1
|
Shu Y, Tian L, Wang X, Meng T, Yu S, Li Y. Decoding serotonin: the molecular symphony behind depression. Front Cell Neurosci 2025; 19:1572462. [PMID: 40342516 PMCID: PMC12058683 DOI: 10.3389/fncel.2025.1572462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/03/2025] [Indexed: 05/11/2025] Open
Abstract
The serotonin (5-hydroxytryptamine) system represents a crucial neurotransmitter network that regulates mood, behavior, and cognitive functions, playing a significant role in the pathogenesis and progression of depression. Although this perspective faces significant challenges, the serotonin system continues to exert substantial modulatory effects on specific aspects of psychological functioning and actively contributes to multiple pathological processes in depression development. Therefore, this review systematically integrates interdisciplinary research advances regarding the relationship between the 5-hydroxytryptamine (5-HT) system and depression. By focusing on core biological processes including serotonin biosynthesis and metabolism, SERT gene regulatory networks, and protein molecular modifications, it aims to elucidate how 5-HT system dysregulation contributes to the development of depression, while providing novel research perspectives and therapeutic targets for innovative antidepressant drug development.
Collapse
Affiliation(s)
- Yue Shu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xing Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Tinyang Meng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Shouyang Yu
- Key Laboratory of Brain Science, Key Laboratory of Anesthesia and Organ Protection of Ministry of Education (In Cultivation), Zunyi Medical University, Zunyi, China
| | - Yulan Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| |
Collapse
|
2
|
Lian M, Li H, Zhang Z, Fang J, Liu X. Gene-level connections between anxiety disorders, ADHD, and head and neck cancer: insights from a computational biology approach. Front Psychiatry 2025; 16:1552815. [PMID: 40182194 PMCID: PMC11967369 DOI: 10.3389/fpsyt.2025.1552815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Background Anxiety disorders (AD), ADHD, and head and neck cancer (HNC) are complex conditions with potential genetic interconnections that remain to be fully elucidated. The purpose of this study is to investigate gene-level connections among ADHD, AD, and HNC. Method A comprehensive literature mining approach identified potential gene-disease relationships from PubMed and bioinformatics databases, analyzing 19,924 genes. An AI-driven computational process constructed a gene-disease relationship table using the Adjusted Binomial Method Algorithm (ABMA) to evaluate association reliability. Overlapping genes were analyzed through protein-protein interaction (PPI) networks, functional annotations, and literature-based pathway analyses to elucidate shared and unique genetic mechanisms linking these diseases. Results The analysis identified 141 significant genes associated with AD, 153 with ADHD, and 1,065 with HNC (q-value < 0.05). These genes demonstrated significant overlap (odds ratio ≥ 1.8; p ≤ 2.58E-2) and high interconnectivity (PPI network density ≥ 0.39, clustering coefficient ≥ 0.76, and diameter ≤ 3). Centrality analysis revealed core genes such as IL-6, MYC, NLRP3, and CXCR4 as critical mediators. Functional enrichment analysis identified key pathways, including serotonergic synapse, inflammatory response, and Toll-like receptor signaling, highlighting the involvement of neuronal and immune mechanisms. Functional pathway analysis demonstrated reciprocal genetic influences among AD, ADHD, and HNC, emphasizing shared and distinct gene-level connections that may underlie their co-occurrence and mutual risk factors. Conclusion This study reveals a complex and interconnected genetic network among AD, ADHD, and HNC, highlighting shared pathways, unique mechanisms, and critical genes, providing valuable insights into the genetic underpinnings of these conditions and potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Meng Lian
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China
| | - Haiyang Li
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China
| | - Zhiyang Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders & Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Jugao Fang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China
| | - Xiaoqin Liu
- Department of Otolaryngology, The Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
TIAN T, XU X, SONG J, ZHANG X, ZHANG D, YUAN H, ZHONG F, LI J, HU Y. Learning and Memory Impairments With Attention-Deficit/Hyperactivity Disorder. Physiol Res 2024; 73:205-216. [PMID: 38710050 PMCID: PMC11081185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/02/2023] [Indexed: 05/08/2024] Open
Abstract
ADHD is a common chronic neurodevelopmental disorder and is characterized by persistent inattention, hyperactivity, impulsivity and are often accompanied by learning and memory impairment. Great evidence has shown that learning and memory impairment of ADHD plays an important role in its executive function deficits, which seriously affects the development of academic, cognitive and daily social skills and will cause a serious burden on families and society. With the increasing attention paid to learning and memory impairment in ADHD, relevant research is gradually increasing. In this article, we will present the current research results of learning and memory impairment in ADHD from the following aspects. Firstly, the animal models of ADHD, which display the core symptoms of ADHD as well as with learning and memory impairment. Secondly, the molecular mechanism of has explored, including some neurotransmitters, receptors, RNAs, etc. Thirdly, the susceptibility gene of ADHD related to the learning and impairment in order to have a more comprehensive understanding of the pathogenesis. Key words: Learning and memory, ADHD, Review.
Collapse
Affiliation(s)
- Tian TIAN
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Xu XU
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Jia SONG
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Xiaoqian ZHANG
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Dan ZHANG
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Hui YUAN
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Fengyu ZHONG
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Jing LI
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Youfang HU
- Department of Children’s Health Care, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Maternal and Child Health Care Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Chen F, Zhang L, Liu Y, Zhang A, Wang W. Resveratrol alleviates perinatal methylmercury-induced neurobehavioral impairments by modulating the gut microbiota composition and neurotransmitter disturbances. ENVIRONMENTAL TOXICOLOGY 2024; 39:329-340. [PMID: 37713589 DOI: 10.1002/tox.23973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/09/2023] [Accepted: 09/01/2023] [Indexed: 09/17/2023]
Abstract
Methylmercury (MeHg), a potent neurotoxic substance, causes adverse health outcomes by modulating metabolites through altered gut microbiota patterns. Among the many metabolites, neurotransmitters play a particularly important role in the nervous system and behavior. Resveratrol (RSV) has been investigated as an antiaging, antioxidant, anti-inflammatory, and neuroprotective agent. The current study evaluated that RSV is protective of neurodevelopmental toxicity induced by MeHg and further explored the underlying mechanisms. Sprague-Dawley rats were treated with 1.2 mg/kg/d of MeHg, and the effects were evaluated after supplementation with RSV (20 mg/kg/d). The results indicated that MeHg had adverse effects on early neurodevelopmental indicators in the experimental group offspring as compared to control pups. Interestingly, RSV significantly improved the MeHg-induced delays in the neurobehavioral reflexes and reduced the total mercury (THg) concentration in the colons of the offspring rats. In agreement, RSV administration improved the gut microbiota diversity and structure by increasing the abundance of probiotics and upregulating the expression of tight junction proteins. It also ameliorated the MeHg-induced abnormalities in the expression profiles of neurotransmitters. Furthermore, eight key bacteria that were strongly linked with the neurotransmitters and neuroreflex parameters were identified. Taken together, these results demonstrate that RSV treatment effectively reduces the occurrence of neurodevelopmental toxicity caused by perinatal MeHg exposure by modulating the intestinal flora and neurotransmitter metabolism. These findings provide a new therapeutic approach for treating MeHg-induced neurotoxicity. The cover image is based on the Research Article Resveratrol alleviates perinatal methylmercury-induced neurobehavioral impairments by modulating the gut microbiota composition and neurotransmitter disturbances by Fang Chen et al., https://doi.org/10.1002/tox.23973.
Collapse
Affiliation(s)
- Fang Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Li Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yi Liu
- Guiyang Maternal and Child Health Care Hospital, Guiyang, China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, China
| |
Collapse
|
5
|
Higuchi Y, Tada T, Nakachi T, Arakawa H. Serotonergic circuit dysregulation underlying autism-related phenotypes in BTBR mouse model of autism. Neuropharmacology 2023:109634. [PMID: 37301467 DOI: 10.1016/j.neuropharm.2023.109634] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/12/2023]
Abstract
The inbred mouse strain, BTBR T+Itpr3tf/J (BTBR), possesses neuronal and circuit abnormalities that underlie atypical behavioral profiles resembling the major symptoms of human autism spectrum disorder (ASD). Forebrain serotonin (5-HT) transmission has been implicated in ASD-related behavioral alterations. In this study, we assessed 5-HT signals and the functional responsiveness in BTBR mice compared to standard C57BL/6J (B6) control mice to elucidate how 5-HT alterations contribute to behavioral abnormalities in BTBR mice. A lower number of 5-HT neurons in the median raphe, but not in the dorsal raphe, was observed in male and female BTBR mice. Acute systemic injection of buspirone, a 5-HT1A receptor agonist, induced c-Fos in several brain regions in both B6 and BTBR mice; however, blunted c-Fos induction in BTBR mice was documented in the cingulate cortex, basolateral amygdala (BLA), and ventral hippocampus (Hipp). Decreased c-Fos responses in these regions are associated with a lack of buspirone effects on anxiety-like behavior in BTBR mice. Analysis of mRNA expression following acute buspirone injection indicated that 5HTR1a gene downregulation (or upregulation) occurred in the BLA and Hipp of B6 mice, respectively, but not BTBR mice. The mRNA expression of factors associated with neurogenesis or the pro-inflammatory state was not consistently altered by acute buspirone injection. Therefore, 5-HT responsivity via 5-HT1A receptors in the BLA and Hipp are linked to anxiety-like behavior, in which circuits are disrupted in BTBR mice. Other distinct 5-HT circuits from the BLA and Hipp that regulate social behavior are restricted but preserved in BTBR mice.
