1
|
Arzuaga AL, Teneqexhi P, Amodeo K, Larson JR, Ragozzino ME. Prenatal stress and fluoxetine exposure in BTBR and B6 mice differentially affects autism-like behaviors in adult male and female offspring. Physiol Behav 2025; 295:114891. [PMID: 40158488 DOI: 10.1016/j.physbeh.2025.114891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Autism spectrum disorder (ASD) is characterized by significant heterogeneity in the variety and severity of symptoms. Prenatal stress and/or exposure to antidepressants may be major contributors to ASD heterogeneity. To date, the effects of prenatal stress or selective serotonin reuptake inhibitor exposure have been primarily examined in common laboratory rat and mouse strains as opposed to in rodent models of autism. The present experiments determined in the BTBR mouse model of autism whether restraint stress (30 min session every 2 days during G4 - G18) and/or exposure to the SSRI, fluoxetine (3 mg/kg during G8 - G18) affects repetitive motor behaviors, anxiety and/or behavioral flexibility in offspring at adulthood. Male and female BTBR mice exhibited elevated grooming behavior compared to that of C57BL/6 J (B6) mice. The prenatal manipulations did not affect grooming in male BTBR mice, but the combination increased rearing and jumping. Prenatal stress, fluoxetine and the combination significantly reduced self-grooming, while concomitantly increasing locomotion in female BTBR mice. These prenatal manipulations also increased rearing and jumping behavior in female BTBR mice. In B6 mice, the prenatal stress conditions increased grooming behavior. In addition, male BTBR mice exposed to prenatal stress and fluoxetine along with female BTBR mice prenatally exposed to fluoxetine were impaired on reversal learning. The prenatal manipulations had no effect on anxiety in either mouse strain. The pattern of results suggest that prenatal exposure to stress and/or a SSRI have long-term effects on autism-like behaviors and may contribute to the heterogeneity and co-morbidity observed in autism.
Collapse
Affiliation(s)
- Anna L Arzuaga
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA.
| | - Pamela Teneqexhi
- Department of Psychology, University of Illinois Chicago, Chicago, IL 60607, USA.
| | - Katelyn Amodeo
- Department of Psychology, University of Illinois Chicago, Chicago, IL 60607, USA.
| | - John R Larson
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA; Department of Psychology, University of Illinois Chicago, Chicago, IL 60607, USA; Department of Psychiatry, University of Illinois Chicago, Chicago, IL 60612, USA.
| | - Michael E Ragozzino
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA; Department of Psychology, University of Illinois Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
2
|
Binder MS, Cauley EB, Cofsky NI, Lemler MO. Neonatal vocalization rate predicts future prosocial behavior in C57 BL/6J mice. Behav Brain Res 2025; 486:115560. [PMID: 40164314 DOI: 10.1016/j.bbr.2025.115560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Neonatal Ultrasonic vocalizations (USVs) are an innate form of mouse communicative behavior that are produced throughout the first two postnatal weeks. While neonatal USVs are commonly assessed, their relationship to future behaviors is largely unknown. In the present study, we addressed this by analyzing vocalizations in C57BL/6 pups throughout development. We then examined each animal's anxiety, locomotion, depressive, prosocial, and aggressive behaviors in adolescence. To analyze the results, we used correlations and also divided the mice into a high and a low group according to quantitative measures of their vocalizations, using a median split design. For call rate, we found a large positive correlation between call rate and sociability, furthermore, high vocalizers were significantly more prosocial than low vocalizers. No other significant differences and significant correlations were found. When we controlled for the relative contribution of the weight, sex, litter size, and sex composition of the litter, as well as the duration, pitch and amplitude of the calls, we found that high vocalizers were still significantly more prosocial than low vocalizers, indicating that this relationship cannot be attributed to these other factors. When the data was split according to the pitch, duration, and amplitude of the vocalizations, no significant adolescent behavioral differences nor correlations were found. Similarly, the types of calls produced had minimal relevance to adolescent behaviors. Altogether, our study elucidated a long-term implication for USVs, finding that the number of USVs produced throughout early development is a significant predictor of an animal's future prosocial behavior.
Collapse
Affiliation(s)
- Matthew S Binder
- Department of Psychology, Trinity University, San Antonio, TX, United States.
| | - Elise B Cauley
- Department of Psychology, Trinity University, San Antonio, TX, United States
| | - Nicole I Cofsky
- Department of Psychology, Trinity University, San Antonio, TX, United States
| | - Morgan O Lemler
- Department of Psychology, Trinity University, San Antonio, TX, United States
| |
Collapse
|
3
|
Perrier L, de Witasse Thézy A, Pradeau A, Schradin C, Greenfield MD, Mathevon N, Levréro F. A cooperatively breeding mouse shows flexible use of its vocal repertoire according to social context. Behav Brain Res 2025; 486:115575. [PMID: 40187690 DOI: 10.1016/j.bbr.2025.115575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Mice exchange information using chemical, visual and acoustic signals. Long ignored, mouse ultrasonic communication is now considered to be an important aspect of their social life, transferring information such as individual identity or stress levels. However, whether and how mice modulate their acoustic communications is largely unknown. Here we show that the cooperatively breeding African striped mouse Rhabdomys pumilio controls its vocal production both qualitatively and quantitatively, depending on naturally relevant social context. By conducting controlled experiments in captivity, we found a vocal repertoire consisting of seven vocalisation types, which it uses differently depending on different types of social interactions. Familiar individuals of the same or different sex vocalise more than two unfamiliar same-sex individuals. The greatest diversity of vocalisations was recorded during the encounter between an unfamiliar female and male, suggesting that certain vocalisations are mainly used for courtship. Our results highlight that familiar mice may alternate their vocalisations while unfamiliar individuals tend to overlap one another. These observations suggest that African striped mice control the production and temporal dynamics of their vocalisations, addressing targeted information to specific receivers via the acoustic channel.
Collapse
Affiliation(s)
- Léo Perrier
- ENES Bioacoustics Research Laboratory, CRNL, CNRS, Inserm, University of Saint-Etienne, Saint-Etienne, France.
| | - Aude de Witasse Thézy
- ENES Bioacoustics Research Laboratory, CRNL, CNRS, Inserm, University of Saint-Etienne, Saint-Etienne, France
| | - Aurélie Pradeau
- ENES Bioacoustics Research Laboratory, CRNL, CNRS, Inserm, University of Saint-Etienne, Saint-Etienne, France
| | - Carsten Schradin
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg F-67000, France; School of Animal, Plant & Environmental Sciences, University of the Witwatersrand, Private Bag 3, WITS 2050, Johannesburg, South Africa
| | - Michael D Greenfield
- ENES Bioacoustics Research Laboratory, CRNL, CNRS, Inserm, University of Saint-Etienne, Saint-Etienne, France; Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS 66045, USA
| | - Nicolas Mathevon
- ENES Bioacoustics Research Laboratory, CRNL, CNRS, Inserm, University of Saint-Etienne, Saint-Etienne, France; Institut universitaire de France, France; Ecole Pratique des Hautes Etudes, CHArt Lab, PSL University, France
| | - Florence Levréro
- ENES Bioacoustics Research Laboratory, CRNL, CNRS, Inserm, University of Saint-Etienne, Saint-Etienne, France; Institut universitaire de France, France
| |
Collapse
|
4
|
Cioccarelli L, Lenihan JA, Erwin LG, Young PW. Differential neuronal functions of LNX1 and LNX2 revealed by behavioural analysis in single and double knockout mice. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:13. [PMID: 40269869 PMCID: PMC12020136 DOI: 10.1186/s12993-025-00276-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Ligand of NUMB protein-X 1 (LNX1) and LNX2 proteins are closely related PDZ domain-containing E3 ubiquitin ligases that interact with and potentially modulate numerous synaptic and neurodevelopmentally important proteins. While both LNX1 and LNX2 are expressed in neurons, it is noteworthy that neuronal LNX1 isoforms lack the catalytic domain responsible for ubiquitination of substrates. Thus, the shared interaction partners of LNX1 and LNX2 might be differentially regulated by these proteins, with LNX1 acting as a stabilizing scaffold while LNX2 may promote their ubiquitination and degradation. Despite the identification of many LNX interacting proteins and substrates, our understanding of the distinct in vivo functions of LNX1 and LNX2 remains very incomplete. RESULTS We previously reported that mice lacking both LNX1 in the central nervous system and LNX2 globally exhibit decreased anxiety-related behaviour. Here we significantly extend this work by examining anxiety-related and risk-taking behaviours in Lnx1-/- and Lnx2-/- single knockout animals for the first time and by analysing previously unexplored aspects of behaviour in both single and double knockout animals. While the absence of both LNX1 and LNX2 contributes to the decreased anxiety-related behaviour of double knockout animals in the open field and elevated plus maze tests, the elimination of LNX2 plays a more prominent role in altered behaviour in the dark-light emergence test and wire beam bridge risk-taking paradigms. By contrast, Lnx knockout mice of all genotypes were indistinguishable from wildtype animals in the marble burying, stress-induced hyperthermia and novel object recognition tests. Analysis of the ultrasonic vocalizations of pups following maternal separation revealed significant differences in call properties and vocal repertoire for Lnx1-/- and Lnx1-/-;Lnx2-/- double knockout animals. Finally, decreased body weight previously noted in double knockout animals could be attributed largely to Lnx1 gene knockout. CONCLUSIONS These results identify specific roles of LNX1 and LNX2 proteins in modulating distinct aspects of anxiety and risk-taking behaviour and social communication in mice. They also reveal an unexpected role for neuronally expressed LNX1 isoforms in determining body weight. These novel insights into the differential neuronal functions of LNX1 and LNX2 proteins provide a foundation for mechanistic studies of these phenomena.
Collapse
Affiliation(s)
- Laura Cioccarelli
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Joan A Lenihan
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Leah G Erwin
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Paul W Young
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| |
Collapse
|
5
|
Mooney C, Parlante A, Canarutto G, Grigoli A, Scattoni ML, Ricceri L, Jimenez-Mateos EM, Sanz-Rodriguez A, Clementi E, Piazza S, Henshall DC, Provenzano G. Deregulated mRNA and microRNA Expression Patterns in the Prefrontal Cortex of the BTBR Mouse Model of Autism. Mol Neurobiol 2025:10.1007/s12035-025-04900-x. [PMID: 40227316 DOI: 10.1007/s12035-025-04900-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition caused by both genetic and environmental factors. Since no single gene variant accounts for more than 1% of the cases, the converging actions of ASD-related genes and other factors, including microRNAs (miRNAs), may contribute to ASD pathogenesis. To date, few studies have simultaneously investigated the mRNA and miRNA profiles in an ASD-relevant model. The BTBR mouse strain displays a range of behaviors with ASD-like features but little is known about the protein-coding and noncoding gene expression landscape that may underlie the ASD-like phenotype. Here we performed parallel mRNA and miRNA profiling using the prefrontal cortex (PFC) of BTBR and C57BL/6 J (B6) mice. This identified 1063 differentially expressed genes and 48 differentially expressed miRNAs. Integration of mRNA and miRNA data identified a strong inverse relationship between upregulated (DEGs) and downregulated miRNAs, and vice versa. Pathway analysis, taking account of the inverse relationship between differentially expressed miRNAs and their target mRNAs highlighted significant shared enrichment in immune signaling, myelination, and neurodevelopmental processes. Notably, miRNA changes were predicted to affect synapse-related functions but we did not find enrichment of protein-coding genes linked to cellular components or biological processes related to synapses in the PFC of BTBR mice, indicating processes may evade miRNA control. In contrast, other miRNAs were predicted to have extensive relationships with DEGs suggesting their role as potential hub coordinators of gene expression. Profiling findings were confirmed via qRT-PCR for representative protein-coding transcripts and miRNAs. Our study underscores the complex interplay between gene expression and miRNA regulation within immune and inflammatory pathways in the BTBR model, offering insights into the neurodevelopmental mechanisms of ASD. These results support the value of the BTBR mouse model and identify strategies that could adjust molecular pathways for therapeutic applications in ASD research.
Collapse
Affiliation(s)
- Catherine Mooney
- Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- School of Computer Science, University College Dublin, Dublin, Ireland
| | - Andrea Parlante
- Computational Biology Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giulia Canarutto
- Computational Biology Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Andrea Grigoli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Maria Luisa Scattoni
- Research Coordination and Promotion Service, Istituto Superiore Di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Laura Ricceri
- Centre for Behavioural Sciences and Mental Health, Istituto Superiore Di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Eva Maria Jimenez-Mateos
- Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Discipline of Physiology, School of Medicine, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - Amaya Sanz-Rodriguez
- Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- FutureNeuro Research Ireland Centre for Translational Brain Science, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Elena Clementi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Silvano Piazza
- Computational Biology Unit, International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - David C Henshall
- Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
- FutureNeuro Research Ireland Centre for Translational Brain Science, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy.
