1
|
Guindon GE, Anzalone A, Burke SG, Murphy CA, Milano ME, Price JC, Tadros S, McFarland AT, Contini FM, Seggio JA. Consumption of dopamine receptor 1 agonist SKF-38393 reduces constant-light-induced hyperactivity, depression-like, and anxiety-like behaviors in a sex specific manner in C57BL/6J mice. Front Behav Neurosci 2025; 19:1537048. [PMID: 40144749 PMCID: PMC11936926 DOI: 10.3389/fnbeh.2025.1537048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Artificial light exposure during nighttime, including constant light (LL), is an increasingly prevalent environmental occurrence linked to impaired mood and cognitive impairments in both humans and animal models. Dopamine and dopamine 1 receptors are well known to modulate circadian rhythms and mood. This study investigated the effects of LL on anxiety-like, depressive-like, and cognitive behaviors in male and female C57BL/6J mice and assessed whether consumption of SKF-38393, a dopamine 1 receptor agonist, can mitigate these negative behavioral outcomes. Mice were exposed to LL or a standard 12:12 light:dark cycle (LD) for 6 weeks, with subgroups receiving either SKF-38393 or water. All mice had their circadian rhythms continuously monitored and were placed within behavioral tests that assayed their anxiety-like, depressive-like, and learning and memory behaviors. Behavioral assays revealed that LL increased hyperactivity and anxiety-like behaviors, which were mitigated by SKF-38393 consumption in both sexes. In addition, male mice exhibited anhedonia under LL, which was alleviated by SKF-38393, whereas female mice were resistant to LL-induced anhedonia. Sex differences emerged in fluid consumption independent of lighting condition, with females consuming more SKF-38393, and in responses to DA on behavior, including novel object recognition and exploration. These results indicate that low dose oral consumption of dopamine 1 receptor agonists can ameliorate some of the negative behavioral effects of LL exposure. This study highlights the complex interplay between chronic light, dopamine, and sex in influencing mood and behavior, suggesting potential modulatory roles for dopamine 1 receptor agonists in regulating behavioral outcomes to circadian disturbances.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Joseph A. Seggio
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, United States
| |
Collapse
|
2
|
Matsunaga D, Nakagawa H, Ishiwata T. Comparison of forced and voluntary exercise types on male rat brain monoamine levels, anxiety-like behaviour, and physiological indexes under light and dark phases. Behav Brain Res 2025; 479:115321. [PMID: 39510330 DOI: 10.1016/j.bbr.2024.115321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE Physical exercise improves physical and mental health; however, the differences between voluntary and forced exercise protocols are unclear. In addition, knowledge regarding the consequences of differences in testing timing, such as light and dark phases, in response to exercise type is limited. We investigated the effects of chronic forced and voluntary wheel running on the changes in brain monoamine levels (5-HT: serotonin, DA: dopamine, NA: noradrenaline), anxiety-like behaviours, and physiological stress responses in the light and dark phases. METHODS Adult male Wistar rats were equally and randomly assigned to four groups: sedentary control, voluntary exercise (free running on a wheel, V-EX), voluntary limited exercise (wheel available only 1 h/day, VL-EX), and forced exercise (running on a motorised wheel, F-EX). Each group was further divided into dark- or light-experimental condition groups. After 4 weeks, the rats underwent an open-field test. The monoamines and their metabolite levels were measured in the major neural cell bodies and the projection areas related to behaviour, cognition, anxiety, and stress in the brain. RESULTS Adrenal hypertrophy and elevated body temperature, except during the exercise period, were observed in the F-EX rats that exhibited anxiety-like behaviour. The levels of monoamines and their metabolites, particularly the 5-HTergic and DAergic systems, in specific areas, were significantly altered in the rats in the V-EX group compared to those in the VL-EX and other groups. These differences were observed only in the dark phase. CONCLUSION The results suggest that V-EX mainly stimulates the 5-HTergic and DAergic systems, while F-EX induces physiological stress and increases anxiety-like behaviour during the dark phase. This study highlights the importance of accounting for exercise types and light/dark phases in behavioural neuroscience experiments.
Collapse
Affiliation(s)
- Daisuke Matsunaga
- Department of Health-Promotion and Sports Science, Osaka Electro-Communication University, 1130-70 Kiyotaki, Shijonawate-shi, Osaka 575-0063, Japan; Graduate School of Community & Human Services, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan.
| | - Hikaru Nakagawa
- College of Sport &Wellness, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan
| | - Takayuki Ishiwata
- Graduate School of Community & Human Services, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan; College of Sport &Wellness, Rikkyo University, 1-2-26 Kitano, Niiza, Saitama 352-8558, Japan
| |
Collapse
|
3
|
Pantiru AD, Van de Sompele S, Ligneul C, Chatelain C, Barrea C, Lerch JP, Filippi BM, Alkan S, De Baere E, Johnston J, Clapcote SJ. Autistic behavior is a common outcome of biallelic disruption of PDZD8 in humans and mice. Mol Autism 2025; 16:14. [PMID: 40016860 PMCID: PMC11866840 DOI: 10.1186/s13229-025-00650-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Intellectual developmental disorder with autism and dysmorphic facies (IDDADF) is a rare syndromic intellectual disability (ID) caused by homozygous disruption of PDZD8 (PDZ domain-containing protein 8), an integral endoplasmic reticulum (ER) protein. All four previously identified IDDADF cases exhibit autistic behavior, with autism spectrum disorder (ASD) diagnosed in three cases. To determine whether autistic behavior is a common outcome of PDZD8 disruption, we studied a third family with biallelic mutation of PDZD8 (family C) and further characterized PDZD8-deficient (Pdzd8tm1b) mice that exhibit stereotyped motor behavior relevant to ASD. METHODS Homozygosity mapping, whole-exome sequencing, and cosegregation analysis were used to identify the PDZD8 variant responsible for IDDADF, including diagnoses of ASD, in consanguineous family C. To assess the in vivo effect of PDZD8 disruption on social responses and related phenotypes, behavioral, structural magnetic resonance imaging, and microscopy analyses were conducted on the Pdzd8tm1b mouse line. Metabolic activity was profiled using sealed metabolic cages. RESULTS The discovery of a third family with IDDADF caused by biallelic disruption of PDZD8 permitted identification of a core clinical phenotype consisting of developmental delay, ID, autism, and facial dysmorphism. In addition to impairments in social recognition and social odor discrimination, Pdzd8tm1b mice exhibit increases in locomotor activity (dark phase only) and metabolic rate (both lights-on and dark phases), and decreased plasma triglyceride in males. In the brain, Pdzd8tm1b mice exhibit increased levels of accessory olfactory bulb volume, primary olfactory cortex volume, dendritic spine density, and ER stress- and mitochondrial fusion-related transcripts, as well as decreased levels of cerebellar nuclei volume and adult neurogenesis. LIMITATIONS The total number of known cases of PDZD8-related IDDADF remains low. Some mouse experiments in the study did not use balanced numbers of males and females. The assessment of ER stress and mitochondrial fusion markers did not extend beyond mRNA levels. CONCLUSIONS Our finding that the Pdzd8tm1b mouse model and all six known cases of IDDADF exhibit autistic behavior, with ASD diagnosed in five cases, identifies this trait as a common outcome of biallelic disruption of PDZD8 in humans and mice. Other abnormalities exhibited by Pdzd8tm1b mice suggest that the range of comorbidities associated with PDZD8 deficiency may be wider than presently recognized.
Collapse
Affiliation(s)
- Andreea D Pantiru
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, M13 9PT, UK
| | - Stijn Van de Sompele
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Clemence Ligneul
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, OX1 3SR, UK
| | - Camille Chatelain
- Department of Human Genetics, University Hospital of Liege, Liege, Belgium
| | - Christophe Barrea
- Autism Resource Centre of Liege, University of Liege, Liege, Belgium
| | - Jason P Lerch
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, OX1 3SR, UK
| | | | - Serpil Alkan
- Department of Human Genetics, University Hospital of Liege, Liege, Belgium
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jamie Johnston
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Steven J Clapcote
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
4
|
Greer D, Lei T, Kryshtal A, Jessen ZF, Schwartz GW. Visual identification of conspecifics shapes social behavior in mice. Curr Biol 2025; 35:287-299.e4. [PMID: 39706174 DOI: 10.1016/j.cub.2024.11.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/07/2024] [Accepted: 11/20/2024] [Indexed: 12/23/2024]
Abstract
Recognizing conspecifics-others of the same species-in order to determine how to interact with them appropriately is a fundamental goal of animal sensory systems. It has undergone selective pressure in nearly all species. Mice have a large repertoire of social behaviors that are the subject of a rapidly growing field of study in neuroscience. Mouse social interactions likely incorporate all available sensory modalities, and the vast majority of studies have not attempted to isolate them. Our understanding of the role of vision in mouse social interactions remains overlooked, given the prominence of olfactory research in this area. To address this, we developed a behavioral platform that allowed us to present a subject mouse with the visual information of stimulus mice in isolation from olfactory, acoustic, and tactile cues. Our results indicate that the visual identification of the sex or individual identity of other mice influences behavior. These findings highlight the underappreciated role of vision in mouse social interactions and open new avenues to study the visual circuits underlying social behavior.
