-arousal but not a low-arousal state. Our study provides proof of concept that neuromodulator monitoring is now possible using depth electrodes in standard clinical use.
Collapse
Affiliation(s)
- Dan Bang
- Center of Functionally Integrative Neuroscience, Aarhus University, 8000 Aarhus, Denmark; Wellcome Centre for Human Neuroimaging, University College London, London WC1N 3BG, UK; Department of Experimental Psychology, University of Oxford, Oxford OX2 6GG, UK; Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA.
| | - Yi Luo
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; Shanghai Key Laboratory of Mental Health and Psychological Crisis Intervention, East China Normal University, Shanghai 200050, China
| | - Leonardo S Barbosa
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; Department of Psychiatry, University of Wisconsin-Madison, Madison, WI 53719, USA
| | - Seth R Batten
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA
| | | | - Thomas Twomey
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA
| | - Natalie Melville
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA
| | - Jason P White
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA
| | - Alexis Torres
- Department of Psychology, Arizona State University, Tempe, AZ 85281, USA
| | - Xavier Celaya
- Department of Psychology, Arizona State University, Tempe, AZ 85281, USA
| | - Priya Ramaiah
- Department of Neurosurgery, Banner University Medical Center, Phoenix, AZ 85006, USA
| | - Samuel M McClure
- Department of Psychology, Arizona State University, Tempe, AZ 85281, USA
| | - Gene A Brewer
- Department of Psychology, Arizona State University, Tempe, AZ 85281, USA
| | - Robert W Bina
- Department of Neurosurgery, Banner University Medical Center, Phoenix, AZ 85006, USA
| | - Terry Lohrenz
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA
| | - Brooks Casas
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; Department of Psychology, Virginia Tech, Blacksburg, VA 24060, USA
| | - Pearl H Chiu
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; Department of Psychology, Virginia Tech, Blacksburg, VA 24060, USA
| | - Marina Vannucci
- Department of Statistics, Rice University, Houston, TX 77005, USA
| | - Kenneth T Kishida
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Mark R Witcher
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; Division of Neurosurgery, Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA
| | - P Read Montague
- Wellcome Centre for Human Neuroimaging, University College London, London WC1N 3BG, UK; Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA 24016, USA; Department of Physics, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
75
|
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
|
76
|
Huang J, Huang W, Yi J, Deng Y, Li R, Chen J, Shi J, Qiu Y, Wang T, Chen X, Zhang X, Xiang AP. Mesenchymal stromal cells alleviate depressive and anxiety-like behaviors via a lung vagal-to-brain axis in male mice. Nat Commun 2023; 14:7406. [PMID: 37973914 PMCID: PMC10654509 DOI: 10.1038/s41467-023-43150-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Major depressive disorder (MDD) is one of the most common and disabling mental disorders, and current strategies remain inadequate. Although mesenchymal stromal cells (MSCs) have shown beneficial effects in experimental models of depression, underlying mechanisms remain elusive. Here, using murine depression models, we demonstrated that MSCs could alleviate depressive and anxiety-like behaviors not due to a reduction in proinflammatory cytokines, but rather activation of dorsal raphe nucleus (DRN) 5-hydroxytryptamine (5-HT) neurons. Mechanistically, peripheral delivery of MSCs activated pulmonary innervating vagal sensory neurons, which projected to the nucleus tractus solitarius, inducing the release of 5-HT in DRN. Furthermore, MSC-secreted brain-derived neurotrophic factor activated lung sensory neurons through tropomyosin receptor kinase B (TrkB), and inhalation of a TrkB agonist also achieved significant therapeutic effects in male mice. This study reveals a role of peripheral MSCs in regulating central nervous system function and demonstrates a potential "lung vagal-to-brain axis" strategy for MDD.
Collapse
Affiliation(s)
- Jing Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Junzhe Yi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yiwen Deng
- Key Laboratory of Medical Transformation of Jiujiang, Jiujiang University, Jiujiang, Jiangxi, 332005, China
| | - Ruijie Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jieying Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Jiahao Shi
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Yuan Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
- Department of Histoembryology and Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
77
|
Chen T, Zhu SJ, Xu S, Wang YQ, Aji A, Zhang C, Wang H, Li FL, Chu YX. Resting-state fMRI reveals changes within the anxiety and social avoidance circuitry of the brain in mice with psoriasis-like skin lesions. Exp Dermatol 2023; 32:1900-1914. [PMID: 37622736 DOI: 10.1111/exd.14914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 08/26/2023]
Abstract
Psoriasis is an autoimmune skin disease that often co-occurs with psychological morbidities such as anxiety and depression, and psychosocial issues also lead psoriasis patients to avoid other people. However, the precise mechanism underlying the comorbidity of psoriasis and anxiety is unknown. Also, whether the social avoidance phenomenon seen in human patients also exists in psoriasis-like animal models remains unknown. In the present study, anxiety-like behaviours and social avoidance-like behaviours were observed in an imiquimod-induced psoriasis-like C57-BL6 mouse model along with typical psoriasis-like dermatitis and itch-like behaviours. The 11.7T resting-state functional magnetic resonance imaging showed differences in brain regions between the model and control group, and voxel-based morphometry showed that the grey matter volume changed in the basal forebrain region, anterior commissure intrabulbar and striatum in the psoriasis-like mice. Seed-based resting state functional connectivity analysis revealed connectivity changes in the amygdala, periaqueductal gray, raphe nuclei and lateral septum. We conclude that the imiquimod-induced psoriasis-like C57-BL6 mouse model is well suited for mechanistic studies and for performing preclinical therapeutic trials for treating anxiety and pathological social avoidance in psoriasis patients.
