1
|
Chen Y, Wang P, Li Z. Exploring genetic and epigenetic markers for predicting or monitoring response to cognitive-behavioral therapy in obsessive-compulsive disorder: A systematic review. Neurosci Biobehav Rev 2025; 174:106192. [PMID: 40324706 DOI: 10.1016/j.neubiorev.2025.106192] [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: 05/30/2024] [Revised: 04/17/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Growing evidence has identified potential biomarkers of cognitive-behavioural therapy (CBT) efficacy in obsessive-compulsive disorder (OCD). Genetic and epigenetic mechanisms (e.g., polymorphisms, DNA methylation) contribute to OCD pathogenesis and CBT response variability, establishing them as a key research focus. To evaluate their associations with CBT outcomes in OCD, we conducted a systematic review of PubMed, Web of Science, CNKI, and Cochrane Library (from inception to January 2025), identifying eight studies that met rigorous inclusion criteria. The identified predictors included: (1) Genetic polymorphisms (BDNF); (2) Epigenetic modifications (DNA methylation of MAOA, SLC6A4, OXTR, PIWIL1, MIR886, PLEKHA1, KCNQ1, TRPM8, HEBP1, HTR7P1, MAPK8IP3, ENAH, RABGGTB (SNORD45C), MYEF2, GALK2, CEP192, and UIMC1). These markers may influence neural plasticity, neurotransmitter regulation, and related processes, providing molecular substrates for the observed treatment effects. Converging evidence suggests that distinct neurocognitive mechanisms may mediate CBT efficacy in OCD, particularly fear extinction learning and goal-directed behaviors (GDBs), which we analyze mechanistically. Future studies should integrate polygenic risk scores (PRS) with functional neuroimaging to dissect individual variability in CBT response, mainly through cortico-striato-thalamo-cortical (CSTC) circuit profiling. To our knowledge, this is the first systematic review synthesizing genetic and epigenetic predictors of CBT response in OCD; these findings provide compelling evidence for biomarkers for CBT personalization in OCD, advancing a novel precision psychiatry framework.
Collapse
Affiliation(s)
- Yu Chen
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Pengchong Wang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Zhanjiang Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Guo WT, Li WX, Liu YC, Zhao YB, Xu L, Zhou QX. Time-Dependent Transcriptional Dynamics of Contextual Fear Memory Retrieval Reveals the Function of Dipeptidyl Peptidase 9 in Reconsolidation. Neurosci Bull 2025; 41:16-32. [PMID: 39621238 PMCID: PMC11748732 DOI: 10.1007/s12264-024-01324-w] [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: 12/31/2023] [Accepted: 08/26/2024] [Indexed: 01/19/2025] Open
Abstract
Numerous studies on the formation and consolidation of memory have shown that memory processes are characterized by phase-dependent and dynamic regulation. Memory retrieval, as the only representation of memory content and an active form of memory processing that induces memory reconsolidation, has attracted increasing attention in recent years. Although the molecular mechanisms specific to memory retrieval-induced reconsolidation have been gradually revealed, an understanding of the time-dependent regulatory mechanisms of this process is still lacking. In this study, we applied a transcriptome analysis of memory retrieval at different time points in the recent memory stage. Differential expression analysis and Short Time-series Expression Miner (STEM) depicting temporal gene expression patterns indicated that most differential gene expression occurred at 48 h, and the STEM cluster showing the greatest transcriptional upregulation at 48 h demonstrated the most significant difference. We then screened the differentially-expressed genes associated with that met the expression patterns of those cluster-identified genes that have been reported to be involved in learning and memory processes in addition to dipeptidyl peptidase 9 (DPP9). Further quantitative polymerase chain reaction verification and pharmacological intervention suggested that DPP9 is involved in 48-h fear memory retrieval and viral vector-mediated overexpression of DPP9 countered the 48-h retrieval-induced attenuation of fear memory. Taken together, our findings suggest that temporal gene expression patterns are induced by recent memory retrieval and provide hitherto undocumented evidence of the role of DPP9 in the retrieval-induced reconsolidation of fear memory.
Collapse
Affiliation(s)
- Wen-Ting Guo
- Key Laboratory of Animal Models and Human Disease Mechanisms, Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Wen-Xing Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Yu-Chen Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms, Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Ya-Bo Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms, Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China.
- CAS Centre for Excellence in Brain Science and Intelligent Technology, Shanghai, 200031, China.
| | - Qi-Xin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms, Laboratory of Learning and Memory, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, 650223, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China.
| |
Collapse
|
3
|
Gattuso JJ, Wilson C, Hannan AJ, Renoir T. Psilocybin as a lead candidate molecule in preclinical therapeutic studies of psychiatric disorders: A systematic review. J Neurochem 2024; 168:1687-1720. [PMID: 38019032 DOI: 10.1111/jnc.16017] [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: 09/03/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023]
Abstract
Psilocybin is the main psychoactive compound found in hallucinogenic/magic mushrooms and can bind to both serotonergic and tropomyosin receptor kinase b (TrkB) receptors. Psilocybin has begun to show efficacy for a range of neuropsychiatric conditions, including treatment-resistant depression and anxiety disorders; however, neurobiological mechanisms are still being elucidated. Clinical research has found that psilocybin can alter functional connectivity patterns in human brains, which is often associated with therapeutic outcomes. However, preclinical research affords the opportunity to assess the potential cellular mechanisms by which psilocybin may exert its therapeutic effects. Preclinical rodent models can also facilitate a more tightly controlled experimental context and minimise placebo effects. Furthermore, where there is a rationale, preclinical researchers can investigate psilocybin administration in neuropsychiatric conditions that have not yet been researched clinically. As a result, we have systematically reviewed the knowledge base, identifying 82 preclinical studies which were screened based on specific criteria. This resulted in the exclusion of 44 articles, with 34 articles being included in the main review and another 2 articles included as Supporting Information materials. We found that psilocybin shows promise as a lead candidate molecule for treating a variety of neuropsychiatric conditions, albeit showing the most efficacy for depression. We discuss the experimental findings, and identify possible mechanisms whereby psilocybin could invoke therapeutic changes. Furthermore, we critically evaluate the between-study heterogeneity and possible future research avenues. Our review suggests that preclinical rodent models can provide valid and translatable tools for researching novel psilocybin-induced molecular and cellular mechanisms, and therapeutic outcomes.
Collapse
Affiliation(s)
- James J Gattuso
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Carey Wilson
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Cheng J, Chen L, Zheng YN, Liu J, Zhang L, Zhang XM, Huang L, Yuan QL. Disfunction of dorsal raphe nucleus-hippocampus serotonergic-HTR3 transmission results in anxiety phenotype of Neuroplastin 65-deficient mice. Acta Pharmacol Sin 2024; 45:1393-1405. [PMID: 38528118 PMCID: PMC11192762 DOI: 10.1038/s41401-024-01252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/26/2024] [Indexed: 03/27/2024]
Abstract
Anxiety disorders are the most common psychiatric condition, but the etiology of anxiety disorders remains largely unclear. Our previous studies have shown that neuroplastin 65 deficiency (NP65-/-) mice exhibit abnormal social and mental behaviors and decreased expression of tryptophan hydroxylase 2 (TPH2) protein. However, whether a causal relationship between TPH2 reduction and anxiety disorders exists needs to be determined. In present study, we found that replenishment of TPH2 in dorsal raphe nucleus (DRN) enhanced 5-HT level in the hippocampus and alleviated anxiety-like behaviors. In addition, injection of AAV-NP65 in DRN significantly increased TPH2 expression in DRN and hippocampus, and reduced anxiety-like behaviors. Acute administration of exogenous 5-HT or HTR3 agonist SR57227A in hippocampus mitigated anxiety-like behaviors in NP65-/- mice. Moreover, replenishment of TPH2 in DRN partly repaired the impairment of long-term potentiation (LTP) maintenance in hippocampus of NP65-/- mice. Finally, we found that loss of NP65 lowered transcription factors Lmx1b expression in postnatal stage and replenishment of NP65 in DRN reversed the decrease in Lmx1b expression of NP65-/- mice. Together, our findings reveal that NP65 deficiency induces anxiety phenotype by downregulating DRN-hippocampus serotonergic-HTR3 transmission. These studies provide a novel and insightful view about NP65 function, suggesting an attractive potential target for treatment of anxiety disorders.
Collapse
Affiliation(s)
- Jie Cheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ling Chen
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ya-Ni Zheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Juan Liu
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Lei Zhang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xiao-Ming Zhang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Liang Huang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiong-Lan Yuan
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
- Department of Human Anatomy, Histology and Embryology, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
5
|
Li Y, Wan LP, Song NN, Ding YQ, Zhao S, Niu J, Mao B, Sheng N, Ma P. RNF220-mediated K63-linked polyubiquitination stabilizes Olig proteins during oligodendroglial development and myelination. SCIENCE ADVANCES 2024; 10:eadk3931. [PMID: 38324685 PMCID: PMC10849602 DOI: 10.1126/sciadv.adk3931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
Maldevelopment of oligodendroglia underlies neural developmental disorders such as leukodystrophy. Precise regulation of the activity of specific transcription factors (TFs) by various posttranslational modifications (PTMs) is required to ensure proper oligodendroglial development and myelination. However, the role of ubiquitination of these TFs during oligodendroglial development is yet unexplored. Here, we find that RNF220, a known leukodystrophy-related E3 ubiquitin ligase, is required for oligodendroglial development. RNF220 depletion in oligodendrocyte lineage cells impedes oligodendrocyte progenitor cell proliferation, differentiation, and (re)myelination, which consequently leads to learning and memory defects. Mechanistically, RNF220 targets Olig1/2 for K63-linked polyubiquitination and stabilization during oligodendroglial development. Furthermore, in a knock-in mouse model of leukodystrophy-related RNF220R365Q mutation, the ubiquitination and stabilization of Olig proteins are deregulated in oligodendroglial cells. This results in pathomimetic oligodendroglial developmental defects, impaired myelination, and abnormal behaviors. Together, our evidence provides an alternative insight into PTMs of oligodendroglial TFs and how this essential process may be implicated in the etiology of leukodystrophy.
Collapse
Affiliation(s)
- Yuwei Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650223, China
| | - Li Pear Wan
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650223, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Shuhua Zhao
- First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Jianqin Niu
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Nengyin Sheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
6
|
Szumiec L, Bugno R, Szumiec L, Przewlocki R. The Differential Influence of PZM21, A Nonrewarding μ-opioid Receptor Agonist With G Protein Bias, on Behavioural Despair and Fear Response in Mice. Behav Brain Res 2023; 449:114466. [PMID: 37146718 DOI: 10.1016/j.bbr.2023.114466] [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: 03/09/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
A growing body of evidence points out the involvement of the µ-opioid receptors in the modulation of stress-related behaviour. It has been suggested that µ-opioid receptor agonists may attenuate behavioural despair following animals' exposure to an acute, inescapable stressor. Moreover, morphine was shown to ameliorate fear memories caused by a traumatic experience. As typical µ-opioid receptor agonists entail a risk of serious side effects and addiction, novel, possibly safer and less addictive agonists of this receptor are currently under investigation. One of them, PZM21, preferentially acting via the G protein signalling pathway, was previously shown to be analgesic, but less addictive than morphine. Here, we aimed to further test this ligand in stress-related behavioural paradigms in mice. The study has shown that, unlike morphine, PZM21 does not decrease immobility in the forced swimming and tail suspension tests. On the other hand, we observed that both mice treated with PZM21 and those receiving morphine presented a slight attenuation of freezing across the consecutive fear memory retrievals in the fear conditioning test. Therefore, our study implies that at the range of tested doses, PZM21, a nonrewarding representative of G protein-biased µ-opioid receptor agonists, may interfere with fear memory consolidation while having no beneficial effects on behavioural despair in mice.