Collapse
Affiliation(s)
- Devon Greer
- Northwestern Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL 60611, USA.
| | - Tianhao Lei
- Northwestern Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL 60611, USA
| | - Anna Kryshtal
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zachary F Jessen
- Northwestern Interdepartmental Neuroscience Graduate Program, Northwestern University, Chicago, IL 60611, USA; Medical Scientist Training Program, Northwestern University, Chicago, IL 60611, USA.
| | - Gregory William Schwartz
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
5
|
Shangase KB, Luvuno M, Mabandla M. Effects of combined postweaning social isolation and ketamine administration on schizophrenia-like behaviour in male Sprague Dawley rats. Behav Brain Res 2025; 476:115214. [PMID: 39182622 DOI: 10.1016/j.bbr.2024.115214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/08/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
The pathophysiology behind negative and cognitive symptoms of schizophrenia is not well understood, thus limiting the effectiveness of treatment on these symptoms. Developing reliable animal model of schizophrenia is vital to advance our understanding on the neurobiological basis of the disorder. Double hit is used to refer to the use of two schizophrenia inducing interventions viz ketamine exposure and social isolation. In this study we aim to investigate the robustness of double hit model of schizophrenia in inducing negative and cognitive symptoms of schizophrenia. On postnatal day (PND) 23, thirty-two male Sprague Dawley rats were randomly grouped into four equal groups as follows: group housed + saline (GH), group housed + ketamine (GHK), isolated + saline (SI), and isolated + ketamine (SIK). A single ketamine dose (16 mg/kg) was administered 3 times a week for four weeks. Isolated animals were housed singly throughout the study. The following behavioural tests were carried out: elevated plus maze, three chamber social interaction, resident intruder tests, and novel object recognition (NOR). The SIK group exhibited high anxiety levels, with increased ACTH, corticosterone and norepinephrine concentration when compared to the other groups. The SIK animals also presented with reduced social interaction and decreased oxytocin concentration. SIK rats were more aggressive towards a juvenile intruder but had low testosterone concentration. The SIK group or double hit model showed impaired visual learning and memory and increased expression of proinflammatory cytokines. This suggest that the double hit model is more robust in inducing negative and cognitive symptoms of schizophrenia than each treatment alone.
Collapse
Affiliation(s)
- Khanyiso Bright Shangase
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa.
| | - Mluleki Luvuno
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Musa Mabandla
- Department of Human Physiology, School of Laboratory Medicine and Medical Science, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| |
Collapse
|
6
|
Wozna‐Wysocka M, Jazurek‐Ciesiolka M, Przybyl L, Wronka D, Misiorek JO, Suszynska‐Zajczyk J, Figura G, Ciesiolka A, Sobieszczanska P, Zeller A, Niemira M, Switonski PM, Fiszer A. Insights into RNA-mediated pathology in new mouse models of Huntington's disease. FASEB J 2024; 38:e70182. [PMID: 39604147 PMCID: PMC11602643 DOI: 10.1096/fj.202401465r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
Huntington's disease (HD) is a neurodegenerative polyglutamine (polyQ) disease resulting from the expansion of CAG repeats located in the ORF of the huntingtin gene (HTT). The extent to which mutant mRNA-driven disruptions contribute to HD pathogenesis, particularly in comparison to the dominant mechanisms related to the gain-of-function effects of the mutant polyQ protein, is still debatable. To evaluate this contribution in vivo, we generated two mouse models through a knock-in strategy at the Rosa26 locus. These models expressed distinct variants of human mutant HTT cDNA fragment: a translated variant (HD/100Q model, serving as a reference) and a nontranslated variant (HD/100CAG model). The cohorts of animals were subjected to a broad spectrum of molecular, behavioral, and cognitive analysis for 21 months. Behavioral testing revealed alterations in both models, with the HD/100Q model exhibiting late disease phenotype. The rotarod, static rod, and open-field tests showed some motor deficits in HD/100CAG and HD/100Q model mice during the light phase, while ActiMot indicated hyperkinesis during the dark phase. Both models also exhibited certain gene deregulations in the striatum that are related to disrupted pathways and phenotype alterations observed in HD. In conclusion, we provide in vivo evidence for a minor contributory role of mutant RNA in HD pathogenesis. The separated effects resulting from the presence of mutant RNA in the HD/100CAG model led to less severe but, to some extent, similar types of impairments as in the HD/100Q model. Increased anxiety was one of the most substantial effects caused by mutant HTT RNA.
Collapse
Affiliation(s)
| | | | - Lukasz Przybyl
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| | - Dorota Wronka
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| | | | | | - Grzegorz Figura
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Present address:
Department of Bioenergetics, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz UniversityPoznanPoland
| | - Adam Ciesiolka
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Present address:
Department of Gene Expression, Institute of Molecular Biology and BiotechnologyAdam Mickiewicz UniversityPoznanPoland
| | | | - Anna Zeller
- Genomics and Epigenomics Laboratory, Clinical Research CentreMedical University of BialystokBialystokPoland
| | - Magdalena Niemira
- Genomics and Epigenomics Laboratory, Clinical Research CentreMedical University of BialystokBialystokPoland
| | | | - Agnieszka Fiszer
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
| |
Collapse
|
7
|
Liu J, Gao J, Wang H, Fan X, Li L, Wang X, Wang X, Lu J, Shi X, Yang P. Acute Neurobehavioral and Glial Responses to Explosion Gas Inhalation in Rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:5099-5111. [PMID: 39092980 DOI: 10.1002/tox.24389] [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: 02/22/2024] [Revised: 04/27/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Military personnel, firefighters, and fire survivors exhibit a higher prevalence of mental health conditions such as depression and post-traumatic stress disorder (PTSD) compared to the general population. While numerous studies have examined the neurological impacts of physical trauma and psychological stress, research on acute neurobehavioral effects of gas inhalation from explosions or fires is limited. This study investigates the early-stage neurobehavioral and neuronal consequences of acute explosion gas inhalation in Sprague-Dawley rats. Rats were exposed to simulated explosive gas and subsequently assessed using behavioral tests and neurobiological analyses. The high-dose exposure group demonstrated significant depression-like behaviors, including reduced mobility and exploration. However, neuronal damage was not evident in histological analyses. Immunofluorescence revealed increased density of radial glia and oligodendrocytes in specific brain regions, suggesting hypoxia and axon damage induced by gas inhalation as a potential mechanism for the observed neurobehavioral changes. These findings underscore the acute impact of explosion gas inhalation on mental health, highlighting the habenula and dentate gyrus of hippocampus as the possible target regions. The findings are expected to support early diagnosis and treatment strategies for brain injuries caused by explosion gas, offering insights into early intervention for depression and PTSD in affected populations.
Collapse
Affiliation(s)
- Jinren Liu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Junhong Gao
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Hong Wang
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Xiaolin Fan
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Liang Li
- Xi'an Key Laboratory of Toxicology and Biological Effects, Institute for Hygiene of Ordnance Industry, Xi'an, China
| | - Xiangni Wang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiying Wang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jiajia Lu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xingmin Shi
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Medical Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Pinglin Yang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
McIlwraith EK, Loganathan N, Mak KWY, He W, Belsham DD. Phoenixin knockout mice show no impairment in fertility or differences in metabolic response to a high-fat diet, but exhibit behavioral differences in an open field test. J Neuroendocrinol 2024; 36:e13398. [PMID: 38733120 DOI: 10.1111/jne.13398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/27/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024]
Abstract
Phoenixin (PNX) is a conserved secreted peptide that was identified 10 years ago with numerous studies published on its pleiotropic functions. PNX is associated with estrous cycle length, protection from a high-fat diet, and reduction of anxiety behavior. However, no study had yet evaluated the impact of deleting PNX in the whole animal. We sought to evaluate a mouse model lacking the PNX parent gene, small integral membrane protein 20 (Smim20), and the resulting effect on reproduction, energy homeostasis, and anxiety. We found that the Smim20 knockout mice had normal fertility and estrous cycle lengths. Consistent with normal fertility, the hypothalamii of the knockout mice showed no changes in the levels of reproduction-related genes, but the male mice had some changes in energy homeostasis-related genes, such as melanocortin receptor 4 (Mc4r). When placed on a high-fat diet, the wildtype and knockout mice responded similarly, but the male heterozygous mice gained slightly less weight. When placed in an open field test box, the female knockout mice traveled less distance in the outer zone, indicating alterations in anxiety or locomotor behavior. In summary, the homozygous knockout of PNX did not alter fertility and modestly alters a few neuroendocrine genes in response to a high-fat diet, especially in the female mice. However, it altered the behavior of mice in an open field test. PNX therefore may not be crucial for reproductive function or weight, however, we cannot rule out possible compensatory mechanisms in the knockout model. Understanding the role of PNX in physiology may ultimately lead to an enhanced understanding of neuroendocrine mechanisms involving this enigmatic peptide.
Collapse
Affiliation(s)
- Emma K McIlwraith
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Neruja Loganathan
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kimberly W Y Mak
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Wenyuan He
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Denise D Belsham
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Mattei D, Ivanov A, Hammer J, Ugursu B, Schalbetter S, Richetto J, Weber-Stadlbauer U, Mueller F, Scarborough J, Wolf SA, Kettenmann H, Wollscheid B, Beule D, Meyer U. Microglia undergo molecular and functional adaptations to dark and light phases in male laboratory mice. Brain Behav Immun 2024; 120:571-583. [PMID: 38986723 DOI: 10.1016/j.bbi.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/20/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024] Open
Abstract
Microglia are increasingly recognized to contribute to brain health and disease. Preclinical studies using laboratory rodents are essential to advance our understanding of the physiological and pathophysiological roles of these cells in the central nervous system. Rodents are nocturnal animals, and they are mostly maintained in a defined light-dark cycle within animal facilities, with many laboratories investigating the molecular and functional profiles of microglia exclusively during the animals' light (sleep) phase. However, only a few studies have considered possible differences in microglial functions between the active and sleep phases. Based on initial evidence suggesting that microglial intrinsic clock genes can affect their phenotypes, we sought to investigate differences in transcriptional, proteotype and functional profiles of microglia between light (sleep) and dark (active) phases, and how these changes are affected in pathological models. We found marked transcriptional and proteotype differences between microglia harvested from male mice during the light or dark phase. Amongst others, these differences related to genes and proteins associated with immune responses, motility, and phagocytosis, which were reflected by functional alterations in microglial synaptic pruning and response to bacterial stimuli. Possibly accounting for such changes, we found RNA and protein regulation in SWI/SNF and NuRD chromatin remodeling complexes between light and dark phases. Importantly, we also show that the time of microglial sample collection influences the nature of microglial transcriptomic changes in a model of immune-mediated neurodevelopmental disorders. Our findings emphasize the importance of considering diurnal factors in studying microglial cells and indicate that implementing a circadian perspective is pivotal for advancing our understanding of their physiological and pathophysiological roles in brain health and disease.