Collapse
Affiliation(s)
- Yuki Higuchi
- Dept. Systems Physiology, Graduate School of Medicine, University of the Ryukyus, Japan
| | - Tomoaki Tada
- Dept. Systems Physiology, Faculty of Medicine, University of the Ryukyus, Japan
| | - Taiga Nakachi
- Dept. Systems Physiology, Faculty of Medicine, University of the Ryukyus, Japan
| | - Hiroyuki Arakawa
- Dept. Systems Physiology, Graduate School of Medicine, University of the Ryukyus, Japan.
| |
Collapse
|
6
|
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] [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
|
7
|
Kameneva P, Melnikova VI, Kastriti ME, Kurtova A, Kryukov E, Murtazina A, Faure L, Poverennaya I, Artemov AV, Kalinina TS, Kudryashov NV, Bader M, Skoda J, Chlapek P, Curylova L, Sourada L, Neradil J, Tesarova M, Pasqualetti M, Gaspar P, Yakushov VD, Sheftel BI, Zikmund T, Kaiser J, Fried K, Alenina N, Voronezhskaya EE, Adameyko I. Serotonin limits generation of chromaffin cells during adrenal organ development. Nat Commun 2022; 13:2901. [PMID: 35614045 PMCID: PMC9133002 DOI: 10.1038/s41467-022-30438-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 04/23/2022] [Indexed: 11/12/2022] Open
Abstract
Adrenal glands are the major organs releasing catecholamines and regulating our stress response. The mechanisms balancing generation of adrenergic chromaffin cells and protecting against neuroblastoma tumors are still enigmatic. Here we revealed that serotonin (5HT) controls the numbers of chromaffin cells by acting upon their immediate progenitor "bridge" cells via 5-hydroxytryptamine receptor 3A (HTR3A), and the aggressive HTR3Ahigh human neuroblastoma cell lines reduce proliferation in response to HTR3A-specific agonists. In embryos (in vivo), the physiological increase of 5HT caused a prolongation of the cell cycle in "bridge" progenitors leading to a smaller chromaffin population and changing the balance of hormones and behavioral patterns in adulthood. These behavioral effects and smaller adrenals were mirrored in the progeny of pregnant female mice subjected to experimental stress, suggesting a maternal-fetal link that controls developmental adaptations. Finally, these results corresponded to a size-distribution of adrenals found in wild rodents with different coping strategies.
Collapse
Affiliation(s)
- Polina Kameneva
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Victoria I Melnikova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Maria Eleni Kastriti
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Anastasia Kurtova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Emil Kryukov
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Aliia Murtazina
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Irina Poverennaya
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Artem V Artemov
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- National Medical Research Center for Endocrinology, Moscow, Russia
| | - Tatiana S Kalinina
- Federal state budgetary institution "Research Zakusov Institute of Pharmacology" (FSBI "Zakusov Institute of Pharmacology"), Russian Academy of Sciences, Moscow, Russia
| | - Nikita V Kudryashov
- Federal state budgetary institution "Research Zakusov Institute of Pharmacology" (FSBI "Zakusov Institute of Pharmacology"), Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
- Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
- Institute for Biology, University of Lübeck, 23562, Lübeck, Germany
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Petr Chlapek
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Lucie Curylova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Lukas Sourada
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jakub Neradil
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
- Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy
| | | | - Vasily D Yakushov
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Boris I Sheftel
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Elena E Voronezhskaya
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria.
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
8
|
Vorhees CV, Williams MT. Issues in the design, analysis, and application of rodent developmental neurotoxicology studies. Neurotoxicol Teratol 2021; 87:107018. [PMID: 34256163 PMCID: PMC8440477 DOI: 10.1016/j.ntt.2021.107018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 12/16/2022]
Abstract
Developmental neurotoxicity (DNT) studies could benefit from revisions to study design, data analysis, and some behavioral test methods to enhance reproducibility. The Environmental Protection Agency (EPA) reviewed 69 studies submitted to the Office of Pesticide Programs. Two of the behavioral tests identified the lowest observable adverse effect level (LOAEL) 20 and 13 times, respectively, while the other two tests identified the LOAEL only 3 and 4 times, respectively. The EPA review showed that the functional observational battery (FOB) was least effective at detecting the LOAEL, whereas tests of learning and memory (L&M) had methodological shortcomings. Human neurodevelopmental toxicity studies over the past 30 years show that most of the adverse effects are on higher cognitive functions such as L&M. The results of human studies together with structure-function relationships from neuroscience, suggest that tests of working memory, spatial navigation/memory, and egocentric navigation/memory should be added to guideline studies. Collectively, the above suggest that EPA and EU DNT studies would better reflect human findings and be more relevant to children by aligning L&M tests to the same domains that are affected in children, removing less useful methods (FOB), and using newer statistical models to better account for random factors of litter and litter × sex. Common issues in study design and data analyses are discussed: sample size, random group assignment, blinding, elimination of subjective rating methods, avoiding confirmation bias, more complete reporting of species, housing, test protocols, age, test order, and litter effects. Litter in DNT studies should at least be included as a random factor in ANOVA models and may benefit from inclusion of litter × sex as random factors.
Collapse
Affiliation(s)
- Charles V Vorhees
- Department of Pediatrics, College of Medicine, University of Cincinnati and Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States of America.
| | - Michael T Williams
- Department of Pediatrics, College of Medicine, University of Cincinnati and Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States of America
| |
Collapse
|
9
|
Schaefer TL, Ashworth AA, Tiwari D, Tomasek MP, Parkins EV, White AR, Snider A, Davenport MH, Grainger LM, Becker RA, Robinson CK, Mukherjee R, Williams MT, Gibson JR, Huber KM, Gross C, Erickson CA. GABA A Alpha 2,3 Modulation Improves Select Phenotypes in a Mouse Model of Fragile X Syndrome. Front Psychiatry 2021; 12:678090. [PMID: 34093287 PMCID: PMC8175776 DOI: 10.3389/fpsyt.2021.678090] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common cause of inherited intellectual disability. FXS is caused by functional loss of the Fragile X Protein (FXP), also known as Fragile X Mental Retardation Protein (FMRP). In humans and animal models, loss of FXP leads to sensory hypersensitivity, increased susceptibility to seizures and cortical hyperactivity. Several components of the GABAergic system, the major inhibitory system in the brain, are dysregulated in FXS, and thus modulation of GABAergic transmission was suggested and tested as a treatment strategy. However, so far, clinical trials using broad spectrum GABAA or GABAB receptor-specific agonists have not yielded broad improvement of FXS phenotypes in humans. Here, we tested a more selective strategy in Fmr1 knockout (KO) mice using the experimental drug BAER-101, which is a selective GABAA α2/α3 agonist. Our results suggest that BAER-101 reduces hyperexcitability of cortical circuits, partially corrects increased frequency-specific baseline cortical EEG power, reduces susceptibility to audiogenic seizures and improves novel object memory. Other Fmr1 KO-specific phenotypes were not improved by the drug, such as increased hippocampal dendritic spine density, open field activity and marble burying. Overall, this work shows that BAER-101 improves select phenotypes in Fmr1 KO mice and encourages further studies into the efficacy of GABAA-receptor subunit-selective agonists for the treatment of FXS.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Amy A Ashworth
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Durgesh Tiwari
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Madison P Tomasek
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Emma V Parkins
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Angela R White
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Andrew Snider
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Matthew H Davenport
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Lindsay M Grainger
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Robert A Becker
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Chandler K Robinson
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Rishav Mukherjee
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Michael T Williams
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Jay R Gibson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kimberly M Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Craig A Erickson
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
10
|
Tiwari D, Schaefer TL, Schroeder-Carter LM, Krzeski JC, Bunk AT, Parkins EV, Snider A, Danzer R, Williams MT, Vorhees CV, Danzer SC, Gross C. The potassium channel Kv4.2 regulates dendritic spine morphology, electroencephalographic characteristics and seizure susceptibility in mice. Exp Neurol 2020; 334:113437. [PMID: 32822706 PMCID: PMC7642025 DOI: 10.1016/j.expneurol.2020.113437] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 01/21/2023]
Abstract
The voltage-gated potassium channel Kv4.2 is a critical regulator of dendritic excitability in the hippocampus and is crucial for dendritic signal integration. Kv4.2 mRNA and protein expression as well as function are reduced in several genetic and pharmacologically induced rodent models of epilepsy and autism. It is not known, however, whether reduced Kv4.2 is just an epiphenomenon or a disease-contributing cause of neuronal hyperexcitability and behavioral impairments in these neurological disorders. To address this question, we used male and female mice heterozygous for a Kv.2 deletion and adult-onset manipulation of hippocampal Kv4.2 expression in male mice to assess the role of Kv4.2 in regulating neuronal network excitability, morphology and anxiety-related behaviors. We observed a reduction in dendritic spine density and reduced proportions of thin and stubby spines but no changes in anxiety, overall activity, or retention of conditioned freezing memory in Kv4.2 heterozygous mice compared with wildtype littermates. Using EEG analyses, we showed elevated theta power and increased spike frequency in Kv4.2 heterozygous mice under basal conditions. In addition, the latency to onset of kainic acid-induced seizures was significantly shortened in Kv4.2 heterozygous mice compared with wildtype littermates, which was accompanied by a significant increase in theta power. By contrast, overexpressing Kv4.2 in wildtype mice through intrahippocampal injection of Kv4.2-expressing lentivirus delayed seizure onset and reduced EEG power. These results suggest that Kv4.2 is an important regulator of neuronal network excitability and dendritic spine morphology, but not anxiety-related behaviors. In the future, manipulation of Kv4.2 expression could be used to alter seizure susceptibility in epilepsy.
Collapse
Affiliation(s)
- Durgesh Tiwari
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Tori L Schaefer
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | - Joseph C Krzeski
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alexander T Bunk
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Emma V Parkins
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew Snider
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Reese Danzer
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael T Williams
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Charles V Vorhees
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Steve C Danzer
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
11
|
Zhang SR, Wu JL, Chen H, Luo R, Chen WJ, Tang LJ, Li XW, Yang JM, Gao TM. ErbB4 knockdown in serotonergic neurons in the dorsal raphe induces anxiety-like behaviors. Neuropsychopharmacology 2020; 45:1698-1706. [PMID: 31905370 PMCID: PMC7419508 DOI: 10.1038/s41386-020-0601-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022]
Abstract
There is a close relationship between serotonergic (5-HT) activity and anxiety. ErbB4, a receptor tyrosine kinase, is expressed in 5-HT neurons. However, whether ErbB4 regulates 5-HT neuronal function and anxiety-related behaviors is unclear. Here, using transgenic and viral approaches, we show that mice with ErbB4 deficiency in 5-HT neurons exhibit heightened anxiety-like behavior and impaired fear extinction, possibly due to an increased excitability of 5-HT neurons in the dorsal raphe nucleus (DRN). Notably, the chemogenetic inhibition of 5-HT neurons in the DRN of ErbB4 mutant mice rescues anxiety-like behaviors. Altogether, our results unravel a previously unknown role of ErbB4 signaling in the regulation of DRN 5-HT neuronal function and anxiety-like behaviors, providing novel insights into the treatment of anxiety disorders.