Collapse
Affiliation(s)
- Lucja Szumiec
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - Ryszard Bugno
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Lukasz Szumiec
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
7
|
Ma J, Wang R, Chen Y, Wang Z, Dong Y. 5-HT attenuates chronic stress-induced cognitive impairment in mice through intestinal flora disruption. J Neuroinflammation 2023; 20:23. [PMID: 36737776 PMCID: PMC9896737 DOI: 10.1186/s12974-023-02693-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The microbiota-gut-brain axis plays an important role in the development of depression. The aim of this study was to investigate the effects of 5-HT on cognitive function, learning and memory induced by chronic unforeseeable mild stress stimulation (CUMS) in female mice. CUMS mice and TPH2 KO mice were used in the study. Lactococcus lactis E001-B-8 fungus powder was orally administered to mice with CUMS. METHODS We used the open field test, Morris water maze, tail suspension test and sucrose preference test to examine learning-related behaviours. In addition, AB-PAS staining, immunofluorescence, ELISA, qPCR, Western blotting and microbial sequencing were employed to address our hypotheses. RESULTS The effect of CUMS was more obvious in female mice than in male mice. Compared with female CUMS mice, extracellular serotonin levels in TPH2 KO CUMS mice were significantly reduced, and cognitive dysfunction was aggravated. Increased hippocampal autophagy levels, decreased neurotransmitter levels, reduced oxidative stress damage, increased neuroinflammatory responses and disrupted gut flora were observed. Moreover, L. lactis E001-B-8 significantly improved the cognitive behaviour of mice. CONCLUSIONS These results strongly suggest that L. lactis E001-B-8 but not FLX can alleviate rodent depressive and anxiety-like behaviours in response to CUMS, which is associated with the improvement of 5-HT metabolism and modulation of the gut microbiome composition.
Collapse
Affiliation(s)
- Junxing Ma
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Yaoxing Chen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Zixu Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yulan Dong
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
8
|
Reich N, Hölscher C. Beyond Appetite: Acylated Ghrelin As A Learning, Memory and Fear Behavior-modulating Hormone. Neurosci Biobehav Rev 2022; 143:104952. [DOI: 10.1016/j.neubiorev.2022.104952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 04/27/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
|
9
|
Ma P, Wan LP, Li Y, He CH, Song NN, Zhao S, Wang H, Ding YQ, Mao B, Sheng N. RNF220 is an E3 ubiquitin ligase for AMPA receptors to regulate synaptic transmission. SCIENCE ADVANCES 2022; 8:eabq4736. [PMID: 36179027 PMCID: PMC9524831 DOI: 10.1126/sciadv.abq4736] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/15/2022] [Indexed: 06/12/2023]
Abstract
The accurate expression of postsynaptic AMPA receptors (AMPARs) is critical for information processing in the brain, and ubiquitination is a key regulator for this biological process. However, the roles of E3 ubiquitin ligases in the regulation of AMPARs are poorly understood. Here, we find that RNF220 directly interacts with AMPARs to meditate their polyubiquitination, and RNF220 knockout specifically increases AMPAR protein levels, thereby enhancing basal synaptic activity while impairing synaptic plasticity. Moreover, depending on its E3 ubiquitin ligase activity, RNF220 represses AMPAR-mediated excitatory synaptic responses and their neuronal surface expression. Furthermore, learning and memory are altered in forebrain RNF220-deficient mice. In addition, two neuropathology-related RNF220 variants fail to repress excitatory synaptic activity because of the incapability to regulate AMPAR ubiquitination due to their attenuated interaction. Together, we identify RNF220 as an E3 ubiquitin ligase for AMPARs and establish its substantial role in excitatory synaptic transmission and brain function.
Collapse
Affiliation(s)
- Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Li Pear Wan
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650223, China
| | - Yuwei Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650223, China
| | - Chun-Hui He
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ning-Ning Song
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
| | - Shiping Zhao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Huishan Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650223, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Nengyin Sheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
10
|
Pharmacological Implications of Adjusting Abnormal Fear Memory: Towards the Treatment of Post-Traumatic Stress Disorder. Pharmaceuticals (Basel) 2022; 15:ph15070788. [PMID: 35890087 PMCID: PMC9322538 DOI: 10.3390/ph15070788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a unique clinical mental abnormality presenting a cluster of symptoms in which patients primarily experience flashbacks, nightmares and uncontrollable thoughts about the event that triggered their PTSD. Patients with PTSD may also have comorbid depression and anxiety in an intractable and long-term course, which makes establishing a comprehensive treatment plan difficult and complicated. The present article reviews current pharmacological manipulations for adjusting abnormal fear memory. The roles of the central monoaminergic systems (including serotonin, norepinephrine and dopamine) within the fear circuit areas and the involvement of the hypothalamic-pituitary-adrenal (HPA) axis and glucocorticoid receptor (GR) are explored based on attempts to integrate current clinical and preclinical basic studies. In this review, we explain how these therapeutic paradigms function based on their connections to stages of the abnormal fear memory process from condition to extinction. This may provide useful translational interpretations for clinicians to manage PTSD.
Collapse
|
11
|
Coray R, Quednow BB. The role of serotonin in declarative memory: A systematic review of animal and human research. Neurosci Biobehav Rev 2022; 139:104729. [PMID: 35691469 DOI: 10.1016/j.neubiorev.2022.104729] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 10/18/2022]
Abstract
The serotonergic system is involved in diverse cognitive functions including memory. Of particular importance to daily life are declarative memories that contain information about personal experiences, general facts, and events. Several psychiatric or neurological diseases, such as depression, attention-deficit-hyperactivity disorder (ADHD), and dementia, show alterations in serotonergic signalling and attendant memory disorders. Nevertheless, understanding serotonergic neurotransmission and its influence on memory remained a challenge until today. In this systematic review, we summarize recent psychopharmacological studies in animals and humans from a psychological memory perspective, in consideration of task-specific requirements. This approach has the advantage that comparisons between serotonin (5-HT)-related neurochemical mechanisms and manipulations are each addressing specific mnemonic circuits. We conclude that applications of the same 5-HT-related treatments can differentially affect unrelated tasks of declarative memories. Moreover, the analysis of specific mnemonic phases (e.g., encoding vs. consolidation) reveals opposing impacts of increased or decreased 5-HT tones, with low 5-HT supporting spatial encoding but impairing the consolidation of objects and verbal memories. Promising targets for protein synthesis-dependent consolidation enhancements include 5-HT4 receptor agonists and 5-HT6 receptor antagonists, with the latter being of special interest for the treatment of age-related decline. Further implications are pointed out as base for the development of novel therapeutic targets for memory impairment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rebecca Coray
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Switzerland.
| | - Boris B Quednow
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and Swiss Federal Institute of Technology Zurich, Switzerland
| |
Collapse
|
12
|
Miyajima K, Sudo Y, Sanechika S, Hara Y, Horiguchi M, Xu F, Suzuki M, Hara S, Tanda K, Inoue KI, Takada M, Yoshioka N, Takebayashi H, Mori-Kojima M, Sugimoto M, Sumi-Ichinose C, Kondo K, Takao K, Miyakawa T, Ichinose H. Perturbation of monoamine metabolism and enhanced fear responses in mice defective in the regeneration of tetrahydrobiopterin. J Neurochem 2022; 161:129-145. [PMID: 35233765 DOI: 10.1111/jnc.15600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/11/2022] [Accepted: 02/23/2022] [Indexed: 11/28/2022]
Abstract
Increasing evidence suggests the involvement of peripheral amino acid metabolism in the pathophysiology of neuropsychiatric disorders, whereas the molecular mechanisms are largely unknown. Tetrahydrobiopterin (BH4) is a cofactor for enzymes that catalyze phenylalanine metabolism, monoamine synthesis, nitric oxide production, and lipid metabolism. BH4 is synthesized from guanosine triphosphate and regenerated by quinonoid dihydropteridine reductase (QDPR), which catalyzes the reduction of quinonoid dihydrobiopterin. We analyzed Qdpr-/- mice to elucidate the physiological significance of the regeneration of BH4. We found that the Qdpr-/- mice exhibited mild hyperphenylalaninemia and monoamine deficiency in the brain, despite the presence of substantial amounts of BH4 in the liver and brain. Hyperphenylalaninemia was ameliorated by exogenously administered BH4, and dietary phenylalanine restriction was effective for restoring the decreased monoamine contents in the brain of the Qdpr-/- mice, suggesting that monoamine deficiency was caused by the secondary effect of hyperphenylalaninemia. Immunohistochemical analysis showed that QDPR was primarily distributed in oligodendrocytes but hardly detectable in monoaminergic neurons in the brain. Finally, we performed a behavioral assessment using a test battery. The Qdpr-/- mice exhibited enhanced fear responses after electrical foot shock. Taken together, our data suggest that the perturbation of BH4 metabolism should affect brain monoamine levels through alterations in peripheral amino acid metabolism, and might contribute to the development of anxiety-related psychiatric disorders. Cover Image for this issue: https://doi.org/10.1111/jnc.15398.
Collapse
Affiliation(s)
- Katsuya Miyajima
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yusuke Sudo
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Sho Sanechika
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yoshitaka Hara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Mieko Horiguchi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Department of Domestic Science, Otsuma Women's University Junior College Division, Tokyo, Japan
| | - Feng Xu
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Minori Suzuki
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Satoshi Hara
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Koichi Tanda
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | - Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masayo Mori-Kojima
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Chiho Sumi-Ichinose
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Kazunao Kondo
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Keizo Takao
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department Behavioral Physiology, Faculty of Medicine, University of Toyama, Toyama, Japan
- Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Tsuyoshi Miyakawa
- Genetic Engineering and Functional Genomics Group, Frontier Technology Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Section of Behavior Patterns, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Division of Systems Medical Science, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Hiroshi Ichinose
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
13
|
Wang YB, Song NN, Zhang L, Ma P, Chen JY, Huang Y, Hu L, Mao B, Ding YQ. Rnf220 is Implicated in the Dorsoventral Patterning of the Hindbrain Neural Tube in Mice. Front Cell Dev Biol 2022; 10:831365. [PMID: 35399523 PMCID: PMC8988044 DOI: 10.3389/fcell.2022.831365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 11/24/2022] Open
Abstract
Rnf220 is reported to regulate the patterning of the ventral spinal neural tube in mice. The brainstem has divergent connections with peripheral and central targets and contains unique internal neuronal groups, but the role of Rnf220 in the early development of the hindbrain has not been explored. In this study, Nestin-Cre-mediated conditional knockout (Rnf220Nestin CKO) mice were used to examine if Rnf220 is involved in the early morphogenesis of the hindbrain. Rnf220 showed restricted expression in the ventral half of ventricular zone (VZ) of the hindbrain at embryonic day (E) 10.5, and as development progressed, Rnf220-expressing cells were also present in the mantle zone outside the VZ at E12.5. In Rnf220Nestin CKO embryos, alterations of progenitor domains in the ventral VZ were observed at E10.5. There were significant reductions of the p1 and p2 domains shown by expression of Dbx1, Olig2, and Nkx6.1, accompanied by a ventral expansion of the Dbx1+ p0 domain and a dorsal expansion of the Nkx2.2+ p3 domain. Different from the case in the spinal cord, the Olig2+ pMN (progenitors of somatic motor neuron) domain shifted and expanded dorsally. Notably, the total range of the ventral VZ and the extent of the dorsal tube were unchanged. In addition, the post-mitotic cells derived from their corresponding progenitor domain, including oligodendrocyte precursor cells (OPCs) and serotonergic neurons (5-HTNs), were also changed in the same trend as the progenitor domains do in the CKO embryos at E12.5. In summary, our data suggest similar functions of Rnf220 in the hindbrain dorsoventral (DV) patterning as in the spinal cord with different effects on the pMN domain. Our work also reveals novel roles of Rnf220 in the development of 5-HTNs and OPCs.