Collapse
Affiliation(s)
- Daniele Mattei
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.
| | - Andranik Ivanov
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin, Berlin, Germany
| | - Jacqueline Hammer
- Institute of Molecular Systems Biology and Department for Health Sciences and Technology, ETH Zürich, Switzerland
| | - Bilge Ugursu
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sina Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Flavia Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Joseph Scarborough
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Susanne A Wolf
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bernd Wollscheid
- Institute of Molecular Systems Biology and Department for Health Sciences and Technology, ETH Zürich, Switzerland
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin, Berlin, Germany
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
Pádua MS, Guil-Guerrero JL, Lopes PA. Behaviour Hallmarks in Alzheimer's Disease 5xFAD Mouse Model. Int J Mol Sci 2024; 25:6766. [PMID: 38928472 PMCID: PMC11204382 DOI: 10.3390/ijms25126766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
The 5xFAD transgenic mouse model widely used in Alzheimer's disease (AD) research recapitulates many AD-related phenotypes with a relatively early onset and aggressive age-dependent progression. Besides developing amyloid peptide deposits alongside neuroinflammation by the age of 2 months, as well as exhibiting neuronal decline by the age of 4 months that intensifies by the age of 9 months, these mice manifest a broad spectrum of behavioural impairments. In this review, we present the extensive repertoire of behavioural dysfunctions in 5xFAD mice, organised into four categories: motor skills, sensory function, learning and memory abilities, and neuropsychiatric-like symptoms. The motor problems, associated with agility and reflex movements, as well as balance and coordination, and skeletal muscle function, typically arise by the time mice reach 9 months of age. The sensory function (such as taste, smell, hearing, and vision) starts to deteriorate when amyloid peptide buildups and neuroinflammation spread into related anatomical structures. The cognitive functions, encompassing learning and memory abilities, such as visual recognition, associative, spatial working, reference learning, and memory show signs of decline from 4 to 6 months of age. Concerning neuropsychiatric-like symptoms, comprising apathy, anxiety and depression, and the willingness for exploratory behaviour, it is believed that motivational changes emerge by approximately 6 months of age. Unfortunately, numerous studies from different laboratories are often contradictory on the conclusions drawn and the identification of onset age, making preclinical studies in rodent models not easily translatable to humans. This variability is likely due to a range of factors associated with animals themselves, housing and husbandry conditions, and experimental settings. In the forthcoming studies, greater clarity in experimental details when conducting behavioural testing in 5xFAD transgenic mice could minimise the inconsistencies and could ensure the reliability and the reproducibility of the results.
Collapse
Affiliation(s)
- Mafalda Soares Pádua
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal;
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
| | - José L. Guil-Guerrero
- Departamento de Tecnología de Alimentos, Universidad de Almería, 04120 Almería, Spain;
| | - Paula Alexandra Lopes
- CIISA—Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal;
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal
| |
Collapse
|
11
|
Mori D, Ikeda R, Sawahata M, Yamaguchi S, Kodama A, Hirao T, Arioka Y, Okumura H, Inami C, Suzuki T, Hayashi Y, Kato H, Nawa Y, Miyata S, Kimura H, Kushima I, Aleksic B, Mizoguchi H, Nagai T, Nakazawa T, Hashimoto R, Kaibuchi K, Kume K, Yamada K, Ozaki N. Phenotypes for general behavior, activity, and body temperature in 3q29 deletion model mice. Transl Psychiatry 2024; 14:138. [PMID: 38453903 PMCID: PMC10920862 DOI: 10.1038/s41398-023-02679-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 03/09/2024] Open
Abstract
Whole genome analysis has identified rare copy number variations (CNV) that are strongly involved in the pathogenesis of psychiatric disorders, and 3q29 deletion has been found to have the largest effect size. The 3q29 deletion mice model (3q29-del mice) has been established as a good pathological model for schizophrenia based on phenotypic analysis; however, circadian rhythm and sleep, which are also closely related to neuropsychiatric disorders, have not been investigated. In this study, our aims were to reevaluate the pathogenesis of 3q29-del by recreating model mice and analyzing their behavior and to identify novel new insights into the temporal activity and temperature fluctuations of the mouse model using a recently developed small implantable accelerometer chip, Nano-tag. We generated 3q29-del mice using genome editing technology and reevaluated common behavioral phenotypes. We next implanted Nano-tag in the abdominal cavity of mice for continuous measurements of long-time activity and body temperature. Our model mice exhibited weight loss similar to that of other mice reported previously. A general behavioral battery test in the model mice revealed phenotypes similar to those observed in mouse models of schizophrenia, including increased rearing frequency. Intraperitoneal implantation of Nano-tag, a miniature acceleration sensor, resulted in hypersensitive and rapid increases in the activity and body temperature of 3q29-del mice upon switching to lights-off condition. Similar to the 3q29-del mice reported previously, these mice are a promising model animals for schizophrenia. Successive quantitative analysis may provide results that could help in treating sleep disorders closely associated with neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan.
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan.
| | - Ryosuke Ikeda
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
- Department of Applied Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Sho Yamaguchi
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Akiko Kodama
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Hirao
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuko Arioka
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Hiroki Okumura
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Chihiro Inami
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Toshiaki Suzuki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yu Hayashi
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidekazu Kato
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Nawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiko Miyata
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Japan
| | - Takanobu Nakazawa
- Laboratory of Molecular Biology, Department of Bioscience, Graduate School of Life Sciences, Tokyo University of Agriculture, Tokyo, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, 1-98 Dengakugakubo, Kusukake-cho, Toyoake, Aichi, Japan
| | - Kazuhiko Kume
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Department of Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Japan
| |
Collapse
|
12
|
Spark DL, Ma S, Nowell CJ, Langmead CJ, Stewart GD, Nithianantharajah J. Sex-Dependent Attentional Impairments in a Subchronic Ketamine Mouse Model for Schizophrenia. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:229-239. [PMID: 38298794 PMCID: PMC10829638 DOI: 10.1016/j.bpsgos.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 02/02/2024] Open
Abstract
Background The development of more effective treatments for schizophrenia targeting cognitive and negative symptoms has been limited, partly due to a disconnect between rodent models and human illness. Ketamine administration is widely used to model symptoms of schizophrenia in both humans and rodents. In mice, subchronic ketamine treatment reproduces key dopamine and glutamate dysfunction; however, it is unclear how this translates into behavioral changes reflecting positive, negative, and cognitive symptoms. Methods In male and female mice treated with either subchronic ketamine or saline, we assessed spontaneous and amphetamine-induced locomotor activity to measure behaviors relevant to positive symptoms, and used a touchscreen-based progressive ratio task of motivation and the rodent continuous performance test of attention to capture specific negative and cognitive symptoms, respectively. To explore neuronal changes underlying the behavioral effects of subchronic ketamine treatment, we quantified expression of the immediate early gene product, c-Fos, in key corticostriatal regions using immunofluorescence. Results We showed that spontaneous locomotor activity was unchanged in male and female subchronic ketamine-treated animals, and amphetamine-induced locomotor response was reduced. Subchronic ketamine treatment did not alter motivation in either male or female mice. In contrast, we identified a sex-specific effect of subchronic ketamine on attentional processing wherein female mice performed worse than control mice due to increased nonselective responding. Finally, we showed that subchronic ketamine treatment increased c-Fos expression in prefrontal cortical and striatal regions, consistent with a mechanism of widespread disinhibition of neuronal activity. Conclusions Our results highlight that the subchronic ketamine mouse model reproduces a subset of behavioral symptoms that are relevant for schizophrenia.
Collapse
Affiliation(s)
- Daisy L. Spark
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Sherie Ma
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron J. Nowell
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Christopher J. Langmead
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Gregory D. Stewart
- Drug Discovery Biology Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jess Nithianantharajah
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
13
|
Schröder JK, Abdel-Hafiz L, Ali AAH, Cousin TC, Hallenberger J, Rodrigues Almeida F, Anstötz M, Lenz M, Vlachos A, von Gall C, Tundo-Lavalle F. Effects of the Light/Dark Phase and Constant Light on Spatial Working Memory and Spine Plasticity in the Mouse Hippocampus. Cells 2023; 12:1758. [PMID: 37443792 PMCID: PMC10340644 DOI: 10.3390/cells12131758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Circadian rhythms in behavior and physiology such as rest/activity and hormones are driven by an internal clock and persist in the absence of rhythmic environmental cues. However, the period and phase of the internal clock are entrained by the environmental light/dark cycle. Consequently, aberrant lighting conditions, which are increasing in modern society, have a strong impact on rhythmic body and brain functions. Mice were exposed to three different lighting conditions, 12 h light/12 h dark cycle (LD), constant darkness (DD), and constant light (LL), to study the effects of the light/dark cycle and aberrant lighting on the hippocampus, a critical structure for temporal and spatial memory formation and navigation. Locomotor activity and plasma corticosterone levels were analyzed as readouts for circadian rhythms. Spatial working memory via Y-maze, spine morphology of Golgi-Cox-stained hippocampi, and plasticity of excitatory synapses, measured by number and size of synaptopodin and GluR1-immunreactive clusters, were analyzed. Our results indicate that the light/dark cycle drives diurnal differences in synaptic plasticity in hippocampus. Moreover, spatial working memory, spine density, and size and number of synaptopodin and GluR1 clusters were reduced in LL, while corticosterone levels were increased. This indicates that acute constant light affects hippocampal function and synaptic plasticity.
