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
|
Kougias DG, Atillasoy E, Southall MD, Scialli AR, Ejaz S, Chu C, Jeminiwa BO, Massarsky A, Unice KM, Schaeffer TH, Kovochich M. A quantitative weight-of-evidence review of preclinical studies examining the potential developmental neurotoxicity of acetaminophen. Crit Rev Toxicol 2025; 55:124-178. [PMID: 39982125 DOI: 10.1080/10408444.2024.2442344] [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: 07/31/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 02/22/2025]
Abstract
Acetaminophen [paracetamol; N-acetyl-para-aminophenol (APAP)] is an antipyretic/analgesic commonly used in the treatment of fever and mild to moderate pain, headache, myalgia, and dysmenorrhea. Recent literature has questioned the safety of acetaminophen use during pregnancy, with an emphasis on whether exposure to the developing nervous system results in behavioral changes consistent with autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and/or other cognitive deficits in the offspring. No previous review has used a fully detailed, quantitative weight-of-evidence (QWoE) approach to critically examine the preclinical acetaminophen data with regards to potential developmental neurotoxicity (DNT). Following regulatory guidance, a QWoE framework using prespecified scoring criteria was developed consistent with previous approaches to characterize potential adverse DNT outcomes with considerations for biological relevance of the response to adverse outcomes (outcome score) and the strength of methods and study design (methods score). Considerations for the methods score included (1) experimental design, (2) details/reliability of measurement(s), (3) data transparency, and (4) translational/methodological relevance. Considerations for the outcome score included response-related (1) statistical significance, (2) dose-response, (3) relevance/reliability/magnitude, (4) plausibility, and (5) translational relevance, including consideration of systemic toxicity/hepatotoxicity and therapeutic and/or non-systemically toxic doses and durations of use. Application of this QWoE framework to the 34 in vivo studies identified that assess the potential DNT of acetaminophen resulted in 188 QWoE entries documented across 11 DNT endpoints: social behavior, stereotypic behavior, behavioral rigidity, attention/impulsivity, hyperactivity, anxiety-like behavior, sensorimotor function, spatial learning/memory, nonspatial learning/memory, neuroanatomy, and neurotransmission. For each endpoint, the mean outcome score and methods score were calculated for total entries and for entries segregated by sex to assist in determining data quality and potential adversity. Informed by all 188 entries, the QWoE analysis demonstrated data of moderate quality showing no consistent evidence of DNT in male and female rodents following exposure to acetaminophen at therapeutic and/or nonsystemically toxic doses. Although some of the DNT endpoints (behavioral rigidity, attention/impulsivity, spatial learning/memory, neuroanatomy, and neurotransmission) generally displayed a more limited dataset and/or relatively lower data quality, similar conclusions were drawn based on results indicating a lack of biological relevance and reliability of reported adverse effects. Overall, this QWoE analysis on the preclinical in vivo data demonstrates no consistent evidence of adverse effects following developmental exposure to acetaminophen at therapeutic and/or non-systemically toxic doses on the structure and function of the nervous system, including neuroanatomical, neurotransmission, and behavioral endpoints.
Collapse
Affiliation(s)
| | - Evren Atillasoy
- Kenvue Medical Clinical and Safety Sciences, Fort Washington, PA, USA
| | | | - Anthony R Scialli
- Reproductive Toxicology Center, A Non-Profit Foundation, Washington, DC, USA
| | - Sadaff Ejaz
- Kenvue Medical Clinical and Safety Sciences, Skillman, NJ, USA
| | - Christopher Chu
- Kenvue Medical Clinical and Safety Sciences, Skillman, NJ, USA
| | | | | | | | | | | |
Collapse
|
3
|
Yang JQ, Yin BQ, Yang CH, Jiang MM, Li Z. A critical period for paired-housing-dependent autistic-like behaviors attenuation in a prenatal valproic acid-induced male mouse model of autism. Front Neurosci 2025; 18:1467047. [PMID: 39897951 PMCID: PMC11782243 DOI: 10.3389/fnins.2024.1467047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 12/13/2024] [Indexed: 02/04/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social communication and the presence of restrictive and repetitive behaviors. Investigating the etiological process and identifying an appropriate therapeutic target remain as formidable challenges to overcome ASD due to numerous risk factors and complex symptoms associated with the disorder. Recent studies have indicated that early rehabilitative intervention can alleviate the symptoms of individuals with ASD. However, there remain unsolved issues of behavioral intervention such as the appropriate time and types of therapies. In this study, we employed a mouse model prenatally exposed to valproic acid to establish a validated ASD mouse model and We found that paired-housing with control mice for 4 week after weaning palliated sociability deficits, anxiety and repetitive behaviors in this model of ASD-like behaviors, while paired-housing with their ASD littermate did not produce this effects. Furthermore, by evaluating different time window of paired-housing, we found that paired-housing during postnatal day 21 (P21) to P35, but not P21 to P28 or P35 to P49 or P28 to P35, is a critical period for the influence of paired-housing on autistic-like behaviors. Finally, paired-housing with control mice improved the impaired GABA system in this model of ASD. So our study demonstrates the therapeutic potential of environmental intervention during a critical period in the treatment of ASD.
Collapse
Affiliation(s)
| | | | | | | | - Zhe Li
- Department of Children's Rehabilitation, Research Center of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
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
|
5
|
Castellano G, Bonnet Da Silva J, Pietropaolo S. The role of gene-environment interactions in social dysfunction: Focus on preclinical evidence from mouse studies. Neuropharmacology 2024; 261:110179. [PMID: 39369849 DOI: 10.1016/j.neuropharm.2024.110179] [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: 07/28/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Human and animal research has demonstrated that genetic and environmental factors can strongly modulate behavioral function, including the expression of social behaviors and their dysfunctionalities. Several genes have been linked to pathologies characterized by alterations in social behaviors, e.g., aggressive/antisocial personality disorder (ASPD), or autism spectrum disorder (ASD). Environmental stimulation (e.g., physical exercise, environmental enrichment) or adversity (e.g., chronic stress, social isolation) may respectively improve or impair social interactions. While the independent contribution of genetic and environmental factors to social behaviors has been assessed in a variety of human and animal studies, the impact of their interactive effects on social functions has been less extensively investigated. Genetic mutations and environmental changes can indeed influence each other through complex mutual effects, e.g., inducing synergistic, antagonistic or interactive behavioral outcomes. This complexity is difficult to be disentangled in human populations, thus encouraging studies in animal models, especially in the mouse species which is the most suitable for genetic manipulations. Here we review the available preclinical evidence on the impact of gene-environment interactions on social behaviors and their dysfunction, focusing on studies in laboratory mice. We included findings combining naturally occurring mutations, selectively bred or transgenic mice with multiple environmental manipulations, including positive (environmental enrichment, physical exercise) and aversive (social isolation, maternal separation, and stress) experiences. The impact of these results is critically discussed in terms of their generalizability across mouse models and social tests, as well as their implications for human studies on social dysfunction.