Collapse
Affiliation(s)
- Sheng-Rong Zhang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Lin Wu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hao Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rong Luo
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Jun Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li-Juan Tang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Collaborative Innovation Center for Brain Science, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
12
|
Abdulla ZI, Pennington JL, Gutierrez A, Skelton MR. Creatine transporter knockout mice (Slc6a8) show increases in serotonin-related proteins and are resilient to learned helplessness. Behav Brain Res 2019; 377:112254. [PMID: 31542396 DOI: 10.1016/j.bbr.2019.112254] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
Abstract
Approximately 20% of adults in the U.S. will experience an affective disorder during their life. While it is well established that serotonin (5-HT) is a crucial factor in mood, impaired cellular bioenergetics are also implicated. Creatine (Cr), through the Cr/Phospho-Cr (PCr) shuttle, maintains high ATP concentrations in the neuron. This system may be implicated in the etiology of affective disorders, as reduced Cr, PCr, and ATP are often seen in the brains of affected patients. To address this issue, Cr transporter (Crt) deficient male mice (Slc6a8-/y) and female mice heterozygous for Crt expression (Slc6a8+/-) were used to evaluate how a Cr deficient system would alter affective-like behaviors. Slc6a8-/y and Slc6a8+/- mice had faster escape latencies in learned helplessness, indicating a potential resilience to behavioral despair. Slc6a8-/y had decrease latency to immobility in the tail-suspension test and Slc6a8+/- had increased open entries in elevated zero maze, but all other variables matched those of wildtype mice, however. Slc6a8-/y mice have increased 5-hydroxyindoleacetic acid content in the hippocampus and striatum and increased monoamine oxidase protein and tryptophan hydroxylase-2 protein content in the hippocampus, while 5-HT levels are unchanged. This indicates an alteration to the 5-HTergic system in Cr deficient mice. Our results indicate that Cr plays a complex role in affective disorders and 5-HT, warranting further investigation.
Collapse
Affiliation(s)
- Zuhair I Abdulla
- Department of Pediatrics, University of Cincinnati College of Medicine, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Jordan L Pennington
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Arnold Gutierrez
- Department of Pediatrics, University of Cincinnati College of Medicine, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Matthew R Skelton
- Department of Pediatrics, University of Cincinnati College of Medicine, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
| |
Collapse
|
13
|
Sargin D, Jeoung HS, Goodfellow NM, Lambe EK. Serotonin Regulation of the Prefrontal Cortex: Cognitive Relevance and the Impact of Developmental Perturbation. ACS Chem Neurosci 2019; 10:3078-3093. [PMID: 31259523 DOI: 10.1021/acschemneuro.9b00073] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The prefrontal cortex is essential for both executive function and emotional regulation. The interrelationships among these behavioral domains are increasingly recognized, as well as their sensitivity to serotonin (5-hydroxytryptamine, 5-HT). Prefrontal cortex receives serotonergic inputs from the dorsal and median raphe nuclei and is modulated by multiple subtypes of 5-HT receptor across its layers and cell types. Extremes of serotonergic modulation alter mood regulation in vulnerable individuals, yet the impact of serotonin under more typical physiological parameters remains unclear. In this regard, new tools are permitting a closer examination of the behavioral impact of the serotonin system. Optogenetic and chemogenetic manipulations of dorsal raphe 5-HT neurons reveal that serotonin has a greater impact on executive function than previously appreciated. Domains that appear sensitive to fluctuations in 5-HT neuronal excitability include patience and cognitive flexibility. This work is broadly consistent with ex vivo research investigating how 5-HT regulates prefrontal cortex and its output projections. A growing literature suggests 5-HT modulation of these prefrontal circuits is unexpectedly flexible to alteration during development by genetic, behavioral, environmental or pharmacological manipulations, with lasting repercussions for cognition and emotional regulation. Here, we review the cellular and circuit mechanisms of prefrontal serotonergic modulation, investigate recent research into the cognitive consequences of the serotonergic system, and probe the lasting consequences of developmental perturbations. Understanding both the complexity of the prefrontal serotonin system and its sensitivity during development are essential to learn more about the vulnerabilities of this system in mood and anxiety disorders and the underappreciated cognitive consequences of these disorders and their treatment.
Collapse
Affiliation(s)
- Derya Sargin
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary AB T2N 1N4, Canada
| | - Ha-Seul Jeoung
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of OBGYN, University of Toronto, Toronto, ON M5G 1E2, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| |
Collapse
|
14
|
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
|
15
|
Adult Brain Serotonin Deficiency Causes Hyperactivity, Circadian Disruption, and Elimination of Siestas. J Neurosci 2017; 36:9828-42. [PMID: 27656022 DOI: 10.1523/jneurosci.1469-16.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/03/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Serotonin (5-HT) is a crucial neuromodulator linked to many psychiatric disorders. However, after more than 60 years of study, its role in behavior remains poorly understood, in part because of a lack of methods to target 5-HT synthesis specifically in the adult brain. Here, we have developed a genetic approach that reproducibly achieves near-complete elimination of 5-HT synthesis from the adult ascending 5-HT system by stereotaxic injection of an adeno-associated virus expressing Cre recombinase (AAV-Cre) into the midbrain/pons of mice carrying a loxP-conditional tryptophan hydroxylase 2 (Tph2) allele. We investigated the behavioral effects of deficient brain 5-HT synthesis and discovered a unique composite phenotype. Surprisingly, adult 5-HT deficiency did not affect anxiety-like behavior, but resulted in a robust hyperactivity phenotype in novel and home cage environments. Moreover, loss of 5-HT led to an altered pattern of circadian behavior characterized by an advance in the onset and a delay in the offset of daily activity, thus revealing a requirement for adult 5-HT in the control of daily activity patterns. Notably, after normalizing for hyperactivity, we found that the normal prolonged break in nocturnal activity (siesta), a period of rapid eye movement (REM) and non-REM sleep, was absent in all animals in which 5-HT deficiency was verified. Our findings identify adult 5-HT as a requirement for siestas, implicate adult 5-HT in sleep-wake homeostasis, and highlight the importance of our adult-specific 5-HT-synthesis-targeting approach in understanding 5-HT's role in controlling behavior. SIGNIFICANCE STATEMENT Serotonin (5-HT) is a crucial neuromodulator, yet its role in behavior remains poorly understood, in part because of a lack of methods to target specifically adult brain 5-HT synthesis. We developed an approach that reproducibly achieves near-complete elimination of 5-HT synthesis from the adult ascending 5-HT system. Using this technique, we discovered that adult 5-HT deficiency led to a novel compound phenotype consisting of hyperactivity, disrupted circadian behavior patterns, and elimination of siestas, a period of increased sleep during the active phase. These findings highlight the importance of our approach in understanding 5-HT's role in behavior, especially in controlling activity levels, circadian behavior, and sleep-wake homeostasis, behaviors that are disrupted in many psychiatric disorders such as attention deficit hyperactivity disorder.
Collapse
|
16
|
Sargassum swartzii extracts ameliorate memory functions by neurochemical modulation in a rat model. Food Sci Biotechnol 2017; 26:1055-1062. [PMID: 30263636 DOI: 10.1007/s10068-017-0133-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/27/2017] [Accepted: 04/20/2017] [Indexed: 12/28/2022] Open
Abstract
Recently, considerable attention has been paid to drug exploration from natural sources for treating memory loss, a major manifestation of various neurodegenerative diseases. Increasing evidences implicate brain serotonin metabolism in learning and memory, supporting the notion that targeting 5-HT (5-hydroxytryptamine) and its receptors would be beneficial in the treatment of cognitive disorders. In the present study, behavioral and neurochemical effects were examined following administration of Sargassum swartzii extracts in albino Wistar rats. Increase in spatial working memory and recognition memory was exhibited by the seaweed-treated rats as compared to controls. Plasma tryptophan, brain 5-HT, and 5-hydroxyindoleacetic acid levels were measured using HPLC-ECD, and a significant increase in brain 5-HT metabolism was observed in the seaweed-treated rats. The increase in memory functions following repeated administration of S. swartzii extracts is suggested to be due to the increased serotonergic neurotransmission in the brain of seaweed-treated rats.