Collapse
Affiliation(s)
- Jane K. Schröder
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
- Department of Pediatric Hematology and Oncology, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Laila Abdel-Hafiz
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Amira A. H. Ali
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, El-Gomhoria St. 1, Mansoura 35516, Egypt
| | - Teresa C. Cousin
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Johanna Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Filipe Rodrigues Almeida
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Max Anstötz
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Maximilian Lenz
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany;
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany;
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany;
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| | - Federica Tundo-Lavalle
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany; (J.K.S.); (L.A.-H.); (A.A.H.A.); (T.C.C.); (J.H.); (F.R.A.); (M.A.); (F.T.-L.)
| |
Collapse
|
14
|
Smith-Osborne L, Duong A, Resendez A, Palme R, Fadok JP. Female dominance hierarchies influence responses to psychosocial stressors. Curr Biol 2023; 33:1535-1549.e5. [PMID: 37003262 PMCID: PMC10321215 DOI: 10.1016/j.cub.2023.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/26/2023] [Accepted: 03/07/2023] [Indexed: 04/03/2023]
Abstract
Social species form dominance hierarchies to ensure survival and promote reproductive success. Traditionally studied in males, rodent hierarchies are considered despotic, and dominant social rank results from a history of winning agonistic encounters. By contrast, female hierarchies are thought to be less despotic, and rank is conferred by intrinsic traits. Both social buffering and elevated social status confer resilience to depression, anxiety, and other consequences of chronic stress. Here, we investigate whether female social hierarchies and individual traits related to social rank likewise influence stress resilience. We observe the formation of dyadic female hierarchies under varying conditions of ambient light and circadian phase and subject mice to two forms of chronic psychosocial stress: social isolation or social instability. We find that stable female hierarchies emerge rapidly in dyads. Individual behavioral and endocrinological traits are characteristic of rank, some of which are circadian phase dependent. Further, female social rank is predicted by behavior and stress status prior to social introduction. Other behavioral characteristics suggest that rank is motivation-based, indicating that female rank identity serves an evolutionarily relevant purpose. Rank is associated with alterations in behavior in response to social instability stress and prolonged social isolation, but the different forms of stress produce disparate rank responses in endocrine status. Histological examination of c-Fos protein expression identified brain regions that respond to social novelty or social reunion following chronic isolation in a rank-specific manner. Collectively, female rank is linked to neurobiology, and hierarchies exert context-specific influence upon stress outcomes.
Collapse
Affiliation(s)
- Lydia Smith-Osborne
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Tulane National Primate Research Center, Covington, LA 70433, USA.
| | - Anh Duong
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Neuroscience Program, Tulane University, New Orleans, LA 70118, USA
| | - Alexis Resendez
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA
| | - Rupert Palme
- Department of Biomedical Sciences, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | - Jonathan P Fadok
- Department of Psychology, Tulane University, New Orleans, LA 70118, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
15
|
Dauchy RT, Blask DE. Vivarium Lighting as an Important Extrinsic Factor Influencing Animal-based Research. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2023; 62:3-25. [PMID: 36755210 PMCID: PMC9936857 DOI: 10.30802/aalas-jaalas-23-000003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 01/22/2023]
Abstract
Light is an extrinsic factor that exerts widespread influence on the regulation of circadian, physiologic, hormonal, metabolic, and behavioral systems of all animals, including those used in research. These wide-ranging biologic effects of light are mediated by distinct photoreceptors, the melanopsin-containing intrinsically photosensitive retinal ganglion cells of the nonvisual system, which interact with the rods and cones of the conventional visual system. Here, we review the nature of light and circadian rhythms, current industry practices and standards, and our present understanding of the neurophysiology of the visual and nonvisual systems. We also consider the implications of this extrinsic factor for vivarium measurement, production, and technological application of light, and provide simple recommendations on artificial lighting for use by regulatory authorities, lighting manufacturers, designers, engineers, researchers, and research animal care staff that ensure best practices for optimizing animal health and wellbeing and, ultimately, improving scientific outcomes.
Collapse
Key Words
- blad, blue-enriched led light at daytime
- clock, circadian locomotor output kaput
- cct, correlated color temperature
- cwf, cool white fluorescent
- iprgc, intrinsically photosensitive retinal ganglion cell
- hiomt, hydroxyindole-o-methyltransferase
- lan, light at night
- led, light-emitting diode
- plr, pupillary light reflex
- scn, suprachiasmatic nuclei
- spd, spectral power distribution
Collapse
Affiliation(s)
- Robert T Dauchy
- Department of Structural and Cellular Biology, Laboratory of Chrono-Neuroendocrine Oncology, Tulane University School of Medicine, New Orleans, Louisiana
| | - David E Blask
- Department of Structural and Cellular Biology, Laboratory of Chrono-Neuroendocrine Oncology, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
16
|
Meyer U. Sources and Translational Relevance of Heterogeneity in Maternal Immune Activation Models. Curr Top Behav Neurosci 2022; 61:71-91. [PMID: 36306055 DOI: 10.1007/7854_2022_398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The epidemiological literature reporting increased risk for neurodevelopmental and psychiatric disorders after prenatal exposure to maternal immune activation (MIA) is still evolving, and so are the attempts to model this association in animals. Epidemiological studies of MIA offer the advantage of directly evaluating human populations but are often limited in their ability to uncover pathogenic mechanisms. Animal models, on the other hand, are limited in their generalizability to psychiatric disorders but have made significant strides toward discovering causal relationships and biological pathways between MIA and neurobiological phenotypes. Like in any other model system, both planned and unplanned sources of variability exist in animal models of MIA. Therefore, the design, implementation, and interpretation of MIA models warrant a careful consideration of these sources, so that appropriate strategies can be developed to handle them satisfactorily. While every research group may have its own strategy to this aim, it is essential to report the methodological details of the chosen MIA model in order to enhance the transparency and comparability of models across research laboratories. Even though it poses a challenge for attempts to compare experimental findings across laboratories, variability does not undermine the utility of MIA models for translational research. In fact, variability and heterogenous outcomes in MIA models offer unique opportunities for new discoveries and developments in this field, including the identification of disease pathways and molecular mechanisms determining susceptibility and resilience to MIA. This review summarizes the most important sources of variability in animal models of MIA and discusses how model variability can be used to investigate neurobiological and immunological factors causing phenotypic heterogeneity in offspring exposed to MIA.
Collapse
Affiliation(s)
- Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
17
|
Hernández-Ramírez S, Salcedo-Tello P, Osorio-Gómez D, Bermúdez-Rattoni F, Pacheco-López G, Ferreira G, Lafenetre P, Guzmán-Ramos KR. Voluntary physical activity improves spatial and recognition memory deficits induced by post-weaning chronic exposure to a high-fat diet. Physiol Behav 2022; 254:113910. [PMID: 35820628 DOI: 10.1016/j.physbeh.2022.113910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/02/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022]
Abstract
Childhood and adolescent exposure to obesogenic environments has contributed to the development of several health disorders, including neurocognitive impairment. Adolescence is a critical neurodevelopmental window highly influenced by environmental factors that affect brain function until adulthood. Post-weaning chronic exposure to a high-fat diet (HFD) adversely affects memory performance; physical activity is one approach to coping with these dysfunctions. Previous studies indicate that voluntary exercise prevents HFD's detrimental effects on memory; however, it remains to evaluate whether it has a remedial/therapeutical effect when introduced after a long-term HFD exposure. This study was conducted on a diet-induced obesity mice model over six months. After three months of HFD exposure (without interrupting the diet) access to voluntary physical activity was provided. HFD produced weight gain, increased adiposity, and impaired glucose tolerance. Voluntary physical exercise ameliorated glucose tolerance and halted weight gain and fat accumulation. Additionally, physical activity mitigated HFD-induced spatial and recognition memory impairments. Our data indicate that voluntary physical exercise starting after several months of periadolescent HFD exposure reverses metabolic and cognitive alterations demonstrating that voluntary exercise, in addition to its known preventive effect, also has a restorative impact on metabolism and cognition dysfunctions associated with obesity.