Collapse
Affiliation(s)
- Giulia Castellano
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, F-33000, Bordeaux, France
| | | | | |
Collapse
|
6
|
Ünal D, Varol AB, Köse TB, Koçak EE. Morphological Correlates of Behavioral Variation in Autism Spectrum Disorder Groups in A Maternal Immune Activation Model. Noro Psikiyatr Ars 2024; 67:195-201. [PMID: 39258126 PMCID: PMC11382561 DOI: 10.29399/npa.28637] [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: 10/09/2023] [Accepted: 11/16/2023] [Indexed: 09/12/2024] Open
Abstract
Introduction Clinical heterogeneity is one of the biggest challenges for researchers studying underlying neurobiological mechanisms in Autism Spectrum Disorder (ASD). We aimed to use polyinosinic-polycytidylic acid [Poly (I:C)] induced maternal immune activation mice model to investigate the behavioral variation and the role of brain circuits related to symptom clusters in ASD. For this purpose, behavioral tests were applied to offsprings and regional thickness was measured from histological brain sections in medial prefrontal cortex, hippocampus and striatum. Methods Pups of intraperitoneal Poly (I:C)-applied mothers (n: 14) and phosphate buffered saline-applied mothers (n: 6) were used for this study. We used three chamber socialization test and social memory test to evaluate social behavior deficit in mice. Marble burying test was used for assessing stereotypic behavior and new object recognition test for learning and cognitive flexibility. Three subgroups (n: 4 for each) were determined according to behavioral test parameters. Regional thickness was measured in medial prefrontal cortex, hippocampus and striatum and compared between subgroups. Results We detected that the behavioral differences were distributed in a spectrum as expected in the clinic and also detected increased hippocampus thickness in the stereotypic behavior dominant Poly (I:C) subgroup. Conclusion Poly (I:C) induced maternal immune activation model creates the behavioral variation and cortical development differences that are seen in relation with symptom groups in ASD.
Collapse
Affiliation(s)
- Dilek Ünal
- Hacettepe University School of Medicine, Department of Child and Adolescent Psychiatry, Ankara, Turkey
| | - Aslıhan Bahadır Varol
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| | - Tansu Bilge Köse
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| | - Emine Eren Koçak
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| |
Collapse
|
7
|
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
|
8
|
Arakawa H. Revisiting sociability: Factors facilitating approach and avoidance during the three-chamber test. Physiol Behav 2023; 272:114373. [PMID: 37805136 DOI: 10.1016/j.physbeh.2023.114373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/07/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
The three-chamber test, the so-called sociability test, has been widely used to assess social deficits based on impaired socially oriented investigations in rodent models. An innate motivation for investigating conspecifics is theoretically a prerequisite for gaining sociability scores in this paradigm. However, several relevant factors mediating investigatory motives, such as familiarity, attractiveness, and aggression, may affect sociability scores, which must be verified to obtain an adequate evaluation of the psychiatric phenotypes exhibited by disease-relevant rodent models. We assessed the social and non-social factors that mediate proximity preference by the three-chamber test with standard C57BL/6 J (B6) mice and low sociability BTBR+ltpr3tf/J (BTBR) mice. Strains of the opponents had no effect. Sexual cues (i.e., opposite sex) increased proximity preference in both strains of mice; in contrast, novel objects induced an approach in B6 mice but avoidance in BTBR mice. Single-housing before testing, stimulated social motive, affected BTBR mice but not B6 mice. BTBR females showed increased proximity preference across the sessions, and BTBR males showed increased preference toward a male B6 stimulus, but not a male BTBR stimulus. The male preference was restored when the male BTBR stimulus was anesthetized. In addition, self-grooming was facilitated by social and non-social novelty cues in both strains. B6 mice predominantly exhibited an investigatory approach toward social or non-social stimuli, whereas BTBR mice recognized social cues but tended to show avoidance. The three-chamber test could evaluate approach-avoidance strategies in target mouse strains that comprise innate social distance between mice.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department Systems Physiology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Alomar HA, Ansari MA, Nadeem A, Attia SM, Bakheet SA, Al-Mazroua HA, Hussein MH, Alqarni SA, Ahmad SF. A potent and selective CXCR2 antagonist improves neuroimmune dysregulation through the inhibition of NF-κB and notch inflammatory signaling in the BTBR mouse model of autism. J Neuroimmunol 2023; 377:578069. [PMID: 36931207 DOI: 10.1016/j.jneuroim.2023.578069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Autism comprises a broad range of neurodevelopmental disorders characterized by social communication deficits and repetitive and stereotyped behaviors. Chemokine receptor CXCR2 is expressed on neurons and is upregulated in neurological disorders. BTBR T+ Itpr3tf/J (BTBR) mice, a model for autism that shows the core features of ASD. Here, we studied the anti-inflammatory effect of a potent and selective CXCR2 antagonist SB332235 in the BTBR mice. The CXCR2 antagonist represents a promising therapeutic agent for several neuroinflammatory disorders. In this study, we investigated the effects of SB332235 administration on NF-κB-, Notch-1-, Notch-3-, GM-CSF-, MCP-1-, IL-6-, and IL-2- and TGF-β1-expressing CD40+ cells in BTBR and C57BL/6 (C57) mice in the spleen cells by flow cytometry. We further assessed the effect of SB332235 treatment on NF-κB, Notch-1, GM-CSF, MCP-1, IL-6, and IL-2 mRNA expression levels in the brain tissue by RT-PCR. We also explored the effect of SB332235 administration on NF-κB, GM-CSF, IL-6, and TGF-β1 protein expression levels in the brain tissue by western blotting. The SB332235-treated BTBR mice significantly decreases in CD40 + NF-κB+, CD40 + Notch-1+, CD40 + Notch-3+, CD40 + GM-CSF+, CD40 + MCP-1+, CD40 + IL-6+, and CD40 + IL-2+, and increases in CD40 + TGF-β1+ in the spleen cells. Our results further demonstrated that BTBR mice treated with SB332235 effectively decreased NF-κB, Notch-1, GM-CSF, MCP-1, IL-6, and IL-2, increasing TGF-β1 mRNA and protein expression levels in the brain tissue. In conclusion, these results indicate that SB332235 elicits an anti-inflammatory response by downregulating the inflammatory mediators and NF-κB/Notch inflammatory signaling in BTBR mice. This could represent a promising novel therapeutic target for autism treatment.