Collapse
|
17
|
Schaefer TL, Davenport MH, Grainger LM, Robinson CK, Earnheart AT, Stegman MS, Lang AL, Ashworth AA, Molinaro G, Huber KM, Erickson CA. Acamprosate in a mouse model of fragile X syndrome: modulation of spontaneous cortical activity, ERK1/2 activation, locomotor behavior, and anxiety. J Neurodev Disord 2017; 9:6. [PMID: 28616095 PMCID: PMC5467053 DOI: 10.1186/s11689-017-9184-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/13/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Fragile X Syndrome (FXS) occurs as a result of a silenced fragile X mental retardation 1 gene (FMR1) and subsequent loss of fragile X mental retardation protein (FMRP) expression. Loss of FMRP alters excitatory/inhibitory signaling balance, leading to increased neuronal hyperexcitability and altered behavior. Acamprosate (the calcium salt of N-acetylhomotaurinate), a drug FDA-approved for relapse prevention in the treatment of alcohol dependence in adults, is a novel agent with multiple mechanisms that may be beneficial for people with FXS. There are questions regarding the neuroactive effects of acamprosate and the significance of the molecule's calcium moiety. Therefore, the electrophysiological, cellular, molecular, and behavioral effects of acamprosate were assessed in the Fmr1-/y (knock out; KO) mouse model of FXS controlling for the calcium salt in several experiments. METHODS Fmr1 KO mice and their wild-type (WT) littermates were utilized to assess acamprosate treatment on cortical UP state parameters, dendritic spine density, and seizure susceptibility. Brain extracellular-signal regulated kinase 1/2 (ERK1/2) activation was used to investigate this signaling molecule as a potential biomarker for treatment response. Additional adult mice were used to assess chronic acamprosate treatment and any potential effects of the calcium moiety using CaCl2 treatment on behavior and nuclear ERK1/2 activation. RESULTS Acamprosate attenuated prolonged cortical UP state duration, decreased elevated ERK1/2 activation in brain tissue, and reduced nuclear ERK1/2 activation in the dentate gyrus in KO mice. Acamprosate treatment modified behavior in anxiety and locomotor tests in Fmr1 KO mice in which control-treated KO mice were shown to deviate from control-treated WT mice. Mice treated with CaCl2 were not different from saline-treated mice in the adult behavior battery or nuclear ERK1/2 activation. CONCLUSIONS These data indicate that acamprosate, and not calcium, improves function reminiscent of reduced anxiety-like behavior and hyperactivity in Fmr1 KO mice and that acamprosate attenuates select electrophysiological and molecular dysregulation that may play a role in the pathophysiology of FXS. Differences between control-treated KO and WT mice were not evident in a recognition memory test or in examination of acoustic startle response/prepulse inhibition which impeded conclusions from being made about the treatment effects of acamprosate in these instances.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Matthew H Davenport
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Lindsay M Grainger
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Chandler K Robinson
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Anthony T Earnheart
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| | - Melinda S Stegman
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229 USA
| | - Anna L Lang
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202 USA
| | - Amy A Ashworth
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA.,Present address: BlackbookHR, Cincinnati, OH 45202 USA
| | - Gemma Molinaro
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Kimberly M Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Craig A Erickson
- Division of Psychiatry, MLC 7004, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039 USA
| |
Collapse
|
18
|
Lundwall RA, Stephenson KG, Neeley-Tass ES, Cox JC, South M, Bigler ED, Anderberg E, Prigge MD, Hansen BD, Lainhart JE, Kellems RO, Petrie JA, Gabrielsen TP. Relationship between brain stem volume and aggression in children diagnosed with autism spectrum disorder. RESEARCH IN AUTISM SPECTRUM DISORDERS 2017; 34:44-51. [PMID: 28966659 PMCID: PMC5617125 DOI: 10.1016/j.rasd.2016.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
BACKGROUND Aggressive behaviors are common in individuals diagnosed with autism spectrum disorder (ASD) and may be phenotypic indicators of different subtypes within ASD. In current research literature for non-ASD samples, aggression has been linked to several brain structures associated with emotion and behavioral control. However, few if any studies exist investigating brain volume differences in individuals with ASD who have comorbid aggression as indicated by standardized diagnostic and behavioral measures. METHOD We examined neuroimaging data from individuals rigorously diagnosed with ASD versus typically developing (TD) controls. We began with data from brain volume regions of interest (ROI) taken from previous literature on aggression including the brainstem, amygdala, orbitofrontal cortex, anterior cingulate cortex, and dorsolateral prefrontal cortex. We defined aggression status using the Irritability subscale of the Aberrant Behavior Checklist and used lasso logistic regression to select among these predictor variables. Brainstem volume was the only variable shown to be a predictor of aggression status. RESULTS We found that smaller brainstem volumes are associated with higher odds of being in the high aggression group. CONCLUSIONS Understanding brain differences in individuals with ASD who engage in aggressive behavior from those with ASD who do not can inform treatment approaches. Future research should investigate brainstem structure and function in ASD to identify possible mechanisms related to arousal and aggression.
Collapse
Affiliation(s)
| | | | | | | | - Mikle South
- Brigham Young University, Provo, UT 84602, USA
| | | | | | - Molly D Prigge
- University of Utah, 201 Presidents Circle, SLC, UT 84112, USA
| | - Blake D Hansen
- University of Utah, 201 Presidents Circle, SLC, UT 84112, USA
| | - Janet E Lainhart
- University of Wisconsin-Madison, 500 Lincoln Drive, Madison, WI 53706, USA
| | - Ryan O Kellems
- University of Wisconsin-Madison, 500 Lincoln Drive, Madison, WI 53706, USA
| | - Jo Ann Petrie
- University of Wisconsin-Madison, 500 Lincoln Drive, Madison, WI 53706, USA
| | | |
Collapse
|
19
|
Neurodevelopmental Effects of Serotonin on the Brainstem Respiratory Network. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:193-216. [DOI: 10.1007/978-3-319-62817-2_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
20
|
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
|
21
|
Wang X, Yang L, Wang Q, Guo Y, Li N, Ma M, Zhou B. The neurotoxicity of DE-71: effects on neural development and impairment of serotonergic signaling in zebrafish larvae. J Appl Toxicol 2016; 36:1605-1613. [PMID: 27001416 DOI: 10.1002/jat.3322] [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: 12/02/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 01/29/2023]
Abstract
The underlying mechanism of polybrominated diphenyl ether (PBDE)-induced neurotoxicity is still a major concern due to its ubiquitous nature and persistence. Here, zebrafish embryos (2 h postfertilization, hpf) were exposed to different concentrations of the commercial PBDE mixture DE-71 (0-100 µg l-1 ) until 120 hpf, and the impact on neural development and serotonergic system was investigated. The in vivo results revealed significantly reduced transcription of genes involved in neurogenesis (fgf8, shha, wnt1), and contents of proteins in neuronal morphogenesis (myelin basic protein, synapsin IIa), suggesting an impairment of neural development in zebrafish embryos. Further results demonstrated a reduction of 5-hydroxytryptamine neuron and a dose-dependent decrease of whole-body serotonin levels, as well as the transcription of genes involved in serotonergic synthesis (tph1, tph2, trhr) and neurotransmission (serta/b, htr1aa/b). In addition, we predicted possible targets of PBDEs by molecular docking, and the results indicated that PBDE congeners showed high binding affinities with fibroblast growth factor 8 other than SHH and HTR1B. Taken together, this study demonstrated that PBDE exposure during embryogenesis could damage neural development and cause impairment of the serotonergic system as secondary effects in the zebrafish larvae. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Xianfeng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Lihua Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
| | - Qiangwei Wang
- Institute of Pesticide and Environmental Toxicology, Zhejiang University, Hangzhou, 310058, China
| | - Yongyong Guo
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Na Li
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Mei Ma
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Bingsheng Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| |
Collapse
|
22
|
Thomas SA. Neuromodulatory signaling in hippocampus-dependent memory retrieval. Hippocampus 2015; 25:415-31. [PMID: 25475876 DOI: 10.1002/hipo.22394] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2014] [Indexed: 12/15/2022]
Abstract
Considerable advances have been made toward understanding the molecular signaling events that underlie memory acquisition and consolidation. In contrast, less is known about memory retrieval, despite its necessity for utilizing learned information. This review focuses on neuromodulatory and intracellular signaling events that underlie memory retrieval mediated by the hippocampus, for which the most information is currently available. Among neuromodulators, adrenergic signaling is required for the retrieval of various types of hippocampus-dependent memory. Although they contribute to acquisition and/or consolidation, cholinergic and dopaminergic signaling are generally not required for retrieval. Interestingly, while not required for retrieval, serotonergic and opioid signaling may actually constrain memory retrieval. Roles for histamine and non-opioid neuropeptides are currently unclear but possible. A critical effector of adrenergic signaling in retrieval is reduction of the slow afterhyperpolarization mediated by β1 receptors, cyclic AMP, protein kinase A, Epac, and possibly ERK. In contrast, stress and glucocorticoids impair retrieval by decreasing cyclic AMP, mediated in part by the activation of β2 -adrenergic receptors. Clinically, alterations in neuromodulatory signaling and in memory retrieval occur in Alzheimer's disease, Down syndrome, depression, and post-traumatic stress disorder, and recent evidence has begun to link changes in neuromodulatory signaling with effects on memory retrieval.
Collapse
Affiliation(s)
- Steven A Thomas
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
23
|
An L, Zhang T. Comparison Impairments of Spatial Cognition and Hippocampal Synaptic Plasticity Between Prenatal and Postnatal Melamine Exposure in Male Adult Rats. Neurotox Res 2015; 29:218-29. [DOI: 10.1007/s12640-015-9578-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/14/2015] [Accepted: 11/17/2015] [Indexed: 12/12/2022]
|
24
|
Reprint of “Value of water mazes for assessing spatial and egocentric learning and memory in rodent basic research and regulatory studies”. Neurotoxicol Teratol 2015; 52:93-108. [DOI: 10.1016/j.ntt.2015.06.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 11/19/2022]
|
25
|
Autism-Like Behavior and Epigenetic Changes Associated with Autism as Consequences of In Utero Exposure to Environmental Pollutants in a Mouse Model. Behav Neurol 2015; 2015:426263. [PMID: 26586927 PMCID: PMC4637446 DOI: 10.1155/2015/426263] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/01/2015] [Accepted: 10/01/2015] [Indexed: 12/31/2022] Open
Abstract
We tested the hypothesis that in utero exposure to heavy metals increases autism-like behavioral phenotypes in adult animals and induces epigenetic changes in genes that have roles in the etiology of autism. Mouse dams were treated with cadmium, lead, arsenate, manganese, and mercury via drinking water from gestational days (E) 1–10. Valproic acid (VPA) injected intraperitoneally once on (E) 8.5 served as a positive control. Young male offspring were tested for behavioral deficits using four standardized behavioral assays. In this study, in utero exposure to heavy metals resulted in multiple behavioral abnormalities that persisted into adulthood. VPA and manganese induced changes in perseverative/impulsive behavior and social dominance behavior, arsenic caused changes only in perseverative/impulsive behavior, and lead induced abnormalities in social interaction in comparison to the control animals. Brain samples from Mn, Pb, and VPA treated and control animals were evaluated for changes in CpG island methylation in promoter regions and associated changes in gene expression. The Chd7 gene, essential for neural crest cell migration and patterning, was found to be hypomethylated in each experimental animal tested compared to water-treated controls. Furthermore, distinct patterns of CpG island methylation yielded novel candidate genes for further investigation.