Collapse
Affiliation(s)
- Yu-Bing Wang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jia-Yin Chen
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ying Huang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ling Hu
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- *Correspondence: Bingyu Mao, ; Yu-Qiang Ding,
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- *Correspondence: Bingyu Mao, ; Yu-Qiang Ding,
| |
Collapse
|
14
|
Peroxiredoxin 6 Knockout Mice Demonstrate Anxiety Behavior and Attenuated Contextual Fear Memory after Receiving Acute Immobilization Stress. Antioxidants (Basel) 2021; 10:antiox10091416. [PMID: 34573048 PMCID: PMC8466988 DOI: 10.3390/antiox10091416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Stress can elicit glucocorticoid release to promote coping mechanisms and influence learning and memory performance. Individual memory performance varies in response to stress, and the underlying mechanism is not clear yet. Peroxiredoxin 6 (PRDX6) is a multifunctional enzyme participating in both physiological and pathological conditions. Several studies have demonstrated the correlation between PRDX6 expression level and stress-related disorders. Our recent finding indicates that lack of the Prdx6 gene leads to enhanced fear memory. However, it is unknown whether PRDX6 is involved in changes in anxiety response and memory performance upon stress. The present study reveals that hippocampal PRDX6 level is downregulated 30 min after acute immobilization stress (AIS) and trace fear conditioning (TFC). In human retinal pigment epithelium (ARPE-19) cells, the PRDX6 expression level decreases after being treated with stress hormone corticosterone. Lack of PRDX6 caused elevated basal H2O2 levels in the hippocampus, basolateral amygdala, and medial prefrontal cortex, brain regions involved in anxiety response and fear memory formation. Additionally, this H2O2 level was still high in the medial prefrontal cortex of the knockout mice under AIS. Anxiety behavior of Prdx6-/- mice was enhanced after immobilization for 30 min. After exposure to AIS before a contextual test, Prdx6-/- mice displayed a contextual fear memory deficit. Our results showed that the memory performance of Prdx6-/- mice was impaired when responding to AIS, accompanied by dysregulated H2O2 levels. The present study helps better understand the function of PRDX6 in memory performance after acute stress.
Collapse
|
15
|
Chai AP, Chen XF, Xu XS, Zhang N, Li M, Li JN, Zhang L, Zhang D, Zhang X, Mao RR, Ding YQ, Xu L, Zhou QX. A Temporal Activity of CA1 Neurons Underlying Short-Term Memory for Social Recognition Altered in PTEN Mouse Models of Autism Spectrum Disorder. Front Cell Neurosci 2021; 15:699315. [PMID: 34335191 PMCID: PMC8319669 DOI: 10.3389/fncel.2021.699315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/21/2021] [Indexed: 11/30/2022] Open
Abstract
Memory-guided social recognition identifies someone from previous encounters or experiences, but the mechanisms of social memory remain unclear. Here, we find that a short-term memory from experiencing a stranger mouse lasting under 30 min interval is essential for subsequent social recognition in mice, but that interval prolonged to hours by replacing the stranger mouse with a familiar littermate. Optogenetic silencing of dorsal CA1 neuronal activity during trials or inter-trial intervals disrupted short-term memory-guided social recognition, without affecting the ability of being sociable or long-term memory-guided social recognition. Postnatal knockdown or knockout of autism spectrum disorder (ASD)-associated phosphatase and tensin homolog (PTEN) gene in dorsal hippocampal CA1 similarly impaired neuronal firing rate in vitro and altered firing pattern during social recognition. These PTEN mice showed deficits in social recognition with stranger mouse rather than littermate and exhibited impairment in T-maze spontaneous alternation task for testing short-term spatial memory. Thus, we suggest that a temporal activity of dorsal CA1 neurons may underlie formation of short-term memory to be critical for organizing subsequent social recognition but that is possibly disrupted in ASD.
Collapse
Affiliation(s)
- An-Ping Chai
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xue-Feng Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
- School of Life Sciences, Yunnan University, Kunming, China
| | - Xiao-Shan Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Na Zhang
- School of Life Sciences, Anhui University, Hefei, China
| | - Meng Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Jin-Nan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Lei Zhang
- Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Dai Zhang
- Institute of Mental Health, The Sixth Hospital of Peking University, Beijing, China
| | - Xia Zhang
- Department of Cellular and Molecular Medicine, Institute of Mental Health Research at the Royal, University of Ottawa, Ottawa, ON, Canada
- Department of Psychiatry, Institute of Mental Health Research at the Royal, University of Ottawa, Ottawa, ON, Canada
| | - Rong-Rong Mao
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| | - Yu-Qiang Ding
- Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
- School of Life Sciences, Yunnan University, Kunming, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai, China
| | - Qi-Xin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, and KIZ-SU Joint Laboratory of Animal Model and Drug Development, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
16
|
Abstract
OBJECTIVE Whereas numerous experimental and clinical studies suggest a complex involvement of serotonin in the regulation of anxiety, it remains to be clarified if the dominating impact of this transmitter is best described as anxiety-reducing or anxiety-promoting. The aim of this study was to assess the impact of serotonin depletion on acquisition, consolidation, and expression of conditioned fear. METHODS Male Sprague-Dawley rats were exposed to foot shocks as unconditioned stimulus and assessed with respect to freezing behaviour when re-subjected to context. Serotonin depletion was achieved by administration of a serotonin synthesis inhibitor, para-chlorophenylalanine (PCPA) (300 mg/kg daily × 3), (i) throughout the period from (and including) acquisition to (and including) expression, (ii) during acquisition but not expression, (iii) after acquisition only, and (iv) during expression only. RESULTS The time spent freezing was significantly reduced in animals that were serotonin-depleted during the entire period from (and including) acquisition to (and including) expression, as well as in those being serotonin-depleted during either acquisition only or expression only. In contrast, PCPA administrated immediately after acquisition, that is during memory consolidation, did not impact the expression of conditioned fear. CONCLUSION Intact serotonergic neurotransmission is important for both acquisition and expression of context-conditioned fear.
Collapse
|
17
|
PTEN in prefrontal cortex is essential in regulating depression-like behaviors in mice. Transl Psychiatry 2021; 11:185. [PMID: 33771972 PMCID: PMC7998021 DOI: 10.1038/s41398-021-01312-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/24/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic stress is an environmental risk factor for depression and causes neuronal atrophy in the prefrontal cortex (PFC) and other brain regions. It is still unclear about the molecular mechanism underlying the behavioral alterations and neuronal atrophy induced by chronic stress. We here report that phosphatase and tensin homolog deleted on chromosome ten (PTEN) is a mediator for chronic stress-induced depression-like behaviors and neuronal atrophy in mice. One-month chronic restraint stress (CRS) up-regulated PTEN signaling pathway in the PFC of mice as indicated by increasing levels of PTEN, p-MEK, and p-ERK but decreasing levels of p-AKT. Over-expression of Pten in the PFC led to an increase of depression-like behaviors, whereas genetic inactivation or knockdown of Pten in the PFC prevented the CRS-induced depression-like behaviors. In addition, systemic administration of PTEN inhibitor was also able to prevent these behaviors. Cellular examination showed that Pten over-expression or the CRS treatment resulted in PFC neuron atrophy, and this atrophy was blocked by genetic inactivation of Pten or systemic administration of PTEN inhibitor. Furthermore, possible causal link between Pten and glucocorticoids was examined. In chronic dexamethasone (Dex, a glucocorticoid agonist) treatment-induced depression model, increased PTEN levels were observed, and depression-like behaviors and PFC neuron atrophy were attenuated by the administration of PTEN inhibitor. Our results indicate that PTEN serves as a key mediator in chronic stress-induced neuron atrophy as well as depression-like behaviors, providing molecular evidence supporting the synaptic plasticity theory of depression.
Collapse
|
18
|
Pten is a key intrinsic factor regulating raphe 5-HT neuronal plasticity and depressive behaviors in mice. Transl Psychiatry 2021; 11:186. [PMID: 33771970 PMCID: PMC7998026 DOI: 10.1038/s41398-021-01303-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 02/20/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin (5-HT)-based antidepressants, selective serotonin reuptake inhibitors (SSRIs) aim to enhance serotonergic activity by blocking its reuptake. We propose PTEN as a target for an alternative approach for regulating 5-HT neuron activity in the brain and depressive behaviors. We show that PTEN is elevated in central 5-HT neurons in the raphe nucleus by chronic stress in mice, and selective deletion of Pten in the 5-HT neurons induces its structural plasticity shown by increases of dendritic branching and density of PSD95-positive puncta in the dendrites. 5-HT levels are elevated and electrical stimulation of raphe neurons evokes more 5-HT release in the brain of condition knockout (cKO) mice with Pten-deficient 5-HT neurons. In addition, the 5-HT neurons remain normal electrophysiological properties but have increased excitatory synaptic inputs. Single-cell RNA sequencing revealed gene transcript alterations that may underlay morphological and functional changes in Pten-deficient 5-HT neurons. Finally, Pten cKO mice and wild-type mice treated with systemic application of PTEN inhibitor display reduced depression-like behaviors. Thus, PTEN is an intrinsic regulator of 5-HT neuron activity, representing a novel therapeutic strategy for producing antidepressant action.
Collapse
|
19
|
Serotonin deficiency induced after brain maturation rescues consequences of early life adversity. Sci Rep 2021; 11:5368. [PMID: 33686115 PMCID: PMC7940624 DOI: 10.1038/s41598-021-83592-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 01/22/2021] [Indexed: 01/31/2023] Open
Abstract
Brain serotonin (5-HT) system dysfunction is implicated in depressive disorders and acute depletion of 5-HT precursor tryptophan has frequently been used to model the influence of 5-HT deficiency on emotion regulation. Tamoxifen (TAM)-induced Cre/loxP-mediated inactivation of the tryptophan hydroxylase-2 gene (Tph2) was used to investigate the effects of provoked 5-HT deficiency in adult mice (Tph2 icKO) previously subjected to maternal separation (MS). The efficiency of Tph2 inactivation was validated by immunohistochemistry and HPLC. The impact of Tph2 icKO in interaction with MS stress (Tph2 icKO × MS) on physiological parameters, emotional behavior and expression of 5-HT system-related marker genes were assessed. Tph2 icKO mice displayed a significant reduction in 5-HT immunoreactive cells and 5-HT concentrations in the rostral raphe region within four weeks following TAM treatment. Tph2 icKO and MS differentially affected food and water intake, locomotor activity as well as panic-like escape behavior. Tph2 icKO prevented the adverse effects of MS stress and altered the expression of the genes previously linked to stress and emotionality. In conclusion, an experimental model was established to study the behavioral and neurobiological consequences of 5-HT deficiency in adulthood in interaction with early-life adversity potentially affecting brain development and the pathogenesis of depressive disorders.
Collapse
|
20
|
Qi CC, Chen XX, Gao XR, Xu JX, Liu S, Ge JF. Impaired Learning and Memory Ability Induced by a Bilaterally Hippocampal Injection of Streptozotocin in Mice: Involved With the Adaptive Changes of Synaptic Plasticity. Front Aging Neurosci 2021; 13:633495. [PMID: 33732137 PMCID: PMC7957014 DOI: 10.3389/fnagi.2021.633495] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/09/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline, psychiatric symptoms and behavioral disorders, resulting in disability, and loss of self-sufficiency. Objective: To establish an AD-like mice model, investigate the behavioral performance, and explore the potential mechanism. Methods: Streptozotocin (STZ, 3 mg/kg) was microinjected bilaterally into the dorsal hippocampus of C57BL/6 mice, and the behavioral performance was observed. The serum concentrations of insulin and nesfatin-1 were measured by ELISA, and the activation of hippocampal microglia and astrocytes was assessed by immunohistochemistry. The protein expression of several molecular associated with the regulation of synaptic plasticity in the hippocampus and the pre-frontal cortex (PFC) was detected via western blotting. Results: The STZ-microinjected model mice showed a slower bodyweight gain and higher serum concentration of insulin and nesfatin-1. Although there was no significant difference between groups with regard to the ability of balance and motor coordination, the model mice presented a decline of spontaneous movement and exploratory behavior, together with an impairment of learning and memory ability. Increased activated microglia was aggregated in the hippocampal dentate gyrus of model mice, together with an increase abundance of Aβ1-42 and Tau in the hippocampus and PFC. Moreover, the protein expression of NMDAR2A, NMDAR2B, SynGAP, PSD95, BDNF, and p-β-catenin/β-catenin were remarkably decreased in the hippocampus and the PFC of model mice, and the expression of p-GSK-3β (ser9)/GSK-3β were reduced in the hippocampus. Conclusion: A bilateral hippocampal microinjection of STZ could induce not only AD-like behavioral performance in mice, but also adaptive changes of synaptic plasticity against neuroinflammatory and endocrinal injuries. The underlying mechanisms might be associated with the imbalanced expression of the key proteins of Wnt signaling pathway in the hippocampus and the PFC.