Collapse
Affiliation(s)
- Hatun A Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Marwa H Hussein
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Alqarni
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
12
|
A comparison of stress reactivity between BTBR and C57BL/6J mice: an impact of early-life stress. Exp Brain Res 2023; 241:687-698. [PMID: 36670311 DOI: 10.1007/s00221-022-06541-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023]
Abstract
Early-life stress (ELS) is associated with hypothalamic-pituitary-adrenal (HPA) axis dysregulation and can increase the risk of psychiatric disorders later in life. The aim of this study was to investigate the influence of ELS on baseline HPA axis functioning and on the response to additional stress in adolescent male mice of strains C57BL/6J and BTBR. As a model of ELS, prolonged separation of pups from their mothers (for 3 h once a day: maternal separation [MS]) was implemented. To evaluate HPA axis activity, we assessed serum corticosterone levels and mRNA expression of corticotropin-releasing hormone (Crh) in the hypothalamus, of steroidogenesis genes in adrenal glands, and of an immediate early gene (c-Fos) in both tissues at baseline and immediately after 1 h of restraint stress. HPA axis activity at baseline did not depend on the history of ELS in mice of both strains. After the exposure to the acute restraint stress, C57BL/6J-MS mice showed less pronounced upregulation of Crh and of corticosterone concentration as compared to the control, indicating a decrease in stress reactivity. By contrast, BTBR-MS mice showed stronger upregulation of c-Fos in the hypothalamus and adrenal glands as compared to controls, thus pointing to greater activation of these organs in response to the acute restraint stress. In addition, we noted that BTBR mice are more stress reactive (than C57BL/6J mice) because they exhibited greater upregulation of corticosterone, c-Fos, and Cyp11a1 in response to the acute restraint stress. Taken together, these results indicate strain-specific and situation-dependent effects of ELS on HPA axis functioning and on c-Fos expression.
Collapse
|
13
|
Cheung A, Konno K, Imamura Y, Matsui A, Abe M, Sakimura K, Sasaoka T, Uemura T, Watanabe M, Futai K. Neurexins in serotonergic neurons regulate neuronal survival, serotonin transmission, and complex mouse behaviors. eLife 2023; 12:85058. [PMID: 36695811 PMCID: PMC9876567 DOI: 10.7554/elife.85058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
Extensive serotonin (5-hydroxytryptamine, 5-HT) innervation throughout the brain corroborates 5-HT's modulatory role in numerous cognitive activities. Volume transmission is the major mode for 5-HT transmission but mechanisms underlying 5-HT signaling are still largely unknown. Abnormal brain 5-HT levels and function have been implicated in autism spectrum disorder (ASD). Neurexin (Nrxn) genes encode presynaptic cell adhesion molecules important for the regulation of synaptic neurotransmitter release, notably glutamatergic and GABAergic transmission. Mutations in Nrxn genes are associated with neurodevelopmental disorders including ASD. However, the role of Nrxn genes in the 5-HT system is poorly understood. Here, we generated a mouse model with all three Nrxn genes disrupted specifically in 5-HT neurons to study how Nrxns affect 5-HT transmission. Loss of Nrxns in 5-HT neurons reduced the number of serotonin neurons in the early postnatal stage, impaired 5-HT release, and decreased 5-HT release sites and serotonin transporter expression. Furthermore, 5-HT neuron-specific Nrxn knockout reduced sociability and increased depressive-like behavior. Our results highlight functional roles for Nrxns in 5-HT neurotransmission, 5-HT neuron survival, and the execution of complex behaviors.
Collapse
Affiliation(s)
- Amy Cheung
- Department of Neurobiology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
- Brudnick Neuropsychiatric Research Institute, University of MassachusettsWorcesterUnited States
- Medical Scientist Training Program, University of MassachusettsWorcesterUnited States
| | - Kotaro Konno
- Department of Anatomy, Faculty of Medicine, Hokkaido UniversitySapporoJapan
| | - Yuka Imamura
- Departments of Pharmacology and Biochemistry & Molecular Biology, Institute for Personalized Medicine, Pennsylvania State University College of Medicine, 500 University DriveHersheyUnited States
| | - Aya Matsui
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Toshikuni Sasaoka
- Department of Comparative and Experimental Medicine, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Takeshi Uemura
- Division of Gene Research, Research Center for Advanced Science, Shinshu UniversityNaganoJapan
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu UniversityNaganoJapan
| | - Masahiko Watanabe
- Department of Anatomy, Faculty of Medicine, Hokkaido UniversitySapporoJapan
| | - Kensuke Futai
- Department of Neurobiology, University of Massachusetts Chan Medical SchoolWorcesterUnited States
- Brudnick Neuropsychiatric Research Institute, University of MassachusettsWorcesterUnited States
| |
Collapse
|
14
|
The live biotherapeutic Blautia stercoris MRx0006 attenuates social deficits, repetitive behaviour, and anxiety-like behaviour in a mouse model relevant to autism. Brain Behav Immun 2022; 106:115-126. [PMID: 35995237 DOI: 10.1016/j.bbi.2022.08.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/27/2022] [Accepted: 08/13/2022] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social behaviour, increased repetitive behaviour, anxiety and gastrointestinal symptoms. The aetiology of ASD is complex and involves an interplay of genetic and environmental factors. Emerging pre-clinical and clinical studies have documented a potential role for the gut microbiome in ASD, and consequently, the microbiota represents a potential target in the development of novel therapeutics for this neurodevelopmental disorder. In this study, we investigate the efficacy of the live biotherapeutic strain, Blautia stercoris MRx0006, in attenuating some of the behavioural deficits in the autism-relevant, genetic mouse model, BTBR T+ Itpr3tf/J (BTBR). We demonstrate that daily oral administration with MRx0006 attenuates social deficits while also decreasing repetitive and anxiety-like behaviour. MRx0006 administration increases the gene expression of oxytocin and its receptor in hypothalamic cells in vitro and increases the expression of hypothalamic arginine vasopressin and oxytocin mRNA in BTBR mice. Additionally at the microbiome level, we observed that MRx0006 administration decreases the abundance of Alistipes putredinis, and modulates the faecal microbial metabolite profile. This alteration in the metabolite profile possibly underlies the observed increase in expression of oxytocin, arginine vasopressin and its receptors, and the consequent improvements in behavioural outcomes. Taken together, these findings suggest that the live biotherapeutic MRx0006 may represent a viable and efficacious treatment option for the management of physiological and behavioural deficits associated with ASD.