Collapse
Affiliation(s)
- Teng Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Sheng-Jie Zhu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuai Xu
- Department of Neurology, Institute of Science and Technology for Brain-Inspired Intelligence, Zhongshan Hospital, Human Phenome Institute, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai, China
| | - Yu-Quan Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Abudula Aji
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Chen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - He Wang
- Department of Neurology, Institute of Science and Technology for Brain-Inspired Intelligence, Zhongshan Hospital, Human Phenome Institute, Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai, China
| | - Fu-Lun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Institute of Acupuncture Research, Institutes of Integrative Medicine, Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| |
Collapse
|
78
|
Wu J, Yu J, Qu K, Yin J, Zhu C, Liu X. Serotonin syndrome caused by a CYP2C19-mediated interaction between low-dose escitalopram and clopidogrel: a case report. Front Psychiatry 2023; 14:1257984. [PMID: 37886119 PMCID: PMC10598681 DOI: 10.3389/fpsyt.2023.1257984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/14/2023] [Indexed: 10/28/2023] Open
Abstract
Background Serotonin syndrome has been recognized as a serious adverse reaction to antidepressants and is characterized by sudden or severe autonomic nerve dysfunction and neuromuscular symptoms. Without an accurate diagnosis and prompt treatment, serotonin syndrome progresses rapidly and can be life-threatening. It is usually related to the dose of 5-hydroxytryptamine drugs, and the dose is the basis for diagnosis. Therefore, serotonin syndrome induced by low-dose antidepressants rarely occurs, and clinicians are more likely to misdiagnose patients who take low-dose antidepressants with similar symptoms. Here, we present a case study of serotonin syndrome caused by a relatively low dose of escitalopram, which is not common in past references. Case summary The patient was a 74-year-old Asian woman with a 42-year history of schizophrenia. After 6 weeks of antidepressant treatment, our patient presented with characteristic myoclonus in the lower limbs and closed eyes with fluttering. Initially, she was misdiagnosed with neuroleptic malignant syndrome (NMS) due to antipsychotic medication and was treated accordingly, even with discontinuation of clozapine. However, her symptoms persisted, and then therapeutic drug monitoring was initiated with the involvement of a clinical pharmacist. Eventually, she was diagnosed with serotonin syndrome due to escitalopram levels reaching the warning level. Subsequently, the patient's treatment was modified, and her clinical outcome was satisfactory without any other serious adverse reactions. Gene detection was also performed, and a cytochrome P450 enzyme (CYP) 2C19-mediated interaction between low-dose escitalopram and clopidogrel seems to be a possible mechanism. Conclusion Data on this is extremely scarce, and to the best of our knowledge, serotonin syndrome caused by low-dose antidepressants has not yet been discussed to any great extent in the literature. Our case provides more clinical experience in the treatment of serotonin syndrome.
Collapse
Affiliation(s)
- Jianhong Wu
- The Affiliated Mental Health Center, Jiangnan University, Wuxi, China
- Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Jiao Yu
- The Affiliated Mental Health Center, Jiangnan University, Wuxi, China
- Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Kankan Qu
- The Affiliated Mental Health Center, Jiangnan University, Wuxi, China
- Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Jiajun Yin
- The Affiliated Mental Health Center, Jiangnan University, Wuxi, China
- Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Chunming Zhu
- The Affiliated Mental Health Center, Jiangnan University, Wuxi, China
- Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Xiaowei Liu
- The Affiliated Mental Health Center, Jiangnan University, Wuxi, China
- Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| |
Collapse
|
79
|
Mays KC, Haiman JH, Janušonis S. An experimental platform for stochastic analyses of single serotonergic fibers in the mouse brain. Front Neurosci 2023; 17:1241919. [PMID: 37869509 PMCID: PMC10587471 DOI: 10.3389/fnins.2023.1241919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
The self-organization of the serotonergic matrix, a massive axon meshwork in all vertebrate brains, is driven by the structural and dynamical properties of its constitutive elements. Each of these elements, a single serotonergic axon (fiber), has a unique trajectory and can be supported by a soma that executes one of the many available transcriptional programs. This "individuality" of serotonergic neurons necessitates the development of specialized methods for single-fiber analyses, both at the experimental and theoretical levels. We developed an integrated platform that facilitates experimental isolation of single serotonergic fibers in brain tissue, including regions with high fiber densities, and demonstrated the potential of their quantitative analyses based on stochastic modeling. Single fibers were visualized using two transgenic mouse models, one of which is the first implementation of the Brainbow toolbox in this system. The trajectories of serotonergic fibers were automatically traced in the three spatial dimensions with a novel algorithm, and their properties were captured with a single parameter associated with the directional von Mises-Fisher probability distribution. The system represents an end-to-end workflow that can be imported into various studies, including those investigating serotonergic dysfunction in brain disorders. It also supports new research directions inspired by single-fiber analyses in the serotonergic matrix, including supercomputing simulations and modeling in physics.
Collapse
Affiliation(s)
| | | | - Skirmantas Janušonis
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|
80
|
Wallace ML, Sabatini BL. Synaptic and circuit functions of multitransmitter neurons in the mammalian brain. Neuron 2023; 111:2969-2983. [PMID: 37463580 PMCID: PMC10592565 DOI: 10.1016/j.neuron.2023.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023]
Abstract
Neurons in the mammalian brain are not limited to releasing a single neurotransmitter but often release multiple neurotransmitters onto postsynaptic cells. Here, we review recent findings of multitransmitter neurons found throughout the mammalian central nervous system. We highlight recent technological innovations that have made the identification of new multitransmitter neurons and the study of their synaptic properties possible. We also focus on mechanisms and molecular constituents required for neurotransmitter corelease at the axon terminal and synaptic vesicle, as well as some possible functions of multitransmitter neurons in diverse brain circuits. We expect that these approaches will lead to new insights into the mechanism and function of multitransmitter neurons, their role in circuits, and their contribution to normal and pathological brain function.