| |
Collapse
|
6
|
Zheng H, Chen D, Zhong Z, Li Z, Yuan M, Zhang Z, Zhou X, Zhu G, Sun H, Sun L. Behavioral tests for the assessment of social hierarchy in mice. Front Behav Neurosci 2025; 19:1549666. [PMID: 40110389 PMCID: PMC11920152 DOI: 10.3389/fnbeh.2025.1549666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025] Open
Abstract
Social hierarchy refers to the set of social ranks in a group of animals where individuals can gain priority access to resources through repeated social interactions. Key mechanisms involved in this process include conflict, social negotiation, prior experience, and physical advantages. The establishment and maintenance of social hierarchies not only promote group stability and well-being but also shape individual social behaviors by fostering cooperation and reducing conflict. Existing research indicates that social hierarchy is closely associated with immune responses, neural regulation, metabolic processes, and endocrine functions. These physiological systems collectively modulate an individual's sensitivity to stress and influence adaptive responses, thereby playing a critical role in the development of psychiatric disorders such as depression and anxiety. This review summarizes the primary behavioral methods used to assess social dominance in mice, evaluates their applicability and limitations, and discusses potential improvements. Additionally, it explores the underlying neural mechanisms associated with these methods to deepen our understanding of their biological basis. By critically assessing existing methodologies and proposing refinements, this study aims to provide a systematic reference framework and methodological guidance for future research, facilitating a more comprehensive exploration of the neural mechanisms underlying social behavior. The role of sex differences in social hierarchy formation remains underexplored. Most studies focus predominantly on males, while the distinct social strategies and physiological mechanisms of females are currently overlooked. Future studies should place greater emphasis on evaluating social hierarchy in female mice to achieve a more comprehensive understanding of sex-specific social behaviors and their impact on group structure and individual health. Advances in automated tracking technologies may help address this gap by improving behavioral assessments in female mice. Future research may also benefit from integrating physiological data (e.g., hormone levels) to gain deeper insights into the relationships between social status, stress regulation, and mental health. Additionally, developments in artificial intelligence and deep learning could enhance individual recognition and behavioral analysis, potentially reducing reliance on chemical markers or implanted devices.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Dantong Chen
- Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Zilong Zhong
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Ziyi Li
- Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Meng Yuan
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Zhenkun Zhang
- Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Xiaoping Zhou
- Network Information Center, Shandong Second Medical University, Weifang, China
| | - Guohui Zhu
- Depression Treatment Center, Weifang Mental Health Center, Weifang, China
| | - Hongwei Sun
- Department of Psychology, Shandong Second Medical University, Weifang, China
| | - Lin Sun
- Department of Psychology, Shandong Second Medical University, Weifang, China
| |
Collapse
|
7
|
Leuner LR, Hurley LM. What matters to a mouse? Effects of internal and external context on male vocal response to female squeaks. PLoS One 2025; 20:e0312789. [PMID: 39970156 PMCID: PMC11838898 DOI: 10.1371/journal.pone.0312789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/11/2025] [Indexed: 02/21/2025] Open
Abstract
House mice adjust their signaling behavior depending on the social context of an interaction, but which aspects of context elicit the strongest responses from these individuals is often difficult to determine. To explore how internal and external contextual factors influence how house mice produce and respond to social signals, we assessed how dominant and subordinate male mice differed in their ultrasonic vocalization (USV) production in response to playback of broadband vocalizations (BBVs, or squeaks) when given limited access to a stimulus female. We used a repeated measures design in which each male was exposed to two types of trials with different odor conditions: either just female odors (Fem condition) or female odors in addition to the odors of potential competitors, other males (Fem+Male condition). The presence of odors from other males in this assay served as a proxy for an "audience" as the male interacted with the stimulus female. These conditions were replicated for two distinct cohorts of individuals: males exposed to the odor of familiar competitors in the Fem+Male condition (Familiar odor cohort), and males exposed to the odor of unfamiliar competitors in the Fem+Male condition (Unfamiliar odor cohort). By assessing dominance status of the focal individual and familiarity of the "audience", we are able to explore how these factors may affect one another as males respond to BBVs. Dominants and subordinates did not differ in their baseline vocal production (vocalizations produced prior to squeak playback) or response to squeaks. However, all groups, regardless of dominance status or odor condition, reduced their vocal production in response to BBV playback. The presence of unfamiliar male odor prompted mice to decrease their baseline level of calling and decrease the complexity of their vocal repertoire compared to trials that only included female odor, and this effect also did not differ across dominance status. Importantly, the presence of male odor did not affect vocal behavior when the male odor was familiar to the focal individual. These findings suggest that mice alter their vocal behavior during courtship interactions in response to cues that indicate the presence of potential competitors, and this response is modulated by the familiarity of these competitor cues.
Collapse
Affiliation(s)
- Lauren R. Leuner
- Department of Biology, Hurley Laboratory, Indiana University, Bloomington, Indiana, United States of America
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, Indiana, United States of America
| | - Laura M. Hurley
- Department of Biology, Hurley Laboratory, Indiana University, Bloomington, Indiana, United States of America
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
8
|
Wingfield KK, Misic T, Jain K, McDermott CS, Abney NM, Richardson KT, Rubman MB, Beierle JA, Miracle SA, Sandago EJ, Baskin BM, Lynch WB, Borrelli KN, Yao EJ, Wachman EM, Bryant CD. The ultrasonic vocalization (USV) syllable profile during neonatal opioid withdrawal and a kappa opioid receptor component to increased USV emissions in female mice. Psychopharmacology (Berl) 2025; 242:427-447. [PMID: 39348003 PMCID: PMC11775077 DOI: 10.1007/s00213-024-06694-7] [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: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024]
Abstract
RATIONALE Opioid use during pregnancy can lead to negative infant health outcomes, including neonatal opioid withdrawal syndrome (NOWS). NOWS comprises gastrointestinal, autonomic nervous system, and neurological dysfunction that manifest during spontaneous withdrawal. Variability in NOWS severity necessitates a more individualized treatment approach. Ultrasonic vocalizations (USVs) in neonatal mice are emitted in isolation as a stress response and are increased during opioid withdrawal, thus modeling a negative affective state that can be utilized to test new treatments. OBJECTIVES We sought to identify the behavioral and USV profile, brainstem transcriptomic adaptations, and role of kappa opioid receptors in USVs during neonatal opioid withdrawal. METHODS We employed a third trimester-approximate opioid exposure model, where neonatal inbred FVB/NJ pups were injected twice-daily with morphine (10mg/kg, s.c.) or saline (0.9%, 20 ul/g, s.c.) from postnatal day(P) 1 to P14. This protocol induces reduced weight gain, hypothermia, thermal hyperalgesia, and increased USVs during spontaneous morphine withdrawal. RESULTS On P14, there were increased USV emissions and altered USV syllables during withdrawal, including an increase in Complex 3 syllables in FVB/NJ females (but not males). Brainstem bulk mRNA sequencing revealed an upregulation of the kappa opioid receptor (Oprk1), which contributes to withdrawal-induced dysphoria. The kappa opioid receptor (KOR) antagonist, nor-BNI (30 mg/kg, s.c.), significantly reduced USVs in FVB/NJ females, but not males during spontaneous morphine withdrawal. Furthermore, the KOR agonist, U50,488h (0.625 mg/kg, s.c.), was sufficient to increase USVs on P10 (both sexes) and P14 (females only) in FVB/NJ mice. CONCLUSIONS We identified an elevated USV syllable, Complex 3, and a female-specific recruitment of the dynorphin/KOR system in increased USVs associated with neonatal opioid withdrawal severity.
Collapse
MESH Headings
- Animals
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/agonists
- Vocalization, Animal/drug effects
- Vocalization, Animal/physiology
- Female
- Mice
- Male
- Morphine/administration & dosage
- Morphine/pharmacology
- Animals, Newborn
- Neonatal Abstinence Syndrome/physiopathology
- Neonatal Abstinence Syndrome/metabolism
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/administration & dosage
- Pregnancy
- Substance Withdrawal Syndrome/physiopathology
- Naltrexone/analogs & derivatives
- Naltrexone/pharmacology
- Narcotic Antagonists/pharmacology
- Brain Stem/metabolism
- Brain Stem/drug effects
- Disease Models, Animal
Collapse
Affiliation(s)
- Kelly K Wingfield
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Teodora Misic
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
| | - Kaahini Jain
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
| | - Carly S McDermott
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
| | - Nalia M Abney
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
| | - Kayla T Richardson
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
- Post-Baccalaureate Research Education Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Mia B Rubman
- NIH/NIDA Summer Undergraduate Fellowship Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jacob A Beierle
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sophia A Miracle
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Emma J Sandago
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
| | - Britahny M Baskin
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
- T32 Training Program on Development of Medications for Substance Use Disorders Fellowship, Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | - William B Lynch
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Kristyn N Borrelli
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Emily J Yao
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA
| | - Elisha M Wachman
- Department of Pediatrics, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Camron D Bryant
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, 140 The Fenway, Boston, MA, USA.
| |
Collapse
|
9
|
Randell AM, Salia S, Fowler LF, Aung T, Puts DA, Swift-Gallant A. A meta-analysis of sex differences in neonatal rodent ultrasonic vocalizations and the implication for the preclinical maternal immune activation model. Biol Sex Differ 2025; 16:4. [PMID: 39863873 PMCID: PMC11762899 DOI: 10.1186/s13293-025-00685-9] [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: 10/16/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
As the earliest measure of social communication in rodents, ultrasonic vocalizations (USVs) in response to maternal separation are critical in preclinical research on neurodevelopmental disorders (NDDs). While sex differences in both USV production and behavioral outcomes are reported, many studies overlook sex as a biological variable in preclinical NDD models. We aimed to evaluate sex differences in USV call parameters and determine if USVs are differently impacted based on sex in the preclinical maternal immune activation (MIA) model. Results indicate that sex differences in USVs vary with developmental stage and are more pronounced in MIA offspring. Specifically, developmental stage is a moderator of sex differences in USV call duration, with control females emitting longer calls than males in early development (up to postnatal day [PND] 8), but this pattern reverses after PND8. MIA leads to a reduction in call numbers for females compared to same-sex controls in early development, with a reversal post-PND8. MIA decreased call duration and increased total call duration in males, but unlike females, developmental stage did not influence these differences. In males, MIA effects varied by species, with decreased call numbers in rats but increased call numbers in mice. MIA timing (gestational day ≤ 12.5 vs. > 12.5) did not significantly affect results. Our findings highlight the importance of considering sex, developmental timing, and species in USVs research. We discuss how analyzing USV call types and incorporating sex as a biological variable can enhance our understanding of neonatal ultrasonic communication and its translational value in NDD research.
Collapse
Affiliation(s)
- Alison M Randell
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Lucas F Fowler
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
- Cognitive and Behavioural Ecology Program, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Toe Aung
- Department of Psychology and Counseling, Immaculata University, Immaculata, PA, USA
| | - David A Puts
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| |
Collapse
|
10
|
Ziętek MM, Jaszczyk A, Stankiewicz AM, Sampino S. Prenatal gene-environment interactions mediate the impact of advanced maternal age on mouse offspring behavior. Sci Rep 2024; 14:31733. [PMID: 39738558 PMCID: PMC11685589 DOI: 10.1038/s41598-024-82070-x] [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: 05/24/2024] [Accepted: 12/02/2024] [Indexed: 01/02/2025] Open
Abstract
Autism spectrum disorders encompass diverse neurodevelopmental conditions marked by alterations in social communication and repetitive behaviors. Advanced maternal age is associated with an increased risk of bearing children affected by autism but the etiological factors underlying this association are not well known. Here, we investigated the effects of advanced maternal age on offspring health and behavior in two genetically divergent mouse strains: the BTBR T+ Itpr3tf/J (BTBR) mouse model of idiopathic autism, and the C57BL/6 J (B6) control strain, as a model of genetic variability. In both strains, advanced maternal age negatively affected female reproductive and pregnancy outcomes, and perturbed placental and fetal growth, and the expression of genes in the fetal brain tissues. Postnatally, advanced maternal age had strain-dependent effects on offspring sociability, learning skills, and the occurrence of perseverative behaviors, varying between male and female offspring. These findings disentangle the relationship between genetic determinants and maternal age-related factors in shaping the emergence of autism-like behaviors in mice, highlighting the interplay between maternal age, genetic variability, and prenatal programming, in the occurrence of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Marta Marlena Ziętek
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
| | - Aneta Jaszczyk
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
| | - Adrian Mateusz Stankiewicz
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland
| | - Silvestre Sampino
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzębiec, Poland.
| |
Collapse
|
11
|
Mercado E, Zhuo J. Do rodents smell with sound? Neurosci Biobehav Rev 2024; 167:105908. [PMID: 39343078 DOI: 10.1016/j.neubiorev.2024.105908] [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: 06/26/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Chemosensation via olfaction is a critical process underlying social interactions in many different species. Past studies of olfaction in mammals often have focused on its mechanisms in isolation from other systems, limiting the generalizability of findings from olfactory research to perceptual processes in other modalities. Studies of chemical communication, in particular, have progressed independently of research on vocal behavior and acoustic communication. Those bioacousticians who have considered how sound production and reception might interact with olfaction often portray odors as cues to the kinds of vocalizations that might be functionally useful. In the olfaction literature, vocalizations are rarely mentioned. Here, we propose that ultrasonic vocalizations may affect what rodents smell by altering the deposition of inhaled particles and that rodents coordinate active sniffing with sound production specifically to enhance reception of pheromones. In this scenario, rodent vocalizations may contribute to a unique mode of active olfactory sensing, in addition to whatever roles they serve as social signals. Consideration of this hypothesis highlights the perceptual advantages that parallel coordination of multiple sensorimotor processes may provide to individuals exploring novel situations and environments, especially those involving dynamic social interactions.
Collapse
Affiliation(s)
- Eduardo Mercado
- University at Buffalo, The State University of New York, USA.
| | | |
Collapse
|
12
|
Möhrle D, Murari K, Rho JM, Cheng N. Vocal communication in asocial BTBR mice is more malleable by a ketogenic diet in juveniles than adults. Neuroscience 2024; 561:43-64. [PMID: 39413868 DOI: 10.1016/j.neuroscience.2024.10.001] [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: 08/28/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024]
Abstract
Deficits in social communication and language development are a hallmark of autism spectrum disorder currently with no effective approaches to reduce the negative impact. Interventional studies using animal models have been very limited in demonstrating improved vocal communication. Autism has been proposed to involve metabolic dysregulation. Ketogenic diet (KD) is a metabolism-based therapy for medically intractable epilepsy, and its applications in other neurological conditions have been increasingly tested. However, how KD would affect vocal communication has not been explored. The BTBR mouse strain is widely used to model asocial phenotypes. They display robust and pronounced deficits in vocalization during social interaction, and have metabolic changes implicated in autism. We investigated the effects of KD on ultrasonic vocalizations (USVs) in juvenile and adult BTBR mice during male-female social encounters. After a brief treatment with KD, the number, spectral bandwidth, and much of the temporal structure of USVs were robustly closer to control levels in both juvenile and adult BTBR mice. Composition of call categories and transitioning between individual call subtypes were more effectively altered to more closely align with the control group in juvenile BTBR mice. Together, our data provide further support to the hypothesis that metabolism-based dietary intervention could modify disease expression, including core symptoms, in autism. Future studies should tease apart the molecular mechanisms of KD's effects on vocalization.
Collapse
Affiliation(s)
- Dorit Möhrle
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.
| | - Kartikeya Murari
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada; Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada.
| | - Jong M Rho
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Ning Cheng
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
13
|
Wingfield KK, Misic T, Miracle SA, McDermott CS, Jain K, Abney NM, Richardson KT, Rubman MB, Beierle JA, Wachman EM, Bryant CD. The acoustic properties, syllable structure, and syllable sequences of ultrasonic vocalizations (USVs) during neonatal opioid withdrawal in FVB/N mouse substrains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622304. [PMID: 39574631 PMCID: PMC11580921 DOI: 10.1101/2024.11.06.622304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Concomitant with the opioid epidemic, there has been a rise in pregnant women diagnosed with opioid use disorder and cases of infants born with neonatal opioid withdrawal syndrome (NOWS). NOWS refers to signs and symptoms following cessation of prenatal opioid exposure that comprise neurological, gastrointestinal, and autonomic system dysfunction. A critical indicator of NOWS severity is excessive, high-pitched crying. However, NOWS evaluation is, in large part, subjective, and additional cry features may not be easily recognized during clinical assessment. Thus, there is a need for more objective measures to determine NOWS severity. We used a third trimester-approximate opioid exposure paradigm to model NOWS traits in genetically similar inbred substrains of FVB/N mice (NJ, NCrl, NHsd, and NTac). Pups were injected twice daily from postnatal day 1 (P1) to P14 with morphine (10 mg/kg, s.c.) or saline (20 ml/g, s.c.). Because there were only very minor substrain differences in spontaneous withdrawal-induced ultrasonic vocalization (USV) profiles, we collapsed across substrains to evaluate the effects of morphine withdrawal on additional USV properties. We identified syllable sequences unique to morphine-withdrawn and saline-control FVB/N pups on P7 and P14. We also observed an effect of spontaneous morphine withdrawal on the acoustic properties of USVs and specific syllables on P7 and P14. Multiple withdrawal traits correlated with some acoustic properties of USVs and syllable type emission in morphine-withdrawn FVB/N pups on P7 and P14. These data provide an in-depth investigation of mouse USV syllable profiles and acoustic features during spontaneous neonatal opioid withdrawal in mice.