Collapse
Affiliation(s)
- Susana Hernández-Ramírez
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Av. de las Garzas No. 10, Lerma de Villada, Estado de México, C.P. 52005, Mexico
| | - Pamela Salcedo-Tello
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana (UAM), Unidad Lerma. Av. de las Garzas No. 10, Col. el Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico
| | - Daniel Osorio-Gómez
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México (UNAM). Circuito Exterior, Ciudad Universitaria, 04510 Mexico City
| | - Federico Bermúdez-Rattoni
- División de Neurociencias. Instituto de Fisiología Celular. Universidad Nacional Autónoma de México (UNAM). Circuito Exterior, Ciudad Universitaria, 04510 Mexico City
| | - Gustavo Pacheco-López
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana (UAM), Unidad Lerma. Av. de las Garzas No. 10, Col. el Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico
| | - Guillaume Ferreira
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro Laboratory, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Pauline Lafenetre
- Université de Bordeaux, Nutrition and Integrative Neurobiology, UMR 1286, Bordeaux, France
| | - Kioko R Guzmán-Ramos
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana (UAM), Unidad Lerma. Av. de las Garzas No. 10, Col. el Panteón, Lerma de Villada, Estado de México, C.P. 52005, Mexico.
| |
Collapse
|
18
|
Mueller FS, Amport R, Notter T, Schalbetter SM, Lin HY, Garajova Z, Amini P, Weber-Stadlbauer U, Markkanen E. Deficient DNA base-excision repair in the forebrain leads to a sex-specific anxiety-like phenotype in mice. BMC Biol 2022; 20:170. [PMID: 35907861 PMCID: PMC9339204 DOI: 10.1186/s12915-022-01377-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 07/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background Neuropsychiatric disorders, such as schizophrenia (SZ) and autism spectrum disorder (ASD), are common, multi-factorial and multi-symptomatic disorders. Ample evidence implicates oxidative stress, deficient repair of oxidative DNA lesions and DNA damage in the development of these disorders. However, it remains unclear whether insufficient DNA repair and resulting DNA damage are causally connected to their aetiopathology, or if increased levels of DNA damage observed in patient tissues merely accumulate as a consequence of cellular dysfunction. To assess a potential causal role for deficient DNA repair in the development of these disorders, we behaviourally characterized a mouse model in which CaMKIIa-Cre-driven postnatal conditional knockout (KO) of the core base-excision repair (BER) protein XRCC1 leads to accumulation of unrepaired DNA damage in the forebrain. Results CaMKIIa-Cre expression caused specific deletion of XRCC1 in the dorsal dentate gyrus (DG), CA1 and CA2 and the amygdala and led to increased DNA damage therein. While motor coordination, cognition and social behaviour remained unchanged, XRCC1 KO in the forebrain caused increased anxiety-like behaviour in males, but not females, as assessed by the light–dark box and open field tests. Conversely, in females but not males, XRCC1 KO caused an increase in learned fear-related behaviour in a cued (Pavlovian) fear conditioning test and a contextual fear extinction test. The relative density of the GABA(A) receptor alpha 5 subunit (GABRA5) was reduced in the amygdala and the dorsal CA1 in XRCC1 KO females, whereas male XRCC1 KO animals exhibited a significant reduction of GABRA5 density in the CA3. Finally, assessment of fast-spiking, parvalbumin-positive (PV) GABAergic interneurons revealed a significant increase in the density of PV+ cells in the DG of male XRCC1 KO mice, while females remained unchanged. Conclusions Our results suggest that accumulation of unrepaired DNA damage in the forebrain alters the GABAergic neurotransmitter system and causes behavioural deficits in relation to innate and learned anxiety in a sex-dependent manner. Moreover, the data uncover a previously unappreciated connection between BER deficiency, unrepaired DNA damage in the hippocampus and a sex-specific anxiety-like phenotype with implications for the aetiology and therapy of neuropsychiatric disorders. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01377-1.
Collapse
Affiliation(s)
- Flavia S Mueller
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - René Amport
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Tina Notter
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, Faculty of Science, University of Zurich, 8057, Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Han-Yu Lin
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Zuzana Garajova
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Parisa Amini
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland. .,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057, Zurich, Switzerland.
| |
Collapse
|
19
|
Schoettner K, Alonso M, Button M, Goldfarb C, Herrera J, Quteishat N, Meyer C, Bergdahl A, Amir S. Characterization of Affective Behaviors and Motor Functions in Mice With a Striatal-Specific Deletion of Bmal1 and Per2. Front Physiol 2022; 13:922080. [PMID: 35755440 PMCID: PMC9216244 DOI: 10.3389/fphys.2022.922080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
The expression of circadian clock genes, either centrally or in the periphery, has been shown to play an integral role in the control of behavior. Brain region-specific downregulation of clock genes revealed behavioral phenotypes associated with neuropsychiatric disorders and neurodegenerative disease. The specific function of the clock genes as well as the underlying mechanisms that contribute to the observed phenotypes, however, are not yet fully understood. We assessed anxiety- and depressive-like behavior and motor functions in male and female mice with a conditional ablation of Bmal1 or Per2 from medium spiny neurons (MSNs) of the striatum as well as mice lacking one copy of Gpr88. Whereas the conditional knockout of Bmal1 and Per2 had mild effects on affective behaviors, a pronounced effect on motor functions was found in Bmal1 knockout mice. Subsequent investigation revealed an attenuated response of Bmal1 knockout mice to dopamine receptor type 1 agonist treatment, independently of the expression of targets of the dopamine signaling pathway or mitochondrial respiration in MSNs. The study thus suggests a potential interaction of Bmal1 within the direct dopamine signaling pathway, which may provide the link to a shared, MSN-dependent mechanism regulating affective behavior and motor function in mice.
Collapse
Affiliation(s)
- Konrad Schoettner
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Mariana Alonso
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Margo Button
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Cassandra Goldfarb
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Juliana Herrera
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Nour Quteishat
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Christiane Meyer
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| | - Andreas Bergdahl
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, QC, Canada
| | - Shimon Amir
- Department of Psychology, Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada
| |
Collapse
|
20
|
Bloch S, Holleran KM, Kash TL, Vazey EM, Rinker JA, Lebonville CL, O'Hara K, Lopez MF, Jones SR, Grant KA, Becker HC, Mulholland PJ. Assessing negative affect in mice during abstinence from alcohol drinking: Limitations and future challenges. Alcohol 2022; 100:41-56. [PMID: 35181404 PMCID: PMC8983487 DOI: 10.1016/j.alcohol.2022.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/09/2022] [Accepted: 02/09/2022] [Indexed: 01/09/2023]
Abstract
Alcohol use disorder (AUD) is frequently comorbid with mood disorders, and these co-occurring neuropsychiatric disorders contribute to the development and maintenance of alcohol dependence and relapse. In preclinical models, mice chronically exposed to alcohol display anxiety-like and depressive-like behaviors during acute withdrawal and protracted abstinence. However, in total, results from studies using voluntary alcohol-drinking paradigms show variable behavioral outcomes in assays measuring negative affective behaviors. Thus, the main objective of this review is to summarize the literature on the variability of negative affective behaviors in mice after chronic alcohol exposure. We compare the behavioral phenotypes that emerge during abstinence across different exposure models, including models of alcohol and stress interactions. The complicated outcomes from these studies highlight the difficulties of assessing negative affective behaviors in mouse models designed for the study of AUD. We discuss new behavioral assays, comprehensive platforms, and unbiased machine-learning algorithms as promising approaches to better understand the interaction between alcohol and negative affect in mice. New data-driven approaches in the understanding of mouse behavior hold promise for improving the identification of mechanisms, cell subtypes, and neurocircuits that mediate negative affect. In turn, improving our understanding of the neurobehavioral basis of alcohol-associated negative affect will provide a platform to test hypotheses in mouse models that aim to improve the development of more effective strategies for treating individuals with AUD and co-occurring mood disorders.
Collapse
Affiliation(s)
- Solal Bloch
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Katherine M Holleran
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Thomas L Kash
- Bowles Center for Alcohol Studies, Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Elena M Vazey
- Department of Biology, University of Massachusetts Amherst, Amherst, MA 01003, United States
| | - Jennifer A Rinker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Christina L Lebonville
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Krysten O'Hara
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Marcelo F Lopez
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Sara R Jones
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, United States
| | - Howard C Becker
- Department of Psychiatry & Behavioral Sciences, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC 29425, United States.
| |
Collapse
|
21
|
von Gall C. The Effects of Light and the Circadian System on Rhythmic Brain Function. Int J Mol Sci 2022; 23:ijms23052778. [PMID: 35269920 PMCID: PMC8911243 DOI: 10.3390/ijms23052778] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 02/06/2023] Open
Abstract
Life on earth has evolved under the influence of regularly recurring changes in the environment, such as the 24 h light/dark cycle. Consequently, organisms have developed endogenous clocks, generating 24 h (circadian) rhythms that serve to anticipate these rhythmic changes. In addition to these circadian rhythms, which persist in constant conditions and can be entrained to environmental rhythms, light drives rhythmic behavior and brain function, especially in nocturnal laboratory rodents. In recent decades, research has made great advances in the elucidation of the molecular circadian clockwork and circadian light perception. This review summarizes the role of light and the circadian clock in rhythmic brain function, with a focus on the complex interaction between the different components of the mammalian circadian system. Furthermore, chronodisruption as a consequence of light at night, genetic manipulation, and neurodegenerative diseases is briefly discussed.
Collapse
Affiliation(s)
- Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich Heine University, 40225 Dusseldorf, Germany
| |
Collapse
|
22
|
Harry GJ, McBride S, Witchey SK, Mhaouty-Kodja S, Trembleau A, Bridge M, Bencsik A. Roadbumps at the Crossroads of Integrating Behavioral and In Vitro Approaches for Neurotoxicity Assessment. FRONTIERS IN TOXICOLOGY 2022; 4:812863. [PMID: 35295216 PMCID: PMC8915899 DOI: 10.3389/ftox.2022.812863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/25/2022] [Indexed: 12/15/2022] Open
Abstract
With the appreciation that behavior represents the integration and complexity of the nervous system, neurobehavioral phenotyping and assessment has seen a renaissance over the last couple of decades, resulting in a robust database on rodent performance within various testing paradigms, possible associations with human disorders, and therapeutic interventions. The interchange of data across behavior and other test modalities and multiple model systems has advanced our understanding of fundamental biology and mechanisms associated with normal functions and alterations in the nervous system. While there is a demonstrated value and power of neurobehavioral assessments for examining alterations due to genetic manipulations, maternal factors, early development environment, the applied use of behavior to assess environmental neurotoxicity continues to come under question as to whether behavior represents a sensitive endpoint for assessment. Why is rodent behavior a sensitive tool to the neuroscientist and yet, not when used in pre-clinical or chemical neurotoxicity studies? Applying new paradigms and evidence on the biological basis of behavior to neurobehavioral testing requires expertise and refinement of how such experiments are conducted to minimize variability and maximize information. This review presents relevant issues of methods used to conduct such test, sources of variability, experimental design, data analysis, interpretation, and reporting. It presents beneficial and critical limitations as they translate to the in vivo environment and considers the need to integrate across disciplines for the best value. It proposes that a refinement of behavioral assessments and understanding of subtle pronounced differences will facilitate the integration of data obtained across multiple approaches and to address issues of translation.