Collapse
|
15
|
Winiarski M, Kondrakiewicz L, Kondrakiewicz K, Jędrzejewska‐Szmek J, Turzyński K, Knapska E, Meyza K. Social deficits in BTBR T+ Itpr3tf/J mice vary with ecological validity of the test. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12814. [PMID: 35621219 PMCID: PMC9744492 DOI: 10.1111/gbb.12814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 12/15/2022]
Abstract
Translational value of mouse models of neuropsychiatric disorders depends heavily on the accuracy with which they replicate symptoms observed in the human population. In mouse models of autism spectrum disorder (ASD) these include, among others, social affiliation, and communication deficits as well as impairments in understanding and perception of others. Most studies addressing these issues in the BTBR T+ Itpr3tf/J mouse, an idiopathic model of ASD, were based on short dyadic interactions of often non-familiar partners placed in a novel environment. In such stressful and variable conditions, the reproducibility of the phenotype was low. Here, we compared physical conditions and the degree of habituation of mice at the time of testing in the three chambered social affiliation task, as well as parameters used to measure social deficits and found that both the level of stress and human bias profoundly affect the results of the test. To minimize these effects, we tested social preference and network dynamics in mice group-housed in the Eco-HAB system. This automated recording allowed for long-lasting monitoring of differences in social repertoire (including interest in social stimuli) in BTBR T+ Itpr3tf/J and normosocial c57BL/6J mice. With these observations we further validate the BTBR T+ Itpr3tf/J mouse as a model for ASD, but at the same time emphasize the need for more ecological testing of social behavior within all constructs of the Systems for Social Processes domain (as defined by the Research Domain Criteria framework).
Collapse
Affiliation(s)
- Maciej Winiarski
- Laboratory of Emotions Neurobiology, BRAINCITY – Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental BiologyPolish Academy of SciencesWarsawPoland
| | - Ludwika Kondrakiewicz
- Laboratory of Emotions Neurobiology, BRAINCITY – Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental BiologyPolish Academy of SciencesWarsawPoland
| | - Kacper Kondrakiewicz
- Laboratory of Emotions Neurobiology, BRAINCITY – Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental BiologyPolish Academy of SciencesWarsawPoland,NeuroElectronics Research FlandersLeuvenBelgium
| | - Joanna Jędrzejewska‐Szmek
- Laboratory of Neuroinformatics, Nencki Institute of Experimental BiologyPolish Academy of SciencesWarsawPoland
| | | | - Ewelina Knapska
- Laboratory of Emotions Neurobiology, BRAINCITY – Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental BiologyPolish Academy of SciencesWarsawPoland
| | - Ksenia Meyza
- Laboratory of Emotions Neurobiology, BRAINCITY – Center of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental BiologyPolish Academy of SciencesWarsawPoland
| |
Collapse
|
16
|
Arakawa H. Implication of the social function of excessive self-grooming behavior in BTBR T +ltpr3 tf/J mice as an idiopathic model of autism. Physiol Behav 2021; 237:113432. [PMID: 33901528 DOI: 10.1016/j.physbeh.2021.113432] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 11/24/2022]
Abstract
Autism spectrum disorder (ASD) is defined by two core behavioral characteristics, namely, restricted repetitive behaviors and impaired social-communicative functioning. BTBR T+ltpr3tf/J (BTBR) mice provide a valuable animal model for ASD to elucidate the underlying mechanisms of these two behavioral characteristics of ASD. This study examined the social function of excessive grooming behavior in BTBR mice as a phenotype of restricted repetitive behaviors. Compared to the control C57BL/6 J (B6) strain, BTBR mice showed increased self-grooming when placed alone in a test apparatus, and this behavior was even more evident when confronted with a stimulus mouse (either B6 or BTBR) in a three-chamber test apparatus. While B6 mice tended to groom their face/snout region on the empty side of the chamber, BTBR mice showed excessive grooming with frequent transitions among grooming body regions on the side of the chamber containing a social stimulus. Acute systemic injection of buspirone,a serotonin 1A receptor agonist, as an anxiolytic, facilitated approach behavior toward social stimuli in the three-chamber setting in both B6 and BTBR mice. However, this treatment did not affect grooming behavior in B6 mice and significantly enhanced self-grooming in BTBR mice. These behaviors in BTBR mice suggest a potential signaling function of grooming in response to social stimuli, in which bodywide grooming of BTBR mice expressed in the proximity of social opponents may stimulate the release of olfactory (possibly dismissive) signals. Consequently, the putative neural mechanisms underlying excessive grooming may differ from those regulating social approaches that are associated with anxiolytic mechanisms.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department of Psychology, Tokiwa University, Mito, Ibaraki, Japan; Department of Systems Physiology, University of the Ryukyus, Faculty of Medicine, Nishihara, Okinawa, Japan.