Collapse
|
26
|
Pagani JH, Williams Avram SK, Cui Z, Song J, Mezey É, Senerth JM, Baumann MH, Young WS. Raphe serotonin neuron-specific oxytocin receptor knockout reduces aggression without affecting anxiety-like behavior in male mice only. GENES BRAIN AND BEHAVIOR 2015; 14:167-76. [PMID: 25677455 DOI: 10.1111/gbb.12202] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/19/2015] [Indexed: 01/10/2023]
Abstract
Serotonin and oxytocin influence aggressive and anxiety-like behaviors, though it is unclear how the two may interact. That the oxytocin receptor is expressed in the serotonergic raphe nuclei suggests a mechanism by which the two neurotransmitters may cooperatively influence behavior. We hypothesized that oxytocin acts on raphe neurons to influence serotonergically mediated anxiety-like, aggressive and parental care behaviors. We eliminated expression of the oxytocin receptor in raphe neurons by crossing mice expressing Cre recombinase under control of the serotonin transporter promoter (Slc6a4) with our conditional oxytocin receptor knockout line. The knockout mice generated by this cross are normal across a range of behavioral measures: there are no effects for either sex on locomotion in an open-field, olfactory habituation/dishabituation or, surprisingly, anxiety-like behaviors in the elevated O and plus mazes. There was a profound deficit in male aggression: only one of 11 raphe oxytocin receptor knockouts showed any aggressive behavior, compared to 8 of 11 wildtypes. In contrast, female knockouts displayed no deficits in maternal behavior or aggression. Our results show that oxytocin, via its effects on raphe neurons, is a key regulator of resident-intruder aggression in males but not maternal aggression. Furthermore, this reduction in male aggression is quite different from the effects reported previously after forebrain or total elimination of oxytocin receptors. Finally, we conclude that when constitutively eliminated, oxytocin receptors expressed by serotonin cells do not contribute to baseline anxiety-like behaviors or maternal care.
Collapse
Affiliation(s)
- J H Pagani
- Section on Neural Gene Regulation, National Institute of Mental Health, National Institutes of Health, DHHS, Bethesda
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Roubertoux PL, Carlier M, Tordjman S. Deficit in Social Relationships and Reduced Field of Interest in Mice. ORGANISM MODELS OF AUTISM SPECTRUM DISORDERS 2015. [DOI: 10.1007/978-1-4939-2250-5_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
Prenatal melamine exposure induces impairments of spatial cognition and hippocampal synaptic plasticity in male adolescent rats. Reprod Toxicol 2014; 49:78-85. [DOI: 10.1016/j.reprotox.2014.07.081] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 07/27/2014] [Accepted: 07/30/2014] [Indexed: 11/19/2022]
|
29
|
Angoa-Pérez M, Kane MJ, Briggs DI, Herrera-Mundo N, Sykes CE, Francescutti DM, Kuhn DM. Mice genetically depleted of brain serotonin do not display a depression-like behavioral phenotype. ACS Chem Neurosci 2014; 5:908-19. [PMID: 25089765 DOI: 10.1021/cn500096g] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Reductions in function within the serotonin (5HT) neuronal system have long been proposed as etiological factors in depression. Selective serotonin reuptake inhibitors (SSRIs) are the most common treatment for depression, and their therapeutic effect is generally attributed to their ability to increase the synaptic levels of 5HT. Tryptophan hydroxylase 2 (TPH2) is the initial and rate-limiting enzyme in the biosynthetic pathway of 5HT in the CNS, and losses in its catalytic activity lead to reductions in 5HT production and release. The time differential between the onset of 5HT reuptake inhibition by SSRIs (minutes) and onset of their antidepressant efficacy (weeks to months), when considered with their overall poor therapeutic effectiveness, has cast some doubt on the role of 5HT in depression. Mice lacking the gene for TPH2 are genetically depleted of brain 5HT and were tested for a depression-like behavioral phenotype using a battery of valid tests for affective-like disorders in animals. The behavior of TPH2(-/-) mice on the sucrose preference test, tail suspension test, and forced swim test and their responses in the unpredictable chronic mild stress and learned helplessness paradigms was the same as wild-type controls. While TPH2(-/-) mice as a group were not responsive to SSRIs, a subset responded to treatment with SSRIs in the same manner as wild-type controls with significant reductions in immobility time on the tail suspension test, indicative of antidepressant drug effects. The behavioral phenotype of the TPH2(-/-) mouse questions the role of 5HT in depression. Furthermore, the TPH2(-/-) mouse may serve as a useful model in the search for new medications that have therapeutic targets for depression that are outside of the 5HT neuronal system.
Collapse
Affiliation(s)
- Mariana Angoa-Pérez
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Michael J. Kane
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Denise I. Briggs
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Nieves Herrera-Mundo
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Catherine E. Sykes
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Dina M. Francescutti
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| | - Donald M. Kuhn
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan 48201, United States
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan 48201, United States
| |
Collapse
|
30
|
Vorhees CV, Williams MT. Value of water mazes for assessing spatial and egocentric learning and memory in rodent basic research and regulatory studies. Neurotoxicol Teratol 2014; 45:75-90. [PMID: 25116937 DOI: 10.1016/j.ntt.2014.07.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 12/12/2022]
Abstract
Maneuvering safely through the environment is central to survival of all animals. The ability to do this depends on learning and remembering locations. This capacity is encoded in the brain by two systems: one using cues outside the organism (distal cues), allocentric navigation, and one using self-movement, internal cues and sometimes proximal cues, egocentric navigation. Allocentric navigation involves the hippocampus, entorhinal cortex, and surrounding structures (e.g., subiculum); in humans this system encodes declarative memory (allocentric, semantic, and episodic, i.e., memory for people, places, things, and events). This form of memory is assessed in laboratory animals by many methods, but predominantly the Morris water maze (MWM). Egocentric navigation involves the dorsal striatum and connected structures; in humans this system encodes routes and integrated paths and when over-learned becomes implicit or procedural memory. Several allocentric methods for rodents are reviewed and compared with the MWM with particular focus on the Cincinnati water maze (CWM). MWM advantages include minimal training, no food deprivation, ease of testing, reliable learning, insensitivity to differences in body weight and appetite, absence of non-performers, control methods for performance effects, repeated testing capability and other factors that make this test well-suited for regulatory studies. MWM limitations are also reviewed. Evidence-based MWM design and testing methods are presented. On balance, the MWM is arguably the preferred test for assessing learning and memory in basic research and regulatory studies and the CWM is recommended if two tests can be accommodated so that both allocentric (MWM) and egocentric (CWM) learning and memory can be effectively and efficiently assessed.
Collapse
Affiliation(s)
- Charles V Vorhees
- Division of Child Neurology, Dept. of Pediatrics, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| | - Michael T Williams
- Division of Child Neurology, Dept. of Pediatrics, Cincinnati Children's Research Foundation and University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States
| |
Collapse
|
31
|
Hautman ER, Kokenge AN, Udobi KC, Williams MT, Vorhees CV, Skelton MR. Female mice heterozygous for creatine transporter deficiency show moderate cognitive deficits. J Inherit Metab Dis 2014; 37:63-8. [PMID: 23716276 PMCID: PMC7025435 DOI: 10.1007/s10545-013-9619-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 04/04/2013] [Accepted: 04/30/2013] [Indexed: 11/29/2022]
Abstract
Creatine transporter (CrT) deficiency (CTD) is an X-linked disorder characterized by intellectual disability and speech delay. There have been reports that show female carriers have clinical symptoms. We have created CrT knockout (CrT(-/y)) mice in which males show severe cognitive deficits as a model of this disorder. The purpose of this study was to examine if the female carrier mice show cognitive deficits. Reductions in Cr levels as well as CrT transcript were observed in the brains of the female CrT(+/-) mice. CrT(+/-) mice show hyperactivity and increased latency to find the cued platform in the Morris water maze (MWM). CrT(+/-) female mice showed deficits in MWM hidden platform acquisition but not during reversal testing. Memory deficits on probe trials were observed during both phases. Novel object recognition memory and contextual fear memory were not affected in female CrT(+/-) mice. Female CrT(+/-) mice show moderate cognitive deficits, which is consistent with some of the human data. Female CrT(+/-) mice could prove to be beneficial in further understanding CTD and testing therapeutic approaches.
Collapse
Affiliation(s)
- Emily R Hautman
- Division of Neurology, MLC 7044 Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH, 45229-3039, USA
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
Maneuvering safely through the environment is central to survival of almost all species. The ability to do this depends on learning and remembering locations. This capacity is encoded in the brain by two systems: one using cues outside the organism (distal cues), allocentric navigation, and one using self-movement, internal cues and nearby proximal cues, egocentric navigation. Allocentric navigation involves the hippocampus, entorhinal cortex, and surrounding structures; in humans this system encodes allocentric, semantic, and episodic memory. This form of memory is assessed in laboratory animals in many ways, but the dominant form of assessment is the Morris water maze (MWM). Egocentric navigation involves the dorsal striatum and connected structures; in humans this system encodes routes and integrated paths and, when overlearned, becomes procedural memory. In this article, several allocentric assessment methods for rodents are reviewed and compared with the MWM. MWM advantages (little training required, no food deprivation, ease of testing, rapid and reliable learning, insensitivity to differences in body weight and appetite, absence of nonperformers, control methods for proximal cue learning, and performance effects) and disadvantages (concern about stress, perhaps not as sensitive for working memory) are discussed. Evidence-based design improvements and testing methods are reviewed for both rats and mice. Experimental factors that apply generally to spatial navigation and to MWM specifically are considered. It is concluded that, on balance, the MWM has more advantages than disadvantages and compares favorably with other allocentric navigation tasks.