Collapse
Affiliation(s)
- Cong-Cong Qi
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Xing-Xing Chen
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xin-Ran Gao
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jing-Xian Xu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Sen Liu
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jin-Fang Ge
- School of Pharmacy, Anhui Medical University, Hefei, China.,Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Anhui Institute of Innovative Drugs, Hefei, China.,The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Phasuk S, Pairojana T, Suresh P, Yang CH, Roytrakul S, Huang SP, Chen CC, Pakaprot N, Chompoopong S, Nudmamud-Thanoi S, Liu IY. Enhanced contextual fear memory in peroxiredoxin 6 knockout mice is associated with hyperactivation of MAPK signaling pathway. Mol Brain 2021; 14:42. [PMID: 33632301 PMCID: PMC7908735 DOI: 10.1186/s13041-021-00754-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Fear dysregulation is one of the symptoms found in post-traumatic stress disorder (PTSD) patients. The functional abnormality of the hippocampus is known to be implicated in the development of such pathology. Peroxiredoxin 6 (PRDX6) belongs to the peroxiredoxin family. This antioxidant enzyme is expressed throughout the brain, including the hippocampus. Recent evidence reveals that PRDX6 plays an important role in redox regulation and the modulation of several signaling molecules involved in fear regulation. Thus, we hypothesized that PRDX6 plays a role in the regulation of fear memory. We subjected a systemic Prdx6 knockout (Prdx6-/-) mice to trace fear conditioning and observed enhanced fear response after training. Intraventricular injection of lentivirus-carried mouse Prdx6 into the 3rd ventricle reduced the enhanced fear response in these knockout mice. Proteomic analysis followed by validation of western blot analysis revealed that several proteins in the MAPK pathway, such as NTRK2, AKT, and phospho-ERK1/2, cPLA2 were significantly upregulated in the hippocampus of Prdx6-/- mice during the retrieval stage of contextual fear memory. The distribution of PRDX6 found in the astrocytes was also observed throughout the hippocampus. This study identifies PRDX6 as a participant in the regulation of fear response. It suggests that PRDX6 and related molecules may have important implications for understanding fear-dysregulation associated disorders like PTSD.
Collapse
Affiliation(s)
- Sarayut Phasuk
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tanita Pairojana
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Pavithra Suresh
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Chee-Hing Yang
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Chien-Chang Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Narawut Pakaprot
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supin Chompoopong
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sutisa Nudmamud-Thanoi
- Department of Anatomy, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
- Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Ingrid Y. Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
22
|
Liu WG, Zhang LM, Yao JQ, Yin YY, Zhang XY, Li YF, Cao JB. Anti-PTSD Effects of Hypidone Hydrochloride (YL-0919): A Novel Combined Selective 5-HT Reuptake Inhibitor/5-HT 1A Receptor Partial Agonist/5-HT 6 Receptor Full Agonist. Front Pharmacol 2021; 12:625547. [PMID: 33643051 PMCID: PMC7902863 DOI: 10.3389/fphar.2021.625547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/06/2021] [Indexed: 01/17/2023] Open
Abstract
Posttraumatic stress disorder (PTSD) is a debilitating trauma and stressor-related disorder that has become a major neuropsychiatric problem, leading to substantial disruptions in individual health and societal costs. Our previous studies have demonstrated that hypidone hydrochloride (YL-0919), a novel combined selective 5-HT reuptake inhibitor/5-HT1A receptor partial agonist/5-HT6 receptor full agonist, exerts notable antidepressant- and anxiolytic-like as well as procognitive effects. However, whether YL-0919 exerts anti-PTSD effects and its underlying mechanisms are still unclear. In the present study, we showed that repeated treatment with YL-0919 caused significant suppression of contextual fear, enhanced anxiety and cognitive dysfunction induced by the time-dependent sensitization (TDS) procedure in rats and by inescapable electric foot-shock in a mouse model of PTSD. Furthermore, we found that repeated treatment with YL-0919 significantly reversed the accompanying decreased expression of the brain-derived neurotrophic factor (BDNF) and the synaptic proteins (synapsin1 and GluA1), and ameliorated the neuroplasticity disruption in the prefrontal cortex (PFC), including the dendritic complexity and spine density of pyramidal neurons. Taken together, the current study indicated that YL-0919 exerts clear anti-PTSD effects, which might be partially mediated by ameliorating the structural neuroplasticity by increasing the expression of BDNF and the formation of synaptic proteins in the PFC.
Collapse
Affiliation(s)
- Wen-Gang Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China.,Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Jun-Qi Yao
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Xiao-Ying Zhang
- Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.,Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jiang-Bei Cao
- Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
23
|
ZFP804A mutant mice display sex-dependent schizophrenia-like behaviors. Mol Psychiatry 2021; 26:2514-2532. [PMID: 33303946 PMCID: PMC8440220 DOI: 10.1038/s41380-020-00972-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 10/20/2020] [Accepted: 11/24/2020] [Indexed: 12/22/2022]
Abstract
Genome-wide association studies uncovered the association of ZNF804A (Zinc-finger protein 804A) with schizophrenia (SZ). In vitro data have indicated that ZNF804A might exert its biological roles by regulating spine and neurite morphogenesis. However, no in vivo data are available for the role of ZNF804A in psychiatric disorders in general, SZ in particular. We generated ZFP804A mutant mice, and they showed deficits in contextual fear and spatial memory. We also observed the sensorimotor gating impairment, as revealed by the prepulse inhibition test, but only in female ZFP804A mutant mice from the age of 6 months. Notably, the PPI difference between the female mutant and control mice was no longer existed with the administration of Clozapine or after the ovariectomy. Hippocampal long-term potentiation was normal in both genders of the mutant mice. Long-term depression was absent in male mutants, but facilitated in the female mutants. Protein levels of hippocampal serotonin-6 receptor and GABAB1 receptor were increased, while those of cortical dopamine 2 receptor were decreased in the female mutants with no obvious changes in the male mutants. Moreover, the spine density was reduced in the cerebral cortex and hippocampus of the mutant mice. Knockdown of ZFP804A impaired the neurite morphogenesis of cortical and hippocampal neurons, while its overexpression enhanced neurite morphogenesis only in the cortical neurons in vitro. Our data collectively support the idea that ZFP804A/ZNF804A plays important roles in the cognitive functions and sensorimotor gating, and its dysfunction may contribute to SZ, particularly in the female patients.
Collapse
|
24
|
Schiele MA, Thiel C, Weidner M, Endres D, Zaudig M, Berberich G, Domschke K. Serotonin transporter gene promoter hypomethylation in obsessive-compulsive disorder - Predictor of impaired response to exposure treatment? J Psychiatr Res 2021; 132:18-22. [PMID: 33035761 DOI: 10.1016/j.jpsychires.2020.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/22/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022]
Abstract
Treatment resistance is common in obsessive-compulsive disorder (OCD) and associated with a significant burden for the individual patient. Accordingly, the identification of biomarkers as early predictors of the clinical response has become a central goal in the search for more efficacious and personalized treatments. Epigenetic mechanisms such as DNA methylation of the serotonin transporter gene (SLC6A4) have been suggested to predict therapy outcome in mental disorders closely related to OCD, but have not yet been investigated as such in OCD. The present therapy-epigenetic study therefore sought to address the potential role of SLC6A4 promoter methylation in the prediction of treatment response for the first time in OCD. Overall, 112 patients with primary OCD were investigated over the course of 8-10-week OCD-specific, cognitive behavioral therapy (CBT) comprising exposure and response prevention/management (phase I) and in vivo exposure exercises ('flooding', phase II). OCD symptoms were measured using the Yale-Brown Obsessive Compulsive Scale (Y-BOCS) at baseline as well as before and after the in vivo exposure phase. SLC6A4 promoter methylation at baseline was analyzed via pyrosequencing of sodium bisulfite-treated DNA extracted from blood cells. Lower baseline SLC6A4 promoter methylation predicted impaired treatment response (defined as reduction in Y-BOCS scores) in phase II (but not phase I) of CBT (β = -0.359, p = .002). SLC6A4 methylation may thus constitute a potential early biomarker predicting biologically mediated clinical changes elicited specifically by exposure treatment. These results carry promise for clinical application and in the future could aid in early treatment modification and personalized treatment efforts.
Collapse
Affiliation(s)
- Miriam A Schiele
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Christiane Thiel
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Magdalena Weidner
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Dominique Endres
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | | | | | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
25
|
Laureano-Melo R, Dos-Santos RC, da Conceição RR, de Souza JS, da Silva Lau R, da Silva Souza Silva S, Marinho BG, Giannocco G, Ahmed RG, da Silva Côrtes W. Perinatal fluoxetine treatment promotes long-term behavioral changes in adult mice. Metab Brain Dis 2020; 35:1341-1351. [PMID: 32827287 DOI: 10.1007/s11011-020-00606-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/04/2020] [Indexed: 01/19/2023]
Abstract
Serotonin exerts a significant role in the mammalian central nervous system embryogenesis and brain ontogeny. Therefore, we investigate the effect of perinatal fluoxetine (FLX), a selective serotonin reuptake inhibitor, administration on the behavioral expression of adult male Swiss mice. For this purpose, two groups (n = 6 each, and ~ 35 g) of pregnant female Swiss mice were mated. Their offspring were treated with FLX (10 mg/Kg, s.c.) from postnatal day (PND) 5 to 15. At PND 16, one male puppy of each litter was euthanized, and the hippocampus was dissected for RNA analysis. At 70 days of life, the male offspring underwent a behavioral assessment in the open field, object recognition task, light-dark box, tail suspension and rotarod test. According to our results, the programmed animals had a decrease in TPH2, 5HT1a, SERT, BDNF, and LMX1B expression. Also, it was observed less time of immobility in tail suspension test and higher grooming time in the open field test. In the light-dark box test, the FLX-treated offspring had less time in the light side than control. We also observed a low cognitive performance in the object recognition task and poor motor skill learning in the rotarod test. These findings suggest that programming with FLX during the neonatal period alters a hippocampal serotonergic system, promoting anxiety and antidepressant behavior in adults, as well as a low mnemonic capacity.
Collapse
Affiliation(s)
- Roberto Laureano-Melo
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Health and Biological Sciences, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil.