| |
Collapse
|
17
|
Kljakic O, Al-Onaizi M, Janíčková H, Chen KS, Guzman MS, Prado MAM, Prado VF. Cholinergic transmission from the basal forebrain modulates social memory in male mice. Eur J Neurosci 2021; 54:6075-6092. [PMID: 34308559 DOI: 10.1111/ejn.15400] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/06/2021] [Accepted: 07/21/2021] [Indexed: 01/02/2023]
Abstract
Disruptions in social behaviour are prevalent in many neuropsychiatric disorders such as schizophrenia, bipolar disorder and autism spectrum disorders. However, the underlying neurochemical regulation of social behaviour is still not well understood. The central cholinergic system has been proposed to contribute to the regulation of social behaviour. For instance, decreased global levels of acetylcholine release in the brain leads to decreased social interaction and an impairment of social memory in mice. Nonetheless, it has been difficult to ascertain the specific brain areas where cholinergic signalling influences social preference and social memory. In this study, we investigated the impact of different forebrain cholinergic regions on social behaviour by examining mouse lines that differ in their regional expression level of the vesicular acetylcholine transporter-the protein that regulates acetylcholine secretion. We found that when cholinergic signalling is highly disrupted in the striatum, hippocampus, cortex and amygdala mice have intact social preference but are impaired in social memory, as they cannot remember a familiar conspecific nor recognize a novel one. A similar pattern emerges when acetylcholine release is disrupted mainly in the striatum, cortex, and amygdala; however, the ability to recognize novel conspecifics is retained. In contrast, cholinergic signalling of the striatum and amygdala does not appear to significantly contribute to the modulation of social memory and social preference. Furthermore, we demonstrated that increasing global cholinergic tone does not increase social behaviours. Together, these data suggest that cholinergic transmission from the hippocampus and cortex are important for regulating social memory.
Collapse
Affiliation(s)
- Ornela Kljakic
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Mohammed Al-Onaizi
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Helena Janíčková
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Neurochemistry, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Kevin S Chen
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Monica S Guzman
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Marco A M Prado
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Vania F Prado
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.,Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
18
|
Amodeo DA, Oliver B, Pahua A, Hitchcock K, Bykowski A, Tice D, Musleh A, Ryan BC. Serotonin 6 receptor blockade reduces repetitive behavior in the BTBR mouse model of autism spectrum disorder. Pharmacol Biochem Behav 2021; 200:173076. [DOI: 10.1016/j.pbb.2020.173076] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 12/18/2022]
|
19
|
Jahan MS, Ito T, Ichihashi S, Masuda T, Bhuiyan MER, Takahashi I, Takamatsu H, Kumanogoh A, Tsuzuki T, Negishi T, Yukawa K. PlexinA1 deficiency in BALB/cAJ mice leads to excessive self-grooming and reduced prepulse inhibition. IBRO Rep 2020; 9:276-289. [PMID: 33163687 PMCID: PMC7607060 DOI: 10.1016/j.ibror.2020.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/20/2020] [Indexed: 11/17/2022] Open
Abstract
PlexinA1 (PlxnA1) is a transmembrane receptor for semaphorins, a large family of proteins that act as axonal guidance cues during nervous system development. However, there are limited studies on PlxnA1 function in neurobehavior. The present study examined if PlxnA1 deficiency leads to behavioral abnormalities in BALB/cAJ mice. PlxnA1 knockout (KO) mice were generated by homologous recombination and compared to wild type (WT) littermates on a comprehensive battery of behavioral tests, including open field assessment of spontaneous ambulation, state anxiety, and grooming, home cage grooming, the wire hang test of muscle strength, motor coordination on the rotarod task, working memory on the Y maze alternation task, cued and contextual fear conditioning, anxiety on the elevated plus maze, sociability to intruders, and sensory processing as measured by prepulse inhibition (PPI). Measures of motor performance, working memory, fear memory, and sociability did not differ significantly between genotypes, while PlxnA1 KO mice displayed excessive self-grooming, impaired PPI, and slightly lower anxiety. These results suggest a crucial role for PlxnA1 in the development and function of brain regions controlling self-grooming and sensory gating. PlxnA1 KO mice may be a valuable model to investigate the repetitive behaviors and information processing deficits characteristic of many neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Mst Sharifa Jahan
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takuji Ito
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Sachika Ichihashi
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takanobu Masuda
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | | | - Ikuko Takahashi
- Radioisotope Center, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Hyota Takamatsu
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Takamasa Tsuzuki
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Takayuki Negishi
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Kazunori Yukawa
- Department of Physiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
- Corresponding author.
| |
Collapse
|
20
|
Kim J, Ciernia AV. Chromatin Dynamics and Genetic Variation Combine to Regulate Innate Immune Memory. JOURNAL OF CLINICAL & CELLULAR IMMUNOLOGY 2020; 11:595. [PMID: 34295572 PMCID: PMC8294664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Recent work by Ciernia et al. (2020) identified how genetic and epigenetic mechanisms interact to regulate innate immune memory in bone marrow derived macrophages. The authors examined the BTBR strain, a naturally occurring mouse model of Autism Spectrum Disorder (ASD) that captures the complex genetics, behavioral and immune dysregulation found in the human disorder. Immune cell cultures from the BTBR strain compared to the standard C57 showed hyper-responsive immune gene expression that was linked to altered chromatin accessibility at sites with genetic differences between the strains. Together, findings from this work demonstrated that multiple levels of gene regulation likely dictate the formation of innate immune memory and are likely disrupted in immune cells in ASD. Future work will be needed to extend these findings to immune gene regulation in the brain and how changes in immune function are related to abnormal behaviors in brain disorders.