Collapse
Affiliation(s)
- Kelly K. Wingfield
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Teodora Misic
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Sophia A. Miracle
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- Graduate Program for Neuroscience, Boston University, Boston, MA USA
| | - Carly S. McDermott
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Kaahini Jain
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Nalia M. Abney
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Kayla T. Richardson
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- Post-Baccalaureate Research Education Program, Boston University Chobanian & Avedisian School of Medicine
| | | | - Jacob A. Beierle
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine
| | - Elisha M. Wachman
- Department of Pediatrics, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston MA USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| |
Collapse
|
14
|
Binder MS, Pranske ZJ, Hodges SL, Womble PD, Kwok EM, Quintero SI, Kim AD, Narvaiz DA, Lugo JN. Agomelatine Is Unable to Attenuate Kainic Acid-Induced Deficits in Early Life Communicative Behavior. Dev Psychobiol 2024; 66:e22543. [PMID: 39205500 PMCID: PMC11376987 DOI: 10.1002/dev.22543] [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: 07/25/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Early life seizures are associated with a variety of behavioral comorbidities. Among the most prevalent of these are deficits in communication. Auditory communicative behaviors in mice, known as ultrasonic vocalizations (USVs), can be used to assess potential treatments. Agomelatine is a melatonin agonist that effectively reduces behavioral comorbidities of seizures in adults; however, its ability to attenuate seizure-induced communicative deficits in neonates is unknown. To address this, we administered C57 mice either saline or kainic acid (KA) on postnatal day (PD) 10. The mice then received either agomelatine or saline 1-h post-status epilepticus. On PD 11, we assessed the quantity of USVs produced, the duration, peak frequency, fundamental frequency, and amplitude of the vocalizations, as well as the call type utilization. We found that KA increased vocal production and reduced USV variability relative to controls. KA also increased USV duration and amplitude and significantly altered the types of calls produced. Agomelatine did not attenuate any of the deficits. Our study is the first to assess agomelatine's efficacy to correct USVs and thus provides an important point of context to the literature, indicating that despite its high therapeutic efficacy to attenuate other behavioral comorbidities of seizures, agomelatine's ability to correct neonatal communicative deficits is limited.
Collapse
Affiliation(s)
- Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Zachary J Pranske
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Samantha L Hodges
- Institute of Biomedical Studies, Baylor University, Waco, Texas, USA
| | - Paige D Womble
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Eliesse M Kwok
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Saul I Quintero
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Andrew D Kim
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - David A Narvaiz
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
- Institute of Biomedical Studies, Baylor University, Waco, Texas, USA
- Department of Biology, Baylor University, Waco, Texas, USA
| |
Collapse
|
15
|
Herdt R, Kinzel L, Maaß JG, Walther M, Fröhlich H, Schubert T, Maass P, Schaaf CP. Enhancing the analysis of murine neonatal ultrasonic vocalizations: Development, evaluation, and application of different mathematical models. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2024; 156:2448-2466. [PMID: 39400270 DOI: 10.1121/10.0030473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
Rodents employ a broad spectrum of ultrasonic vocalizations (USVs) for social communication. As these vocalizations offer valuable insights into affective states, social interactions, and developmental stages of animals, various deep learning approaches have aimed at automating both the quantitative (detection) and qualitative (classification) analysis of USVs. So far, no notable efforts have been made to determine the most suitable architecture. We present the first systematic evaluation of different types of neural networks for USV classification. We assessed various feedforward networks, including a custom-built, fully-connected network, a custom-built convolutional neural network, several residual neural networks, an EfficientNet, and a Vision Transformer. Our analysis concluded that convolutional networks with residual connections specifically adapted to USV data, are the most suitable architecture for analyzing USVs. Paired with a refined, entropy-based detection algorithm (achieving recall of 94.9 % and precision of 99.3 %), the best architecture (achieving 86.79 % accuracy) was integrated into a fully automated pipeline capable of analyzing extensive USV datasets with high reliability. In ongoing projects, our pipeline has proven to be a valuable tool in studying neonatal USVs. By comparing these distinct deep learning architectures side by side, we have established a solid foundation for future research.
Collapse
Affiliation(s)
- Rudolf Herdt
- Center for Industrial Mathematics, University of Bremen, Bremen 28334, Germany
| | - Louisa Kinzel
- Center for Industrial Mathematics, University of Bremen, Bremen 28334, Germany
| | - Johann Georg Maaß
- Institute of Human Genetics, University of Heidelberg, Heidelberg 69120, Germany
- Interdisciplinary Neurobehavioral Core, University of Heidelberg, Heidelberg, 69120, Germany
| | - Marvin Walther
- Institute of Electrodynamics and Microelectronics, University of Bremen, Bremen 28334, Germany
| | - Henning Fröhlich
- Institute of Human Genetics, University of Heidelberg, Heidelberg 69120, Germany
| | - Tim Schubert
- Institute of Human Genetics, University of Heidelberg, Heidelberg 69120, Germany
| | - Peter Maass
- Center for Industrial Mathematics, University of Bremen, Bremen 28334, Germany
| | | |
Collapse
|
16
|
Rutovskaya MV, Volodin IA, Feoktistova NY, Surov AV, Gureeva AV, Volodina EV. Acoustic complexity of pup isolation calls in Mongolian hamsters: 3-frequency phenomena and chaos. Curr Zool 2024; 70:559-574. [PMID: 39463689 PMCID: PMC11502153 DOI: 10.1093/cz/zoad036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 07/13/2023] [Indexed: 10/29/2024] Open
Abstract
Studying pup isolation calls of wild rodents provides background for developing new early-life animal models for biomedical research and drug testing. This study discovered a highly complex acoustic phenotype of pup isolation calls in 4-5-day-old Mongolian hamsters Allocricetulus curtatus. We analyzed the acoustic structure of 5,010 isolation calls emitted in the broad range of frequencies (sonic, below 20 kHz, and ultrasonic, from 20 to 128 kHz) by 23 pups during 2-min isolation test trials, 1 trial per pup. In addition, we measured 5 body size parameters and the body weight of each pup. The calls could contain up to 3 independent fundamental frequencies in their spectra, the low (f0), the medium (g0), and the high (h0), or purely consisted of chaos in which the fundamental frequency could not be tracked. By presence/absence of the 3 fundamental frequencies or their combinations and chaos, we classified calls into 6 distinctive categories (low-frequency [LF]-f0, LF-chaos, high-frequency [HF]-g0, HF-h0, HF-g0 + h0, and HF-chaos) and estimated the relative abundance of calls in each category. Between categories, we compared acoustic parameters and estimated their relationship with pup body size index. We discuss the results of this study with data on the acoustics of pup isolation calls reported for other species of rodents. We conclude that such high complexity of Mongolian hamster pup isolation calls is unusual for rodents. Decreased acoustic complexity serves as a good indicator of autism spectrum disorders in knockout mouse models, which makes knockout hamster models prospective new wild animal model of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Marina V Rutovskaya
- Department of Behaviour and Behavioural Ecology of Mammals, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow 119071, Russia
| | - Ilya A Volodin
- Department of Behaviour and Behavioural Ecology of Mammals, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow 119071, Russia
- Department of Vertebrate Zoology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia
| | - Natalia Y Feoktistova
- Department of Comparative Ethology and Biocommunication, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow 119071, Russia
| | - Alexey V Surov
- Department of Comparative Ethology and Biocommunication, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow 119071, Russia
| | - Anna V Gureeva
- Department of Comparative Ethology and Biocommunication, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow 119071, Russia
| | - Elena V Volodina
- Department of Behaviour and Behavioural Ecology of Mammals, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow 119071, Russia
| |
Collapse
|
17
|
Ornoy A, Echefu B, Becker M. Animal Models of Autistic-like Behavior in Rodents: A Scoping Review and Call for a Comprehensive Scoring System. Int J Mol Sci 2024; 25:10469. [PMID: 39408797 PMCID: PMC11477392 DOI: 10.3390/ijms251910469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Appropriate animal models of human diseases are a cornerstone in the advancement of science and medicine. To create animal models of neuropsychiatric and neurobehavioral diseases such as autism spectrum disorder (ASD) necessitates the development of sufficient neurobehavioral measuring tools to translate human behavior to expected measurable behavioral features in animals. If possible, the severity of the symptoms should also be assessed. Indeed, at least in rodents, adequate neurobehavioral and neurological tests have been developed. Since ASD is characterized by a number of specific behavioral trends with significant severity, animal models of autistic-like behavior have to demonstrate the specific characteristic features, namely impaired social interactions, communication deficits, and restricted, repetitive behavioral patterns, with association to several additional impairments such as somatosensory, motor, and memory impairments. Thus, an appropriate model must show behavioral impairment of a minimal number of neurobehavioral characteristics using an adequate number of behavioral tests. The proper animal models enable the study of ASD-like-behavior from the etiologic, pathogenetic, and therapeutic aspects. From the etiologic aspects, models have been developed by the use of immunogenic substances like polyinosinic-polycytidylic acid (PolyIC), lipopolysaccharide (LPS), and propionic acid, or other well-documented immunogens or pathogens, like Mycobacterium tuberculosis. Another approach is the use of chemicals like valproic acid, polychlorinated biphenyls (PCBs), organophosphate pesticides like chlorpyrifos (CPF), and others. These substances were administered either prenatally, generally after the period of major organogenesis, or, especially in rodents, during early postnatal life. In addition, using modern genetic manipulation methods, genetic models have been created of almost all human genetic diseases that are manifested by autistic-like behavior (i.e., fragile X, Rett syndrome, SHANK gene mutation, neuroligin genes, and others). Ideally, we should not only evaluate the different behavioral modes affected by the ASD-like behavior, but also assess the severity of the behavioral deviations by an appropriate scoring system, as applied to humans. We therefore propose a scoring system for improved assessment of ASD-like behavior in animal models.
Collapse
Affiliation(s)
- Asher Ornoy
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
- Hadassah Academic College, Jerusalem 9101001, Israel
- Hadassah Medical School, Hebrew University, Jerusalem 9112102, Israel
| | - Boniface Echefu
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| | - Maria Becker
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| |
Collapse
|
18
|
Binder MS, Escobar I, Xu Y, Sokolov AM, Zhang L, Bordey A. Reducing Filamin A Restores Cortical Synaptic Connectivity and Early Social Communication Following Cellular Mosaicism in Autism Spectrum Disorder Pathways. J Neurosci 2024; 44:e1245232024. [PMID: 39164108 PMCID: PMC11426378 DOI: 10.1523/jneurosci.1245-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 06/17/2024] [Accepted: 06/27/2024] [Indexed: 08/22/2024] Open
Abstract
Communication in the form of nonverbal, social vocalization, or crying is evolutionary conserved in mammals and is impaired early in human infants that are later diagnosed with autism spectrum disorder (ASD). Defects in infant vocalization have been proposed as an early sign of ASD that may exacerbate ASD development. However, the neural mechanisms associated with early communicative deficits in ASD are not known. Here, we expressed a constitutively active mutant of Rheb (RhebS16H), which is known to upregulate two ASD core pathways, mTOR complex 1 (mTORC1) and ERK1/2, in Layer (L) 2/3 pyramidal neurons of the neocortex of mice of either sex. We found that cellular mosaic expression of RhebS16H in L2/3 pyramidal neurons altered the production of isolation calls from neonatal mice. This was accompanied by an expected misplacement of neurons and dendrite overgrowth, along with an unexpected increase in spine density and length, which was associated with increased excitatory synaptic activity. This contrasted with the known decrease in spine density in RhebS16H neurons of 1-month-old mice. Reducing the levels of the actin cross-linking and adaptor protein filamin A (FLNA), known to be increased downstream of ERK1/2, attenuated dendrite overgrowth and fully restored spine properties, synaptic connectivity, and the production of pup isolation calls. These findings suggest that upper-layer cortical pyramidal neurons contribute to communicative deficits in a condition known to affect two core ASD pathways and that these mechanisms are regulated by FLNA.
Collapse
Affiliation(s)
- Matthew S Binder
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, Connecticut 06520-8082
| | - Iris Escobar
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, Connecticut 06520-8082
| | - Youfen Xu
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, Connecticut 06520-8082
| | - Aidan M Sokolov
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, Connecticut 06520-8082
| | - Longbo Zhang
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, Connecticut 06520-8082
| | - Angélique Bordey
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, Connecticut 06520-8082
| |
Collapse
|
19
|
Wingfield KK, Misic T, Jain K, McDermott CS, Abney NM, Richardson KT, Rubman MB, Beierle JA, Miracle SA, Sandago EJ, Baskin BM, Lynch WB, Borrelli KN, Yao EJ, Wachman EM, Bryant CD. The ultrasonic vocalization (USV) syllable profile during neonatal opioid withdrawal and a kappa opioid receptor component to increased USV emissions in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601766. [PMID: 39005445 PMCID: PMC11244951 DOI: 10.1101/2024.07.02.601766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Rationale Opioid use during pregnancy can lead to negative infant health outcomes, including neonatal opioid withdrawal syndrome (NOWS). NOWS comprises gastrointestinal, autonomic nervous system, and neurological dysfunction that manifest during spontaneous withdrawal. Variability in NOWS severity necessitates a more individualized treatment approach. Ultrasonic vocalizations (USVs) in neonatal mice are emitted in isolation as a stress response and are increased during opioid withdrawal, thus modeling a negative affective state that can be utilized to test new treatments. Objectives We sought to identify the behavioral and USV profile, brainstem transcriptomic adaptations, and role of kappa opioid receptors in USVs during neonatal opioid withdrawal. Methods We employed a third trimester-approximate opioid exposure model, where neonatal inbred FVB/NJ pups were injected twice-daily with morphine (10mg/kg, s.c.) or saline (0.9%, 20 ul/g, s.c.) from postnatal day(P) 1 to P14. This protocol induces reduced weight gain, hypothermia, thermal hyperalgesia, and increased USVs during spontaneous morphine withdrawal. Results On P14, there were increased USV emissions and altered USV syllables during withdrawal, including an increase in Complex 3 syllables in FVB/NJ females (but not males). Brainstem bulk mRNA sequencing revealed an upregulation of the kappa opioid receptor (Oprk1), which contributes to withdrawal-induced dysphoria. The kappa opioid receptor (KOR) antagonist, nor-BNI (30 mg/kg, s.c.), significantly reduced USVs in FVB/NJ females, but not males during spontaneous morphine withdrawal. Furthermore, the KOR agonist, U50,488h (0.625 mg/kg, s.c.), was sufficient to increase USVs on P10 (both sexes) and P14 (females only) in FVB/NJ mice. Conclusions We identified an elevated USV syllable, Complex 3, and a female-specific recruitment of the dynorphin/KOR system in increased USVs associated with neonatal opioid withdrawal severity.