Collapse
Affiliation(s)
- Michael L Wallace
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Bernardo L Sabatini
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
81
|
Li Z, Shang Z, Liu J, Zhen H, Zhu E, Zhong S, Sturgess RN, Zhou Y, Hu X, Zhao X, Wu Y, Li P, Lin R, Ren J. D-LMBmap: a fully automated deep-learning pipeline for whole-brain profiling of neural circuitry. Nat Methods 2023; 20:1593-1604. [PMID: 37770711 PMCID: PMC10555838 DOI: 10.1038/s41592-023-01998-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 08/02/2023] [Indexed: 09/30/2023]
Abstract
Recent proliferation and integration of tissue-clearing methods and light-sheet fluorescence microscopy has created new opportunities to achieve mesoscale three-dimensional whole-brain connectivity mapping with exceptionally high throughput. With the rapid generation of large, high-quality imaging datasets, downstream analysis is becoming the major technical bottleneck for mesoscale connectomics. Current computational solutions are labor intensive with limited applications because of the exhaustive manual annotation and heavily customized training. Meanwhile, whole-brain data analysis always requires combining multiple packages and secondary development by users. To address these challenges, we developed D-LMBmap, an end-to-end package providing an integrated workflow containing three modules based on deep-learning algorithms for whole-brain connectivity mapping: axon segmentation, brain region segmentation and whole-brain registration. D-LMBmap does not require manual annotation for axon segmentation and achieves quantitative analysis of whole-brain projectome in a single workflow with superior accuracy for multiple cell types in all of the modalities tested.
Collapse
Affiliation(s)
- Zhongyu Li
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Zengyi Shang
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Liu
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Haotian Zhen
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Entao Zhu
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Shilin Zhong
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Robyn N Sturgess
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Yitian Zhou
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Xuemeng Hu
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Xingyue Zhao
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Yi Wu
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Peiqi Li
- School of Software Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing, China
| | - Jing Ren
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
82
|
Wang J, Li W, Li Z, Xue Z, Zhang Y, Yuan Y, Shi Y, Shan S, Han W, Li F, Qiu Z. Taok1 haploinsufficiency leads to autistic-like behaviors in mice via the dorsal raphe nucleus. Cell Rep 2023; 42:113078. [PMID: 37656623 DOI: 10.1016/j.celrep.2023.113078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/11/2023] [Accepted: 08/16/2023] [Indexed: 09/03/2023] Open
Abstract
Strong evidence from human genetic studies associates the thousand and one amino acid kinase 1 (TAOK1) gene with autism spectrum disorder (ASD). In this work, we discovered a de novo frameshifting mutation in TAOK1 within a Chinese ASD cohort. We found that Taok1 haploinsufficiency induces autistic-like behaviors in mice. Importantly, we observed a significant enrichment of Taok1 in the dorsal raphe nucleus (DRN). The haploinsufficiency of Taok1 considerably restrained the activation of DRN neurons during social interactions, leading to the aberrant phosphorylation of numerous proteins. Intriguingly, the genetic deletion of Taok1 in VGlut3-positive neurons of DRN resulted in mice exhibiting autistic-like behaviors. Ultimately, reintroducing wild-type Taok1, but not its kinase-dead variant, into the DRN of adult mice effectively mitigated the autistic-like behaviors associated with Taok1 haploinsufficiency. This work suggests that Taok1, through its influence in the DRN, regulates social interaction behaviors, providing critical insights into the etiology of ASD.
Collapse
Affiliation(s)
- Jincheng Wang
- Songjiang Research Institute, Songjiang District Central Hospital, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Weike Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zimeng Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhenyu Xue
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuefang Zhang
- Songjiang Research Institute, Songjiang District Central Hospital, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiting Yuan
- Songjiang Research Institute, Songjiang District Central Hospital, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhan Shi
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Shifang Shan
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenjian Han
- Songjiang Research Institute, Songjiang District Central Hospital, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- MOE-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zilong Qiu
- Songjiang Research Institute, Songjiang District Central Hospital, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China; MOE-Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Clinic Neuroscience Center, Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
83
|
Mich JK, Sunil S, Johansen N, Martinez RA, Leytze M, Gore BB, Mahoney JT, Ben-Simon Y, Bishaw Y, Brouner K, Campos J, Canfield R, Casper T, Dee N, Egdorf T, Gary A, Gibson S, Goldy J, Groce EL, Hirschstein D, Loftus L, Lusk N, Malone J, Martin NX, Monet D, Omstead V, Opitz-Araya X, Oster A, Pom CA, Potekhina L, Reding M, Rimorin C, Ruiz A, Sedeño-Cortés AE, Shapovalova NV, Taormina M, Taskin N, Tieu M, Valera Cuevas NJ, Weed N, Way S, Yao Z, McMillen DA, Kunst M, McGraw M, Thyagarajan B, Waters J, Bakken TE, Yao S, Smith KA, Svoboda K, Podgorski K, Kojima Y, Horwitz GD, Zeng H, Daigle TL, Lein ES, Tasic B, Ting JT, Levi BP. Enhancer-AAVs allow genetic access to oligodendrocytes and diverse populations of astrocytes across species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558718. [PMID: 37790503 PMCID: PMC10542530 DOI: 10.1101/2023.09.20.558718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Proper brain function requires the assembly and function of diverse populations of neurons and glia. Single cell gene expression studies have mostly focused on characterization of neuronal cell diversity; however, recent studies have revealed substantial diversity of glial cells, particularly astrocytes. To better understand glial cell types and their roles in neurobiology, we built a new suite of adeno-associated viral (AAV)-based genetic tools to enable genetic access to astrocytes and oligodendrocytes. These oligodendrocyte and astrocyte enhancer-AAVs are highly specific (usually > 95% cell type specificity) with variable expression levels, and our astrocyte enhancer-AAVs show multiple distinct expression patterns reflecting the spatial distribution of astrocyte cell types. To provide the best glial-specific functional tools, several enhancer-AAVs were: optimized for higher expression levels, shown to be functional and specific in rat and macaque, shown to maintain specific activity in epilepsy where traditional promoters changed activity, and used to drive functional transgenes in astrocytes including Cre recombinase and acetylcholine-responsive sensor iAChSnFR. The astrocyte-specific iAChSnFR revealed a clear reward-dependent acetylcholine response in astrocytes of the nucleus accumbens during reinforcement learning. Together, this collection of glial enhancer-AAVs will enable characterization of astrocyte and oligodendrocyte populations and their roles across species, disease states, and behavioral epochs.