Collapse
|
33
|
Convergent effects of mouse Pet-1 deletion and human PET-1 variation on amygdala fear and threat processing. Exp Neurol 2013; 250:260-9. [PMID: 24100022 DOI: 10.1016/j.expneurol.2013.09.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 08/10/2013] [Accepted: 09/24/2013] [Indexed: 12/31/2022]
Abstract
Serotonin is critical for shaping the development of neural circuits regulating emotion. Pet-1 (FEV-1) is an ETS-domain transcription factor essential for differentiation and forebrain targeting of serotonin neurons. Constitutive Pet-1 knockout (KO) causes major loss of serotonin neurons and forebrain serotonin availability, and behavioral abnormalities. We phenotyped Pet-1 KO mice for fear conditioning and extinction, and on a battery of assays for anxiety- and depression-related behaviors. Morphology of Golgi-stained neurons in basolateral amygdala (BLA) and prelimbic cortex was examined. Using human imaging genetics, a common variant (rs860573) in the PET-1 (FEV) gene was tested for effects on threat-related amygdala reactivity and psychopathology in 88 Asian-ancestry subjects. Pet-1 KO mice exhibited increased acquisition and expression of fear, and elevated fear recovery following extinction, relative to wild-type (WT). BLA dendrites of Pet-1 KO mice were significantly longer than in WT. Human PET-1 variation associated with differences in amygdala threat processing and psychopathology. This novel evidence for the role of Pet-1 in fear processing and dendritic organization of amygdala neurons and in human amygdala threat processing extends a growing literature demonstrating the influence of genetic variation in the serotonin system on emotional regulation via effects on structure and function of underlying corticolimbic circuitry.
Collapse
|
34
|
Korade Z, Folkes OM, Harrison FE. Behavioral and serotonergic response changes in the Dhcr7-HET mouse model of Smith-Lemli-Opitz syndrome. Pharmacol Biochem Behav 2013; 106:101-8. [PMID: 23541496 DOI: 10.1016/j.pbb.2013.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/13/2013] [Accepted: 03/16/2013] [Indexed: 01/22/2023]
Abstract
Smith-Lemli-Opitz syndrome (SLOS) is a developmental disorder resulting from mutations to the Dhcr7 gene, which is required for cholesterol synthesis. Patients with SLOS typically exhibit a number of severe behavioral deficits and many are diagnosed with autistic spectrum disorder. Although the molecular pathophysiology underlying behavioral changes in SLOS and autism spectrum disorders is poorly understood, there is evidence for the involvement of the serotonergic system in SLOS and autism in general. Behavioral testing was undertaken to ascertain the basal behavioral differences between Dhcr7-heterozygous (HET) and wild-type control mice and explore the utility of a Dhcr7-HET mouse line in the development of new treatments for this disorder. Dhcr7-HET mice did not differ from wild-type control mice on basic measures of locomotor activity, anxiety and neuromuscular ability. However, female Dhcr7-HET mice at 6 months of age or older were significantly more likely to win on the social dominance tube test against an unfamiliar mouse. Pharmacological testing, using the 5-HT2A agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI), showed increased head-twitch response in Dhcr7-HET mice, which was apparent from 6 months of age. No differences were found between the genotypes in testing for 5-HT1A agonist 8-OH-DPAT-induced hypothermia. These data indicate an underlying dysfunction of the 5-HT2A receptors in Dhcr7-HET mice that warrants further investigation to establish how this may relate to behavioral disturbances in human patients carrying Dhcr7 mutations.
Collapse
Affiliation(s)
- Z Korade
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN 37232-0475, USA
| | | | | |
Collapse
|
35
|
Lesch KP, Araragi N, Waider J, van den Hove D, Gutknecht L. Targeting brain serotonin synthesis: insights into neurodevelopmental disorders with long-term outcomes related to negative emotionality, aggression and antisocial behaviour. Philos Trans R Soc Lond B Biol Sci 2012; 367:2426-43. [PMID: 22826343 DOI: 10.1098/rstb.2012.0039] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aggression, which comprises multi-faceted traits ranging from negative emotionality to antisocial behaviour, is influenced by an interaction of biological, psychological and social variables. Failure in social adjustment, aggressiveness and violence represent the most detrimental long-term outcome of neurodevelopmental disorders. With the exception of brain-specific tryptophan hydroxylase-2 (Tph2), which generates serotonin (5-HT) in raphe neurons, the contribution of gene variation to aggression-related behaviour in genetically modified mouse models has been previously appraised (Lesch 2005 Novartis Found Symp. 268, 111-140; Lesch & Merschdorf 2000 Behav. Sci. Law 18, 581-604). Genetic inactivation of Tph2 function in mice led to the identification of phenotypic changes, ranging from growth retardation and late-onset obesity, to enhanced conditioned fear response, increased aggression and depression-like behaviour. This spectrum of consequences, which are amplified by stress-related epigenetic interactions, are attributable to deficient brain 5-HT synthesis during development and adulthood. Human data relating altered TPH2 function to personality traits of negative emotionality and neurodevelopmental disorders characterized by deficits in cognitive control and emotion regulation are based on genetic association and are therefore not as robust as the experimental mouse results. Mouse models in conjunction with approaches focusing on TPH2 variants in humans provide unexpected views of 5-HT's role in brain development and in disorders related to negative emotionality, aggression and antisocial behaviour.
Collapse
Affiliation(s)
- Klaus-Peter Lesch
- Division of Molecular Psychiatry (MP), Laboratory of Translational Neuroscience (LTN), Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Wuerzburg, , Fuechsleinstrasse 15, 97080 Wuerzburg, Germany.
| | | | | | | | | |
Collapse
|
36
|
Waider J, Proft F, Langlhofer G, Asan E, Lesch KP, Gutknecht L. GABA concentration and GABAergic neuron populations in limbic areas are differentially altered by brain serotonin deficiency in Tph2 knockout mice. Histochem Cell Biol 2012; 139:267-81. [PMID: 23052836 DOI: 10.1007/s00418-012-1029-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2012] [Indexed: 12/21/2022]
Abstract
While tryptophan hydroxylase-2 (Tph2) null mutant (Tph2(-/-)) mice are completely deficient in brain serotonin (5-HT) synthesis, the formation of serotonergic neurons and pathfinding of their projections are not impaired. However, 5-HT deficiency, during development and in the adult, might affect morphological and functional parameters of other neural systems. To assess the influence of 5-HT deficiency on γ-amino butyric acid (GABA) systems, we carried out measurements of GABA concentrations in limbic brain regions of adult male wildtype (wt), heterozygous (Tph2(+/-)) and Tph2(-/-) mice. In addition, unbiased stereological estimation of GABAergic interneuron numbers and density was performed in subregions of amygdala and hippocampus. Amygdala and prefrontal cortex displayed significantly increased and decreased GABA concentrations, respectively, exclusively in Tph2(+/-) mice while no changes were detected between Tph2(-/-) and wt mice. In contrast, in the hippocampus, increased GABA concentrations were found in Tph2(-/-) mice. While total cell density in the anterior basolateral amygdala did not differ between genotypes, the number and density of the GABAergic interneurons were significantly decreased in Tph2(-/-) mice, with the group of parvalbumin (PV)-immunoreactive (ir) interneurons contributing somewhat less to the decrease than that of non-PV-ir GABAergic interneurons. Major morphological changes were also absent in the dorsal hippocampus, and only a trend toward reduced density of PV-ir cells was observed in the CA3 region of Tph2(-/-) mice. Our findings are the first to document that life-long reduction or complete lack of brain 5-HT transmission causes differential changes of GABA systems in limbic regions which are key players in emotional learning and memory processes. The changes likely reflect a combination of developmental alterations and functional adaptations of emotion circuits to balance the lack of 5-HT, and may underlie altered emotional behavior in 5-HT-deficient mice. Taken together, our findings provide further insight into the mechanisms how life-long 5-HT deficiency impacts the pathogenesis of anxiety- and fear-related disorders.
Collapse
Affiliation(s)
- Jonas Waider
- Laboratory of Translational Neuroscience, Division of Molecular Psychiatry, Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Wuerzburg, Fuechsleinstrasse 15, 97080 Wuerzburg, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Chen Y, Curran CP, Nebert DW, Patel KV, Williams MT, Vorhees CV. Effect of chronic glutathione deficiency on the behavioral phenotype of Gclm-/- knockout mice. Neurotoxicol Teratol 2012; 34:450-7. [PMID: 22580179 DOI: 10.1016/j.ntt.2012.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 04/27/2012] [Accepted: 04/30/2012] [Indexed: 12/12/2022]
Abstract
Enhanced oxidative stress or deficient oxidative stress response in the brain is associated with neurodegenerative disorders and behavioral abnormalities. Previously we generated a knockout mouse line lacking the gene encoding glutamate-cysteine ligase modifier subunit (GCLM). Gclm(-/-) knockout (KO) mice are viable and fertile, yet exhibit only 9-35% of wild-type levels of reduced glutathione (GSH) in tissues, making them a useful model for chronic GSH depletion. Having the global absence of this gene, KO mice--from the time of conception and throughout postnatal life--experience chronic oxidative stress in all tissues, including brain. Between postnatal day (P) 60 and P100, we carried out behavioral phenotyping tests in adults, comparing male and female Gclm(-/-) with Gclm(-/-) wild-type (WT) littermates. Compared with WT, KO mice exhibited: subnormal anxiety in the elevated zero maze; normal overall exploratory open-field activity, but slightly more activity in the peripheral zones; normal acoustic startle and prepulse inhibition reactions; normal novel object recognition with increased time attending to the stimulus objects; slightly reduced latencies to reach a random marked platform in the Morris water maze; normal spatial learning and memory in multiple phases of the Morris water maze; and significantly greater hyperactivity in response to methamphetamine in the open field. These findings are generally in agreement with two prior studies on these mice and suggest that the brain is remarkably resilient to lowered GSH levels, implying significant reserve capacity to regulate reactive oxygen species-but with regional differences such that anxiety and stimulated locomotor control brain regions might be more vulnerable.