Collapse
Affiliation(s)
- Jennifer Kim
- Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Department of Biochemistry and Molecular Biology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
21
|
Rein B, Yan Z, Wang Z. Diminished social interaction incentive contributes to social deficits in mouse models of autism spectrum disorder. GENES BRAIN AND BEHAVIOR 2019; 19:e12610. [DOI: 10.1111/gbb.12610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/09/2019] [Accepted: 09/21/2019] [Indexed: 01/27/2023]
Affiliation(s)
- Benjamin Rein
- Department of Physiology and Biophysics State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences Buffalo New York
| | - Zhen Yan
- Department of Physiology and Biophysics State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences Buffalo New York
| | - Zi‐Jun Wang
- Department of Physiology and Biophysics State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences Buffalo New York
| |
Collapse
|
22
|
Eissa N, Azimullah S, Jayaprakash P, Jayaraj RL, Reiner D, Ojha SK, Beiram R, Stark H, Łażewska D, Kieć-Kononowicz K, Sadek B. The dual-active histamine H3 receptor antagonist and acetylcholine esterase inhibitor E100 ameliorates stereotyped repetitive behavior and neuroinflammmation in sodium valproate induced autism in mice. Chem Biol Interact 2019; 312:108775. [DOI: 10.1016/j.cbi.2019.108775] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/17/2019] [Accepted: 07/29/2019] [Indexed: 01/03/2023]
|
23
|
Marshall BL, Liu Y, Farrington MJ, Mao J, Helferich WG, Schenk AK, Bivens NJ, Sarma SJ, Lei Z, Sumner LW, Joshi T, Rosenfeld CS. Early genistein exposure of California mice and effects on the gut microbiota-brain axis. J Endocrinol 2019; 242:139-157. [PMID: 31189133 PMCID: PMC6885123 DOI: 10.1530/joe-19-0214] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022]
Abstract
Human offspring encounter high amounts of phytoestrogens, such as genistein (GEN), through maternal diet and soy-based formulas. Such chemicals can exert estrogenic activity and thereby disrupt neurobehavioral programming. Besides inducing direct host effects, GEN might cause gut dysbiosis and alter gut metabolites. To determine whether exposure to GEN affects these parameters, California mice (Peromyscus californicus) dams were placed 2 weeks prior to breeding and throughout gestation and lactation on a diet supplemented with GEN (250 mg/kg feed weight) or AIN93G phytoestrogen-free control diet (AIN). At weaning, offspring socio-communicative behaviors, gut microbiota and metabolite profiles were assayed. Exposure of offspring to GEN-induced sex-dependent changes in gut microbiota and metabolites. GEN exposed females were less likely to investigate a novel female mouse when tested in a three-chamber social test. When isolated, GEN males and females exhibited increased latency to elicit their first call, suggestive of reduced motivation to communicate with other individuals. Correlation analyses revealed interactions between GEN-induced microbiome, metabolome and socio-communicative behaviors. Comparison of GEN males with AIN males revealed the fraction of calls above 20 kHz was associated with daidzein, α-tocopherol, Flexispira spp. and Odoribacter spp. Results suggest early GEN exposure disrupts normal socio-communicative behaviors in California mice, which are otherwise evident in these social rodents. Such effects may be due to GEN disruptions on neural programming but might also be attributed to GEN-induced microbiota shifts and resultant changes in gut metabolites. Findings indicate cause for concern that perinatal exposure to GEN may detrimentally affect the offspring microbiome-gut-brain axis.
Collapse
Affiliation(s)
- Brittney L Marshall
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Yang Liu
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Informatics Institute, University of Missouri, Columbia, Missouri, USA
| | - Michelle J Farrington
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Jiude Mao
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - William G Helferich
- Food Science and Human Nutrition, University of Illinois, Urbana, Illinois, USA
| | | | - Nathan J Bivens
- DNA Core Facility, University of Missouri, Columbia, Missouri, USA
| | - Saurav J Sarma
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- MU Metabolomics Center, University of Missouri, Columbia, Missouri, USA
| | - Zhentian Lei
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- MU Metabolomics Center, University of Missouri, Columbia, Missouri, USA
- Department of Biochemistry, University of Missouri, Columbia, Missouri, USA
| | - Lloyd W Sumner
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- MU Metabolomics Center, University of Missouri, Columbia, Missouri, USA
- Department of Biochemistry, University of Missouri, Columbia, Missouri, USA
| | - Trupti Joshi
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Informatics Institute, University of Missouri, Columbia, Missouri, USA
- Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
- Informatics Institute, University of Missouri, Columbia, Missouri, USA
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, Missouri, USA
- Genetics Area Program, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
24
|
Amodeo DA, Pahua AE, Zarate M, Taylor JA, Peterson S, Posadas R, Oliver BL, Amodeo LR. Differences in the expression of restricted repetitive behaviors in female and male BTBR T + tf/J mice. Behav Brain Res 2019; 372:112028. [PMID: 31212059 DOI: 10.1016/j.bbr.2019.112028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 03/04/2019] [Accepted: 06/08/2019] [Indexed: 01/28/2023]
Abstract
Autism spectrum disorder (ASD) is characterized by the expression of restricted repetitive behaviors (RRBs) and impairments in social recognition and communication. Epidemiological studies demonstrate males are three times more likely than females to be affected. Although this is the case, more recent studies suggest females may be underrepresented in these numbers due to standard clinical measures of RRBs and social behaviors. In addition, many studies examining mouse models of ASD exclude females due to the sex disparity in diagnoses. The present study examined how female and male BTBR T + Itpr3tf /J (BTBR) compare to control C57BL/6J mice on tests of RRBs (probabilistic reversal learning, repetitive grooming, spontaneous alternation, and marble burying) and social behaviors (three chambered social approach task). Utilizing a spatial reversal learning test with 80/20 probabilistic feedback, in which ASD individuals have exhibited deficits, we find that female BTBR mice do not show the same impairment found in male BTBR mice. Interestingly, control female C57BL/6J mice required more trials to reach criterion. Female BTBR mice expressed comparable rates of repetitive grooming, marble burying and spontaneous alternation compared to female C57BL/6J mice. Male BTBR mice expressed higher rates of grooming behavior and locomotor activity compared to male C57BL/6J mice, as found in previous studies. Similarly, male BTBR mice showed a reduction in both measures of social approach compared to controls. Both male and female BTBR mice showed a reduction in sniff time for the stranger mouse compared to controls. Together these findings demonstrate how female BTBR mice do not display the RRB profile expressed by male BTBR mice. Testing of repetitive behaviors in ASD needs to better reflect the sex differences in how RRBs manifest in females compared to their extensively researched male counterparts.
Collapse
Affiliation(s)
- Dionisio A Amodeo
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States.
| | - Alma E Pahua
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Marta Zarate
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Jordan A Taylor
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Sophie Peterson
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Rebekah Posadas
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Brandon L Oliver
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| | - Leslie R Amodeo
- Department of Psychology, California State University San Bernardino, 5500 University Parkway, San Bernardino, CA, 92407, United States
| |
Collapse
|
25
|
Ball NJ, Mercado E, Orduña I. Enriched Environments as a Potential Treatment for Developmental Disorders: A Critical Assessment. Front Psychol 2019; 10:466. [PMID: 30894830 PMCID: PMC6414413 DOI: 10.3389/fpsyg.2019.00466] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/15/2019] [Indexed: 12/18/2022] Open
Abstract
The beneficial effects of enriched environments have been established through a long history of research. Enrichment of the living conditions of captive animals in the form of larger cages, sensory stimulating objects, and opportunities for social interaction and physical exercise, has been shown to reduce emotional reactivity, ameliorate abnormal behaviors, and enhance cognitive functioning. Recently, environmental enrichment research has been extended to humans, in part due to growing interest in its potential therapeutic benefits for children with neurodevelopmental disorders (NDDs). This paper reviews the history of enriched environment research and the use of enriched environments as a developmental intervention in studies of both NDD animal models and children. We argue that while environmental enrichment may sometimes benefit children with NDDs, several methodological factors need to be more closely considered before the efficacy of this approach can be adequately evaluated, including: (i) operationally defining and standardizing enriched environment treatments across studies; (ii) use of control groups and better control over potentially confounding variables; and (iii) a comprehensive theoretical framework capable of predicting when and how environmental enrichment will alter the trajectory of NDDs.