- Department of Veterinary Medicine, Barra Mansa University Center, Rio de Janeiro, Brazil.
| | - Raoni Conceição Dos-Santos
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Health and Biological Sciences, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Rodrigo Rodrigues da Conceição
- Molecular and Translational Endocrinology Laboratory, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Janaina Sena de Souza
- Molecular and Translational Endocrinology Laboratory, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Raphael da Silva Lau
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Health and Biological Sciences, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Samantha da Silva Souza Silva
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Health and Biological Sciences, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Bruno Guimarães Marinho
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Health and Biological Sciences, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| | - Gisele Giannocco
- Molecular and Translational Endocrinology Laboratory, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - R G Ahmed
- Division of Anatomy and Embryology, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Wellington da Silva Côrtes
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Institute of Health and Biological Sciences, Universidade Federal Rural do Rio de Janeiro, Seropedica, Brazil
| |
Collapse
|
26
|
Inactivation of the GATA Cofactor ZFPM1 Results in Abnormal Development of Dorsal Raphe Serotonergic Neuron Subtypes and Increased Anxiety-Like Behavior. J Neurosci 2020; 40:8669-8682. [PMID: 33046550 DOI: 10.1523/jneurosci.2252-19.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
Serotonergic neurons in the dorsal raphe (DR) nucleus are associated with several psychiatric disorders including depression and anxiety disorders, which often have a neurodevelopmental component. During embryonic development, GATA transcription factors GATA2 and GATA3 operate as serotonergic neuron fate selectors and regulate the differentiation of serotonergic neuron subtypes of DR. Here, we analyzed the requirement of GATA cofactor ZFPM1 in the development of serotonergic neurons using Zfpm1 conditional mouse mutants. Our results demonstrated that, unlike the GATA factors, ZFPM1 is not essential for the early differentiation of serotonergic precursors in the embryonic rhombomere 1. In contrast, in perinatal and adult male and female Zfpm1 mutants, a lateral subpopulation of DR neurons (ventrolateral part of the DR) was lost, whereas the number of serotonergic neurons in a medial subpopulation (dorsal region of the medial DR) had increased. Additionally, adult male and female Zfpm1 mutants had reduced serotonin concentration in rostral brain areas and displayed increased anxiety-like behavior. Interestingly, female Zfpm1 mutant mice showed elevated contextual fear memory that was abolished with chronic fluoxetine treatment. Altogether, these results demonstrate the importance of ZFPM1 for the development of DR serotonergic neuron subtypes involved in mood regulation. It also suggests that the neuronal fate selector function of GATAs is modulated by their cofactors to refine the differentiation of neuronal subtypes.SIGNIFICANCE STATEMENT Predisposition to anxiety disorders has both a neurodevelopmental and a genetic basis. One of the brainstem nuclei involved in the regulation of anxiety is the dorsal raphe, which contains different subtypes of serotonergic neurons. We show that inactivation of a transcriptional cofactor ZFPM1 in mice results in a developmental failure of laterally located dorsal raphe serotonergic neurons and changes in serotonergic innervation of rostral brain regions. This leads to elevated anxiety-like behavior and contextual fear memory, alleviated by chronic fluoxetine treatment. Our work contributes to understanding the neurodevelopmental mechanisms that may be disturbed in the anxiety disorder.
Collapse
|
27
|
Hessel M, Pape HC, Seidenbecher T. Stimulation of 5-HT receptors in anterodorsal BNST guides fear to predictable and unpredictable threat. Eur Neuropsychopharmacol 2020; 39:56-69. [PMID: 32873441 DOI: 10.1016/j.euroneuro.2020.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 01/31/2023]
Abstract
Through pharmacological manipulation of the serotonergic (5-Hydroxytryptamin, 5-HT) system, combined with behavioral analysis, we tested the hypothesis that fear responses to predictable and unpredictable threat are regulated through stimulation of 5-HT receptors (5-HT-R) in the anterodorsal section of the bed nucleus of the stria terminalis (adBNST). Local adBNST application of 5-HT1A-R antagonist WAY100635 and 5-HT1B-R antagonist NAS-181 before fear retrieval enhanced freezing, 24 h after predictable fear conditioning. In contrast, increased fear responses to unpredictable threat were blocked by 5-HT1A-R agonist Buspirone (given before conditioning or retrieval) and 5-HT1B-R agonist CP-94253 (applied before training). Prolonged fear responses were also blocked by local application of the 5-HT2A-R antagonist R-96544 before fear retrieval, and conversely, local application of the 5-HT2A-R agonist NBOH-2C-CN hydrochloride before fear retrieval enhanced freezing 24 h after predictable conditioning, indicating augmented fear responses. Activation of inhibitory 5-HT1A- or 5-HT1B-Rs and the blockade of the excitatory 5-HT2A-R before unpredictable fear conditioning significantly reduced freezing during retrieval. The results from this study suggest that modulation of inhibitory 5-HT1A/1B-R and/or excitatory 5-HT2A-R activity in the adBNST may represent potential targets for the development of new treatment strategies in anxiety disorders. In addition, this study supports the validity and reliability of the mouse model of modulated fear to predictable and unpredictable threats to study mechanisms of fear and anxiety in combination with pharmacological manipulations.
Collapse
Affiliation(s)
- Margarita Hessel
- Institute of Physiology I, Westfälische Wilhelms-University Münster, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische Wilhelms-University Münster, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Thomas Seidenbecher
- Institute of Physiology I, Westfälische Wilhelms-University Münster, Robert-Koch-Str. 27a, D-48149 Münster, Germany.
| |
Collapse
|
28
|
Chen GY, Zhang S, Li CH, Qi CC, Wang YZ, Chen JY, Wang G, Ding YQ, Su CJ. Mediator Med23 Regulates Adult Hippocampal Neurogenesis. Front Cell Dev Biol 2020; 8:699. [PMID: 32850819 PMCID: PMC7403405 DOI: 10.3389/fcell.2020.00699] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/09/2020] [Indexed: 11/13/2022] Open
Abstract
Mammalian Mediator (Med) is a key regulator of gene expression by linking transcription factors to RNA polymerase II (Pol II) transcription machineries. The Mediator subunit 23 (Med23) is a member of the conserved Med protein complex and plays essential roles in diverse biological processes including adipogenesis, carcinogenesis, osteoblast differentiation, and T-cell activation. However, its potential functions in the nervous system remain unknown. We report here that Med23 is required for adult hippocampal neurogenesis in mouse. Deletion of Med23 in adult hippocampal neural stem cells (NSCs) was achieved in Nestin-CreER:Med23flox/flox mice by oral administration of tamoxifen. We found an increased number of proliferating NSCs shown by pulse BrdU-labeling and immunostaining of MCM2 and Ki67, which is possibly due to a reduction in cell cycle length, with unchanged GFAP+/Sox2+ NSCs and Tbr2+ progenitors. On the other hand, neuroblasts and immature neurons indicated by NeuroD and DCX were decreased in number in the dentate gyrus (DG) of Med23-deficient mice. In addition, these mice also displayed defective dendritic morphogenesis, as well as a deficiency in spatial and contextual fear memory. Gene ontology (GO) analysis of hippocampal NSCs revealed an enrichment in genes involved in cell proliferation, Pol II-associated transcription, Notch signaling pathway and apoptosis. These results demonstrate that Med23 plays roles in regulating adult brain neurogenesis and functions.
Collapse
Affiliation(s)
- Guo-Yan Chen
- Department of Neurology, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi'an, China.,Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Shuai Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - Chong-Hui Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Cong-Cong Qi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, and Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Ya-Zhou Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jia-Yin Chen
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
| | - Gang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Sciences, Fudan University, Shanghai, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Brain Science, and Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Chang-Jun Su
- Department of Neurology, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi'an, China
| |
Collapse
|
29
|
Guo KF, Dai M, Liu YM, Zhang JC, Chen YM, Ye H, Li MB, Mao RR, Cao J. Acute Administration of Methyleugenol Impairs Hippocampus-Dependent Contextual Fear Memory and Increases Anxiety-like Behavior in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:7490-7497. [PMID: 32551566 DOI: 10.1021/acs.jafc.0c01863] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Methyleugenol (ME) as a natural essential oil in many plant species is widely used in human food and beverage for its fragrance and possible beneficial health effects. Previous chronic or subacute studies in rodents show that ME mainly causes liver toxicity. However, whether and how acute ME affects the central nervous system still remain elusive. Here, we found that ME administrated into the hippocampus impaired the acquisition of hippocampus-dependent contextual fear memory in mice (ME vs control: repeated-measures two-way ANOVA, F(5,70) = 2.937, p < 0.05; Fisher test, p < 0.05, respectively, 53 ± 5.2% vs 73 ± 7.6% during trial 4 and 46.8 ± 6% vs 74.5 ± 9.3% during trial 5). Meanwhile, acute ME impaired hippocampal CA1 long-term potentiation (LTP; ME vs control: independent t-test, p < 0.01, 110.6 ± 1.8% vs 133.3 ± 5.6%) while facilitated long-term depression (LTD; p < 0.01, 75.7 ± 3.4% vs 88.6 ± 1.7%) in mice brain slices and inducing a decrease in learning-dependent phosphorylation of Ser831 (ME vs control: independent t-test, p < 0.001, 0.87 ± 0.03 vs 1.23 ± 0.03) and Ser845 (p < 0.01, 0.42 ± 0.07 vs 0.97 ± 0.14) sites of excitatory glutamate AMPA receptor subunit 1 (GluA1) in the hippocampus, which may be the underlying mechanisms of impairment of hippocampus-dependent learning. In addition, intrahippocampal infusion of ME also increased anxiety-like behaviors in mice. These results suggested that acute ME impaired the hippocampus function at behavioral, cellular, and molecular levels, indicating the potential risks of ME on the central nervous system.
Collapse
Affiliation(s)
- Kai-Fei Guo
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
- School of Agriculture, Yunnan University, Kunming, Yunnan 650504, China
| | - Man Dai
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
| | - Yi-Miao Liu
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
| | - Ji-Chuan Zhang
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yan-Mei Chen
- Department of Basic Medicine, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Hui Ye
- School of Agriculture, Yunnan University, Kunming, Yunnan 650504, China
| | - Man-Bi Li
- Yunnan Institute of Environmental Sciences, Kunming, Yunnan 650034, China
| | - Rong-Rong Mao
- Department of Pathology and Pathophysiology, School of Basic Medical Science, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Jun Cao
- Yunnan Key Laboratory of Plant Reproductive Adaption and Evolutionary Ecology and School of Ecology and Environmental Science, Yunnan University, Kunming, Yunnan 650504, China
| |
Collapse
|
30
|
Forero A, Ku HP, Malpartida AB, Wäldchen S, Alhama-Riba J, Kulka C, Aboagye B, Norton WHJ, Young AMJ, Ding YQ, Blum R, Sauer M, Rivero O, Lesch KP. Serotonin (5-HT) neuron-specific inactivation of Cadherin-13 impacts 5-HT system formation and cognitive function. Neuropharmacology 2020; 168:108018. [PMID: 32113967 DOI: 10.1016/j.neuropharm.2020.108018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/15/2020] [Accepted: 02/23/2020] [Indexed: 02/06/2023]
Abstract
Genome-wide screening approaches identified the cell adhesion molecule Cadherin-13 (CDH13) as a risk factor for neurodevelopmental disorders, nevertheless the contribution of CDH13 to the disease mechanism remains obscure. CDH13 is involved in neurite outgrowth and axon guidance during early brain development and we previously provided evidence that constitutive CDH13 deficiency influences the formation of the raphe serotonin (5-HT) system by modifying neuron-radial glia interaction. Here, we dissect the specific impact of CDH13 on 5-HT system development and function using a 5-HT neuron-specific Cdh13 knockout mouse model (conditional Cdh13 knockout, Cdh13 cKO). Our results show that exclusive inactivation of CDH13 in 5-HT neurons selectively increases 5-HT neuron density in the embryonic dorsal raphe, with persistence into adulthood, and serotonergic innervation of the developing prefrontal cortex. At the behavioral level, adult Cdh13 cKO mice display delayed acquisition of several learning tasks and a subtle impulsive-like phenotype, with decreased latency in a sociability paradigm alongside with deficits in visuospatial memory. Anxiety-related traits were not observed in Cdh13 cKO mice. Our findings further support the critical role of CDH13 in the development of dorsal raphe 5-HT circuitries, a mechanism that may underlie specific clinical features observed in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Andrea Forero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.
| | - Hsing-Ping Ku
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Ana Belén Malpartida
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Sina Wäldchen
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Judit Alhama-Riba
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Christina Kulka
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Benjamin Aboagye
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - William H J Norton
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Andrew M J Young
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Yu-Qiang Ding
- Institute of Brain Sciences, Fudan University, Shanghai, 200031, China
| | - Robert Blum
- Institute of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Olga Rivero
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
31
|
Jiang L, Wang L, Yin Y, Huo M, Liu C, Zhou Q, Yu D, Xu L, Mao R. Spaced Training Enhances Contextual Fear Memory via Activating Hippocampal 5-HT2A Receptors. Front Mol Neurosci 2020; 12:317. [PMID: 32038159 PMCID: PMC6992649 DOI: 10.3389/fnmol.2019.00317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/11/2019] [Indexed: 11/20/2022] Open
Abstract
Spaced training is robustly superior to massed training, which is a well-documented phenomenon in humans and animals. However, the mechanisms underlying the spacing effect still remain unclear. We have reported previously that spacing training exerts memory-enhancing effects by inhibiting forgetting via decreasing hippocampal Rac1 activity. Here, using contextual fear conditioning in rat, we found that spaced but not massed training increased hippocampal 5-HT2A receptors' expression. Furthermore, hippocampal administration of 5-HT2A receptor antagonist MDL11939 before spaced training blocked the enhanced memory, while hippocampal administration of 5-HT2A receptor agonist TCB-2 before massed training promoted the memory. Moreover, MDL11939 activated hippocampal Rac1, while TCB-2 decreased hippocampal Rac1 activity in naïve rats. These results indicated the possibility of interaction between 5-HT2A receptors and Rac1, which was demonstrated by co-immunoprecipitation experiments. Our study first demonstrates that activation of hippocampal 5-HT2A is a mechanism underlying the spacing effect, and forgetting related molecular Rac1 is engaged in this process through interacting with 5-HT2A receptors, which suggest a promising strategy to modulate abnormal learning in cognitive disorders.