Collapse
Affiliation(s)
- G. Jean Harry
- Neurotoxicology Group, Molecular Toxicology Branch, Division National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Sandra McBride
- Social & Scientific Systems, Inc., a DLH Holdings Company, Durham, NC, United States
| | - Shannah K. Witchey
- Division National Toxicology Program, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Sakina Mhaouty-Kodja
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Paris, France
| | - Alain Trembleau
- Sorbonne Université, CNRS UMR8246, Inserm U1130, Institut de Biologie Paris Seine (IBPS), Neuroscience Paris Seine (NPS), Paris, France
| | - Matthew Bridge
- Social & Scientific Systems, Inc., a DLH Holdings Company, Durham, NC, United States
| | - Anna Bencsik
- Anses Laboratoire de Lyon, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), Université de Lyon 1, Lyon, France
| |
Collapse
|
23
|
Nigri M, Åhlgren J, Wolfer DP, Voikar V. Role of Environment and Experimenter in Reproducibility of Behavioral Studies With Laboratory Mice. Front Behav Neurosci 2022; 16:835444. [PMID: 35250504 PMCID: PMC8895324 DOI: 10.3389/fnbeh.2022.835444] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
Behavioral phenotyping of mice has received a great deal of attention during the past three decades. However, there is still a pressing need to understand the variability caused by environmental and biological factors, human interference, and poorly standardized experimental protocols. The inconsistency of results is often attributed to the inter-individual difference between the experimenters and environmental conditions. The present work aims to dissect the combined influence of the experimenter and the environment on the detection of behavioral traits in two inbred strains most commonly used in behavioral genetics due to their contrasting phenotypes, the C57BL/6J and DBA/2J mice. To this purpose, the elevated O-maze, the open field with object, the accelerating rotarod and the Barnes maze tests were performed by two experimenters in two diverse laboratory environments. Our findings confirm the well-characterized behavioral differences between these strains in exploratory behavior, motor performance, learning and memory. Moreover, the results demonstrate how the experimenter and the environment influence the behavioral tests with a variable-dependent effect, often with mutually exclusive contributions. In this context, our study highlights how both the experimenter and the environment can have an impact on the strain effect size without altering the direction of the conclusions. Importantly, the general agreement on the results is reached by converging evidence from multiple measures addressing the same trait. In conclusion, the present work elucidates the contribution of both the experimenter and the laboratory environment in the intricate field of reproducibility in mouse behavioral phenotyping.
Collapse
Affiliation(s)
- Martina Nigri
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland
- *Correspondence: Martina Nigri,
| | - Johanna Åhlgren
- Laboratory Animal Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - David P. Wolfer
- Faculty of Medicine, Institute of Anatomy, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland
| | - Vootele Voikar
- Laboratory Animal Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
- Vootele Voikar,
| |
Collapse
|
24
|
Francois M, Canal Delgado I, Shargorodsky N, Leu CS, Zeltser L. Assessing the effects of stress on feeding behaviors in laboratory mice. eLife 2022; 11:e70271. [PMID: 35167441 PMCID: PMC8846584 DOI: 10.7554/elife.70271] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022] Open
Abstract
Stress often affects eating behaviors, increasing caloric intake in some individuals and decreasing it in others. The determinants of feeding responses to stress are unknown, in part because this issue is rarely studied in rodents. We focused our efforts on the novelty-suppressed feeding (NSF) assay, which uses latency to eat as readout of anxiety-like behavior, but rarely assesses feeding per se. We explored how key variables in experimental paradigms - estrous and diurnal cyclicity, age and duration of social isolation, prandial state, diet palatability, and elevated body weight - influence stress-induced anxiety-like behavior and food intake in male and female C57BL/6J mice. Latency to eat in the novel environment is increased in both sexes across most of the conditions tested, while effects on caloric intake are variable. In the common NSF assay (i.e., lean mice in the light cycle), sex-specific effects of the length of social isolation, and not estrous cyclicity, are the main source of variability. Under conditions that are more physiologically relevant for humans (i.e., overweight mice in the active phase), the novel stress now elicits robust hyperphagia in both sexes . This novel model of stress eating can be used to identify underlying neuroendocrine and neuronal substrates. Moreover, these studies can serve as a framework to integrate cross-disciplinary studies of anxiety and feeding related behaviors in rodents.
Collapse
Affiliation(s)
- Marie Francois
- Naomi Berrie Diabetes Center, Division of Molecular Genetics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Isabella Canal Delgado
- Naomi Berrie Diabetes Center, Division of Molecular Genetics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Nikolay Shargorodsky
- Naomi Berrie Diabetes Center, Division of Molecular Genetics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Cheng-Shiun Leu
- Naomi Berrie Diabetes Center, Division of Molecular Genetics, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Biostatistics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Lori Zeltser
- Naomi Berrie Diabetes Center, Division of Molecular Genetics, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia University Irving Medical CenterNew YorkUnited States
| |
Collapse
|
25
|
Tsao CH, Flint J, Huang GJ. Influence of diurnal phase on behavioral tests of sensorimotor performance, anxiety, learning and memory in mice. Sci Rep 2022; 12:432. [PMID: 35013366 PMCID: PMC8748730 DOI: 10.1038/s41598-021-03155-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
Behavioral measurements in mice are critical tools used to evaluate the effects of interventions. Whilst mice are nocturnal animals, many studies conduct behavioral tests during the day. To better understand the effects of diurnal rhythm on mouse behaviors, we compared the results from behavioral tests conducted in the active and inactive phases. C57BL/6 mice were used in this study; we focus on sensorimotor performance, anxiety, learning and memory. Overall, our results show mice exhibit slightly higher cutaneous sensitivity, better long-term contextual memory, and a greater active avoidance escape response during the active phase. We did not observe significant differences in motor coordination, anxiety, or spatial learning and memory. Furthermore, apart from the elevated-O-maze, there was no remarkable sex effect among these tests. This study provides information on the effects of different diurnal phases on types of behavior and demonstrates the importance of the circadian cycle on learning and memory. Although we did not detect differences in anxiety and spatial learning/memory, diurnal rhythm may interact with other factors to influence these behaviors.
Collapse
Affiliation(s)
- Chi-Hui Tsao
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Jonathan Flint
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Guo-Jen Huang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 33302, Taiwan.
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Taoyuan 333, Taiwan.
- Healthy Aging Research Center, Chang Gung University, Taoyuan, 33302, Taiwan.
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, 33302, Taiwan.
| |
Collapse
|
26
|
Spatial working memory is disparately interrelated with social status through different developmental stages in rats. Behav Brain Res 2022; 416:113547. [PMID: 34437940 DOI: 10.1016/j.bbr.2021.113547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/15/2021] [Accepted: 08/21/2021] [Indexed: 02/02/2023]
Abstract
Social life necessitates cognitive competence to meet the dynamic demands of social development. The formation of dominance hierarchy is a general phenomenon in social groups. As an essential element of executive and cognitive function, working memory could influence and be influenced by social status in a dominance hierarchy. However, the direction and degree of the association between them through different developmental stages remain unclear. To address this issue and clarify the "cause or consequence" problem, we investigated the spatial working memory performance in a Y-maze and Morris water maze in home-caged sibling Wistar rats (N = 26 cages, three rats/cage) through three stages of their life: before (week 7), during (week 10), and after (week 20) assumed timings of the social dominance hierarchy formation (SDHF). We used the social dominance tube test during the assumed time of hierarchy formation (weeks 9-11) to measure the relative dominance status in each cage. Here, we found that higher working memory index before SDHF could be predictive of later acquisition of higher social status. Working memory performance declined for all animals during SDHF, in which agonistic conflicts are increased. However, living within an established hierarchical social network for several weeks deteriorated the working memory performance of dominant and middle-ranked animals, while the performance of subordinates improved and got significantly better than higher-ranked animals. In conclusion, while working memory and social status were correlated positively before dominance hierarchy formation, there was a trade-off between them after the formation of it. In contrast to the common view, these results highlight the adverse effect of higher social status on cognitive behavior.
Collapse
|
27
|
Yates AG, Weglinski CM, Ying Y, Dunstan IK, Strekalova T, Anthony DC. Nafamostat reduces systemic inflammation in TLR7-mediated virus-like illness. J Neuroinflammation 2022; 19:8. [PMID: 34991643 PMCID: PMC8734544 DOI: 10.1186/s12974-021-02357-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The serine protease inhibitor nafamostat has been proposed as a treatment for COVID-19, by inhibiting TMPRSS2-mediated viral cell entry. Nafamostat has been shown to have other, immunomodulatory effects, which may be beneficial for treatment, however animal models of ssRNA virus infection are lacking. In this study, we examined the potential of the dual TLR7/8 agonist R848 to mimic the host response to an ssRNA virus infection and the associated behavioural response. In addition, we evaluated the anti-inflammatory effects of nafamostat in this model. METHODS CD-1 mice received an intraperitoneal injection of R848 (200 μg, prepared in DMSO, diluted 1:10 in saline) or diluted DMSO alone, and an intravenous injection of either nafamostat (100 μL, 3 mg/kg in 5% dextrose) or 5% dextrose alone. Sickness behaviour was determined by temperature, food intake, sucrose preference test, open field and forced swim test. Blood and fresh liver, lung and brain were collected 6 h post-challenge to measure markers of peripheral and central inflammation by blood analysis, immunohistochemistry and qPCR. RESULTS R848 induced a robust inflammatory response, as evidenced by increased expression of TNF, IFN-γ, CXCL1 and CXCL10 in the liver, lung and brain, as well as a sickness behaviour phenotype. Exogenous administration of nafamostat suppressed the hepatic inflammatory response, significantly reducing TNF and IFN-γ expression, but had no effect on lung or brain cytokine production. R848 administration depleted circulating leukocytes, which was restored by nafamostat treatment. CONCLUSIONS Our data indicate that R848 administration provides a useful model of ssRNA virus infection, which induces inflammation in the periphery and CNS, and virus infection-like illness. In turn, we show that nafamostat has a systemic anti-inflammatory effect in the presence of the TLR7/8 agonist. Therefore, the results indicate that nafamostat has anti-inflammatory actions, beyond its ability to inhibit TMPRSS2, that might potentiate its anti-viral actions in pathologies such as COVID-19.