Collapse
|
84
|
Maddaloni G, Chang YJ, Senft RA, Dymecki SM. A brain circuit and neuronal mechanism for decoding and adapting to change in daylength. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557218. [PMID: 37745319 PMCID: PMC10515809 DOI: 10.1101/2023.09.11.557218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Changes in daylight amount (photoperiod) drive pronounced alterations in physiology and behaviour1,2. Adaptive responses to seasonal photoperiods are vital to all organisms - dysregulation is associated with disease, from affective disorders3 to metabolic syndromes4. Circadian rhythm circuitry has been implicated5,6 yet little is known about the precise neural and cellular substrates that underlie phase synchronization to photoperiod change. Here we present a previously unknown brain circuit and novel system of axon branch-specific and reversible neurotransmitter deployment that together prove critical for behavioural and sleep adaptation to photoperiod change. We found that the recently defined neuron type called mrEn1-Pet17 located in the mouse brainstem Median Raphe Nucleus (MRN) segregates serotonin versus VGLUT3 (here proxy for the neurotransmitter glutamate) to different axonal branches innervating specific brain regions involved in circadian rhythm and sleep/wake timing8,9. We found that whether measured during the light or dark phase of the day this branch-specific neurotransmitter deployment in mrEn1-Pet1 neurons was indistinguishable; however, it strikingly reorganizes on photoperiod change. Specifically, axonal boutons but not cell soma show a shift in neurochemical phenotype upon change away from equinox light/dark conditions that reverses upon return to equinox. When we genetically disabled the deployment of VGLUT3 in mrEn1-Pet1 neurons, we found that sleep/wake periods and voluntary activity failed to synchronize to the new photoperiod or was significantly delayed. Combining intersectional rabies virus tracing and projection-specific neuronal silencing in vivo, we delineated a Preoptic Area-to-mrEn1Pet1 connection responsible for decoding the photoperiodic inputs, driving the neurochemical shift and promoting behavioural synchronization. Our results reveal a previously unrecognized brain circuit along with a novel form of periodic, branch-specific neurotransmitter deployment that together regulate organismal adaptation to photoperiod changes.
Collapse
Affiliation(s)
- G Maddaloni
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| | - Y J Chang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| | - R A Senft
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| | - S M Dymecki
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115 MA, USA
| |
Collapse
|
85
|
Tao C, Zhang GW, Huang JJ, Li Z, Tao HW, Zhang LI. The medial preoptic area mediates depressive-like behaviors induced by ovarian hormone withdrawal through distinct GABAergic projections. Nat Neurosci 2023; 26:1529-1540. [PMID: 37524978 PMCID: PMC11037266 DOI: 10.1038/s41593-023-01397-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 06/26/2023] [Indexed: 08/02/2023]
Abstract
Fluctuations in reproductive hormone levels are associated with mood disruptions in women, such as in postpartum and perimenopausal depression. However, the neural circuit mechanisms remain unclear. Here we report that medial preoptic area (MPOA) GABAergic neurons mediate multifaceted depressive-like behaviors in female mice after ovarian hormone withdrawal (HW), which can be attributed to downregulation of activity in Esr1 (estrogen receptor-1)-expressing GABAergic neurons. Enhancing activity of these neurons ameliorates depressive-like behaviors in HW-treated mice, whereas reducing their activity results in expression of these behaviors. Two separate subpopulations mediate different symptoms: a subpopulation projecting to the ventral tegmental area (VTA) mediates anhedonia and another projecting to the periaqueductal gray mediates immobility. These projections enhance activity of dopaminergic neurons in the VTA and serotonergic neurons in the dorsal raphe, respectively, with increased release of dopamine and serotonin, possibly through disinhibition mechanisms. Thus, the MPOA is a hub that mediates depressive-like behaviors resulting from transitions in reproductive hormone levels.
Collapse
Affiliation(s)
- Can Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Junxiang J Huang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Graduate Programs in Biological and Biomedical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhong Li
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
86
|
Guan W, Li B. Diverse Roles of Serotonergic Projections to the Basolateral Amygdala. Neurosci Bull 2023; 39:1463-1465. [PMID: 37029325 PMCID: PMC10465427 DOI: 10.1007/s12264-023-01061-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/05/2023] [Indexed: 04/09/2023] Open
Affiliation(s)
- Wuqiang Guan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
| |
Collapse
|
87
|
Yip KYT, Gräff J. Tissue clearing applications in memory engram research. Front Behav Neurosci 2023; 17:1181818. [PMID: 37700912 PMCID: PMC10493294 DOI: 10.3389/fnbeh.2023.1181818] [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: 03/07/2023] [Accepted: 05/26/2023] [Indexed: 09/14/2023] Open
Abstract
A memory engram is thought to be the physical substrate of the memory trace within the brain, which is generally depicted as a neuronal ensemble activated by learning to fire together during encoding and retrieval. It has been postulated that engram cell ensembles are functionally interconnected across multiple brain regions to store a single memory as an "engram complex", but visualizing this engram complex across the whole brain has for long been hindered by technical limitations. With the recent development of tissue clearing techniques, advanced light-sheet microscopy, and automated 3D image analysis, it has now become possible to generate a brain-wide map of engram cells and thereby to visualize the "engram complex". In this review, we first provide a comprehensive summary of brain-wide engram mapping studies to date. We then compile a guide on implementing the optimal tissue clearing technique for engram tagging approaches, paying particular attention to visualize engram reactivation as a critical mnemonic property, for which whole-brain multiplexed immunostaining becomes a challenging prerequisite. Finally, we highlight the potential of tissue clearing to simultaneously shed light on both the circuit connectivity and molecular underpinnings of engram cells in a single snapshot. In doing so, novel brain regions and circuits can be identified for subsequent functional manipulation, thus providing an opportunity to robustly examine the "engram complex" underlying memory storage.