Collapse
Affiliation(s)
- Lizhu Jiang
- Department of Neuropsychopathy, Clinical Medical School, Dali University, Dali, China
- Key Lab of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- Department of Central Laboratory, The Third People’s Hospital of Yunnan Province, Kunming, China
| | - Liping Wang
- Key Lab of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Yan Yin
- Department of Central Laboratory, The Third People’s Hospital of Yunnan Province, Kunming, China
| | - Mengke Huo
- Department of Neuropsychopathy, Clinical Medical School, Dali University, Dali, China
| | - Chao Liu
- Key Lab of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- Key Lab of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Qixin Zhou
- Key Lab of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Dafu Yu
- Department of Nuclear Medicine, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Lin Xu
- Key Lab of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, China
- Key Lab of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Rongrong Mao
- Department of Pathology and Pathophysiology, School of Basic Medical Science, Kunming Medical University, Kunming, China
| |
Collapse
|
32
|
Donovan LJ, Spencer WC, Kitt MM, Eastman BA, Lobur KJ, Jiao K, Silver J, Deneris ES. Lmx1b is required at multiple stages to build expansive serotonergic axon architectures. eLife 2019; 8:e48788. [PMID: 31355748 PMCID: PMC6685705 DOI: 10.7554/elife.48788] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/27/2019] [Indexed: 01/18/2023] Open
Abstract
Formation of long-range axons occurs over multiple stages of morphological maturation. However, the intrinsic transcriptional mechanisms that temporally control different stages of axon projection development are unknown. Here, we addressed this question by studying the formation of mouse serotonin (5-HT) axons, the exemplar of long-range profusely arborized axon architectures. We report that LIM homeodomain factor 1b (Lmx1b)-deficient 5-HT neurons fail to generate axonal projections to the forebrain and spinal cord. Stage-specific targeting demonstrates that Lmx1b is required at successive stages to control 5-HT axon primary outgrowth, selective routing, and terminal arborization. We show a Lmx1b→Pet1 regulatory cascade is temporally required for 5-HT arborization and upregulation of the 5-HT axon arborization gene, Protocadherin-alphac2, during postnatal development of forebrain 5-HT axons. Our findings identify a temporal regulatory mechanism in which a single continuously expressed transcription factor functions at successive stages to orchestrate the progressive development of long-range axon architectures enabling expansive neuromodulation.
Collapse
Affiliation(s)
- Lauren J Donovan
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - William C Spencer
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Meagan M Kitt
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Brent A Eastman
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Katherine J Lobur
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Kexin Jiao
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Jerry Silver
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| | - Evan S Deneris
- Department of NeurosciencesSchool of Medicine, Case Western Reserve UniversityClevelandUnited States
| |
Collapse
|
33
|
Miao Y, Wang N, Shao W, Xu Z, Yang Z, Wang L, Ju C, Zhang R, Zhang F. Overexpression of TIPE2, a Negative Regulator of Innate and Adaptive Immunity, Attenuates Cognitive Deficits in APP/PS1 Mice. J Neuroimmune Pharmacol 2019; 14:519-529. [DOI: 10.1007/s11481-019-09861-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/30/2019] [Indexed: 12/29/2022]
|
34
|
Wang J, Xie R, Kou X, Liu Y, Qi C, Liu R, You W, Gao J, Gao X. A protein phosphatase 2A deficit in the hippocampal CA1 area impairs memory extinction. Mol Brain 2019; 12:51. [PMID: 31113458 PMCID: PMC6528246 DOI: 10.1186/s13041-019-0469-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/30/2019] [Indexed: 01/17/2023] Open
Abstract
Protein phosphorylation plays an important role in learning and memory. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase involved in the regulation of neural synaptic plasticity. Here, to determine if PP2A is necessary for successful learning and memory, we have utilized a Tg (Camk2a-cre) T29–2Stl mice to specific knock down the expression of hippocampal PP2A in mice. By analysing behavioural, we observed that loss of PP2A in the hippocampal CA1 area did not affect the formation of memory but impaired contextual fear memory extinction. We use the electrophysiological recording to find the synaptic mechanisms. The results showed that the basic synapse transmission and synaptic plasticity of PP2A conditional knockout (CKO) mice were impaired. Moreover, PP2A CKO mice exhibited a saturating long-term potentiation inducted by strong theta burst stimulation but no depotentiation after low-frequency stimulation. Taken together, our results provide the evidence that PP2A is involved in synaptic transmission and hippocampus-dependent memory extinction.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ran Xie
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xiaolin Kou
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yu Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Cui Qi
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Rui Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Weiyan You
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jun Gao
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Xiang Gao
- Model Animal Research Center and MOE Key Laboratory of Model Animals for Disease Study, Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
35
|
Waider J, Popp S, Mlinar B, Montalbano A, Bonfiglio F, Aboagye B, Thuy E, Kern R, Thiel C, Araragi N, Svirin E, Schmitt-Böhrer AG, Corradetti R, Lowry CA, Lesch KP. Serotonin Deficiency Increases Context-Dependent Fear Learning Through Modulation of Hippocampal Activity. Front Neurosci 2019; 13:245. [PMID: 31068767 PMCID: PMC6491456 DOI: 10.3389/fnins.2019.00245] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 03/01/2019] [Indexed: 12/21/2022] Open
Abstract
Brain serotonin (5-hydroxytryptamine, 5-HT) system dysfunction is implicated in exaggerated fear responses triggering various anxiety-, stress-, and trauma-related disorders. However, the underlying mechanisms are not well understood. Here, we investigated the impact of constitutively inactivated 5-HT synthesis on context-dependent fear learning and extinction using tryptophan hydroxylase 2 (Tph2) knockout mice. Fear conditioning and context-dependent fear memory extinction paradigms were combined with c-Fos imaging and electrophysiological recordings in the dorsal hippocampus (dHip). Tph2 mutant mice, completely devoid of 5-HT synthesis in brain, displayed accelerated fear memory formation and increased locomotor responses to foot shock. Furthermore, recall of context-dependent fear memory was increased. The behavioral responses were associated with increased c-Fos expression in the dHip and resistance to foot shock-induced impairment of hippocampal long-term potentiation (LTP). In conclusion, increased context-dependent fear memory resulting from brain 5-HT deficiency involves dysfunction of the hippocampal circuitry controlling contextual representation of fear-related behavioral responses.
Collapse
Affiliation(s)
- Jonas Waider
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Sandy Popp
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Boris Mlinar
- Department of Neuroscience, Psychology, Drug Research, and Child Health, University of Florence, Florence, Italy
| | - Alberto Montalbano
- Department of Neuroscience, Psychology, Drug Research, and Child Health, University of Florence, Florence, Italy
| | - Francesco Bonfiglio
- Department of Neuroscience, Psychology, Drug Research, and Child Health, University of Florence, Florence, Italy
| | - Benjamin Aboagye
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Elisabeth Thuy
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Raphael Kern
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Christopher Thiel
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Naozumi Araragi
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Evgeniy Svirin
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Angelika G Schmitt-Böhrer
- Department of Psychiatry, Psychosomatics, and Psychotherapy, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research, and Child Health, University of Florence, Florence, Italy
| | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, United States
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Translational Psychiatry, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
36
|
Park SC, Kim YK. A Novel Bio-Psychosocial-Behavioral Treatment Model of Panic Disorder. Psychiatry Investig 2019; 16:4-15. [PMID: 30301303 PMCID: PMC6354044 DOI: 10.30773/pi.2018.08.21.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/09/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022] Open
Abstract
To conceptualize a novel bio-psychosocial-behavioral treatment model of panic disorder (PD), it is necessary to completely integrate behavioral, psychophysiological, neurobiological, and genetic data. Molecular genetic research on PD is specifically focused on neurotransmitters, including serotonin, neuropeptides, glucocorticoids, and neurotrophins. Although pharmacological interventions for PD are currently available, the need for more effective, faster-acting, and more tolerable pharmacological interventions is unmet. Thus, glutamatergic receptor modulators, orexin receptor antagonists, corticotrophin-releasing factor 1 receptor antagonists, and other novel mechanism-based anti-panic therapeutics have been proposed. Research on the neural correlates of PD is focused on the dysfunctional "cross-talk" between emotional drive (limbic structure) and cognitive inhibition (prefrontal cortex) and the fear circuit, which includes the amygdala-hippocampus-prefrontal axis. The neural perspective regarding PD supports the idea that cognitive-behavioral therapy normalizes alterations in top-down cognitive processing, including increased threat expectancy and attention to threat. Consistent with the concept of "personalized medicine," it is speculated that Research Domain Criteria can enlighten further treatments targeting dysfunctions underlying PD more precisely and provide us with better definitions of moderators used to identify subgroups according to different responses to treatment. Structuring of the "negative valence systems" domain, which includes fear/anxiety, is required to define PD. Therefore, targeting glutamate- and orexin-related molecular mechanisms associated with the fear circuit, which includes the amygdala-hippocampus-prefrontal cortex axis, is required to define a novel bio-psychosocial-behavioral treatment model of PD.
Collapse
Affiliation(s)
- Seon-Cheol Park
- Department of Psychiatry, Inje University College of Medicine and Haeundae Paik Hospital, Busan, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Ansan Hospital, Ansan, Republic of Korea
| |
Collapse
|
37
|
Lin LW, Tsai FS, Yang WT, Lai SC, Shih CC, Lee SC, Wu CR. Differential change in cortical and hippocampal monoamines, and behavioral patterns in streptozotocin-induced type 1 diabetic rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:1026-1034. [PMID: 30524676 PMCID: PMC6281071 DOI: 10.22038/ijbms.2018.29810.7197] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Objectives Diabetes mellitus (DM) is a widespread metabolic disorder worldwide. Clinical physicians have found diabetic patients have mild to middle cognitive dysfunction and an alteration of brain monoaminergic function. This study explored the change in various patterns of behavioral models and brain monoamine function under streptozotocin (STZ)-induced type 1 diabetes. Materials and Methods We established a type 1 DM model via intravenous injection with STZ (65 mg/kg) in rats. Three weeks after the STZ injection, various behavioral measurements including the inhibitory avoidance test, active avoidance test and Morris water maze were conducted. Finally, all rats were dissected and the concentrations of monoamines and their metabolites in cortex and hippocampus were measured by high performance liquid chromatography with electrochemical detection. Results We found that STZ induced type 1 diabetes (hyperglycemia and lack of insulin) in rats. STZ-induced diabetic rats had cognitive impairment in acquisition sessions and long-term retention of the active avoidance test. STZ-induced diabetic rats also had cognitive impairment in spatial learning, reference and working memory of the Morris water maze. STZ significantly reduced concentrations of norepinephrine (NE) in the cortex and dopamine (DA) in the hippocampus, but increased concentrations of DA and serotonin (5-HT) in the cortex 35 days after injection. The concentration of 5-HT in the hippocampus was also significantly increased. Conclusion The data suggested that this cognitive impairment after a short-term period of STZ injection might be related to cortical NE dysfunction, differential alteration of cortical and hippocampal DA function, and brain 5-HT hyperfunction.