Collapse
Affiliation(s)
- Abi G Yates
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Caroline M Weglinski
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
| | - Yuxin Ying
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
| | - Isobel K Dunstan
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK
| | - Tatyana Strekalova
- Sechenov First Moscow State Medical University, Moscow, Russia
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Daniel C Anthony
- Department of Pharmacology, The University of Oxford, Mansfield Road, Oxford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russia.
- University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
28
|
Silva S, Bicker J, Falcão A, Fortuna A. Antidepressants and Circadian Rhythm: Exploring Their Bidirectional Interaction for the Treatment of Depression. Pharmaceutics 2021; 13:1975. [PMID: 34834391 PMCID: PMC8624696 DOI: 10.3390/pharmaceutics13111975] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 01/07/2023] Open
Abstract
Scientific evidence that circadian rhythms affect pharmacokinetics and pharmacodynamics has highlighted the importance of drug dosing-time. Circadian oscillations alter drug absorption, distribution, metabolism, and excretion (ADME) as well as intracellular signaling systems, target molecules (e.g., receptors, transporters, and enzymes), and gene transcription. Although several antidepressant drugs are clinically available, less than 50% of depressed patients respond to first-line pharmacological treatments. Chronotherapeutic approaches to enhance the effectiveness of antidepressants are not completely known. Even so, experimental results found until this day suggest a positive influence of drug dosing-time on the efficacy of depression therapy. On the other hand, antidepressants have also demonstrated to modulate circadian rhythmicity and sleep-wake cycles. This review aims to evidence the potential of chronotherapy to improve the efficacy and/or safety of antidepressants. It includes pre-clinical and clinical studies that demonstrate the relevance of determining the most appropriate time of administration for antidepressant drugs. In parallel, their positive influence on the resynchronization of disrupted circadian rhythms is also herein discussed. It is expected that this review will promote the investigation of chronotherapy for the treatment of depression, contribute to a better understanding of the relationship between antidepressants and circadian rhythms, and consequently promote the development of new therapeutics.
Collapse
Affiliation(s)
- Soraia Silva
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal; (S.S.); (A.F.); (A.F.)
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Azinhaga Sta. Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
29
|
Schalbetter SM, Mueller FS, Scarborough J, Richetto J, Weber-Stadlbauer U, Meyer U, Notter T. Oral application of clozapine-N-oxide using the micropipette-guided drug administration (MDA) method in mouse DREADD systems. Lab Anim (NY) 2021; 50:69-75. [PMID: 33619409 DOI: 10.1038/s41684-021-00723-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 01/19/2021] [Indexed: 01/31/2023]
Abstract
The designer receptor exclusively activated by designer drugs (DREADD) system is one of the most widely used chemogenetic techniques to modulate the activity of cell populations in the brains of behaving animals. DREADDs are activated by acute or chronic administration of their ligand, clozapine-N-oxide (CNO). There is, however, a current lack of a non-invasive CNO administration technique that can control for drug timing and dosing without inducing substantial distress for the animals. Here, we evaluated whether the recently developed micropipette-guided drug administration (MDA) method, which has been used as a non-invasive and minimally stressful alternative to oral gavages, may be applied to administer CNO orally to activate DREADDs in a dosing- and timing-controlled manner. Unlike standard intraperitoneal injections, administration of vehicle substances via MDA did not elevate plasma levels of the major stress hormone, corticosterone, and did not attenuate exploratory activity in the open field test. At the same time, however, administration of CNO via MDA or intraperitoneally was equally efficient in activating hM3DGq-expressing neurons in the medial prefrontal cortex, as evident by time-dependent increases in mRNA levels of neuronal immediate early genes (cFos, Arc and Zif268) and cFos-immunoreactive neurons. Compared to vehicle given via MDA, oral administration of CNO via MDA was also found to potently increase locomotor activity in mice that express hM3DGq in prefrontal neurons. Taken together, our study confirms the effectiveness of CNO given orally via MDA and provides a novel method for non-stressful, yet well controllable CNO treatments in mouse DREADD systems.
Collapse
Affiliation(s)
- Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Joseph Scarborough
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Tina Notter
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, Wales, UK.
| |
Collapse
|
30
|
Neuronal activity increases translocator protein (TSPO) levels. Mol Psychiatry 2021; 26:2025-2037. [PMID: 32398717 PMCID: PMC8440208 DOI: 10.1038/s41380-020-0745-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022]
Abstract
The mitochondrial protein, translocator protein (TSPO), is a widely used biomarker of neuroinflammation, but its non-selective cellular expression pattern implies roles beyond inflammatory processes. In the present study, we investigated whether neuronal activity modifies TSPO levels in the adult central nervous system. First, we used single-cell RNA sequencing to generate a cellular landscape of basal TSPO gene expression in the hippocampus of adult (12 weeks old) C57BL6/N mice, followed by confocal laser scanning microscopy to verify TSPO protein in neuronal and non-neuronal cell populations. We then quantified TSPO mRNA and protein levels after stimulating neuronal activity with distinct stimuli, including designer receptors exclusively activated by designer drugs (DREADDs), exposure to a novel environment and acute treatment with the psychostimulant drug, amphetamine. Single-cell RNA sequencing demonstrated a non-selective and multi-cellular gene expression pattern of TSPO at basal conditions in the adult mouse hippocampus. Confocal laser scanning microscopy confirmed that TSPO protein is present in neuronal and non-neuronal (astrocytes, microglia, vascular endothelial cells) cells of cortical (medial prefrontal cortex) and subcortical (hippocampus) brain regions. Stimulating neuronal activity through chemogenetic (DREADDs), physiological (novel environment exposure) or psychopharmacological (amphetamine treatment) approaches led to consistent increases in TSPO gene and protein levels in neurons, but not in microglia or astrocytes. Taken together, our findings show that neuronal activity has the potential to modify TSPO levels in the adult central nervous system. These findings challenge the general assumption that altered TSPO expression or binding unequivocally mirrors ongoing neuroinflammation and emphasize the need to consider non-inflammatory interpretations in some physiological or pathological contexts.
Collapse
|
31
|
Elsilä LV, Korhonen N, Hyytiä P, Korpi ER. Acute Lysergic Acid Diethylamide Does Not Influence Reward-Driven Decision Making of C57BL/6 Mice in the Iowa Gambling Task. Front Pharmacol 2020; 11:602770. [PMID: 33343373 PMCID: PMC7745734 DOI: 10.3389/fphar.2020.602770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/09/2020] [Indexed: 12/25/2022] Open
Abstract
While interest in psychedelic drugs in the fields of psychiatry and neuroscience has re-emerged in recent last decades, the general understanding of the effects of these drugs remains deficient. In particular, there are gaps in knowledge on executive functions and goal-directed behaviors both in humans and in commonly used animal models. The effects of acute doses of psychedelic lysergic acid diethylamide (LSD) on reward-driven decision making were explored using the mouse version of the Iowa Gambling Task. A total of 15 mice were trained to perform in a touch-screen adaptation of the rodent version of the Iowa Gambling Task, after which single acute doses of LSD (0.025, 0.1, 0.2, 0.4 mg/kg), serotonin 2A receptor-selective agonist 25CN-NBOH (1.5 mg/kg), d-amphetamine (2.0 mg/kg), and saline were administered before the trial. 25CN-NBOH and the three lowest doses of LSD showed no statistically significant changes in option selection or in general functioning during the gambling task trials. The highest dose of LSD (0.4 mg/kg) significantly decreased premature responding and increased the omission rate, but had no effect on option selection in comparison with the saline control. Amphetamine significantly decreased the correct responses and premature responding while increasing the omission rate. In conclusion, mice can perform previously learned, reward-driven decision-making tasks while under the acute influence of LSD at a commonly used dose range.
Collapse
Affiliation(s)
- Lauri V Elsilä
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nuppu Korhonen
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Petri Hyytiä
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
32
|
Cross-site Reproducibility of Social Deficits in Group-housed BTBR Mice Using Automated Longitudinal Behavioural Monitoring. Neuroscience 2020; 445:95-108. [DOI: 10.1016/j.neuroscience.2020.04.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/16/2022]
|
33
|
Hanifin JP, Dauchy RT, Blask DE, Hill SM, Brainard GC. Relevance of Electrical Light on Circadian, Neuroendocrine, and Neurobehavioral Regulation in Laboratory Animal Facilities. ILAR J 2020; 60:150-158. [PMID: 33094817 DOI: 10.1093/ilar/ilaa010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 03/25/2020] [Accepted: 04/15/2020] [Indexed: 02/03/2023] Open
Abstract
Light is a key extrinsic factor to be considered in operations and design of animal room facilities. Over the past four decades, many studies on typical laboratory animal populations have demonstrated impacts on neuroendocrine, neurobehavioral, and circadian physiology. These effects are regulated independently from the defined physiology for the visual system. The range of physiological responses that oscillate with the 24 hour rhythm of the day include sleep and wakefulness, body temperature, hormonal secretion, and a wide range of other physiological parameters. Melatonin has been the chief neuroendocrine hormone studied, but acute light-induced effects on corticosterone as well as other hormones have also been observed. Within the last two decades, a new photosensory system in the mammalian eye has been discovered. A small set of retinal ganglion cells, previously thought to function as a visual output neuron, have been shown to be directly photosensitive and act differently from the classic photoreceptors of the visual system. Understanding the effects of light on mammalian physiology and behavior must take into account how the classical visual photoreceptors and the newly discovered ipRGC photoreceptor systems interact. Scientists and facility managers need to appreciate lighting impacts on circadian, neuroendocrine, and neurobehavioral regulation in order to improve lighting of laboratory facilities to foster optimum health and well-being of animals.