Collapse
Affiliation(s)
| | - Johannes Gräff
- Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
88
|
Wang XY, Jia WB, Xu X, Chen R, Wang LB, Su XJ, Xu PF, Liu XQ, Wen J, Song XY, Liu YY, Zhang Z, Liu XF, Zhang Y. A glutamatergic DRN-VTA pathway modulates neuropathic pain and comorbid anhedonia-like behavior in mice. Nat Commun 2023; 14:5124. [PMID: 37612268 PMCID: PMC10447530 DOI: 10.1038/s41467-023-40860-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Chronic pain causes both physical suffering and comorbid mental symptoms such as anhedonia. However, the neural circuits and molecular mechanisms underlying these maladaptive behaviors remain elusive. Here using a mouse model, we report a pathway from vesicular glutamate transporter 3 neurons in the dorsal raphe nucleus to dopamine neurons in the ventral tegmental area (VGluT3DRN→DAVTA) wherein population-level activity in response to innocuous mechanical stimuli and sucrose consumption is inhibited by chronic neuropathic pain. Mechanistically, neuropathic pain dampens VGluT3DRN → DAVTA glutamatergic transmission and DAVTA neural excitability. VGluT3DRN → DAVTA activation alleviates neuropathic pain and comorbid anhedonia-like behavior (CAB) by releasing glutamate, which subsequently promotes DA release in the nucleus accumbens medial shell (NAcMed) and produces analgesic and anti-anhedonia effects via D2 and D1 receptors, respectively. In addition, VGluT3DRN → DAVTA inhibition produces pain-like reflexive hypersensitivity and anhedonia-like behavior in intact mice. These findings reveal a crucial role for VGluT3DRN → DAVTA → D2/D1NAcMed pathway in establishing and modulating chronic pain and CAB.
Collapse
Affiliation(s)
- Xin-Yue Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Wen-Bin Jia
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Xiang Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Rui Chen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Liang-Biao Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Xiao-Jing Su
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Peng-Fei Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Xiao-Qing Liu
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, 230027, Hefei, China
| | - Jie Wen
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Xiao-Yuan Song
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, 230026, Hefei, China
| | - Yuan-Yuan Liu
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Zhi Zhang
- Hefei National Research Center for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, 230026, Hefei, China.
| | - Xin-Feng Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| | - Yan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| |
Collapse
|
89
|
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
|
90
|
Ielpo D, Guzzo SM, Porcheddu GF, Viscomi MT, Catale C, Reverte I, Cabib S, Cifani C, Antonucci G, Ventura R, Lo Iacono L, Marchetti C, Andolina D. GABAergic miR-34a regulates Dorsal Raphè inhibitory transmission in response to aversive, but not rewarding, stimuli. Proc Natl Acad Sci U S A 2023; 120:e2301730120. [PMID: 37523544 PMCID: PMC10410731 DOI: 10.1073/pnas.2301730120] [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: 01/31/2023] [Accepted: 06/26/2023] [Indexed: 08/02/2023] Open
Abstract
The brain employs distinct circuitries to encode positive and negative valence stimuli, and dysfunctions of these neuronal circuits have a key role in the etiopathogenesis of many psychiatric disorders. The Dorsal Raphè Nucleus (DRN) is involved in various behaviors and drives the emotional response to rewarding and aversive experiences. Whether specific subpopulations of neurons within the DRN encode these behaviors with different valence is still unknown. Notably, microRNA expression in the mammalian brain is characterized by tissue and neuronal specificity, suggesting that it might play a role in cell and circuit functionality. However, this specificity has not been fully exploited. Here, we demonstrate that microRNA-34a (miR-34a) is selectively expressed in a subpopulation of GABAergic neurons of the ventrolateral DRN. Moreover, we report that acute exposure to both aversive (restraint stress) and rewarding (chocolate) stimuli reduces GABA release in the DRN, an effect prevented by the inactivation of DRN miR-34a or its genetic deletion in GABAergic neurons in aversive but not rewarding conditions. Finally, miR-34a inhibition selectively reduced passive coping with severe stressors. These data support a role of miR-34a in regulating GABAergic neurotransmitter activity and behavior in a context-dependent manner and suggest that microRNAs could represent a functional signature of specific neuronal subpopulations with valence-specific activity in the brain.
Collapse
Affiliation(s)
- Donald Ielpo
- Department of Psychology, Sapienza University, Rome00184, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
| | - Serafina M. Guzzo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino62032, Italy
| | - Giovanni F. Porcheddu
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome00161, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health Section of Histology and Embryology, Università Cattolica del Sacro Cuore, Rome00168, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, Istituto di Ricovero e Cura a Carattere Scientifico, Rome00168, Italy
| | - Clarissa Catale
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
| | - Ingrid Reverte
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome00185, Italy
| | - Simona Cabib
- Department of Psychology, Sapienza University, Rome00184, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino62032, Italy
| | - Gabriella Antonucci
- Department of Psychology, Sapienza University, Rome00184, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
| | - Rossella Ventura
- Department of Psychology, Sapienza University, Rome00184, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
- San Raffaele Istituto di Ricovero e Cura a Carattere Scientifico, Rome00166, Italy
| | - Luisa Lo Iacono
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
| | - Cristina Marchetti
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome00161, Italy
- Institute of Molecular Biology and Pathology, National Research Council, Rome00185, Italy
| | - Diego Andolina
- Department of Psychology, Sapienza University, Rome00184, Italy
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma00143, Italy
| |
Collapse
|
91
|
Conde K, Fang S, Xu Y. Unraveling the serotonin saga: from discovery to weight regulation and beyond - a comprehensive scientific review. Cell Biosci 2023; 13:143. [PMID: 37550777 PMCID: PMC10408233 DOI: 10.1186/s13578-023-01091-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023] Open
Abstract
The prevalence of obesity is rapidly increasing worldwide, while the development of effective obesity therapies lags behind. Although new therapeutic targets to alleviate obesity are identified every day, and drug efficacy is improving, adverse side effects and increased health risks remain serious issues facing the weight-loss industry. Serotonin, also known as 5-HT, has been extensively studied in relation to appetite reduction and weight loss. As a result, dozens of upstream and downstream neural targets of 5-HT have been identified, revealing a multitude of neural circuits involved in mediating the anorexigenic effect of 5-HT. Despite the rise and fall of several 5-HT therapeutics in recent decades, the future of 5-HT as a therapeutic target for weight-loss therapy looks promising. This review focuses on the history of serotonin, the state of current central serotonin research, previous serotonergic therapies, and the future of serotonin for treating individuals with obesity.