Collapse
Affiliation(s)
- Kelly K. Wingfield
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Teodora Misic
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Kaahini Jain
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Carly S. McDermott
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Nalia M. Abney
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Kayla T. Richardson
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- Post-Baccalaureate Research Education Program, Boston University Chobanian & Avedisian School of Medicine
| | | | - Jacob A. Beierle
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine
| | - Sophia A. Miracle
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- Graduate Program for Neuroscience, Boston University, Boston, MA USA
| | - Emma J. Sandago
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Britahny M. Baskin
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- T32 Training Program on Development of Medications for Substance Use Disorders Fellowship, Center for Drug Discovery, Northeastern University
| | - William B. Lynch
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine
- Graduate Program for Neuroscience, Boston University, Boston, MA USA
| | - Kristyn N. Borrelli
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
- T32 Biomolecular Pharmacology Training Program, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
- Transformative Training Program in Addiction Science, Boston University Chobanian & Avedisian School of Medicine
- Graduate Program for Neuroscience, Boston University, Boston, MA USA
| | - Emily J. Yao
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| | - Elisha M. Wachman
- Department of Pediatrics, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston MA USA
| | - Camron D. Bryant
- Laboratory of Addiction Genetics, Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA USA
| |
Collapse
|
20
|
Mottolese N, Coiffard O, Ferraguto C, Manolis A, Ciani E, Pietropaolo S. Autistic-relevant behavioral phenotypes of a mouse model of cyclin-dependent kinase-like 5 deficiency disorder. Autism Res 2024; 17:1742-1759. [PMID: 39234879 DOI: 10.1002/aur.3226] [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: 05/18/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a neurodevelopmental disease caused by mutations in the X-linked CDKL5 gene and characterized by early-onset epilepsy, intellectual disability, and autistic features. To date, the etiological mechanisms underlying CDD are largely unknown and no effective therapies are available. The Cdkl5 knock-out (KO) mouse has been broadly employed in preclinical studies on CDD; Cdkl5-KO mice display neurobehavioral abnormalities recapitulating most CDD symptoms, including alterations in motor, sensory, cognitive, and social abilities. However, most available preclinical studies have been carried out on adult Cdkl5-KO mice, so little is known about the phenotypic characteristics of this model earlier during development. Furthermore, major autistic-relevant phenotypes, for example, social and communication deficits, have been poorly investigated and mostly in male mutants. Here, we assessed the autistic-relevant behavioral phenotypes of Cdkl5-KO mice during the first three post-natal weeks and in adulthood. Males and females were tested, the latter including both heterozygous and homozygous mutants. Cdkl5 mutant pups showed qualitative and quantitative alterations in ultrasonic communication, detected first at 2 weeks of age and confirmed later in adulthood. Increased levels of anxiety-like behaviors were observed in mutants at 3 weeks and in adulthood, when stereotypies, reduced social interaction and memory deficits were also observed. These behavioral effects of the mutation were evident in both sexes, being more marked and varied in homozygous than heterozygous females. These findings provide novel evidence for the autistic-relevant behavioral profile of the Cdkl5 mouse model, thus supporting its use in future preclinical studies investigating CDD pathology and autism spectrum disorders.
Collapse
Affiliation(s)
- Nicola Mottolese
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
- CNRS, EPHE, INCIA, Univ. Bordeaux, Bordeaux, France
| | | | | | | | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | | |
Collapse
|
21
|
Maeda K, Tanimura M, Masago Y, Horiyama T, Takemoto H, Sasaki T, Koyama R, Ikegaya Y, Ogawa K. Development of an in vitro compound screening system that replicate the in vivo spine phenotype of idiopathic ASD model mice. Front Pharmacol 2024; 15:1455812. [PMID: 39286633 PMCID: PMC11403255 DOI: 10.3389/fphar.2024.1455812] [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: 06/27/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a developmental condition characterized by core symptoms including social difficulties, repetitive behaviors, and sensory abnormalities. Aberrant morphology of dendritic spines within the cortex has been documented in genetic disorders associated with ASD and ASD-like traits. We hypothesized that compounds that ameliorate abnormalities in spine dynamics might have the potential to ameliorate core symptoms of ASD. Because the morphology of the spine is influenced by signal inputs from other neurons and various molecular interactions, conventional single-molecule targeted drug discovery methods may not suffice in identifying compounds capable of ameliorating spine morphology abnormalities. In this study, we focused on spine phenotypes in the cortex using BTBR T + Itpr3 tf /J (BTBR) mice, which have been used as a model for idiopathic ASD in various studies. We established an in vitro compound screening system using primary cultured neurons from BTBR mice to faithfully represent the spine phenotype. The compound library mainly comprised substances with known target molecules and established safety profiles, including those approved or validated through human safety studies. Following screening of this specialized library containing 181 compounds, we identified 15 confirmed hit compounds. The molecular targets of these hit compounds were largely focused on the 5-hydroxytryptamine receptor (5-HTR). Furthermore, both 5-HT1AR agonist and 5-HT3R antagonist were common functional profiles in hit compounds. Vortioxetine, possessing dual attributes as a 5-HT1AR agonist and 5-HT3R antagonist, was administered to BTBR mice once daily for a period of 7 days. This intervention not only ameliorated their spine phenotype but also alleviated their social behavior abnormality. These results of vortioxetine supports the usefulness of a spine phenotype-based assay system as a potent drug discovery platform targeting ASD core symptoms.
Collapse
Affiliation(s)
- Kazuma Maeda
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi and Co., Ltd., Osaka, Japan
| | - Miki Tanimura
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi and Co., Ltd., Osaka, Japan
| | - Yusaku Masago
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi and Co., Ltd., Osaka, Japan
| | - Tsukasa Horiyama
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi and Co., Ltd., Osaka, Japan
| | - Hiroshi Takemoto
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi and Co., Ltd., Osaka, Japan
| | - Takuya Sasaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Koichi Ogawa
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi and Co., Ltd., Osaka, Japan
| |
Collapse
|
22
|
Miranda R, Ceschi L, Le Verger D, Nagapin F, Edeline JM, Chaussenot R, Vaillend C. Social and emotional alterations in mice lacking the short dystrophin-gene product, Dp71. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:21. [PMID: 39182120 PMCID: PMC11344925 DOI: 10.1186/s12993-024-00246-x] [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: 03/13/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The Duchenne and Becker muscular dystrophies (DMD, BMD) are neuromuscular disorders commonly associated with diverse cognitive and behavioral comorbidities. Genotype-phenotype studies suggest that severity and risk of central defects in DMD patients increase with cumulative loss of different dystrophins produced in CNS from independent promoters of the DMD gene. Mutations affecting all dystrophins are nevertheless rare and therefore the clinical evidence on the contribution of the shortest Dp71 isoform to cognitive and behavioral dysfunctions is limited. In this study, we evaluated social, emotional and locomotor functions, and fear-related learning in the Dp71-null mouse model specifically lacking this short dystrophin. RESULTS We demonstrate the presence of abnormal social behavior and ultrasonic vocalization in Dp71-null mice, accompanied by slight changes in exploratory activity and anxiety-related behaviors, in the absence of myopathy and alterations of learning and memory of aversive cue-outcome associations. CONCLUSIONS These results support the hypothesis that distal DMD gene mutations affecting Dp71 may contribute to the emergence of social and emotional problems that may relate to the autistic traits and executive dysfunctions reported in DMD. The present alterations in Dp71-null mice may possibly add to the subtle social behavior problems previously associated with the loss of the Dp427 dystrophin, in line with the current hypothesis that risk and severity of behavioral problems in patients increase with cumulative loss of several brain dystrophin isoforms.
Collapse
Affiliation(s)
- Rubén Miranda
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France.
- Department of Psychobiology and Methodology in Behavioral Sciences, Universidad Complutense de Madrid, Ciudad Universitaria, 28040, Madrid, Spain.
| | - Léa Ceschi
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Delphine Le Verger
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Flora Nagapin
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Jean-Marc Edeline
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Rémi Chaussenot
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France.
| |
Collapse
|
23
|
Santana-Coelho D, Pranske ZJ, Nolan SO, Hodges SL, Binder MS, Womble PD, Narvaiz DA, Muhammad I, Lugo JN. Neonatal immune stimulation results in sex-specific changes in ultrasonic vocalizations but does not affect seizure susceptibility in neonatal mice. Int J Dev Neurosci 2024; 84:381-391. [PMID: 38712612 DOI: 10.1002/jdn.10333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/08/2024] Open
Abstract
Neuroinflammation during the neonatal period has been linked to disorders such as autism and epilepsy. In this study, we investigated the early life behavioral consequences of a single injection of lipopolysaccharide (LPS) at postnatal day 10 (PD10) in mice. To assess deficits in communication, we performed the isolation-induced ultrasonic vocalizations (USVs) test at PD12. To determine if early life immune stimulus could alter seizure susceptibility, latency to flurothyl-induced generalized seizures was measured at 4 hours (hrs), 2 days, or 5 days after LPS injections. LPS had a sex-dependent effect on USV number. LPS-treated male mice presented significantly fewer USVs than LPS-treated female mice. However, the number of calls did not significantly differ between control and LPS for either sex. In male mice, we found that downward, short, and composite calls were significantly more prevalent in the LPS treatment group, while upward, chevron, and complex calls were less prevalent than in controls (p < 0.05). Female mice that received LPS presented a significantly higher proportion of short, frequency steps, two-syllable, and composite calls in their repertoire when compared with female control mice (p < 0.05). Seizure latency was not altered by early-life inflammation at any of the time points measured. Our findings suggest that early-life immune stimulation at PD10 disrupts vocal development but does not alter the susceptibility to flurothyl-induced seizures during the neonatal period. Additionally, the effect of inflammation in the disruption of vocalization is sex-dependent.
Collapse
Affiliation(s)
| | - Zachary J Pranske
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Suzanne O Nolan
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | | | - Matthew S Binder
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Paige D Womble
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - David A Narvaiz
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Ilyasah Muhammad
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
| | - Joaquin N Lugo
- Department of Psychology and Neuroscience, Baylor University, Waco, Texas, USA
- Institute of Biomedical Studets, Waco, Texas, USA
- Department of Biology, Baylor University, Waco, Texas, USA
| |
Collapse
|
24
|
Pilipenko T, Premoli M, Gnutti A, Bonini SA, Leonardi R, Memo M, Migliorati P. Exploring ultrasonic communication in mice treated with Cannabis sativa oil: Audio data processing and correlation study with different behaviours. Eur J Neurosci 2024; 60:4244-4253. [PMID: 38816916 DOI: 10.1111/ejn.16433] [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: 09/27/2023] [Revised: 04/18/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024]
Abstract
Studying ultrasonic vocalizations (USVs) plays a crucial role in understanding animal communication, particularly in the field of ethology and neuropharmacology. Communication is associated with social behaviour; so, USVs study is a valid assay in behavioural readout and monitoring in this context. This paper delved into an investigation of ultrasonic communication in mice treated with Cannabis sativa oil (CS mice), which has been demonstrated having a prosocial effect on behaviour of mice, versus control mice (vehicle-treated, VH mice). To conduct this study, we created a dataset by recording audio-video files and annotating the duration of time that test mice spent engaging in social activities, along with categorizing the types of emitted USVs. The analysis encompassed the frequency of individual sounds as well as more complex sequences of consecutive syllables (patterns). The primary goal was to examine the extent and nature of diversity in ultrasonic communication patterns emitted by these two groups of mice. As a result, we observed statistically significant differences for each considered pattern length between the two groups of mice. Additionally, the study extended its research by considering specific behaviours, aiming to ascertain whether dissimilarities in ultrasonic communication between CS and VH mice are more pronounced or subtle within distinct behavioural contexts. Our findings suggest that while there is variation in USV communication between the two groups of mice, the degree of this diversity may vary depending on the specific behaviour being observed.
Collapse
Affiliation(s)
- Tatiana Pilipenko
- Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Marika Premoli
- Department of Molecular and Translational, Medicine, University of Brescia, Brescia, Italy
| | - Alessandro Gnutti
- Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Sara Anna Bonini
- Department of Molecular and Translational, Medicine, University of Brescia, Brescia, Italy
| | - Riccardo Leonardi
- Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Maurizio Memo
- Department of Molecular and Translational, Medicine, University of Brescia, Brescia, Italy
| | | |
Collapse
|
25
|
Zheng L, Jiao Y, Zhong H, Tan Y, Yin Y, Liu Y, Liu D, Wu M, Wang G, Huang J, Wang P, Qin M, Wang M, Xiao Y, Lv T, Luo Y, Hu H, Hou ST, Kui L. Human-derived fecal microbiota transplantation alleviates social deficits of the BTBR mouse model of autism through a potential mechanism involving vitamin B 6 metabolism. mSystems 2024; 9:e0025724. [PMID: 38780265 PMCID: PMC11237617 DOI: 10.1128/msystems.00257-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition characterized by social communication deficiencies and stereotypic behaviors influenced by hereditary and/or environmental risk factors. There are currently no approved medications for treating the core symptoms of ASD. Human fecal microbiota transplantation (FMT) has emerged as a potential intervention to improve autistic symptoms, but the underlying mechanisms are not fully understood. In this study, we evaluated the effects of human-derived FMT on behavioral and multi-omics profiles of the BTBR mice, an established model for ASD. FMT effectively alleviated the social deficits in the BTBR mice and normalized their distinct plasma metabolic profile, notably reducing the elevated long-chain acylcarnitines. Integrative analysis linked these phenotypic changes to specific Bacteroides species and vitamin B6 metabolism. Indeed, vitamin B6 supplementation improved the social behaviors in BTBR mice. Collectively, these findings shed new light on the interplay between FMT and vitamin B6 metabolism and revealed a potential mechanism underlying the therapeutic role of FMT in ASD.IMPORTANCEAccumulating evidence supports the beneficial effects of human fecal microbiota transplantation (FMT) on symptoms associated with autism spectrum disorder (ASD). However, the precise mechanism by which FMT induces a shift in the microbiota and leads to symptom improvement remains incompletely understood. This study integrated data from colon-content metagenomics, colon-content metabolomics, and plasma metabolomics to investigate the effects of FMT treatment on the BTBR mouse model for ASD. The analysis linked the amelioration of social deficits following FMT treatment to the restoration of mitochondrial function and the modulation of vitamin B6 metabolism. Bacterial species and compounds with beneficial roles in vitamin B6 metabolism and mitochondrial function may further contribute to improving FMT products and designing novel therapies for ASD treatment.