Collapse
Affiliation(s)
- John P Hanifin
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Robert T Dauchy
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane, Louisiana
| | - David E Blask
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane, Louisiana
| | - Steven M Hill
- Department of Structural and Cellular Biology, Tulane University School of Medicine, Tulane, Louisiana
| | - George C Brainard
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Reproducibility of animal research in light of biological variation. Nat Rev Neurosci 2020; 21:384-393. [PMID: 32488205 DOI: 10.1038/s41583-020-0313-3] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2020] [Indexed: 12/16/2022]
Abstract
Context-dependent biological variation presents a unique challenge to the reproducibility of results in experimental animal research, because organisms' responses to experimental treatments can vary with both genotype and environmental conditions. In March 2019, experts in animal biology, experimental design and statistics convened in Blonay, Switzerland, to discuss strategies addressing this challenge. In contrast to the current gold standard of rigorous standardization in experimental animal research, we recommend the use of systematic heterogenization of study samples and conditions by actively incorporating biological variation into study design through diversifying study samples and conditions. Here we provide the scientific rationale for this approach in the hope that researchers, regulators, funders and editors can embrace this paradigm shift. We also present a road map towards better practices in view of improving the reproducibility of animal research.
Collapse
|
35
|
Bikovski L, Robinson L, Konradsson-Geuken A, Kullander K, Viereckel T, Winberg S, Roman E, Tsoory M. Lessons, insights and newly developed tools emerging from behavioral phenotyping core facilities. J Neurosci Methods 2020; 334:108597. [PMID: 31987912 DOI: 10.1016/j.jneumeth.2020.108597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 01/24/2023]
Abstract
Scientific investigations, in general, and research in neuroscience, in particular, are becoming ever more complex and require the integration of different techniques. Behavioral assays, which are among the most frequently used methodologies in neuroscience, nowadays rely on advanced, sophisticated technologies that require proficient application. Therefore, behavioral core facilities are becoming essential support units, as they provide the specialized expert research services needed to conduct advanced neuroscience. We here review the lessons learned and insights gathered from managing behavioral core facilities in different academic research institutes. This review addresses several issues, including: the advantages of behavioral core facilities, considerations for establishing a behavioral core facility, and the methodological advances made through calibration and standardization of assay protocols and the development of new assays. Collectively, the review highlights the benefits of both working within and collaborating with behavioral core facility units and emphasizes the potential progress in neuro-phenotyping that such facilities provide.
Collapse
Affiliation(s)
- Lior Bikovski
- The Myers Neuro-Behavioral Core Facility, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; School of Behavioral Sciences, Netanya Academic College, 4223587, Netanya, Israel.
| | - Lianne Robinson
- Behavioural Neuroscience Core Facility, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK; Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | | | - Klas Kullander
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Thomas Viereckel
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Svante Winberg
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Erika Roman
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden; Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
36
|
Kim YM, Snijders AM, Brislawn CJ, Stratton KG, Zink EM, Fansler SJ, Metz TO, Mao JH, Jansson JK. Light-Stress Influences the Composition of the Murine Gut Microbiome, Memory Function, and Plasma Metabolome. Front Mol Biosci 2019; 6:108. [PMID: 31681796 PMCID: PMC6813214 DOI: 10.3389/fmolb.2019.00108] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome plays an important role in the mammalian host and when in proper balance helps protect health and prevent disease. Host environmental stress and its influence on the gut microbiome, health, and disease is an emerging area of research. Exposures to unnatural light cycles are becoming increasingly common due to travel and shift work. However, much remains unknown about how these changes influence the microbiome and host health. This information is needed to understand and predict the relationship between the microbiome and host response to altered sleep cycles. In the present study, we exposed three cohorts of mice to different light cycle regimens for 12 consecutive weeks; including continuous light, continuous dark, and a standard light dark regimen consisting of 12 h light followed by 12 h of dark. After exposure, motor and memory behavior, and the composition of the fecal microbiome and plasma metabolome were measured. Memory potential was significantly reduced in mice exposed to continuous light, whereas rotarod performance was minimally affected. The overall composition of the microbiome was relatively constant over time. However, Bacteroidales Rikenellaceae was relatively more abundant in mice exposed to continuous dark, while Bacteroidales S24-7 was relatively more abundant in mice exposed to continuous light. The plasma metabolome after the continuous dark exposure differed from the other exposure conditions. Several plasma metabolites, including glycolic acid, tryptophan, pyruvate, and several unidentified metabolites, were correlated to continuous dark and light exposure conditions. Networking analyses showed that serotonin was positively correlated with three microbial families (Rikenellaceae, Ruminococcaceae, and Turicibacteraceae), while tryptophan was negatively correlated with abundance of Bacteroidales S24-7 based on light exposure. This study provides the foundation for future studies into the mechanisms underlying the role of the gut microbiome on the murine host during light-dark stress.
Collapse
Affiliation(s)
- Young-Mo Kim
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Colin J Brislawn
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Kelly G Stratton
- Computing and Analytics Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Erika M Zink
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Sarah J Fansler
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Janet K Jansson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| |
Collapse
|
37
|
Gururajan A, Reif A, Cryan JF, Slattery DA. The future of rodent models in depression research. Nat Rev Neurosci 2019; 20:686-701. [DOI: 10.1038/s41583-019-0221-6] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2019] [Indexed: 12/15/2022]
|
38
|
Weber-Stadlbauer U, Meyer U. Challenges and opportunities of a-priori and a-posteriori variability in maternal immune activation models. Curr Opin Behav Sci 2019. [DOI: 10.1016/j.cobeha.2019.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
39
|
Kawai H, Iwadate R, Ishibashi T, Kudo N, Kawashima Y, Mitsumoto A. Antidepressants with different mechanisms of action show different chronopharmacological profiles in the tail suspension test in mice. Chronobiol Int 2019; 36:1194-1207. [PMID: 31198056 DOI: 10.1080/07420528.2019.1625360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The circadian system regulates sleep/wake cycles, metabolism, mood, and other functions. It also influences medication efficacy. In this study, we studied the chronopharmacological profiles of antidepressants with various modes of action. We also investigated the effects of dosing time on the pharmacological activity of several antidepressants acting on serotonergic, noradrenergic, and/or dopaminergic neurons. C57BL/6 mice were intraperitoneally administered fluoxetine, imipramine, venlafaxine, or bupropion at 08:00 h (morning), 14:00 h (mid-day), 20:00 h (evening), or 02:00 h (mid-night). Antidepressant activity was evaluated by the tail suspension test. All antidepressants reduced immobility, and their activities varied according to the dosing time. Fluoxetine and imipramine induced relatively strong rhythms with high amplitudes. Their maximal effects were observed in the morning and evening, respectively. Venlafaxine and bupropion induced weak rhythms with maximal effects in the evening and dawn, respectively. These results suggest that the antidepressant activity is associated with circadian fluctuation, and antidepressants with different modes of action have different chronopharmacological profiles. They affect locomotor activity in animals placed in novel (unfamiliar) environments. Fluoxetine, imipramine, and venlafaxine reduced locomotor activity, whereas bupropion increased it. The effects on locomotor activity also vary with circadian rhythm, and the tested drugs showed a maximal effect during the light phase. The peak time was different from that in TST. Plasma and brain levels of all drugs were slightly higher in the morning than in the evening. The dosing time dependency of the antidepressant activity did not correlate with the sedative/stimulatory activity or tissue drug level. Therefore, these latter two factors may have only a small impact on circadian antidepressant activity fluctuations. The relative activity of the serotonergic, noradrenergic, and dopaminergic systems may determine the chronopharmacological profiles of each drug. These results suggest the possibility that drug therapy be optimized by considering the dosing time when the antidepressant activity is high and other pharmacological activities leading to adverse effects are low. Further studies using animal models of depression and in clinical settings are necessary to confirm the effects of dosing time on depressed subjects.
Collapse
Affiliation(s)
- Hiroshi Kawai
- a Faculty of Pharmacy and Pharmaceutical Sciences, Josai University , Sakado , Saitama , Japan
| | - Reiko Iwadate
- a Faculty of Pharmacy and Pharmaceutical Sciences, Josai University , Sakado , Saitama , Japan
| | - Takuya Ishibashi
- b Faculty of Pharmaceutical Sciences, Josai International University , Togane , Chiba , Japan
| | - Naomi Kudo
- a Faculty of Pharmacy and Pharmaceutical Sciences, Josai University , Sakado , Saitama , Japan
| | - Yoichi Kawashima
- a Faculty of Pharmacy and Pharmaceutical Sciences, Josai University , Sakado , Saitama , Japan
| | - Atsushi Mitsumoto
- b Faculty of Pharmaceutical Sciences, Josai International University , Togane , Chiba , Japan
| |
Collapse
|