Collapse
Affiliation(s)
- Kristine Conde
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, USA.
| | - Shuzheng Fang
- College of Art and Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA.
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
92
|
Deng H, Wu Y, Gao P, Kong D, Pan C, Xu S, Tang D, Jiao Y, Wen D, Yu W. Preoperative Pain Facilitates Postoperative Cognitive Dysfunction via Periaqueductal Gray Matter-Dorsal Raphe Circuit. Neuroscience 2023; 524:209-219. [PMID: 36958595 DOI: 10.1016/j.neuroscience.2023.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a medically induced, rapidly occurring postoperative disease, which is hard to recover and seriously threatens the quality of life, especially for elderly patients, so it is important to identify the risk factors for POCD and apply early intervention to prevent POCD. As we have known, pain can impair cognition, and many surgery patients experience different preoperative pain, but it is still unknown whether these patients are vulnerable for POCD. Here we found that chronic pain (7 days, but not 1 day acute pain) induced by Complete Freund's Adjuvant (CFA) injected in the hind paw of rats could easily induce spatial cognition and memory impairment after being exposed to sevoflurane anesthesia. Next, for the mechanisms, we focused on the Periaqueductal Gray Matter (PAG), a well-known pivotal nucleus in pain process. It was detected the existence of neural projection from ventrolateral PAG (vlPAG) to adjacent nucleus Dorsal Raphe (DR), the origin of serotonergic projection for the whole cerebrum, through virus tracing and patch clamp recordings. The Immunofluorescence staining and western blot results showed that Tryptophan Hydroxylase 2 (TPH2) for serotonin synthesis in the DR was increased significantly in the rats treated with CFA for 7 days and sevoflurane for 3 hours, while chemo-genetic inhibition of the vlPAG-DR projection induced obvious spatial learning and memory impairment. Our study suggests that preoperative chronic pain may facilitate cognitive function impairment after receiving anesthesia through the PAG-DR neural circuit, and preventative analgesia should be a considerable measure to reduce the incidence of POCD.
Collapse
Affiliation(s)
- Haoyue Deng
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Yi Wu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Po Gao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Dexu Kong
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Chao Pan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Saihong Xu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China.
| | - Daxiang Wen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China.
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, P.O. Box 200127, No. 160 Pujian Road, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China.
| |
Collapse
|
93
|
Jang S, Park I, Choi M, Kim J, Yeo S, Huh SO, Choi JW, Moon C, Choe HK, Choe Y, Kim K. Impact of the circadian nuclear receptor REV-ERBα in dorsal raphe 5-HT neurons on social interaction behavior, especially social preference. Exp Mol Med 2023; 55:1806-1819. [PMID: 37537215 PMCID: PMC10474013 DOI: 10.1038/s12276-023-01052-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 08/05/2023] Open
Abstract
Social interaction among conspecifics is essential for maintaining adaptive, cooperative, and social behaviors, along with survival among mammals. The 5-hydroxytryptamine (5-HT) neuronal system is an important neurotransmitter system for regulating social behaviors; however, the circadian role of 5-HT in social interaction behaviors is unclear. To investigate whether the circadian nuclear receptor REV-ERBα, a transcriptional repressor of the rate-limiting enzyme tryptophan hydroxylase 2 (Tph2) gene in 5-HT biosynthesis, may affect social interaction behaviors, we generated a conditional knockout (cKO) mouse by targeting Rev-Erbα in dorsal raphe (DR) 5-HT neurons (5-HTDR-specific REV-ERBα cKO) using the CRISPR/Cas9 gene editing system and assayed social behaviors, including social preference and social recognition, with a three-chamber social interaction test at two circadian time (CT) points, i.e., at dawn (CT00) and dusk (CT12). The genetic ablation of Rev-Erbα in DR 5-HTergic neurons caused impaired social interaction behaviors, particularly social preference but not social recognition, with no difference between the two CT points. This deficit of social preference induced by Rev-Erbα in 5-HTDR-specific mice is functionally associated with real-time elevated neuron activity and 5-HT levels at dusk, as determined by fiber-photometry imaging sensors. Moreover, optogenetic inhibition of DR to nucleus accumbens (NAc) 5-HTergic circuit restored the impairment of social preference in 5-HTDR-specific REV-ERBα cKO mice. These results suggest the significance of the circadian regulation of 5-HT levels by REV-ERBα in regulating social interaction behaviors.