Collapse
Affiliation(s)
- Ying Chen
- Department of Environmental Health, and Center for Environmental Genetics-CEG, University of Cincinnati Medical Center, Cincinnati, OH 45267-0056, USA
| | | | | | | | | | | |
Collapse
|
38
|
Mosienko V, Bert B, Beis D, Matthes S, Fink H, Bader M, Alenina N. Exaggerated aggression and decreased anxiety in mice deficient in brain serotonin. Transl Psychiatry 2012; 2:e122. [PMID: 22832966 PMCID: PMC3365263 DOI: 10.1038/tp.2012.44] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Serotonin is a major neurotransmitter in the central nervous system (CNS). Dysregulation of serotonin transmission in the CNS is reported to be related to different psychiatric disorders in humans including depression, impulsive aggression and anxiety disorders. The most frequently prescribed antidepressants and anxiolytics target the serotonergic system. However, these drugs are not effective in 20-30% of cases. The causes of this failure as well as the molecular mechanisms involved in the origin of psychological disorders are poorly understood. Biosynthesis of serotonin in the CNS is initiated by tryptophan hydroxylase 2 (TPH2). In this study, we used Tph2-deficient (Tph2(-/-)) mice to evaluate the impact of serotonin depletion in the brain on mouse behavior. Tph2(-/-) mice exhibited increased depression-like behavior in the forced swim test but not in the tail suspension test. In addition, they showed decreased anxiety-like behavior in three different paradigms: elevated plus maze, marble burying and novelty-suppressed feeding tests. These phenotypes were accompanied by strong aggressiveness observed in the resident-intruder paradigm. Despite carrying only one copy of the gene, heterozygous Tph2(+/-) mice showed only 10% reduction in brain serotonin, which was not sufficient to modulate behavior in the tested paradigms. Our findings provide unequivocal evidence on the pivotal role of central serotonin in anxiety and aggression.
Collapse
Affiliation(s)
- V Mosienko
- Department of Molecular Biology of Peptide Hormones, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany,Faculty of Mathematics and Natural Sciences, Department of Biology, Humboldt-Universität Berlin, Berlin, Germany
| | - B Bert
- Institute of Pharmacology and Toxicology, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - D Beis
- Department of Molecular Biology of Peptide Hormones, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany,Faculty of Mathematics and Natural Sciences, Department of Biology, Humboldt-Universität Berlin, Berlin, Germany
| | - S Matthes
- Department of Molecular Biology of Peptide Hormones, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany,Faculty of Mathematics and Natural Sciences, Department of Biology, Humboldt-Universität Berlin, Berlin, Germany
| | - H Fink
- Institute of Pharmacology and Toxicology, School of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - M Bader
- Department of Molecular Biology of Peptide Hormones, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - N Alenina
- Department of Molecular Biology of Peptide Hormones, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany,Department of Molecular Biology of Peptide Hormones, Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle Street 10, 13125 Berlin, Germany. E-mail:
| |
Collapse
|
39
|
Angoa-Pérez M, Kane MJ, Briggs DI, Sykes CE, Shah MM, Francescutti DM, Rosenberg DR, Thomas DM, Kuhn DM. Genetic depletion of brain 5HT reveals a common molecular pathway mediating compulsivity and impulsivity. J Neurochem 2012; 121:974-84. [PMID: 22443164 DOI: 10.1111/j.1471-4159.2012.07739.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neuropsychiatric disorders characterized by behavioral disinhibition, including disorders of compulsivity (e.g. obsessive-compulsive disorder; OCD) and impulse-control (e.g. impulsive aggression), are severe, highly prevalent and chronically disabling. Treatment options for these diseases are extremely limited. The pathophysiological bases of disorders of behavioral disinhibition are poorly understood but it has been suggested that serotonin dysfunction may play a role. Mice lacking the gene encoding brain tryptophan hydroxylase 2 (Tph2-/-), the initial and rate-limiting enzyme in the synthesis of serotonin, were tested in numerous behavioral assays that are well known for their utility in modeling human neuropsychiatric diseases. Mice lacking Tph2 (and brain 5HT) show intense compulsive and impulsive behaviors to include extreme aggression. The impulsivity is motor in form and not cognitive because Tph2-/- mice show normal acquisition and reversal learning on a spatial learning task. Restoration of 5HT levels by treatment of Tph2-/- mice with its immediate precursor 5-hydroxytryptophan attenuated compulsive and impulsive-aggressive behaviors. Surprisingly, in Tph2-/- mice, the lack of 5HT was not associated with anxiety-like behaviors. The results indicate that 5HT mediates behavioral disinhibition in the mammalian brain independent of anxiogenesis.
Collapse
Affiliation(s)
- Mariana Angoa-Pérez
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Piechal A, Blecharz-Klin K, Wyszogrodzka E, Kołomańska P, Rok-Bujko P, Krząścik P, Kostowski W, Widy-Tyszkiewicz E, Filip M, Stefański R. Neonatal serotonin (5-HT) depletion does not affect spatial learning and memory in rats. Pharmacol Rep 2012; 64:266-74. [DOI: 10.1016/s1734-1140(12)70764-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 11/21/2011] [Indexed: 01/28/2023]
|
41
|
Fournet V, Schweitzer A, Chevarin C, Deloulme JC, Hamon M, Giros B, Andrieux A, Martres MP. The deletion of STOP/MAP6 protein in mice triggers highly altered mood and impaired cognitive performances. J Neurochem 2012; 121:99-114. [PMID: 22146001 DOI: 10.1111/j.1471-4159.2011.07615.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The microtubule-associated Stable Tubulie Only Polypeptide (STOP; also known as MAP6) protein plays a key role in neuron architecture and synaptic plasticity, the dysfunctions of which are thought to be implicated in the pathophysiology of psychiatric diseases. The deletion of STOP in mice leads to severe disorders reminiscent of several schizophrenia-like symptoms, which are also associated with differential alterations of the serotonergic tone in somas versus terminals. In STOP knockout (KO) compared with wild-type mice, serotonin (5-HT) markers are found to be markedly accumulated in the raphe nuclei and, in contrast, deeply depleted in all serotonergic projection areas. In the present study, we carefully examined whether the 5-HT imbalance would lead to behavioral consequences evocative of mood and/or cognitive disorders. We showed that STOP KO mice exhibited depression-like behavior, associated with a decreased anxiety-status in validated paradigms. In addition, although STOP KO mice had a preserved very short-term memory, they failed to perform well in all other learning and memory tasks. We also showed that STOP KO mice exhibited regional imbalance of the norepinephrine tone as observed for 5-HT. As a consequence, mutant mice were hypersensitive to acute antidepressants with different selectivity. Altogether, these data indicate that the deletion of STOP protein in mice caused deep alterations in mood and cognitive performances and that STOP protein might have a crucial role in the 5-HT and norepinephrine networks development.
Collapse
Affiliation(s)
- Vincent Fournet
- INSERM UMRS 952, CNRS UMR 7224, Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
42
|
The influence of neonatal serotonin depletion on emotional and exploratory behaviours in rats. Behav Brain Res 2012; 226:87-95. [DOI: 10.1016/j.bbr.2011.08.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 07/29/2011] [Accepted: 08/23/2011] [Indexed: 11/21/2022]
|
43
|
Powell SB, Weber M, Geyer MA. Genetic models of sensorimotor gating: relevance to neuropsychiatric disorders. Curr Top Behav Neurosci 2012; 12:251-318. [PMID: 22367921 PMCID: PMC3357439 DOI: 10.1007/7854_2011_195] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sensorimotor gating, or the ability of a sensory event to suppress a motor response, can be measured operationally via prepulse inhibition (PPI) of the startle response. PPI is deficient in schizophrenia patients as well as other neuropsychiatric disorders, can be measured across species, and has been used widely as a translational tool in preclinical neuropharmacological and genetic research. First developed to assess drug effects in pharmacological and developmental models, PPI has become one of the standard behavioral measures in genetic models of schizophrenia and other neuropsychiatric disorders that exhibit PPI deficits. In this chapter we review the literature on genetic models of sensorimotor gating and discuss the utility of PPI as a tool in phenotyping mutant mouse models. We highlight the approaches to genetic mouse models of neuropsychiatric disease, discuss some of the important caveats to these approaches, and provide a comprehensive table covering the more recent genetic models that have evaluated PPI.
Collapse
Affiliation(s)
- Susan B. Powell
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0804, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Martin Weber
- Department of Neuroscience, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080-4990, USA
| | - Mark A. Geyer
- Department of Psychiatry, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093-0804, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
44
|
Fernandez SP, Gaspar P. Investigating anxiety and depressive-like phenotypes in genetic mouse models of serotonin depletion. Neuropharmacology 2011; 62:144-54. [PMID: 21945798 DOI: 10.1016/j.neuropharm.2011.08.049] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 08/25/2011] [Accepted: 08/29/2011] [Indexed: 01/24/2023]
Abstract
Emotional disorders such as depression, panic attacks, generalized anxiety, phobias and post-traumatic stress have been associated to decreased serotonin (5-HT) function, based on the positive effects of treatments that enhance 5-HT neurotransmission. However, it has been difficult to establish a primary role for 5-HT deficiency in these diseases, making preclinical models particularly useful. Over the last ten years a variety of genetic mouse models of 5-HT depletion have been produced, complementing previous pharmacologically-based models. Initial models hindered the differentiation of the raphe 5-HT neurons, while more recently produced models suppressed 5-HT production or incapacitated 5-HT vesicular packaging and release in normally developed raphe neurons. Here, we provide an overview of 11 genetic mouse models with lowered 5-HT transmission and summarize the available behavioural investigations concerning their anxiety and depression phenotypes. Although these studies are still ongoing, some common anxiety-related traits and behavioural phenotypes have emerged. Most studies have reported decreased innate anxiety to novelty but heightened fear responses to conditioned aversive cues. This complex phenotype is in general agreement with the proposed dual function of 5-HT in modulating different defensive behaviours. Surprisingly, the depressive-like behaviours have been less studied and, so far, did not yield a consistent phenotype in standard tests. Future studies should be conducted using more ethological relevant models to conclude on the causal role of 5-HT depletion in depression. This review also describes the differences in level and regional distribution of 5-HT depletion among the available mouse models, which could contribute to the diverse phenotypes observed. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
|
45
|
Deneris ES. Molecular genetics of mouse serotonin neurons across the lifespan. Neuroscience 2011; 197:17-27. [PMID: 21920412 DOI: 10.1016/j.neuroscience.2011.08.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 08/25/2011] [Accepted: 08/26/2011] [Indexed: 11/25/2022]
Abstract
New molecular genetics approaches have been developed over the past several years to study brain serotonin (5-HT) neuron development and the roles of 5-HT neurons in behavior and physiology. These approaches were enabled by manipulation of the gene encoding the Pet-1 ETS transcription factor whose expression in the brain is restricted to developing and adult 5-HT neurons. Targeting of the Pet-1 gene led to the development of a mouse line with a severe and stable deficiency of embryonic 5-HT-synthesizing neurons. The Pet-1 transcription regulatory region has been used to create several new 5-HT neuron-type transgenic tools that have greatly increased the experimental accessibility of the small number of brain 5-HT neurons. Permanent and specific marking of 5-HT neurons with Pet-1-based transgenic tools have now been used for flow cytometry, whole cell electrophysiological recordings, progenitor fate mapping, and live time lapse imaging of these neurons. Additional tools provide multiple strategies for conditional temporal targeting of gene expression in 5-HT neurons at different stages of life. Pet-1-based approaches have led to advances in understanding the role of 5-HT neurons in respiration, thermoregulation, emotional behaviors, maternal behavior, and the mechanism of antipsychotic drug actions. In addition, these approaches have begun to reveal the molecular basis of 5-HT neuron heterogeneity and the transcriptional mechanisms that direct 5-HT neuron-type identity, maturation, and maintenance.