Collapse
Affiliation(s)
- Lifeng Zheng
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
- Xbiome Co. Ltd., Shenzhen, China
| | - Yinming Jiao
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Haolin Zhong
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Yan Tan
- Xbiome Co. Ltd., Shenzhen, China
| | | | | | - Ding Liu
- Xbiome Co. Ltd., Shenzhen, China
| | - Manli Wu
- Xbiome Co. Ltd., Shenzhen, China
| | - Guoyun Wang
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | | | - Ping Wang
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Meirong Qin
- Shenzhen Institute for Drug Control, Shenzhen, China
| | - Mingbang Wang
- Microbiome Therapy Center, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children’s Hospital of Fudan University, National Center for Children’s Health, Shanghai, China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| | - Tiying Lv
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yangzi Luo
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Han Hu
- Xbiome Co. Ltd., Shenzhen, China
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Neuroscience, Southern University of Science and Technology, Shenzhen, China
| | - Ling Kui
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
26
|
Harding CD, Walker KMM, Hackett TD, Herwig A, Peirson SN, Vyazovskiy VV. Ultrasonic vocalisation rate tracks the diurnal pattern of activity in winter phenotype Djungarian hamsters (Phodopus sungorus). J Comp Physiol B 2024; 194:383-401. [PMID: 38733409 PMCID: PMC11233387 DOI: 10.1007/s00360-024-01556-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024]
Abstract
Vocalisations are increasingly being recognised as an important aspect of normal rodent behaviour yet little is known of how they interact with other spontaneous behaviours such as sleep and torpor, particularly in a social setting. We obtained chronic recordings of the vocal behaviour of adult male and female Djungarian hamsters (Phodopus sungorus) housed under short photoperiod (8 h light, 16 h dark, square wave transitions), in different social contexts. The animals were kept in isolation or in same-sex sibling pairs, separated by a grid which allowed non-physical social interaction. On approximately 20% of days hamsters spontaneously entered torpor, a state of metabolic depression that coincides with the rest phase of many small mammal species in response to actual or predicted energy shortages. Animals produced ultrasonic vocalisations (USVs) with a peak frequency of 57 kHz in both social and asocial conditions and there was a high degree of variability in vocalisation rate between subjects. Vocalisation rate was correlated with locomotor activity across the 24-h light cycle, occurring more frequently during the dark period when the hamsters were more active and peaking around light transitions. Solitary-housed animals did not vocalise whilst torpid and animals remained in torpor despite overlapping with vocalisations in social-housing. Besides a minor decrease in peak USV frequency when isolated hamsters were re-paired with their siblings, changing social contexts did not influence vocalisation behaviour or structure. In rare instances, temporally overlapping USVs occurred when animals were socially-housed and were grouped in such a way that could indicate coordination. We did not observe broadband calls (BBCs) contemporaneous with USVs in this paradigm, corroborating their correlation with physical aggression which was absent from our experiment. Overall, we find little evidence to suggest a direct social function of hamster USVs. We conclude that understanding the effects of vocalisations on spontaneous behaviours, such as sleep and torpor, will inform experimental design of future studies, especially where the role of social interactions is investigated.
Collapse
Affiliation(s)
- Christian D Harding
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK.
- Division of Pulmonary, Critical Care, Sleep Medicine and Physiology, University of California San Diego, San Diego, USA.
| | - Kerry M M Walker
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | - Annika Herwig
- Institute of Neurobiology, Ulm University, Ulm, Germany
| | - Stuart N Peirson
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- The Kavli Institute for Nanoscience Discovery, Oxford, UK
| | - Vladyslav V Vyazovskiy
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
- Sir Jules Thorn Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, UK
- The Kavli Institute for Nanoscience Discovery, Oxford, UK
| |
Collapse
|
27
|
Pereira MI, Laranjo M, Gomes M, Edfawy M, Peça J. Maternal behaviours disrupted by Gprasp2 deletion modulate neurodevelopmental trajectory in progeny. Sci Rep 2024; 14:12484. [PMID: 38816497 PMCID: PMC11139669 DOI: 10.1038/s41598-024-62088-x] [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: 09/05/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Autism spectrum disorders (ASDs) are known to present sex-specific differences. At the same time, understanding how maternal behaviours are affected by pathogenic mutations is crucial to translate research efforts since rearing may recursively modulate neurodevelopment phenotype of the progeny. In this work, we focused on the effects of Gprasp2 deletion in females and its impact in progeny care and development. Female mice, wild-type (WT), Gprasp2+/- (HET) or Gprasp2-/- (KO) mutants and their progeny were used and behavioural paradigms targeting anxiety, memory, maternal care, and other social behaviours were performed. Analysis of communication was carried out through daily recordings of ultrasonic vocalizations in isolated pups and cross-fostering experiments were performed to understand the effect of maternal genotype in pup development. We found that Gprasp2-/- females presented striking impairments in social and working memory. Females also showed disruptions in maternal care, as well as physiological and molecular alterations in the reproductive system and hypothalamus, such as the structure of the mammary gland and the expression levels of oxytocin receptor (OxtR) in nulliparous versus primiparous females. We observed alterations in pup communication, particularly a reduced number of calls in Gprasp2 KO pups, which resulted from an interaction effect of the dam and pup genotype. Cross-fostering mutant pups with wild-type dams rescued some of the early defects shown in vocalizations, however, this effect was not bidirectional, as rearing WT pups with Gprasp2-/- dams was not sufficient to induce significant phenotypical alterations. Our results suggest Gprasp2 mutations perturb social and working memory in a sex-independent manner, but impact female-specific behaviours towards progeny care, female physiology, and gene expression. These changes in mutant dams contribute to a disruption in early stages of progeny development. More generally, our results highlight the need to better understand GxE interactions in the context of ASDs, when female behaviour may present a contributing factor in postnatal neurodevelopmental trajectory.
Collapse
Affiliation(s)
- Marta I Pereira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- PhDOC PhD Program, CIBB, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Mariana Laranjo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Marcos Gomes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Mohamed Edfawy
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal
- HEMEX AG, Liestal, Switzerland
| | - João Peça
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, 3000-456, Coimbra, Portugal.
| |
Collapse
|
28
|
Zambon A, Rico LC, Herman M, Gundacker A, Telalovic A, Hartenberger LM, Kuehn R, Romanov RA, Hussaini SA, Harkany T, Pollak DD. Gestational immune activation disrupts hypothalamic neurocircuits of maternal care behavior. Mol Psychiatry 2024; 29:859-873. [PMID: 35581295 PMCID: PMC9112243 DOI: 10.1038/s41380-022-01602-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/27/2022]
Abstract
Immune activation is one of the most common complications during pregnancy, predominantly evoked by viral infections. Nevertheless, how immune activation affects mother-offspring relationships postpartum remains unknown. Here, by using the polyinosinic-polycytidylic acid (Poly I:C) model of gestational infection we show that viral-like immune activation at mid-gestation persistently changes hypothalamic neurocircuit parameters in mouse dams and, consequently, is adverse to parenting behavior. Poly I:C-exposed dams favor non-pup-directed exploratory behavior at the expense of pup retrieval. These behavioral deficits are underlain by dendrite pruning and lesser immediate early gene activation in Galanin (Gal)+ neurons with dam-specific transcriptional signatures that reside in the medial preoptic area (mPOA). Reduced activation of an exclusively inhibitory contingent of these distal-projecting Gal+ neurons allows for increased feed-forward inhibition onto putative dopaminergic neurons in the ventral tegmental area (VTA) in Poly I:C-exposed dams. Notably, destabilized VTA output specifically accompanies post-pup retrieval epochs. We suggest that gestational immunogenic insults bias both threat processing and reward perception, manifesting as disfavored infant caregiving.
Collapse
Affiliation(s)
- Alice Zambon
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Laura Cuenca Rico
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Mathieu Herman
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Amina Telalovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lisa-Marie Hartenberger
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rebekka Kuehn
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Roman A Romanov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - S Abid Hussaini
- Department of Pathology and Cell Biology, Taub Institute, Columbia University Irving Medical Center, New York, NY, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
29
|
Lee SH, Cooke ME, Duan KZ, Williams Avram SK, Song J, Elkahloun AG, McGrady G, Howley A, Samal B, Young WS. Investigation of the Fasciola Cinereum, Absent in BTBR mice, and Comparison with the Hippocampal Area CA2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586108. [PMID: 38883723 PMCID: PMC11178005 DOI: 10.1101/2024.03.21.586108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The arginine vasopressin 1b receptor (Avpr1b) plays an important role in social behaviors including social learning, memory, and aggression, and is known to be a specific marker for the cornu ammonis area 2 (CA2) regions of the hippocampus. The fasciola cinereum (FC) is an anatomical region in which Avpr1b expressing neurons are prominent, but the functional roles of the FC have yet to be investigated. Surprisingly, the FC is absent in the inbred BTBR T+tf/J (BTBR) mouse strain used to study core behavioral deficits of autism. Here, we characterized and compared transcriptomic expression profiles using single nucleus RNA sequencing and identified 7 different subpopulations and heterogeneity within the dorsal CA2 (dCA2) and FC. Mef2c, involved in autism spectrum disorder, is more highly expressed in the FC. Using Hiplex in situ hybridization, we examined the neuroanatomical locations of these subpopulations in the proximal and distal regions of the hippocampus. Anterograde tracing of Avpr1b neurons specific for the FC showed projections to the IG, dCA2, lacunosum molecular layer of CA1, dorsal fornix, septofibrial nuclei, and intermediate lateral septum (iLS). In contrast to the dCA2, inhibition of Avpr1b neurons in the FC by the inhibitory DREADD system during behavioral testing did not impair social memory. We performed single nucleus RNA sequencing in the dCA2 region and compared between wildtype (WT) and BTBR mice. We found that transcriptomic profiles of dCA2 neurons between BTBR and WT mice are very similar as they did not form any unique clusters; yet, we found there were differentially expressed genes between the dCA2s of BTBR and WT mice. Overall, this is a comprehensive study of the comparison of Avpr1b neuronal subpopulations between the FC and dCA2. The fact that FC is absent in BTBR mice, a mouse model for autism spectrum disorder, suggests that the FC may play a role in understanding neuropsychiatric disease.
Collapse
|
30
|
Potenzieri A, Uccella S, Preiti D, Pisoni M, Rosati S, Lavarello C, Bartolucci M, Debellis D, Catalano F, Petretto A, Nobili L, Fellin T, Tucci V, Ramenghi LA, Savardi A, Cancedda L. Early IGF-1 receptor inhibition in mice mimics preterm human brain disorders and reveals a therapeutic target. SCIENCE ADVANCES 2024; 10:eadk8123. [PMID: 38427732 PMCID: PMC10906931 DOI: 10.1126/sciadv.adk8123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Besides recent advances in neonatal care, preterm newborns still develop sex-biased behavioral alterations. Preterms fail to receive placental insulin-like growth factor-1 (IGF-1), a major fetal growth hormone in utero, and low IGF-1 serum levels correlate with preterm poor neurodevelopmental outcomes. Here, we mimicked IGF-1 deficiency of preterm newborns in mice by perinatal administration of an IGF-1 receptor antagonist. This resulted in sex-biased brain microstructural, functional, and behavioral alterations, resembling those of ex-preterm children, which we characterized performing parallel mouse/human behavioral tests. Pharmacological enhancement of GABAergic tonic inhibition by the U.S. Food and Drug Administration-approved drug ganaxolone rescued functional/behavioral alterations in mice. Establishing an unprecedented mouse model of prematurity, our work dissects the mechanisms at the core of abnormal behaviors and identifies a readily translatable therapeutic strategy for preterm brain disorders.
Collapse
Affiliation(s)
- Alberto Potenzieri
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
- Università degli Studi di Genova, via Balbi, 5, 16126 Genoa, Italy
| | - Sara Uccella
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Deborah Preiti
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Matteo Pisoni
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Silvia Rosati
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Chiara Lavarello
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Martina Bartolucci
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Federico Catalano
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, IRCCS Istituto Giannina Gaslini, via Gerolamo Gaslini 5, 16147 Genoa, Italy
| | - Lino Nobili
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Child Neuropsychiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Valter Tucci
- Genetics and Epigenetics of Behavior (GEB) Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Luca A. Ramenghi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy
- Patologia Neonatale, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Annalisa Savardi
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| | - Laura Cancedda
- Brain Development and Disease Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genoa, Italy
| |
Collapse
|
31
|
Hood KE, Hurley LM. Listening to your partner: serotonin increases male responsiveness to female vocal signals in mice. Front Hum Neurosci 2024; 17:1304653. [PMID: 38328678 PMCID: PMC10847236 DOI: 10.3389/fnhum.2023.1304653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/28/2023] [Indexed: 02/09/2024] Open
Abstract
The context surrounding vocal communication can have a strong influence on how vocal signals are perceived. The serotonergic system is well-positioned for modulating the perception of communication signals according to context, because serotonergic neurons are responsive to social context, influence social behavior, and innervate auditory regions. Animals like lab mice can be excellent models for exploring how serotonin affects the primary neural systems involved in vocal perception, including within central auditory regions like the inferior colliculus (IC). Within the IC, serotonergic activity reflects not only the presence of a conspecific, but also the valence of a given social interaction. To assess whether serotonin can influence the perception of vocal signals in male mice, we manipulated serotonin systemically with an injection of its precursor 5-HTP, and locally in the IC with an infusion of fenfluramine, a serotonin reuptake blocker. Mice then participated in a behavioral assay in which males suppress their ultrasonic vocalizations (USVs) in response to the playback of female broadband vocalizations (BBVs), used in defensive aggression by females when interacting with males. Both 5-HTP and fenfluramine increased the suppression of USVs during BBV playback relative to controls. 5-HTP additionally decreased the baseline production of a specific type of USV and male investigation, but neither drug treatment strongly affected male digging or grooming. These findings show that serotonin modifies behavioral responses to vocal signals in mice, in part by acting in auditory brain regions, and suggest that mouse vocal behavior can serve as a useful model for exploring the mechanisms of context in human communication.