Collapse
Affiliation(s)
- Sangwon Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Inah Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Mijung Choi
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Jihoon Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Seungeun Yeo
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, Hallym University, Chuncheon, 24252, Republic of Korea
| | - Ji-Woong Choi
- Department of Electrical Engineering and Computer Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Cheil Moon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Han Kyoung Choe
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Kyungjin Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| |
Collapse
|
94
|
Liu X, Li S, Yu Y, Hu J, Xu Y. Changes in Plasma TPH2, GDNF, Trk-b, BDNF, and proBDNF in People Who Died by Suicide. Brain Sci 2023; 13:1096. [PMID: 37509026 PMCID: PMC10377529 DOI: 10.3390/brainsci13071096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Recent studies have shown that neuropeptides and neurotrophic factors may be involved in the pathophysiological mechanisms of suicide. However, the current research on this aspect is still insufficient. Our study aimed to explore the biological patterns of suicide deaths, including levels of BDNF, proBDNF, BDNF/proBDNF, Trk-b, GDNF, and TPH2. The researchers selected 25 normal control patients matched by age with 30 suicide deaths. We used enzyme-linked immunosorbent assays to detect the levels of BDNF, proBDNF, BDNF/proBDNF, Trk-b, GDNF, and TPH2 in the plasma of suicide and control subjects. proBDNF, BDNF/proBDNF, Trk-b, GDNF, and TPH2 levels are shown as the median (25th-75th percentile). BDNF levels are shown as the mean (standard error of the mean). (1) The levels of plasma TPH2 and proBDNF in people who died by suicide were significantly higher than those in the control group. (2) The plasma levels of GDNF and BDNF/proBDNF in the suicide group were obviously lower than those in the control group. (3) There was no significant difference in plasma BDNF or Trk-b concentrations between the suicide group and the control group.Plasma TPH2, GDNF, and proBDNF levels are related to suicide. Plasma neurotrophic factor markers may predict suicide risk.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shangda Li
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yueran Yu
- Department of Infectious Diseases, Tongde Hospital of Zhejiang Province, Hangzhou 310003, China
| | - Jianbo Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
- Brain Research Institute of Zijingang Campus of Zhejiang University, Hangzhou 310003, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou 310003, China
| | - Yi Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
- Brain Research Institute of Zijingang Campus of Zhejiang University, Hangzhou 310003, China
- Zhejiang Engineering Center for Mathematical Mental Health, Hangzhou 310003, China
| |
Collapse
|
95
|
Dag U, Nwabudike I, Kang D, Gomes MA, Kim J, Atanas AA, Bueno E, Estrem C, Pugliese S, Wang Z, Towlson E, Flavell SW. Dissecting the functional organization of the C. elegans serotonergic system at whole-brain scale. Cell 2023; 186:2574-2592.e20. [PMID: 37192620 PMCID: PMC10484565 DOI: 10.1016/j.cell.2023.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/07/2023] [Accepted: 04/17/2023] [Indexed: 05/18/2023]
Abstract
Serotonin influences many aspects of animal behavior. But how serotonin acts on its diverse receptors across the brain to modulate global activity and behavior is unknown. Here, we examine how serotonin release in C. elegans alters brain-wide activity to induce foraging behaviors, like slow locomotion and increased feeding. Comprehensive genetic analyses identify three core serotonin receptors (MOD-1, SER-4, and LGC-50) that induce slow locomotion upon serotonin release and others (SER-1, SER-5, and SER-7) that interact with them to modulate this behavior. SER-4 induces behavioral responses to sudden increases in serotonin release, whereas MOD-1 induces responses to persistent release. Whole-brain imaging reveals widespread serotonin-associated brain dynamics, spanning many behavioral networks. We map all sites of serotonin receptor expression in the connectome, which, together with synaptic connectivity, helps predict which neurons show serotonin-associated activity. These results reveal how serotonin acts at defined sites across a connectome to modulate brain-wide activity and behavior.
Collapse
Affiliation(s)
- Ugur Dag
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ijeoma Nwabudike
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Di Kang
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew A Gomes
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jungsoo Kim
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam A Atanas
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA; Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric Bueno
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cassi Estrem
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah Pugliese
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ziyu Wang
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emma Towlson
- Department of Computer Science, Department of Physics and Astronomy, Hotchkiss Brain Institute, Alberta Children's Research Hospital, University of Calgary, Calgary, AB, Canada
| | - Steven W Flavell
- Picower Institute for Learning & Memory, Department of Brain & Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
96
|
Huang Y, Zhang Y, Hodges S, Li H, Yan Z, Liu X, Hou X, Chen W, Chai-Zhang T, Kong J, Liu B. The modulation effects of repeated transcutaneous auricular vagus nerve stimulation on the functional connectivity of key brainstem regions along the vagus nerve pathway in migraine patients. Front Mol Neurosci 2023; 16:1160006. [PMID: 37333617 PMCID: PMC10275573 DOI: 10.3389/fnmol.2023.1160006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023] Open
Abstract
Background Previous studies have shown a significant response to acute transcutaneous vagus nerve stimulation (taVNS) in regions of the vagus nerve pathway, including the nucleus tractus solitarius (NTS), raphe nucleus (RN) and locus coeruleus (LC) in both healthy human participants and migraine patients. This study aims to investigate the modulation effect of repeated taVNS on these brainstem regions by applying seed-based resting-state functional connectivity (rsFC) analysis. Methods 70 patients with migraine were recruited and randomized to receive real or sham taVNS treatments for 4 weeks. fMRI data were collected from each participant before and after 4 weeks of treatment. The rsFC analyses were performed using NTS, RN and LC as the seeds. Results 59 patients (real group: n = 33; sham group: n = 29) completed two fMRI scan sessions. Compared to sham taVNS, real taVNS was associated with a significant reduction in the number of migraine attack days (p = 0.024) and headache pain intensity (p = 0.008). The rsFC analysis showed repeated taVNS modulated the functional connectivity between the brain stem regions of the vagus nerve pathway and brain regions associated with the limbic system (bilateral hippocampus), pain processing and modulation (bilateral postcentral gyrus, thalamus, and mPFC), and basal ganglia (putamen/caudate). In addition, the rsFC change between the RN and putamen was significantly associated with the reduction in the number of migraine days. Conclusion Our findings suggest that taVNS can significantly modulate the vagus nerve central pathway, which may contribute to the potential treatment effects of taVNS for migraine.Clinical Trial Registration: http://www.chictr.org.cn/hvshowproject.aspx?id=11101, identifier ChiCTR-INR-17010559.