Collapse
Affiliation(s)
- Li-Wei Lin
- School of Chinese Medicines for Post-Baccal aureate, I-Shou University, Kaohsiung 82445, Taiwan
| | - Fan-Shiu Tsai
- School of Chinese Medicines for Post-Baccal aureate, I-Shou University, Kaohsiung 82445, Taiwan
| | - Wen-Ta Yang
- Taichung Hospital, Ministry of Health and Welfare, Taichung 402, Taiwan
| | - Shang-Chih Lai
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97071, Taiwan
| | - Chun-Chuan Shih
- School of Chinese Medicines for Post-Baccal aureate, I-Shou University, Kaohsiung 82445, Taiwan
| | - Sheng-Chi Lee
- Pintung Branch, Kaohsiung Veterans General Hospital, Pintung 91245, Taiwan
| | - Chi-Rei Wu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung 402, Taiwan
| |
Collapse
|
38
|
Xi TF, Li DN, Li YY, Qin Y, Wang HH, Song NN, Zhang Q, Ding YQ, Shi XZ, Xie DP. Central 5-hydroxytryptamine (5-HT) mediates colonic motility by hypothalamus oxytocin-colonic oxytocin receptor pathway. Biochem Biophys Res Commun 2018; 508:959-964. [PMID: 30545636 DOI: 10.1016/j.bbrc.2018.11.122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 12/18/2022]
Abstract
Gut-derived 5-hydroxytryptamine (5-HT) is well known for its role in mediating colonic motility function. However, it is not very clear whether brain-derived 5-HT is involved in the regulation of colonic motility. In this study, we used central 5-HT knockout (KO) mice to investigate whether brain-derived 5-HT mediates colonic motility, and if so, whether it involves oxytocin (OT) production in the hypothalamus and OT receptor in the colon. Colon transit time was prolonged in KO mice. The OT levels in the hypothalamus and serum were decreased significantly in the KO mice compared to wild-type (WT) controls. OT increased colonic smooth muscle contraction in both KO and WT mice, and the effects were blocked by OT receptor antagonist and tetrodotoxin but not by hexamethonium or atropine. Importantly, the OT-induced colonic smooth muscle contraction was decreased significantly in the KO mice relative to WT. The OT receptor expression of colon was detected in colonic myenteric plexus of mice. Central 5-HT is involved in the modulation of colonic motility which may modulate through its regulation of OT synthesis in the hypothalamus. Our results reveal a central 5-HT - hypothalamus OT - colonic OT receptor axis, providing a new target for the treatment of brain-gut dysfunction.
Collapse
Affiliation(s)
- Tao-Fang Xi
- Department of Physiology and Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Dan-Ni Li
- Department of Physiology and Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yu-Yian Li
- Department of Physiology and Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ying Qin
- Department of Physiology and Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Hai-Hong Wang
- Department of Physiology and Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ning-Ning Song
- Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiong Zhang
- Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yu-Qiang Ding
- Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xuan-Zheng Shi
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Dong-Ping Xie
- Department of Physiology and Shanghai Pudong New Area Mental Health Center, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
39
|
Microarray Analysis of Gene Expression Changes in Neuroplastin 65-Knockout Mice: Implications for Abnormal Cognition and Emotional Disorders. Neurosci Bull 2018; 34:779-788. [PMID: 29974341 PMCID: PMC6129239 DOI: 10.1007/s12264-018-0251-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 04/26/2018] [Indexed: 02/08/2023] Open
Abstract
Neuroplastin 65 (Np65) is an immunoglobulin superfamily cell adhesion molecule involved in synaptic formation and plasticity. Our recent study showed that Np65-knockout (KO) mice exhibit abnormal cognition and emotional disorders. However, the underlying mechanisms remain unclear. In this study, we found 588 differentially-expressed genes in Np65-KO mice by microarray analysis. RT-PCR analysis also revealed the altered expression of genes associated with development and synaptic structure, such as Cdh1, Htr3a, and Kcnj9. In addition, the expression of Wnt-3, a Wnt protein involved in development, was decreased in Np65-KO mice as evidenced by western blotting. Surprisingly, MRI and DAPI staining showed a significant reduction in the lateral ventricular volume of Np65-KO mice. Together, these findings suggest that ablation of Np65 influences gene expression, which may contribute to abnormal brain development. These results provide clues to the mechanisms underlying the altered brain functions of Np65-deficient mice.
Collapse
|
40
|
Shah R, Courtiol E, Castellanos FX, Teixeira CM. Abnormal Serotonin Levels During Perinatal Development Lead to Behavioral Deficits in Adulthood. Front Behav Neurosci 2018; 12:114. [PMID: 29928194 PMCID: PMC5997829 DOI: 10.3389/fnbeh.2018.00114] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/17/2018] [Indexed: 11/18/2022] Open
Abstract
Serotonin (5-HT) is one of the best-studied modulatory neurotransmitters with ubiquitous presynaptic release and postsynaptic reception. 5-HT has been implicated in a wide variety of brain functions, ranging from autonomic regulation, sensory perception, feeding and motor function to emotional regulation and cognition. The role of this neuromodulator in neuropsychiatric diseases is unquestionable with important neuropsychiatric medications, e.g., most antidepressants, targeting this system. Importantly, 5-HT modulates neurodevelopment and changes in its levels during development can have life-long consequences. In this mini-review, we highlight that exposure to both low and high serotonin levels during the perinatal period can lead to behavioral deficits in adulthood. We focus on three exogenous factors that can change 5-HT levels during the critical perinatal period: dietary tryptophan depletion, exposure to serotonin-selective-reuptake-inhibitors (SSRIs) and poor early life care. We discuss the effects of each of these on behavioral deficits in adulthood.
Collapse
Affiliation(s)
- Relish Shah
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Emmanuelle Courtiol
- CNRS UMR 5292 - INSERM U1028, Lyon Neuroscience Research Center, Université Lyon 1, Lyon, France
| | - Francisco X Castellanos
- Department of Child and Adolescent Psychiatry, Hassenfeld Children's Hospital at NYU Langone, New York, NY, United States.,Division of Clinical Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States
| | - Catia M Teixeira
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States.,Department of Child and Adolescent Psychiatry, Hassenfeld Children's Hospital at NYU Langone, New York, NY, United States
| |
Collapse
|
41
|
The interhemispheric CA1 circuit governs rapid generalisation but not fear memory. Nat Commun 2017; 8:2190. [PMID: 29259187 PMCID: PMC5736595 DOI: 10.1038/s41467-017-02315-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/20/2017] [Indexed: 12/30/2022] Open
Abstract
Encoding specificity theory predicts most effective recall by the original conditions at encoding, while generalization endows recall flexibly under circumstances which deviate from the originals. The CA1 regions have been implicated in memory and generalization but whether and which locally separated mechanisms are involved is not clear. We report here that fear memory is quickly formed, but generalization develops gradually over 24 h. Generalization but not fear memory is impaired by inhibiting ipsilateral (ips) or contralateral (con) CA1, and by optogenetic silencing of the ipsCA1 projections onto conCA1. By contrast, in vivo fEPSP recordings reveal that ipsCA1–conCA1 synaptic efficacy is increased with delay over 24 h when generalization is formed but it is unchanged if generalization is disrupted. Direct excitation of ipsCA1–conCA1 synapses using chemogenetic hM3Dq facilitates generalization formation. Thus, rapid generalization is an active process dependent on bilateral CA1 regions, and encoded by gradual synaptic learning in ipsCA1–conCA1 circuit. Previous work has documented a slow form of memory generalization although a rapid one is demanded. Here the authors elucidate the role of the interhemispheric CA1-CA1 projection in a form of rapid generalization of contextual fear memory via gradual potentiation of these synapses over 24 h.
Collapse
|
42
|
Song NN, Huang Y, Yu X, Lang B, Ding YQ, Zhang L. Divergent Roles of Central Serotonin in Adult Hippocampal Neurogenesis. Front Cell Neurosci 2017; 11:185. [PMID: 28713247 PMCID: PMC5492328 DOI: 10.3389/fncel.2017.00185] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/16/2017] [Indexed: 11/13/2022] Open
Abstract
The central serotonin (5-HT) system is the main target of selective serotonin reuptake inhibitors (SSRIs), the first-line antidepressants widely used in current general practice. One of the prominent features of chronic SSRI treatment in rodents is the enhanced adult neurogenesis in the hippocampus, which has been proposed to contribute to antidepressant effects. Therefore, tremendous effort has been made to decipher how central 5-HT regulates adult hippocampal neurogenesis. In this paper, we review how changes in the central serotonergic system alter adult hippocampal neurogenesis. We focus on data obtained from three categories of genetically engineered mouse models: (1) mice with altered central 5-HT levels from embryonic stages, (2) mice with deletion of 5-HT receptors from embryonic stages, and (3) mice with altered central 5-HT system exclusively in adulthood. These recent findings provide unique insights to interpret the multifaceted roles of central 5-HT on adult hippocampal neurogenesis and its associated effects on depression.
Collapse
Affiliation(s)
- Ning-Ning Song
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Ying Huang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Xin Yu
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Bing Lang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China.,Mental Health Institute of the Second Xiangya Hospital, National Clinical Research Center on Mental Disorders, National Technology Institute on Mental Disorders, Key Laboratory of Psychiatry and Mental Health of Hunan Province, Central South UniversityChangsha, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, Tongji University School of MedicineShanghai, China.,Department of Anatomy and Neurobiology, Tongji University School of MedicineShanghai, China
| |
Collapse
|
43
|
Enhanced dendritic morphogenesis of adult hippocampal newborn neurons in central 5-HT-deficient mice. Stem Cell Res 2017; 19:6-11. [DOI: 10.1016/j.scr.2016.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 11/24/2016] [Accepted: 12/09/2016] [Indexed: 11/20/2022] Open
|
44
|
Fernandez SP, Muzerelle A, Scotto-Lomassese S, Barik J, Gruart A, Delgado-García JM, Gaspar P. Constitutive and Acquired Serotonin Deficiency Alters Memory and Hippocampal Synaptic Plasticity. Neuropsychopharmacology 2017; 42:512-523. [PMID: 27461084 PMCID: PMC5399229 DOI: 10.1038/npp.2016.134] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/07/2016] [Accepted: 07/17/2016] [Indexed: 02/06/2023]
Abstract
Serotonin (5-HT) deficiency occurs in a number of brain disorders that affect cognitive function. However, a direct causal relationship between 5-HT hypo-transmission and memory and underlying mechanisms has not been established. We used mice with a constitutive depletion of 5-HT brain levels (Pet1KO mice) to analyze the contribution of 5-HT to different forms of learning and memory. Pet1KO mice exhibited a striking deficit in novel object recognition memory, a hippocampal-dependent task. No alterations were found in tasks for social recognition, procedural learning, or fear memory. Viral delivery of designer receptors exclusively activated by designer drugs was used to selectively silence the activity of 5-HT neurons in the raphe. Inhibition of 5-HT neurons in the median raphe, but not the dorsal raphe, was sufficient to impair object recognition in adult mice. In vivo electrophysiology in behaving mice showed that long-term potentiation in the hippocampus of 5-HT-deficient mice was altered, and administration of the 5-HT1A agonist 8-OHDPAT rescued the memory deficits. Our data suggest that hyposerotonergia selectively affects declarative hippocampal-dependent memory. Serotonergic projections from the median raphe are necessary to regulate object memory and hippocampal synaptic plasticity processes, through an inhibitory control mediated by 5-HT1A receptors.