Collapse
Affiliation(s)
- Kayleigh E. Hood
- Hurley Lab, Department of Biology, Indiana University, Bloomington, IN, United States
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, United States
| | - Laura M. Hurley
- Hurley Lab, Department of Biology, Indiana University, Bloomington, IN, United States
- Center for the Integrative Study of Animal Behavior, Indiana University, Bloomington, IN, United States
| |
Collapse
|
32
|
McAdams ZL, Gustafson KL, Russell AL, Self R, Petry AL, Lever TE, Ericsson AC. Supplier-origin gut microbiomes affect host body weight and select autism-related behaviors. Gut Microbes 2024; 16:2385524. [PMID: 39679617 DOI: 10.1080/19490976.2024.2385524] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 12/17/2024] Open
Abstract
Autism spectrum disorders (ASD) are complex human neurodiversities increasing in prevalence within the human population. In search of therapeutics to improve quality-of-life for ASD patients, the gut microbiome (GM) has become a promising target as a growing body of work supports roles for the complex community of microorganisms in influencing host behavior via the gut-brain-axis. However, whether naturally-occurring microbial diversity within the host GM affects these behaviors is often overlooked. Here, we applied a model of population-level differences in the GM to a classic ASD model - the BTBR T+ Itpr3tf/J mouse - to assess how complex GMs affect host behavior. Leveraging the naturally occurring differences between supplier-origin GMs, our data demonstrate that differing, complex GMs selectively effect host ASD-related behavior - especially neonatal ultrasonic communication - and reveal a male-specific effect on behavior not typically observed in this strain. We then identified that the body weight of BTBR mice is influenced by the postnatal GM which was potentially mediated by microbiome-dependent effects on energy harvest in the gut. These data provide insight into how variability within the GM affects host behavior and growth, thereby emphasizing the need to incorporate microbial diversity within the host GM as an experimental factor in biomedical research.
Collapse
Affiliation(s)
- Zachary L McAdams
- Molecular Pathogenesis & Therapeutics Program, University of Missouri, Columbia, MO, USA
- MU Metagenomics Center, University of Missouri, Columbia, MO, USA
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Kevin L Gustafson
- MU Metagenomics Center, University of Missouri, Columbia, MO, USA
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Amber L Russell
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Rachel Self
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Amy L Petry
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Teresa E Lever
- Department of Otolaryngology, School of Medicine, University of Missouri, Columbia, MO, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Aaron C Ericsson
- Molecular Pathogenesis & Therapeutics Program, University of Missouri, Columbia, MO, USA
- MU Metagenomics Center, University of Missouri, Columbia, MO, USA
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
33
|
Morgan AT, Scerri TS, Vogel AP, Reid CA, Quach M, Jackson VE, McKenzie C, Burrows EL, Bennett MF, Turner SJ, Reilly S, Horton SE, Block S, Kefalianos E, Frigerio-Domingues C, Sainz E, Rigbye KA, Featherby TJ, Richards KL, Kueh A, Herold MJ, Corbett MA, Gecz J, Helbig I, Thompson-Lake DGY, Liégeois FJ, Morell RJ, Hung A, Drayna D, Scheffer IE, Wright DK, Bahlo M, Hildebrand MS. Stuttering associated with a pathogenic variant in the chaperone protein cyclophilin 40. Brain 2023; 146:5086-5097. [PMID: 37977818 PMCID: PMC10689913 DOI: 10.1093/brain/awad314] [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: 05/12/2023] [Revised: 07/26/2023] [Accepted: 08/10/2023] [Indexed: 11/19/2023] Open
Abstract
Stuttering is a common speech disorder that interrupts speech fluency and tends to cluster in families. Typically, stuttering is characterized by speech sounds, words or syllables which may be repeated or prolonged and speech that may be further interrupted by hesitations or 'blocks'. Rare variants in a small number of genes encoding lysosomal pathway proteins have been linked to stuttering. We studied a large four-generation family in which persistent stuttering was inherited in an autosomal dominant manner with disruption of the cortico-basal-ganglia-thalamo-cortical network found on imaging. Exome sequencing of three affected family members revealed the PPID c.808C>T (p.Pro270Ser) variant that segregated with stuttering in the family. We generated a Ppid p.Pro270Ser knock-in mouse model and performed ex vivo imaging to assess for brain changes. Diffusion-weighted MRI in the mouse revealed significant microstructural changes in the left corticospinal tract, as previously implicated in stuttering. Quantitative susceptibility mapping also detected changes in cortico-striatal-thalamo-cortical loop tissue composition, consistent with findings in affected family members. This is the first report to implicate a chaperone protein in the pathogenesis of stuttering. The humanized Ppid murine model recapitulates network findings observed in affected family members.
Collapse
Affiliation(s)
- Angela T Morgan
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Department of Audiology and Speech Pathology, University of Melbourne, Parkville 3052, Australia
| | - Thomas S Scerri
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
| | - Adam P Vogel
- Department of Audiology and Speech Pathology, University of Melbourne, Parkville 3052, Australia
- Centre for Neuroscience of Speech, The University of Melbourne, Parkville 3053, Australia
- Clinical Trials, Redenlab Inc., Melbourne 3000, Australia
| | - Christopher A Reid
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, 3052, Parkville 3052, Australia
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Heidelberg 3084, Australia
| | - Mara Quach
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne 3004, Australia
| | - Victoria E Jackson
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
| | - Chaseley McKenzie
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, 3052, Parkville 3052, Australia
| | - Emma L Burrows
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, 3052, Parkville 3052, Australia
| | - Mark F Bennett
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Heidelberg 3084, Australia
| | | | - Sheena Reilly
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Menzies Health Institute Queensland, Griffith University, 4215 Southport, Australia
| | - Sarah E Horton
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Department of Audiology and Speech Pathology, University of Melbourne, Parkville 3052, Australia
| | - Susan Block
- Discipline of Speech Pathology, School of Allied Health, La Trobe University, Bundoora 3086, Australia
| | - Elaina Kefalianos
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Department of Audiology and Speech Pathology, University of Melbourne, Parkville 3052, Australia
| | - Carlos Frigerio-Domingues
- Laboratory of Communication Disorders, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892-2320, USA
| | - Eduardo Sainz
- Laboratory of Communication Disorders, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892-2320, USA
| | - Kristin A Rigbye
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Heidelberg 3084, Australia
| | - Travis J Featherby
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, 3052, Parkville 3052, Australia
| | - Kay L Richards
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, 3052, Parkville 3052, Australia
| | - Andrew Kueh
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
| | - Mark A Corbett
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Neurogenetics Research Program, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
| | - Jozef Gecz
- Adelaide Medical School, The University of Adelaide, Adelaide 5000, Australia
- Neurogenetics Research Program, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
| | - Ingo Helbig
- Department of Neurology, Children’s Hospital, Philadelphia, PA 19104, USA
| | - Daisy G Y Thompson-Lake
- Developmental Neurosciences Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Frédérique J Liégeois
- Developmental Neurosciences Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Robert J Morell
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne 3001, Australia
| | - Dennis Drayna
- Laboratory of Communication Disorders, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892-2320, USA
| | - Ingrid E Scheffer
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, 3052, Parkville 3052, Australia
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Heidelberg 3084, Australia
- Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville 3052, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne 3004, Australia
| | - Melanie Bahlo
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville 3052, Australia
- School of Mathematics and Statistics, University of Melbourne, 3010 Parkville, Australia
| | - Michael S Hildebrand
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- Department of Medicine, Epilepsy Research Centre, University of Melbourne, Heidelberg 3084, Australia
| |
Collapse
|
34
|
Biosca-Brull J, Basaure P, Guardia-Escote L, Cabré M, Blanco J, Morales-Navas M, Sánchez-Santed F, Colomina MT. Environmental exposure to chlorpyrifos during gestation, APOE polymorphism and the risk on autistic-like behaviors. ENVIRONMENTAL RESEARCH 2023; 237:116969. [PMID: 37659636 DOI: 10.1016/j.envres.2023.116969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/04/2023] [Accepted: 08/22/2023] [Indexed: 09/04/2023]
Abstract
Autism spectrum disorder (ASD) encompasses several neurodevelopmental conditions characterized by communication and social impairment, as well as repetitive patterns of behavior. However, it can co-occur with other mental conditions such as anxiety. The massive use of chlorpyrifos (CPF) has been linked to the increased prevalence of developmental disorders. Likewise, ASD has also been closely linked to a wide variety of genetic factors. The aims of the present investigation are to study how gestational CPF exposure and APOE polymorphism affects communication skills, early development and mid-term anxiety-like behaviors, as well as, changes in gene expression related to the cholinergic system. C57BL/6J and humanized apoE3 and apoE4 homozygous mice were exposed to 0 or 1 mg/kg/day of CPF through the diet, from gestational day (GD) 12-18. In addition, a group of C57BL/6J females were injected subcutaneously with 300 mg/kg/day of valproic acid (VPA) on GD 12 and 13. This group was used as a positive control for studying some core and associated autism-like behaviors. Communication skills by means of ultrasonic vocalizations and physical/motor development were assessed during the preweaning period, whereas locomotor activity, anxiety-like behaviors and the gene expression of cholinergic elements were evaluated during adolescence. Our results showed that C57BL/6J mice prenatally exposed to CPF or VPA showed a decrease in body weight and a delay in eye opening. Communication and anxiety behavior were affected differently depending on treatment, while gene expression was altered by sex and treatment. In addition, none of the parameters evaluated in apoE transgenic mice exposed to CPF were affected, but there were differences between genotypes. Therefore, we suggest that prenatal CPF exposure and VPA produce divergent effects on communication and anxiety.
Collapse
Affiliation(s)
- Judit Biosca-Brull
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Center of Environmental, Food and Toxicological Technology (TECNATOX), Reus, Spain.
| | - Pia Basaure
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain
| | - Laia Guardia-Escote
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain
| | - Maria Cabré
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, Tarragona, Spain
| | - Jordi Blanco
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Center of Environmental, Food and Toxicological Technology (TECNATOX), Reus, Spain; Universitat Rovira i Virgili, Department of Basic Medical Sciences, Reus, Spain
| | - Miguel Morales-Navas
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - Fernando Sánchez-Santed
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - Maria Teresa Colomina
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Center of Environmental, Food and Toxicological Technology (TECNATOX), Reus, Spain.
| |
Collapse
|
35
|
Dugger SA, Dhindsa RS, Sampaio GDA, Ressler AK, Rafikian EE, Petri S, Letts VA, Teoh J, Ye J, Colombo S, Peng Y, Yang M, Boland MJ, Frankel WN, Goldstein DB. Neurodevelopmental deficits and cell-type-specific transcriptomic perturbations in a mouse model of HNRNPU haploinsufficiency. PLoS Genet 2023; 19:e1010952. [PMID: 37782669 PMCID: PMC10569524 DOI: 10.1371/journal.pgen.1010952] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 10/12/2023] [Accepted: 09/01/2023] [Indexed: 10/04/2023] Open
Abstract
Heterozygous de novo loss-of-function mutations in the gene expression regulator HNRNPU cause an early-onset developmental and epileptic encephalopathy. To gain insight into pathological mechanisms and lay the potential groundwork for developing targeted therapies, we characterized the neurophysiologic and cell-type-specific transcriptomic consequences of a mouse model of HNRNPU haploinsufficiency. Heterozygous mutants demonstrated global developmental delay, impaired ultrasonic vocalizations, cognitive dysfunction and increased seizure susceptibility, thus modeling aspects of the human disease. Single-cell RNA-sequencing of hippocampal and neocortical cells revealed widespread, yet modest, dysregulation of gene expression across mutant neuronal subtypes. We observed an increased burden of differentially-expressed genes in mutant excitatory neurons of the subiculum-a region of the hippocampus implicated in temporal lobe epilepsy. Evaluation of transcriptomic signature reversal as a therapeutic strategy highlights the potential importance of generating cell-type-specific signatures. Overall, this work provides insight into HNRNPU-mediated disease mechanisms and provides a framework for using single-cell RNA-sequencing to study transcriptional regulators implicated in disease.
Collapse
Affiliation(s)
- Sarah A. Dugger
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Ryan S. Dhindsa
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute of Texas Children’s Hospital, Houston, Texas, United States of America
| | - Gabriela De Almeida Sampaio
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Andrew K. Ressler
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Elizabeth E. Rafikian
- Mouse Neurobehavioral Core Facility, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Sabrina Petri
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Verity A. Letts
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - JiaJie Teoh
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Junqiang Ye
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, New York, United States of America
- Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, New York, United States of America
| | - Sophie Colombo
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Yueqing Peng
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Mu Yang
- Mouse Neurobehavioral Core Facility, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Michael J. Boland
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Wayne N. Frankel
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York, United States of America
| | - David B. Goldstein
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York, United States of America
| |
Collapse
|
36
|
Gan-Or B, London M. Cortical circuits modulate mouse social vocalizations. SCIENCE ADVANCES 2023; 9:eade6992. [PMID: 37774030 PMCID: PMC10541007 DOI: 10.1126/sciadv.ade6992] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 08/30/2023] [Indexed: 10/01/2023]
Abstract
Vocalizations provide a means of communication with high fidelity and information rate for many species. Diencephalon and brainstem neural circuits have been shown to control mouse vocal production; however, the role of cortical circuits in this process is debatable. Using electrical and optogenetic stimulation, we identified a cortical region in the anterior cingulate cortex in which stimulation elicits ultrasonic vocalizations. Moreover, fiber photometry showed an increase in Ca2+ dynamics preceding vocal initiation, whereas optogenetic suppression in this cortical area caused mice to emit fewer vocalizations. Last, electrophysiological recordings indicated a differential increase in neural activity in response to female social exposure dependent on vocal output. Together, these results indicate that the cortex is a key node in the neuronal circuits controlling vocal behavior in mice.
Collapse
Affiliation(s)
- Benjamin Gan-Or
- Edmond and Lily Safra Center for Brain Sciences and Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | |
Collapse
|
37
|
Binder MS, Bordey A. Semi-natural housing rescues social behavior and reduces repetitive exploratory behavior of BTBR autistic-like mice. Sci Rep 2023; 13:16260. [PMID: 37758896 PMCID: PMC10533821 DOI: 10.1038/s41598-023-43558-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 09/26/2023] [Indexed: 09/29/2023] Open
Abstract
Environmental enrichment confers numerous benefits when implemented in murine models and can reduce behavioral symptomatology in models of disease, such as autism spectrum disorder (ASD). However, previous work did not examine the impact of early-life environmental enrichment on each core feature of ASD. We thus implemented a social and physical enrichment at birth, modeling a semi-natural housing, and examined its impact on communicative, social, sensory, and repetitive behaviors using BTBR (autistic-like) and C57BL/6 J (B6, wildtype) mice, comparing them to standard housing conditions. We found that environmental enrichment alleviated the social deficit of juvenile BTBR mice and reduced their repetitive exploratory behavior but did not affect their rough versus smooth texture preference nor the number of maternal isolation-induced pup calls. Environmental enrichment only affected the call characteristics of B6 mice. One interpretation of these data is that early-life environmental enrichment has significant therapeutic potential to treat selective core features of ASD. Another interpretation is that reducing environmental complexity causes selective behavioral deficits in ASD-prone mice suggesting that current standard housing may be suboptimal. Overall, our data illustrate the extent to which the environment influences behavior and highlights the importance of considering housing conditions when designing experiments and interpreting behavioral results.