Collapse
Affiliation(s)
- Yiting Huang
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Yue Zhang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sierra Hodges
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Hui Li
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhaoxian Yan
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xian Liu
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyan Hou
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weicui Chen
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Thalia Chai-Zhang
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Jian Kong
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Bo Liu
- Department of Radiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
97
|
Wang R, Khan KM, Balasubramanian N, James T, Pushpavathi SG, Kim D, Pierson S, Wu Q, Niciu MJ, Hefti MM, Marcinkiewcz CA. Alcohol inhibits sociability via serotonin inputs to the nucleus accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542761. [PMID: 37398335 PMCID: PMC10312479 DOI: 10.1101/2023.05.29.542761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Social interaction is a core component of motivational behavior that is perturbed across multiple neuropsychiatric disorders, including alcohol use disorder (AUD). Positive social bonds are neuroprotective and enhance recovery from stress, so reduced social interaction in AUD may delay recovery and lead to alcohol relapse. We report that chronic intermittent ethanol (CIE) induces social avoidance in a sex-dependent manner and is associated with hyperactivity of serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN). While 5-HT DRN neurons are generally thought to enhance social behavior, recent evidence suggests that specific 5-HT pathways can be aversive. Using chemogenetic iDISCO, the nucleus accumbens (NAcc) was identified as one of 5 regions that were activated by 5-HT DRN stimulation. We then employed an array of molecular genetic tools in transgenic mice to show that 5-HT DRN inputs to NAcc dynorphin neurons drive social avoidance in male mice after CIE by activating 5-HT 2C receptors. NAcc dynorphin neurons also inhibit dopamine release during social interaction, reducing the motivational drive to engage with social partners. This study reveals that excessive serotonergic drive after chronic alcohol can promote social aversion by inhibiting accumbal dopamine release. Drugs that boost brain serotonin levels may be contraindicated for individuals with AUD.
Collapse
|
98
|
Oubraim S, Shen RY, Haj-Dahmane S. Oxytocin excites dorsal raphe serotonin neurons and bidirectionally gates their glutamate synapses. iScience 2023; 26:106707. [PMID: 37250336 PMCID: PMC10214716 DOI: 10.1016/j.isci.2023.106707] [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: 02/20/2023] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Oxytocin (OXT) modulates wide spectrum of social and emotional behaviors via modulation of numerous neurotransmitter systems, including serotonin (5-HT). However, how OXT controls the function of dorsal raphe nucleus (DRN) 5-HT neurons remains unknown. Here, we reveal that OXT excites and alters the firing pattern of 5-HT neurons via activation of postsynaptic OXT receptors (OXTRs). In addition, OXT induces cell-type-specific depression and potentiation of DRN glutamate synapses by two retrograde lipid messengers, 2-arachidonoylglycerol (2-AG) and arachidonic acid (AA), respectively. Neuronal mapping demonstrates that OXT preferentially potentiates glutamate synapses of 5-HT neurons projecting to medial prefrontal cortex (mPFC) and depresses glutamatergic inputs to 5-HT neurons projecting to lateral habenula (LHb) and central amygdala (CeA). Thus, by engaging distinct retrograde lipid messengers, OXT exerts a target-specific gating of glutamate synapses on the DRN. As such, our data uncovers the neuronal mechanisms by which OXT modulates the function of DRN 5-HT neurons.
Collapse
Affiliation(s)
- Saida Oubraim
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Roh-Yu Shen
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
- University at Buffalo Neuroscience Program, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, State University of New York, 1021 Main Street, Buffalo, NY 14203, USA
| |
Collapse
|
99
|
Janušonis S, Haiman JH, Metzler R, Vojta T. Predicting the distribution of serotonergic axons: a supercomputing simulation of reflected fractional Brownian motion in a 3D-mouse brain model. Front Comput Neurosci 2023; 17:1189853. [PMID: 37265780 PMCID: PMC10231035 DOI: 10.3389/fncom.2023.1189853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
Abstract
The self-organization of the brain matrix of serotonergic axons (fibers) remains an unsolved problem in neuroscience. The regional densities of this matrix have major implications for neuroplasticity, tissue regeneration, and the understanding of mental disorders, but the trajectories of its fibers are strongly stochastic and require novel conceptual and analytical approaches. In a major extension to our previous studies, we used a supercomputing simulation to model around one thousand serotonergic fibers as paths of superdiffusive fractional Brownian motion (FBM), a continuous-time stochastic process. The fibers produced long walks in a complex, three-dimensional shape based on the mouse brain and reflected at the outer (pial) and inner (ventricular) boundaries. The resultant regional densities were compared to the actual fiber densities in the corresponding neuroanatomically-defined regions. The relative densities showed strong qualitative similarities in the forebrain and midbrain, demonstrating the predictive potential of stochastic modeling in this system. The current simulation does not respect tissue heterogeneities but can be further improved with novel models of multifractional FBM. The study demonstrates that serotonergic fiber densities can be strongly influenced by the geometry of the brain, with implications for brain development, plasticity, and evolution.
Collapse
Affiliation(s)
- Skirmantas Janušonis
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Justin H. Haiman
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Ralf Metzler
- Institute of Physics and Astronomy, University of Potsdam, Potsdam, Germany
- Asia Pacific Center for Theoretical Physics, Pohang, South Korea
| | - Thomas Vojta
- Department of Physics, Missouri University of Science and Technology, Rolla, MO, United States
| |
Collapse
|
100
|
Li HQ, Jiang W, Ling L, Gupta V, Chen C, Pratelli M, Godavarthi SK, Spitzer NC. Generalized fear following acute stress is caused by change in co-transmitter identity of serotonergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540268. [PMID: 37214936 PMCID: PMC10197626 DOI: 10.1101/2023.05.10.540268] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Overgeneralization of fear to harmless situations is a core feature of anxiety disorders resulting from acute stress, yet the mechanisms by which fear becomes generalized are poorly understood. Here we show that generalized fear in mice in response to footshock results from a transmitter switch from glutamate to GABA in serotonergic neurons of the lateral wings of the dorsal raphe. We observe a similar change in transmitter identity in the postmortem brains of PTSD patients. Overriding the transmitter switch in mice using viral tools prevents the acquisition of generalized fear. Corticosterone release and activation of glucocorticoid receptors trigger the switch, and prompt antidepressant treatment blocks the co-transmitter switch and generalized fear. Our results provide new understanding of the plasticity involved in fear generalization.
Collapse
Affiliation(s)
- Hui-Quan Li
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Wuji Jiang
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Lily Ling
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Vaidehi Gupta
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Cong Chen
- Department of Cellular and Molecular Medicine, University of California, San Diego
| | - Marta Pratelli
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Swetha K Godavarthi
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| | - Nicholas C Spitzer
- Neurobiology Department and Kavli Institute for Brain and Mind, University of California, San Diego
| |
Collapse
|