Collapse
Affiliation(s)
- E S Deneris
- Case Western Reserve University, School of Medicine, Department of Neurosciences, Cleveland, OH 44106, USA.
| |
Collapse
|
46
|
Gutleb AC, Lilienthal H, Erhard HW, Zimmer KE, Skaare JU, Ropstad E. Effects of pre- and postnatal polychlorinated biphenyl exposure on emotional reactivity observed in lambs before weaning. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2011; 74:1396-1401. [PMID: 21450342 DOI: 10.1016/j.ecoenv.2011.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 02/27/2011] [Accepted: 03/05/2011] [Indexed: 05/30/2023]
Abstract
Humans and animals are exposed to PCBs and influences on developmental and endocrine processes are among the most pronounced effects. In the present study it was hypothesised that exposure to PCBs may interfere with sexually dimorphic behaviour. To test this hypothesis, behavioural studies in developmentally exposed sheep were conducted. Ewes were orally administered PCB 153 (98 μg/kg bw day), PCB 118 (49 μg/kg bw day) or corn oil from conception until delivery. However, because of accidental cross-contamination occurring twice causing a mixed exposure scenario in all three groups, the focus of this paper is to compare three distinct groups of lambs with different PCB levels (PCB 153 high-PCB 153 h, PCB 118 high-PCB 118 h, and low combined group-LC) rather than comparing animals exposed to single PCB congeners to those of a control group. Lambs were tested between 2 and 6 weeks of age. When LC males started the light/dark choice test in a dark box, they spent significantly more time in the dark part of the pen than LC females. This gender-related difference was not found in groups exposed to PCBs. A significant inhibitory effect on the activity level of males exposed to stress of confinement was found in the PCB 118 h group. In a high stress situation females from PCB 118 h and males from PCB 153 h were less active than their gender counterparts. The results support the hypothesis that intrauterine exposure to PCBs can alter sexually dimorphic behaviour of offspring.
Collapse
Affiliation(s)
- Arno C Gutleb
- Department of Production Animal Clinical Science, Norwegian School of Veterinary Science, POB 8146 Dep., N-0033 Oslo, Norway
| | | | | | | | | | | |
Collapse
|
47
|
A genetically defined morphologically and functionally unique subset of 5-HT neurons in the mouse raphe nuclei. J Neurosci 2011; 31:2756-68. [PMID: 21414898 DOI: 10.1523/jneurosci.4080-10.2011] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Heterogeneity of central serotonin (5-HT) raphe neurons is suggested by numerous lines of evidence, but its genetic basis remains elusive. The transcription factor Pet1 is required for the acquisition of serotonergic identity in a majority of neurons in the raphe nuclei. Nevertheless, a subset of 5-HT neurons differentiates in Pet1 knock-out mice. We show here that these residual 5-HT neurons outline a unique subpopulation of raphe neurons with highly selective anatomical targets and characteristic synaptic differentiations. In Pet1 knock-out mice, 5-HT innervation strikingly outlines the brain areas involved in stress responses with dense innervation to the basolateral amygdala, the paraventricular nucleus of the hypothalamus, and the intralaminar thalamic nuclei. In these regions, 5-HT terminals establish asymmetric synaptic junctions. This target selectivity could not be related to altered growth of the remaining 5-HT neurons, as indicated by axon tracing and cell culture analyses. The residual 5-HT axon terminals are functional with maintained release properties in vitro and in vivo. The functional consequence of this uneven distribution of 5-HT innervation on behavior was characterized. Pet1 knock-out mice showed decreased anxiety behavior in novelty exploration and increased fear responses to conditioned aversive cues. Overall, our findings lead us to propose the existence of Pet1-dependent and Pet1-resistant 5-HT neurons targeting different brain centers that might delineate the anatomical basis for a dual serotonergic control on stress responses.
Collapse
|
48
|
Skelton MR, Schaefer TL, Graham DL, Degrauw TJ, Clark JF, Williams MT, Vorhees CV. Creatine transporter (CrT; Slc6a8) knockout mice as a model of human CrT deficiency. PLoS One 2011; 6:e16187. [PMID: 21249153 PMCID: PMC3020968 DOI: 10.1371/journal.pone.0016187] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 12/09/2010] [Indexed: 11/18/2022] Open
Abstract
Mutations in the creatine (Cr) transporter (CrT; Slc6a8) gene lead to absence of brain Cr and intellectual disabilities, loss of speech, and behavioral abnormalities. To date, no mouse model of CrT deficiency exists in which to understand and develop treatments for this condition. The purpose of this study was to generate a mouse model of human CrT deficiency. We created mice with exons 2–4 of Slc6a8 flanked by loxP sites and crossed these to Cre:CMV mice to create a line of ubiquitous CrT knockout expressing mice. Mice were tested for learning and memory deficits and assayed for Cr and neurotransmitter levels. Male CrT−/y (affected) mice lack Cr in the brain and muscle with significant reductions of Cr in other tissues including heart and testes. CrT−/y mice showed increased path length during acquisition and reversal learning in the Morris water maze. During probe trials, CrT−/y mice showed increased average distance from the platform site. CrT−/y mice showed reduced novel object recognition and conditioned fear memory compared to CrT+/y. CrT−/y mice had increased serotonin and 5-hydroxyindole acetic acid in the hippocampus and prefrontal cortex. Ubiquitous CrT knockout mice have learning and memory deficits resembling human CrT deficiency and this model should be useful in understanding this disorder.
Collapse
Affiliation(s)
- Matthew R Skelton
- Division of Neurology, Cincinnati Children's Research Foundation, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America.
| | | | | | | | | | | | | |
Collapse
|
49
|
Kriegebaum CB, Gutknecht L, Bartke L, Reif A, Buttenschon HN, Mors O, Lesch KP, Schmitt AG. The expression of the transcription factor FEV in adult human brain and its association with affective disorders. J Neural Transm (Vienna) 2010; 117:831-6. [PMID: 20480378 DOI: 10.1007/s00702-010-0405-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Accepted: 04/18/2010] [Indexed: 11/28/2022]
Abstract
A wide range of physiological processes and neuronal functioning is modulated by the serotonergic system. Serotonin (5-HT) plays an important role during early brain development. Moreover, dysfunction of the 5-HT system is implicated in psychiatric disorders, especially in affective disorders. Little is known, however, about the transcriptional mechanisms leading to a functional 5-HT system in humans. The Fifth Ewing Variant (FEV), an E-twenty-six (ETS) transcription factor, is assumed to be involved in the transcription of gene(s) in the serotonergic pathway and to play a role in early brain development. To investigate its specificity, we performed an expression analysis of FEV in different human brain regions utilizing quantitative real-time polymerase chain reaction. Our results demonstrate that FEV is not exclusively expressed in serotonergic neurons, but, on the contrary, also in several non-serotonergic brain regions such as locus coeruleus, caudate nucleus and putamen. In the latter two regions, FEV expression levels actually were higher when compared with the pons and the medulla oblongata, which contain the raphe nuclei. Additionally, we examined whether genetic variance in the FEV gene contributes to the susceptibility towards affective disorders. Direct re-sequencing, however, did not provide evidence for FEV mutations in patients, and neither were non-coding single nucleotide polymorphisms associated with disease. FEV therefore might not account for the genetic risk towards depression or bipolar disorder. Furthermore, the specificity of FEV for the serotonergic system should be reconsidered.
Collapse
Affiliation(s)
- Claudia B Kriegebaum
- Molecular and Clinical Psychobiology, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Füchsleinstr. 15, 97080, Würzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Pollak DD, Monje FJ, Lubec G. The learned safety paradigm as a mouse model for neuropsychiatric research. Nat Protoc 2010; 5:954-62. [PMID: 20431541 DOI: 10.1038/nprot.2010.64] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fear conditioning is one of the most widely used animal models for studying the neurobiological basis of fear and anxiety states. Conditioned inhibition of fear (or learned safety), however, is a relatively unexplored behavioral paradigm addressing the aspect of regulation of fear, which is central to survival and mental health. Although fear conditioning is achieved by pairing a previously neutral, conditioned stimulus (CS) with an aversive, unconditioned stimulus (US), learned safety training consists of a series of explicitly unpaired CS-US presentations. Animals are trained for 3 d, one session per day, and learn to associate the CS with protection from the impending danger of the aversive events. The entire procedure can be completed within 7 d. The protocol has been successfully used to study the molecular underpinnings of a behavioral intervention for depression. This paradigm complements currently used animal tests in neuropsychiatric research addressing the dysregulation of emotional behaviors in genetic, pharmacological or environmental mouse models of human affective disorders.
Collapse
Affiliation(s)
- Daniela D Pollak
- Department of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| | | | | |
Collapse
|