Collapse
Affiliation(s)
- Matthew S Binder
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8082, USA
| | - Angelique Bordey
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8082, USA.
| |
Collapse
|
38
|
Fertan E, Wong AA, Montbrun TSGD, Purdon MK, Roddick KM, Yamamoto T, Brown RE. Early postnatal development of the MDGA2 +/- mouse model of synaptic dysfunction. Behav Brain Res 2023; 452:114590. [PMID: 37499910 DOI: 10.1016/j.bbr.2023.114590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/13/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023]
Abstract
Synaptic dysfunction underlies many neurodevelopmental disorders (NDDs). The membrane-associated mucin domain-containing glycosylphosphatidylinositol anchor proteins (MDGAs) regulate synaptic development by modulating neurexin-neuroligin complex formation. Since understanding the neurodevelopmental profile and the sex-based differences in the manifestation of the symptoms of NDDs is important for their early diagnosis, we tested a mouse model haploinsufficient for MDGA2 (MDGA2+/-) on a neurodevelopmental test battery, containing sensory, motor, and cognitive measures, as well as ultrasonic vocalizations. When male and female MDGA2+/- and wildtype (WT) C57BL/6 J mice were examined from 2 to 23 days of age using this test battery, genotype and sex differences in body weight, sensory-motor processes, and ultrasonic vocalizations were observed. The auditory startle reflex appeared earlier in the MDGA2+/- than in WT mice and the MDGA2+/- mice produced fewer ultrasonic vocalizations. The MDGA2+/- mice showed reduced locomotion and rearing than WT mice in the open field after 17 days of age and spent less time investigating a novel object than WT mice at 21 days of age. Female MDGA2+/- mice weighed less than WT females and showed lower grip strength, indicating a delay in sensory-motor development in MDGA2+/- mice, which appears to be more pronounced in females than males. The behavioural phenotypes resulting from MDGA2 haploinsufficiency suggests that it shows delayed development of motor behaviour, grip strength and exploratory behaviour, non-social phenotypes of NDDs.
Collapse
Affiliation(s)
- Emre Fertan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Aimée A Wong
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | | | - Michaela K Purdon
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kyle M Roddick
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kagawa 761-0793, Japan
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
39
|
Binder MS, Bordey A. The Novel Somatosensory Nose-Poke Adapted Paradigm (SNAP) Is an Effective Tool to Assess Differences in Tactile Sensory Preferences in Autistic-Like Mice. eNeuro 2023; 10:ENEURO.0478-22.2023. [PMID: 37596047 PMCID: PMC10470849 DOI: 10.1523/eneuro.0478-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023] Open
Abstract
One of the most prevalent deficits in autism spectrum disorder (ASD) are sensitivities to sensory stimuli. Despite the prevalence of sensory deficits in autism, there are few paradigms capable of easily assessing sensory behaviors in ASD-like mouse models. We addressed this need by creating the Somatosensory Nose-poke Adapted Paradigm (SNAP), which consists of an elevated platform with 6 holes in the center, half of which are lined with sandpaper and half are smooth, requiring mice to use their whiskers to sense the texture. The SNAP paradigm assesses tactile sensory preferences as well as stereotypy, anxiety, and locomotion. We used two wild-type (neurotypical) mouse strains, C57BL/6J (C57) inbred and CD-1 outbred mice, and two ASD mouse models, BTBR (a model of idiopathic ASD) and Cntnap2 -/- mice (a model of syndromic ASD). We found that both ASD models produced more nose pokes into the rough condition than the smooth condition, suggesting an increased preference for complex tactile stimulation when compared with the neurotypical groups, wherein no differences were observed. Furthermore, we found increased stereotypy and time spent in the center, suggestive of decreased anxiety, only for BTBR mice compared with the other mouse strains. Overall, SNAP is an easy to implement task to assess the degree of preference for complex tactile stimulation in ASD mouse models that can be further modified to exclude possible confounding effects of novelty or anxiety on the sensory preferences.
Collapse
Affiliation(s)
- Matthew S Binder
- Departments of Neurosurgery and Cellular and Molecular Physiology, Wu Tsai Institute, Yale School of Medicine, New Haven, CT 06520-8082
| | - Angelique Bordey
- Departments of Neurosurgery and Cellular and Molecular Physiology, Wu Tsai Institute, Yale School of Medicine, New Haven, CT 06520-8082
| |
Collapse
|
40
|
Becker M, Gorobets D, Shmerkin E, Weinstein-Fudim L, Pinhasov A, Ornoy A. Prenatal SAMe Treatment Changes via Epigenetic Mechanism/s USVs in Young Mice and Hippocampal Monoamines Turnover at Adulthood in a Mouse Model of Social Hierarchy and Depression. Int J Mol Sci 2023; 24:10721. [PMID: 37445911 PMCID: PMC10361211 DOI: 10.3390/ijms241310721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
The role of hippocampal monoamines and their related genes in the etiology and pathogenesis of depression-like behavior, particularly in impaired sociability traits and the meaning of changes in USVs emitted by pups, remains unknown. We assessed the effects of prenatal administration of S-adenosyl-methionine (SAMe) in Sub mice that exhibit depressive-like behavior on serotonergic, dopaminergic and noradrenergic metabolism and the activity of related genes in the hippocampus (HPC) in adulthood in comparison to saline-treated control Sub mice. During postnatal days 4 and 8, we recorded and analyzed the stress-induced USVs emitted by the pups and tried to understand how the changes in the USVs' calls may be related to the changes in the monoamines and the activity of related genes. The recordings of the USVs showed that SAMe induced a reduction in the emitted flat and one-frequency step-up call numbers in PND4 pups, whereas step-down type calls were significantly increased by SAMe in PND8 pups. The reduction in the number of calls induced by SAMe following separation from the mothers implies a reduction in anxiety, which is an additional sign of decreased depressive-like behavior. Prenatal SAMe increased the concentrations of serotonin in the HPC in both male and female mice without any change in the levels of 5HIAA. It also decreased the level of the dopamine metabolite DOPAC in females. There were no changes in the levels of norepinephrine and metabolites. Several changes in the expression of genes associated with monoamine metabolism were also induced by prenatal SAMe. The molecular and biochemical data obtained from the HPC studies are generally in accordance with our previously obtained data from the prefrontal cortex of similarly treated Sub mice on postnatal day 90. The changes in both monoamines and their gene expression observed 2-3 months after SAMe treatment are associated with the previously recorded behavioral improvement and seem to demonstrate that SAMe is effective via an epigenetic mechanism.
Collapse
Affiliation(s)
- Maria Becker
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Denis Gorobets
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Elena Shmerkin
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Liza Weinstein-Fudim
- Department of Medical Neurobiology Hebrew, University Hadassah Medical School, Jerusalem 9112102, Israel
| | - Albert Pinhasov
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Asher Ornoy
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
- Department of Medical Neurobiology Hebrew, University Hadassah Medical School, Jerusalem 9112102, Israel
| |
Collapse
|
41
|
Albertini G, D'Andrea I, Druart M, Béchade C, Nieves-Rivera N, Etienne F, Le Magueresse C, Rebsam A, Heck N, Maroteaux L, Roumier A. Serotonin sensing by microglia conditions the proper development of neuronal circuits and of social and adaptive skills. Mol Psychiatry 2023; 28:2328-2342. [PMID: 37217677 DOI: 10.1038/s41380-023-02048-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 05/24/2023]
Abstract
The proper maturation of emotional and sensory circuits requires fine-tuning of serotonin (5-HT) level during early postnatal development. Consistently, dysfunctions of the serotonergic system have been associated with neurodevelopmental psychiatric diseases, including autism spectrum disorders (ASD). However, the mechanisms underlying the developmental effects of 5-HT remain partially unknown, one obstacle being the action of 5-HT on different cell types. Here, we focused on microglia, which play a role in brain wiring refinement, and we investigated whether the control of these cells by 5-HT is relevant for neurodevelopment and spontaneous behaviors in mice. Since the main 5-HT sensor in microglia is the 5-HT2B receptor subtype, we prevented 5-HT signaling specifically in microglia by conditional invalidation of the Htr2b gene in these cells. We observed that abrogating the serotonergic control of microglia during early postnatal development affects the phagolysosomal compartment of these cells and their proximity to dendritic spines and perturbs neuronal circuits maturation. Furthermore, this early ablation of microglial 5-HT2B receptors leads to adult hyperactivity in a novel environment and behavioral defects in sociability and flexibility. Importantly, we show that these behavioral alterations result from a developmental effect, since they are not observed when microglial Htr2b invalidation is induced later, at P30 onward. Thus, a primary alteration of 5-HT sensing in microglia, during a critical time window between birth and P30, is sufficient to impair social and flexibility skills. This link between 5-HT and microglia may explain the association between serotonergic dysfunctions and behavioral traits like impaired sociability and inadaptability to novelty, which are prominent in psychiatric disorders such as ASD.
Collapse
Affiliation(s)
- Giulia Albertini
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France
| | - Ivana D'Andrea
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France
| | - Mélanie Druart
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France
| | - Catherine Béchade
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France
| | | | - Fanny Etienne
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France
| | | | - Alexandra Rebsam
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012, Paris, France
| | - Nicolas Heck
- Sorbonne Université, CNRS, INSERM, Neurosciences Paris Seine, Institut de Biologie Paris Seine, F-75005, Paris, France
| | - Luc Maroteaux
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France
| | - Anne Roumier
- Sorbonne Université, INSERM, Institut du Fer à Moulin, F-75005, Paris, France.
| |
Collapse
|
42
|
Matthiesen M, Khlaifia A, Steininger CFD, Dadabhoy M, Mumtaz U, Arruda-Carvalho M. Maturation of nucleus accumbens synaptic transmission signals a critical period for the rescue of social deficits in a mouse model of autism spectrum disorder. Mol Brain 2023; 16:46. [PMID: 37226266 DOI: 10.1186/s13041-023-01028-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/20/2023] [Indexed: 05/26/2023] Open
Abstract
Social behavior emerges early in development, a time marked by the onset of neurodevelopmental disorders featuring social deficits, including autism spectrum disorder (ASD). Although social deficits are at the core of the clinical diagnosis of ASD, very little is known about their neural correlates at the time of clinical onset. The nucleus accumbens (NAc), a brain region extensively implicated in social behavior, undergoes synaptic, cellular and molecular alterations in early life, and is particularly affected in ASD mouse models. To explore a link between the maturation of the NAc and neurodevelopmental deficits in social behavior, we compared spontaneous synaptic transmission in NAc shell medium spiny neurons (MSNs) between the highly social C57BL/6J and the idiopathic ASD mouse model BTBR T+Itpr3tf/J at postnatal day (P) 4, P6, P8, P12, P15, P21 and P30. BTBR NAc MSNs display increased spontaneous excitatory transmission during the first postnatal week, and increased inhibition across the first, second and fourth postnatal weeks, suggesting accelerated maturation of excitatory and inhibitory synaptic inputs compared to C57BL/6J mice. BTBR mice also show increased optically evoked medial prefrontal cortex-NAc paired pulse ratios at P15 and P30. These early changes in synaptic transmission are consistent with a potential critical period, which could maximize the efficacy of rescue interventions. To test this, we treated BTBR mice in either early life (P4-P8) or adulthood (P60-P64) with the mTORC1 antagonist rapamycin, a well-established intervention for ASD-like behavior. Rapamycin treatment rescued social interaction deficits in BTBR mice when injected in infancy, but did not affect social interaction in adulthood.
Collapse
Affiliation(s)
- Melina Matthiesen
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Abdessattar Khlaifia
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | | | - Maryam Dadabhoy
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Unza Mumtaz
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, M1C1A4, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S3G5, Canada.
| |
Collapse
|
43
|
Morel C, Martinez Sanchez I, Cherifi Y, Chartrel N, Diaz Heijtz R. Perturbation of maternal gut microbiota in mice during a critical perinatal window influences early neurobehavioral outcomes in offspring. Neuropharmacology 2023; 229:109479. [PMID: 36870672 DOI: 10.1016/j.neuropharm.2023.109479] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The gut microbiota is increasingly recognized as a key environmental factor that shapes host development and physiology, including neural circuits formation and function. Concurrently, there has been growing concern that early-life antibiotic exposure may alter brain developmental trajectories, increasing the risk for neurodevelopmental disorders such as autism spectrum disorder (ASD). Here, we assessed whether perturbation of the maternal gut microbiota in mice during a narrow critical perinatal window (last week of pregnancy and first three postnatal days), induced by exposure to a commonly used broad-spectrum oral antibiotic (ampicillin), influences offspring neurobehavioral outcomes relevant to ASD. Our results demonstrate that neonatal offspring from antibiotic-treated dams display an altered pattern of ultrasonic communication, which was more pronounced in males. Moreover, juvenile male, but not female, offspring from antibiotic-treated dams showed reduced social motivation and social interaction, as well as context-dependent anxiety-like behavior. However, no changes were observed in locomotor or exploratory activity. This behavioral phenotype of exposed juvenile males was associated with reduced gene expression of the oxytocin receptor (OXTR) and several tight-junction proteins in the prefrontal cortex, a key region involved in the regulation of social and emotional behaviors, as well as a mild inflammatory response in the colon. Further, juvenile offspring from exposed dams also showed distinct alterations in several gut bacterial species, including, Lactobacillus murinus, and Parabacteroides goldsteinii. Overall, this study highlights the importance of the maternal microbiome in early-life, and how its perturbation by a widely used antibiotic could contribute to atypical social and emotional development of offspring in a sex-dependent manner.