Collapse
Affiliation(s)
- Sebastian P Fernandez
- Institut du Fer à Moulin, Paris, France,INSERM, UMRS-839, Paris, France,University Pierre and Marie Curie, Paris, France,Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Université Côte d'Azur, CNRS UMR 7275, Valbonne, 0656, France, Tel: +33 4 93 95 34 41, Fax: +33 4 93 95 34 08, E-mail:
| | - Aude Muzerelle
- Institut du Fer à Moulin, Paris, France,INSERM, UMRS-839, Paris, France,University Pierre and Marie Curie, Paris, France
| | - Sophie Scotto-Lomassese
- Institut du Fer à Moulin, Paris, France,INSERM, UMRS-839, Paris, France,University Pierre and Marie Curie, Paris, France
| | - Jacques Barik
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, Seville, Spain
| | | | - Patricia Gaspar
- Institut du Fer à Moulin, Paris, France,INSERM, UMRS-839, Paris, France,University Pierre and Marie Curie, Paris, France
| |
Collapse
|
45
|
5-HT7 receptor-mediated fear conditioning and possible involvement of extracellular signal-regulated kinase. Neurosci Lett 2017; 638:69-75. [DOI: 10.1016/j.neulet.2016.11.065] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/25/2016] [Accepted: 11/29/2016] [Indexed: 10/20/2022]
|
46
|
Ambrogini P, Betti M, Galati C, Di Palma M, Lattanzi D, Savelli D, Galli F, Cuppini R, Minelli A. α-Tocopherol and Hippocampal Neural Plasticity in Physiological and Pathological Conditions. Int J Mol Sci 2016; 17:E2107. [PMID: 27983697 PMCID: PMC5187907 DOI: 10.3390/ijms17122107] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/01/2016] [Accepted: 12/09/2016] [Indexed: 12/25/2022] Open
Abstract
Neuroplasticity is an "umbrella term" referring to the complex, multifaceted physiological processes that mediate the ongoing structural and functional modifications occurring, at various time- and size-scales, in the ever-changing immature and adult brain, and that represent the basis for fundamental neurocognitive behavioral functions; in addition, maladaptive neuroplasticity plays a role in the pathophysiology of neuropsychiatric dysfunctions. Experiential cues and several endogenous and exogenous factors can regulate neuroplasticity; among these, vitamin E, and in particular α-tocopherol (α-T), the isoform with highest bioactivity, exerts potent effects on many plasticity-related events in both the physiological and pathological brain. In this review, the role of vitamin E/α-T in regulating diverse aspects of neuroplasticity is analyzed and discussed, focusing on the hippocampus, a brain structure that remains highly plastic throughout the lifespan and is involved in cognitive functions. Vitamin E-mediated influences on hippocampal synaptic plasticity and related cognitive behavior, on post-natal development and adult hippocampal neurogenesis, as well as on cellular and molecular disruptions in kainate-induced temporal seizures are described. Besides underscoring the relevance of its antioxidant properties, non-antioxidant functions of vitamin E/α-T, mainly involving regulation of cell signaling molecules and their target proteins, have been highlighted to help interpret the possible mechanisms underlying the effects on neuroplasticity.
Collapse
Affiliation(s)
- Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Michele Betti
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Claudia Galati
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Michael Di Palma
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Davide Lattanzi
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - David Savelli
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy.
| | - Riccardo Cuppini
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| | - Andrea Minelli
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy.
| |
Collapse
|
47
|
Riva G. Neurobiology of Anorexia Nervosa: Serotonin Dysfunctions Link Self-Starvation with Body Image Disturbances through an Impaired Body Memory. Front Hum Neurosci 2016; 10:600. [PMID: 27932968 PMCID: PMC5121233 DOI: 10.3389/fnhum.2016.00600] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/10/2016] [Indexed: 12/21/2022] Open
Abstract
The etiology of anorexia nervosa (AN) is still unclear, despite that it is a critical and potentially mortal illness. A recent neurobiological model considers AN as the outcome of dysfunctions in the neuronal processes related to appetite and emotionality (Kaye et al., 2009, 2013). However, this model still is not able to answer a critical question: What is behind body image disturbances (BIDs) in AN? The article starts its analysis from reviewing some of the studies exploring the effects of the serotonin systems in memory (episodic, working, and spatial) and its dysfunctions. The review suggests that serotonin disturbances may: (a) facilitate the encoding of third person (allocentric) episodic memories; (b) facilitate the consolidation of emotional episodic memories (e.g., teasing), if preceded by repeated stress; (c) reduce voluntary inhibition of mnestic contents; (d) impair allocentric spatial memory. If we discuss these results within the interpretative frame suggested by the “Allocentric Lock Hypothesis” (Riva, 2012, 2014), we can hypothesize that altered serotoninergic activity in AN patients: (i) improves their ability to store and consolidate negative autobiographical memories, including those of their body, in allocentric perspective; (ii) impairs their ability to trigger voluntary inhibition of the previously stored negative memory of the body; (iii) impairs their capacity to retrieve/update allocentric information. Taken together, these points suggest a possible link between serotonin dysfunctions, memory impairments and BIDs: the impossibility of updating a disturbed body memory using real time experiential data—I'm locked to a wrong body stored in long term memory—pushes AN patients to control body weight and shape even when underweight.
Collapse
Affiliation(s)
- Giuseppe Riva
- Applied Technology for Neuro-Psychology Lab, Istituto Auxologico ItalianoMilan, Italy; Centro Studi e Ricerche di Psicologia della Comunicazione, Università Cattolica del Sacro CuoreMilano, Italy
| |
Collapse
|
48
|
Niu XY, Huang HJ, Zhang JB, Zhang C, Chen WG, Sun CY, Ding YQ, Liao M. Deletion of autophagy-related gene 7 in dopaminergic neurons prevents their loss induced by MPTP. Neuroscience 2016; 339:22-31. [PMID: 27693472 DOI: 10.1016/j.neuroscience.2016.09.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 09/21/2016] [Accepted: 09/21/2016] [Indexed: 10/20/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease caused by a gradual loss of midbrain dopaminergic (mDA) neurons in the substantia nigra pars compacta (SNpc) during aging. 1-Methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) is one of the neurotoxins used widely to induce PD-like symptoms in PD animal models, including rodents and non-human primates. It has been reported that deletion of autophagy-related gene 7 (Atg7) in the brain results in a reduction of mDA neurons in adulthood. In this study, we used tyrosine hydroxylase (TH)-Cre mice to generate conditional knockout (CKO) mice with the specific deletion of Atg7 in mDA neurons. Consistent with previous reports, adult Atg7 CKO mice contained fewer TH-positive mDA neurons compared with wild-type (WT) controls. TH-expressing neurons containing puncta-like structures with p62 and ubiquitin immunoreactivity were observed in the midbrain of Atg7 CKO mice but were not detected in control mice. However, MPTP-induced loss of mDA neurons was not observed in Atg7 CKO mice. Our results indicate that Atg7-involved autophagy is required not only for the survival of mDA neurons in the mouse brain, but also for MPTP-induced mDA neuron degeneration.
Collapse
Affiliation(s)
- Xue-Yuan Niu
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hou-Ju Huang
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jin-Bao Zhang
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chan Zhang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wei-Guang Chen
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chen-You Sun
- Department of Human Anatomy, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yu-Qiang Ding
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Department of Anatomy and Neurobiology, and Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai 200092, China.
| | - Min Liao
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
49
|
Mlinar B, Montalbano A, Piszczek L, Gross C, Corradetti R. Firing Properties of Genetically Identified Dorsal Raphe Serotonergic Neurons in Brain Slices. Front Cell Neurosci 2016; 10:195. [PMID: 27536220 PMCID: PMC4971071 DOI: 10.3389/fncel.2016.00195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/22/2016] [Indexed: 11/13/2022] Open
Abstract
Tonic spiking of serotonergic neurons establishes serotonin levels in the brain. Since the first observations, slow regular spiking has been considered as a defining feature of serotonergic neurons. Recent studies, however, have revealed the heterogeneity of serotonergic neurons at multiple levels, comprising their electrophysiological properties, suggesting the existence of functionally distinct cellular subpopulations. In order to examine in an unbiased manner whether serotonergic neurons of the dorsal raphe nucleus (DRN) are heterogeneous, we used a non-invasive loose-seal cell-attached method to record α1 adrenergic receptor-stimulated spiking of a large sample of neurons in brain slices obtained from transgenic mice lines that express fluorescent marker proteins under the control of serotonergic system-specific Tph2 and Pet-1 promoters. We found wide homogeneous distribution of firing rates, well fitted by a single Gaussian function (r (2) = 0.93) and independent of anatomical location (P = 0.45), suggesting that in terms of intrinsic firing properties, serotonergic neurons in the DRN represent a single cellular population. Characterization of the population in terms of spiking regularity was hindered by its dependence on the firing rate. For instance, the coefficient of variation of the interspike intervals (ISI), a common measure of spiking irregularity, is of limited usefulness since it correlates negatively with the firing rate (r = -0.33, P < 0.0001). Nevertheless, the majority of neurons exhibited regular, pacemaker-like activity, with coefficient of variance of the ISI lower than 0.5 in ~97% of cases. Unexpectedly, a small percentage of neurons (~1%) exhibited a particular spiking pattern, characterized by low frequency (~0.02-0.1 Hz) oscillations in the firing rate. Transitions between regular and oscillatory firing were observed, suggesting that the oscillatory firing is an alternative firing pattern of serotonergic neurons.
Collapse
Affiliation(s)
- Boris Mlinar
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| | - Alberto Montalbano
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| | - Lukasz Piszczek
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Cornelius Gross
- Mouse Biology Unit, European Molecular Biology Laboratory Monterotondo, Italy
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Children's Health, University of Florence Florence, Italy
| |
Collapse
|
50
|
Pettersson R, Hagsäter SM, Eriksson E. Serotonin depletion eliminates sex differences with respect to context-conditioned immobility in rat. Psychopharmacology (Berl) 2016; 233:1513-21. [PMID: 26905688 DOI: 10.1007/s00213-016-4246-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE Previous studies have shown that male rats display more anxiety-like behavior than females as assessed using the elevated plus maze and that serotonin depletion abolishes this difference by exerting an anxiolytic-like effect in males only. OBJECTIVES To compare male and female rats with respect to immobility and startle responses to sudden noise bursts after contextual fear conditioning and to explore to what extent any possible sex difference in this regard is influenced by serotonin depletion during testing (but not acquisition). RESULTS In line with previous studies, males displayed more immobility following contextual conditioning induced by previous exposure to foot shocks than females. In males but not females, the immobility response was reduced by administration of the serotonin synthesis inhibitor para-chlorophenylalanine (PCPA) between shock exposure and testing, the consequence being that males and females no longer differed in this regard. Untreated males but not females displayed a negative correlation between fear-conditioned startle and immobility, suggesting that the latter behavior, when excessive, interferes with the former. In line with this assumption, the reduction in immobility following administration of PCPA in males coincided with an increase in startle that was not observed in females, hence revealing a sex difference in startle not seen in untreated controls. CONCLUSION The greater display of context-conditioned immobility in males compared with females appears to be serotonin-dependent.
Collapse
Affiliation(s)
- Robert Pettersson
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 431, SE-405 30, Gothenburg, Sweden
| | - Sven Melker Hagsäter
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 431, SE-405 30, Gothenburg, Sweden
| | - Elias Eriksson
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 431, SE-405 30, Gothenburg, Sweden.
| |
Collapse
|