Collapse
Affiliation(s)
- Natalie J Ball
- Neural and Cognitive Plasticity Laboratory, Department of Psychology, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Eduardo Mercado
- Neural and Cognitive Plasticity Laboratory, Department of Psychology, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Itzel Orduña
- Department of School and Counseling Psychology, University at Buffalo, The State University of New York, Buffalo, NY, United States
| |
Collapse
|
26
|
Rhine MA, Parrott JM, Schultz MN, Kazdoba TM, Crawley JN. Hypothesis-driven investigations of diverse pharmacological targets in two mouse models of autism. Autism Res 2019; 12:401-421. [PMID: 30653853 PMCID: PMC6402976 DOI: 10.1002/aur.2066] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/13/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental syndrome diagnosed primarily by persistent deficits in social interactions and communication, unusual sensory reactivity, motor stereotypies, repetitive behaviors, and restricted interests. No FDA‐approved medical treatments exist for the diagnostic symptoms of autism. Here we interrogate multiple pharmacological targets in two distinct mouse models that incorporate well‐replicated autism‐relevant behavioral phenotypes. Compounds that modify inhibitory or excitatory neurotransmission were selected to address hypotheses based on previously published biological abnormalities in each model. Shank3B is a genetic model of a mutation found in autism and Phelan‐McDermid syndrome, in which deficits in excitatory neurotransmission and synaptic plasticity have been reported. BTBR is an inbred strain model of forms of idiopathic autism in which reduced inhibitory neurotransmission and excessive mTOR signaling have been reported. The GABA‐A receptor agonist gaboxadol significantly reduced repetitive self‐grooming in three independent cohorts of BTBR. The TrkB receptor agonist 7,8‐DHF improved spatial learning in Shank3B mice, and reversed aspects of social deficits in BTBR. CX546, a positive allosteric modulator of the glutamatergic AMPA receptor, and d‐cycloserine, a partial agonist of the glycine site on the glutamatergic NMDA receptor, did not rescue aberrant behaviors in Shank3B mice. The mTOR inhibitor rapamycin did not ameliorate social deficits or repetitive behavior in BTBR mice. Comparison of positive and negative pharmacological outcomes, on multiple phenotypes, evaluated for replicability across independent cohorts, enhances the translational value of mouse models of autism for therapeutic discovery. GABA agonists present opportunities for personalized interventions to treat components of autism spectrum disorder. Autism Res 2019, 12: 401–421 © 2019 The Authors. Autism Research published by International Society for Autism Research published by Wiley Periodicals, Inc. Lay Summary Many of the risk genes for autism impair synapses, the connections between nerve cells in the brain. A drug that reverses the synaptic effects of a mutation could offer a precision therapy. Combining pharmacological and behavioral therapies could reduce symptoms and improve the quality of life for people with autism. Here we report reductions in repetitive behavior by a GABA‐A receptor agonist, gaboxadol, and improvements in social and cognitive behaviors by a TrkB receptor agonist, in mouse models of autism.
Collapse
Affiliation(s)
- Maya A Rhine
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Jennifer M Parrott
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Maria N Schultz
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| |
Collapse
|
27
|
Makinodan M, Okumura K, Ikawa D, Yamashita Y, Yamamuro K, Toritsuka M, Kimoto S, Yamauchi T, Komori T, Kayashima Y, Yoshino H, Wanaka A, Kishimoto T. Effects of cross-rearing with social peers on myelination in the medial prefrontal cortex of a mouse model with autism spectrum disorder. Heliyon 2017; 3:e00468. [PMID: 29234739 PMCID: PMC5717317 DOI: 10.1016/j.heliyon.2017.e00468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/01/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social interaction, poor communication skills, and repetitive/restrictive behaviors. Recent studies have indicated that early rehabilitative intervention can alleviate the symptoms of individuals with ASD. However, it remains unknown whether rehabilitative intervention can restore brain structures such as myelin, which generally shows abnormalities in individuals with ASD. Therefore, in the present study, we used a mouse model of ASD (BTBR mice) that demonstrated asocial behaviors and hypomyelination in the medial prefrontal cortex (mPFC) to investigate whether interaction with social peers (C57BL/6J mice) has an effect on myelination. We found that housing with C57BL/6J mice after weaning through adulthood increased the myelin thickness in mPFC, but not in the motor cortex, of BTBR mice. There was no effect of cross-rearing with C57BL/6J mice on axon diameter in mPFC of BTBR mice. This finding suggests that early rehabilitative intervention may alleviate myelin abnormalities in mPFC as well as clinical symptoms in individuals with ASD.
Collapse
Affiliation(s)
- Manabu Makinodan
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Kazuki Okumura
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Daisuke Ikawa
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Yasunori Yamashita
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Kazuhiko Yamamuro
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Michihiro Toritsuka
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Takahira Yamauchi
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Takashi Komori
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Yoshinori Kayashima
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Hiroki Yoshino
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Akio Wanaka
- Department of Anatomy and Neuroscience, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| | - Toshifumi Kishimoto
- Department of Psychiatry, Nara Medical University School of Medicine, 840 Shijo-cho, Kashihara, Nara 634-8522, Japan
| |
Collapse
|
28
|
Kim S, Lee B, Choi JH, Kim JH, Kim CH, Shin HS. Deficiency of a brain-specific chemokine-like molecule, SAM3, induces cardinal phenotypes of autism spectrum disorders in mice. Sci Rep 2017; 7:16503. [PMID: 29184127 PMCID: PMC5705707 DOI: 10.1038/s41598-017-16769-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/12/2017] [Indexed: 12/29/2022] Open
Abstract
Chemokines are small secreted signaling proteins produced by a broad range of cells, including immune cells. Several studies have recently suggested potential roles of chemokines and their receptors in the pathophysiology of autism spectrum disorders (ASDs). SAM3 is a novel brain-specific chemokine-like molecule with an unknown physiological function. We explored the relevance of chemokines in the development of ASD in mice, with a focus on SAM3. We generated Sam3 gene knockout (KO) mice and characterized their behavioral phenotypes, with a focus on those relevant to ASD. Sam3-deficient mice displayed all three core phenotypes of ASD: impaired responses to social novelty, defects in social communication, and increased repetitive behavior. In addition, they showed increased anxiety. Interestingly, gender differences were identified for several behaviors: only male Sam3 KO mice exhibited increased anxiety and increased repetitive behaviors. Sam3 KO mice did not exhibit changes in other behaviors, including locomotor activities, fear learning and memory, and object recognition memory. These findings indicate that a deficiency of SAM3, a novel brain-specific chemokine-like molecule, may lead to the pathogenesis of ASDs and suggest the possibility that SAM3, a soluble factor, could be a novel therapeutic target for ASD treatment.