Collapse
Affiliation(s)
- Cassandre Morel
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden; University of Rouen Normandy, INSERM, NorDIC, UMR, 1239, F-76000, Rouen, France
| | | | - Yamina Cherifi
- University of Rouen Normandy, INSERM, NorDIC, UMR, 1239, F-76000, Rouen, France
| | - Nicolas Chartrel
- University of Rouen Normandy, INSERM, NorDIC, UMR, 1239, F-76000, Rouen, France
| | | |
Collapse
|
44
|
Rashid M, Olson EC. Delayed cortical development in mice with a neural specific deletion of β1 integrin. Front Neurosci 2023; 17:1158419. [PMID: 37250402 PMCID: PMC10213249 DOI: 10.3389/fnins.2023.1158419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
The adhesion systems employed by migrating cortical neurons are not well understood. Genetic deletion studies of focal adhesion kinase (FAK) and paxillin in mice suggested that these classical focal adhesion molecules control the morphology and speed of cortical neuron migration, but whether β1 integrins also regulate migration morphology and speed is not known. We hypothesized that a β1 integrin adhesion complex is required for proper neuronal migration and for proper cortical development. To test this, we have specifically deleted β1 integrin from postmitotic migrating and differentiating neurons by crossing conditional β1 integrin floxed mice into the NEX-Cre transgenic line. Similar to our prior findings with conditional paxillin deficiency, we found that both homozygous and heterozygous deletion of β1 integrin causes transient mispositioning of cortical neurons in the developing cortex when analyzed pre- and perinatally. Paxillin and β1 integrin colocalize in the migrating neurons and deletion of paxillin in the migrating neuron causes an overall reduction of the β1 integrin immunofluorescence signal and reduction in the number of activated β1 integrin puncta in the migrating neurons. These findings suggest that these molecules may form a functional complex in migrating neurons. Similarly, there was an overall reduced number of paxillin+ puncta in the β1 integrin deficient neurons, despite the normal distribution of FAK and Cx26, a connexin required for cortical migration. The double knockout of paxillin and β1 integrin produces a cortical malpositioning phenotype similar to the paxillin or β1 integrin single knockouts, as would be expected if paxillin and β1 integrin function on a common pathway. Importantly, an isolation-induced pup vocalization test showed that β1 integrin mutants produced a significantly smaller number of calls compared to their littermate controls when analyzed at postnatal day 4 (P4) and revealed a several days trend in reduced vocalization development compared to controls. The current study establishes a role for β1 integrin in cortical development and suggests that β1 integrin deficiency leads to migration and neurodevelopmental delays.
Collapse
Affiliation(s)
- Mamunur Rashid
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, United States
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, United States
| | - Eric C. Olson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
45
|
Lin CW, Ellegood J, Tamada K, Miura I, Konda M, Takeshita K, Atarashi K, Lerch JP, Wakana S, McHugh TJ, Takumi T. An old model with new insights: endogenous retroviruses drive the evolvement toward ASD susceptibility and hijack transcription machinery during development. Mol Psychiatry 2023; 28:1932-1945. [PMID: 36882500 PMCID: PMC10575786 DOI: 10.1038/s41380-023-01999-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 03/09/2023]
Abstract
The BTBR T+Itpr3tf/J (BTBR/J) strain is one of the most valid models of idiopathic autism, serving as a potent forward genetics tool to dissect the complexity of autism. We found that a sister strain with an intact corpus callosum, BTBR TF/ArtRbrc (BTBR/R), showed more prominent autism core symptoms but moderate ultrasonic communication/normal hippocampus-dependent memory, which may mimic autism in the high functioning spectrum. Intriguingly, disturbed epigenetic silencing mechanism leads to hyperactive endogenous retrovirus (ERV), a mobile genetic element of ancient retroviral infection, which increases de novo copy number variation (CNV) formation in the two BTBR strains. This feature makes the BTBR strain a still evolving multiple-loci model toward higher ASD susceptibility. Furthermore, active ERV, analogous to virus infection, evades the integrated stress response (ISR) of host defense and hijacks the transcriptional machinery during embryonic development in the BTBR strains. These results suggest dual roles of ERV in the pathogenesis of ASD, driving host genome evolution at a long-term scale and managing cellular pathways in response to viral infection, which has immediate effects on embryonic development. The wild-type Draxin expression in BTBR/R also makes this substrain a more precise model to investigate the core etiology of autism without the interference of impaired forebrain bundles as in BTBR/J.
Collapse
Affiliation(s)
- Chia-Wen Lin
- Laboratory for Mental Biology, RIKEN Brain Science Institute, Wako, 351-0198, Saitama, Japan
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wako, 351-0198, Saitama, Japan
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, 650-0017, Kobe, Japan
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, M5T 3H7, Canada
| | - Kota Tamada
- Laboratory for Mental Biology, RIKEN Brain Science Institute, Wako, 351-0198, Saitama, Japan
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, 650-0017, Kobe, Japan
| | - Ikuo Miura
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Mikiko Konda
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, 160-8582, Tokyo, Japan
| | - Kozue Takeshita
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, 160-8582, Tokyo, Japan
| | - Koji Atarashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, 160-8582, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Tsurumi, 230-0045, Yokohama, Japan
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, M5T 3H7, Canada
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, Oxfordshire, OX39DU, UK
| | - Shigeharu Wakana
- Technology and Development Team for Mouse Phenotype Analysis, Japan Mouse Clinic, RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Thomas J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wako, 351-0198, Saitama, Japan
| | - Toru Takumi
- Laboratory for Mental Biology, RIKEN Brain Science Institute, Wako, 351-0198, Saitama, Japan.
- Department of Physiology and Cell Biology, Kobe University School of Medicine, Chuo, 650-0017, Kobe, Japan.
- RIKEN Center for Biosystems Dynamics Research, Chuo, 650-0047, Kobe, Japan.
| |
Collapse
|
46
|
Higuchi Y, Tachigori SI, Arakawa H. Faded neural projection from the posterior bed nucleus of the stria terminalis to the lateral habenula contributes to social signaling deficit in male BTBR mice as a mouse model of autism. Psychoneuroendocrinology 2023; 149:106004. [PMID: 36543023 DOI: 10.1016/j.psyneuen.2022.106004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/11/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
BTBR T+ Itpr3tf/J (BTBR) mice display several behavioral characteristics, including social deficits resembling the core symptoms of human autism. Atypical social behaviors include sequential processes of assembled cognitive-behavior components, such as recognition, investigatory assessment, and signaling response. This study aimed to elucidate the neural circuits responsible for the regulation of the social signaling response, as shown by scent marking behavior in male mice. We first assessed the recognition and investigatory patterns of male BTBR mice compared to those of C57BL/6 J (B6) mice. Next, we examined their scent-marking behavior as innate social signaling responses adjusted to a confronted feature of social stimuli and situations, along with the expression of c-Fos as a marker of neuronal activity in selected brain areas involved in the regulation of social behavior. The function of the targeted brain area was confirmed by chemogenetic manipulation. We also examined the social peptides, oxytocin and vasopressin neurons of the major brain regions that are associated with the regulation of social behavior. Our data indicate that male BTBR mice are less responsive to the presentation of social stimuli and the expression of social signaling responses, which is paralleled by blunted c-Fos responsivity and vasopressin neurons morphological changes in selected brain areas, including the posterior bed nucleus of the stria terminalis (pBnST) and lateral habenula (LHb) in BTBR mice. Further investigation of LHb function revealed that chemogenetic inhibition and activation of LHb activity can induce a change in scent marking responses in both B6 and BTBR mice. Our elucidation of the downstream LHb circuits controlling scent marking behavior indicates intact function in BTBR mice. The altered morphological characteristics of oxytocin neurons in the paraventricular nucleus of the hypothalamus and vasopressin-positive neurons and axonal projections in the pBnST and LHb appear to underlie the dysfunction of scent marking responses in BTBR mice. (300/300 words).
Collapse
Affiliation(s)
- Yuki Higuchi
- Department of Systems Physiology, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Shun-Ichi Tachigori
- Department of Systems Physiology, University of the Ryukyus, Faculty of Medicine, Okinawa, Japan
| | - Hiroyuki Arakawa
- Department of Systems Physiology, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan.
| |
Collapse
|
47
|
Möhrle D, Yuen M, Zheng A, Haddad FL, Allman BL, Schmid S. Characterizing maternal isolation-induced ultrasonic vocalizations in a gene-environment interaction rat model for autism. GENES, BRAIN, AND BEHAVIOR 2023:e12841. [PMID: 36751016 DOI: 10.1111/gbb.12841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/09/2023]
Abstract
Deficits in social communication and language development belong to the earliest diagnostic criteria of autism spectrum disorders. Of the many risk factors for autism spectrum disorder, the contactin-associated protein-like 2 gene, CNTNAP2, is thought to be important for language development. The present study used a rat model to investigate the potential compounding effects of autism spectrum disorder risk gene mutation and environmental challenges, including breeding conditions or maternal immune activation during pregnancy, on early vocal communication in the offspring. Maternal isolation-induced ultrasonic vocalizations from Cntnap2 wildtype and knockout rats at selected postnatal days were analyzed for their acoustic, temporal and syntax characteristics. Cntnap2 knockout pups from heterozygous breeding showed normal numbers and largely similar temporal structures of ultrasonic vocalizations to wildtype controls, whereas both parameters were affected in homozygously bred knockouts. Homozygous breeding further exacerbated altered pitch and transitioning between call types found in Cntnap2 knockout pups from heterozygous breeding. In contrast, the effect of maternal immune activation on the offspring's vocal communication was confined to call type syntax, but left ultrasonic vocalization acoustic and temporal organization intact. Our results support the "double-hit hypothesis" of autism spectrum disorder risk gene-environment interactions and emphasize that complex features of vocal communication are a useful tool for identifying early autistic-like features in rodent models.
Collapse
Affiliation(s)
- Dorit Möhrle
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Megan Yuen
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Alice Zheng
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Faraj L Haddad
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Brian L Allman
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
48
|
Chen Y, Xiang Z, Su Q, Qin J, Liu Q. Vocal signals with different social or non-social contexts in two wild rodent species (Mus caroli and Rattus losea). Anim Cogn 2023; 26:963-972. [PMID: 36683113 DOI: 10.1007/s10071-023-01745-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
The ultrasonic vocalizations (USVs) of rodents play a substantial role in the communication and interaction between individuals; exhibit a high degree of complexity; and are influenced by a multitude of developmental, environmental, and phylogenetic factors. The functions of USVs are mainly studied in laboratory mice or rats. However, the behavioral relevance of USVs in wild rodents is poorly studied. In this work, we systematically investigated the vocal repertoire of the wild mouse Mus caroli and wild rat Rattus losea in multiple social or non-social contexts, e.g., pup-isolation, juvenile-play, paired opposite-sex encounter, female-female interaction, social-exploring, or foot-shock treatment. Unlike the laboratory mice, M. caroli, whose USVs were recorded during pup-isolation and courtship behaviors, did not produce any vocal sounds during juvenile-play and female-female interactions. R. losea, similar to laboratory rats, emitted USVs in all test situations. We found higher peak frequencies of USVs in both these two wild rodent species than in laboratory animals. Moreover, the parameters and structures of USVs varied significantly across different social or non-social contexts even within each species, confirming the context-sensitivity and complexity of vocal signals in rodents. We also noted a striking difference in call types between these two species: no downward type occurred in M. caroli, but no upward type occurred in R. losea, thereby highlighting the interspecific difference of vocal signals among rodents. Thus, the present study presents behavioral foundations of the vocalization context in wild mice and wild rats, and contributes to revealing the behavioral significance of widely used USVs in rodents.
Collapse
Affiliation(s)
- Yi Chen
- College of Forestry, Central South University of Forestry and Technology, Changsha, China.,Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Zuofu Xiang
- College of Forestry, Central South University of Forestry and Technology, Changsha, China
| | - Qianqian Su
- College of Forestry, Central South University of Forestry and Technology, Changsha, China.,Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiao Qin
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Quansheng Liu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China.
| |
Collapse
|
49
|
Giannoccaro S, Ferraguto C, Petroni V, Marcelly C, Nogues X, Campuzano V, Pietropaolo S. Early Neurobehavioral Characterization of the CD Mouse Model of Williams-Beuren Syndrome. Cells 2023; 12:cells12030391. [PMID: 36766733 PMCID: PMC9913557 DOI: 10.3390/cells12030391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
Williams-Beuren syndrome (WBS) is a neurodevelopmental disorder caused by a chromosomic microdeletion (7q11.23). WBS has been modeled by a mouse line having a complete deletion (CD) of the equivalent mouse locus. This model has been largely used to investigate the etiopathological mechanisms of WBS, although pharmacological therapies have not been identified yet. Surprisingly, CD mice were so far mainly tested in adulthood, despite the developmental nature of WBS and the critical relevance of early timing for potential treatments. Here we provide for the first time a phenotypic characterization of CD mice of both sexes during infancy and adolescence, i.e., between birth and 7 weeks of age. CD pups of both sexes showed reduced body growth, delayed sensory development, and altered patterns of ultrasonic vocalizations and exploratory behaviors. Adolescent CD mice showed reduced locomotion and acoustic startle response, and altered social interaction and communication, the latter being more pronounced in female mice. Juvenile CD mutants of both sexes also displayed reduced brain weight, cortical and hippocampal dendritic length, and spine density. Our findings highlight the critical relevance of early neurobehavioral alterations as biomarkers of WBS pathology, underlying the importance of adolescence for identifying novel therapeutic targets for this neurological disorder.
Collapse
Affiliation(s)
| | - Celeste Ferraguto
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Valeria Petroni
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Coline Marcelly
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000 Bordeaux, France
| | | | - Victoria Campuzano
- Departament de Biomedicina, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Susanna Pietropaolo
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
50
|
Winters C, Gorssen W, Wöhr M, D’Hooge R. BAMBI: A new method for automated assessment of bidirectional early-life interaction between maternal behavior and pup vocalization in mouse dam-pup dyads. Front Behav Neurosci 2023; 17:1139254. [PMID: 36935889 PMCID: PMC10020184 DOI: 10.3389/fnbeh.2023.1139254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Vital early-life dyadic interaction in mice requires a pup to signal its needs adequately, and a dam to recognize and respond to the pup's cues accurately and timely. Previous research might have missed important biological and/or environmental elements of this complex bidirectional interaction, because it often focused on one dyadic member only. In laboratory rodents, the Pup Retrieval Test (PRT) is the leading procedure to assess pup-directed maternal care. The present study describes BAMBI (Bidirectional Automated Mother-pup Behavioral Interaction test), a novel automated PRT methodology based on synchronous video recording of maternal behavior and audio recording of pup vocalizations, which allows to assess bidirectional dam-pup dyadic interaction. We were able to estimate pup retrieval and pup vocalization parameters accurately in 156 pups from 29 dams on postnatal days (PND) 5, 7, 9, 11, and 13. Moreover, we showed an association between number of emitted USVs and retrieval success, indicating dyadic interdependency and bidirectionality. BAMBI is a promising new automated home-cage behavioral method that can be applied to both basic and preclinical studies investigating complex phenotypes related to early-life social development.
Collapse
Affiliation(s)
- Carmen Winters
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- *Correspondence: Carmen Winters, ,
| | - Wim Gorssen
- Department of Biosystems, Center for Animal Breeding and Genetics, KU Leuven, Leuven, Belgium
| | - Markus Wöhr
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Social and Affective Neuroscience Research Group, Laboratory of Biological Psychology, Research Unit Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, Leuven, Belgium
- Behavioral Neuroscience, Experimental and Biological Psychology, Faculty of Psychology, Philipps-University Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, Philipps-University Marburg, Marburg, Germany
| | - Rudi D’Hooge
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|