Collapse
Affiliation(s)
- Sujin Kim
- Center for Cognition and Sociality, Institute for Basic Science, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Basic Science, IBS School, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Boyoung Lee
- Center for Cognition and Sociality, Institute for Basic Science, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jung-Hwa Choi
- Department of Biology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jong-Hyun Kim
- Center for Cognition and Sociality, Institute for Basic Science, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02797, Republic of Korea
- Laboratory of Cell Death and Human Diseases, Department of Life Sciences, School of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Hee-Sup Shin
- Center for Cognition and Sociality, Institute for Basic Science, Yuseong-gu, Daejeon, 34141, Republic of Korea.
- Basic Science, IBS School, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
29
|
Chadman KK. Animal models for autism in 2017 and the consequential implications to drug discovery. Expert Opin Drug Discov 2017; 12:1187-1194. [PMID: 28971687 DOI: 10.1080/17460441.2017.1383982] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is characterized by deficits in social communication and restricted interests/repetitive behaviors, for which there are currently no approved drug treatments. The core symptoms of ASD vary widely in severity and are often accompanied by other neuropsychiatric disorders. Drug discovery has been challenging because of the lack of understanding of the underlying pathophysiology of ASD as well as the heterogeneity of symptoms and symptom severity. Areas covered: In this review, the author discusses animal models of ASD used as targets for drug discovery, focusing primarily on non-syndromic models, primarily rodents. They highlight the wide range of drug targets examined in animal models. While very little of this work has resulted in drug therapy for the behavioral symptoms of ASD yet, it has increased our knowledge of the biology of ASD that is critical for driving drug discovery and has already provided many new drug targets for investigation. Expert opinion: The information gathered from the animal models of ASD is increasing our understanding of the underlying pathophysiology for ASD and is leading to better therapeutic targets. However, the issue of small sample size, heterogeneity within clinical samples, and a lack of replicable outcome measures must be addressed to move forward.
Collapse
Affiliation(s)
- Kathryn K Chadman
- a Behavioral Pharmacology Laboratory , NYS Office for People with Developmental Disabilities, Institute for Basic Research in Developmental Disabilities , Staten Island , NY , USA
| |
Collapse
|
30
|
Martin B, Wang R, Cong WN, Daimon CM, Wu WW, Ni B, Becker KG, Lehrmann E, Wood WH, Zhang Y, Etienne H, van Gastel J, Azmi A, Janssens J, Maudsley S. Altered learning, memory, and social behavior in type 1 taste receptor subunit 3 knock-out mice are associated with neuronal dysfunction. J Biol Chem 2017; 292:11508-11530. [PMID: 28522608 DOI: 10.1074/jbc.m116.773820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 05/03/2017] [Indexed: 12/19/2022] Open
Abstract
The type 1 taste receptor member 3 (T1R3) is a G protein-coupled receptor involved in sweet-taste perception. Besides the tongue, the T1R3 receptor is highly expressed in brain areas implicated in cognition, including the hippocampus and cortex. As cognitive decline is often preceded by significant metabolic or endocrinological dysfunctions regulated by the sweet-taste perception system, we hypothesized that a disruption of the sweet-taste perception in the brain could have a key role in the development of cognitive dysfunction. To assess the importance of the sweet-taste receptors in the brain, we conducted transcriptomic and proteomic analyses of cortical and hippocampal tissues isolated from T1R3 knock-out (T1R3KO) mice. The effect of an impaired sweet-taste perception system on cognition functions were examined by analyzing synaptic integrity and performing animal behavior on T1R3KO mice. Although T1R3KO mice did not present a metabolically disrupted phenotype, bioinformatic interpretation of the high-dimensionality data indicated a strong neurodegenerative signature associated with significant alterations in pathways involved in neuritogenesis, dendritic growth, and synaptogenesis. Furthermore, a significantly reduced dendritic spine density was observed in T1R3KO mice together with alterations in learning and memory functions as well as sociability deficits. Taken together our data suggest that the sweet-taste receptor system plays an important neurotrophic role in the extralingual central nervous tissue that underpins synaptic function, memory acquisition, and social behavior.
Collapse
Affiliation(s)
- Bronwen Martin
- From the Metabolism Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Rui Wang
- From the Metabolism Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Wei-Na Cong
- From the Metabolism Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Caitlin M Daimon
- From the Metabolism Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Wells W Wu
- From the Metabolism Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Bin Ni
- the Receptor Pharmacology Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Kevin G Becker
- the Gene Expression and Genomics Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Elin Lehrmann
- the Gene Expression and Genomics Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - William H Wood
- the Gene Expression and Genomics Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Yongqing Zhang
- the Gene Expression and Genomics Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Harmonie Etienne
- the Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, AN-2610 Antwerp, Belgium, and.,the Department of Biomedical Sciences, University of Antwerp, AN-2610 Antwerp, Belgium
| | - Jaana van Gastel
- the Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, AN-2610 Antwerp, Belgium, and.,the Department of Biomedical Sciences, University of Antwerp, AN-2610 Antwerp, Belgium
| | - Abdelkrim Azmi
- the Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, AN-2610 Antwerp, Belgium, and.,the Department of Biomedical Sciences, University of Antwerp, AN-2610 Antwerp, Belgium
| | - Jonathan Janssens
- the Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, AN-2610 Antwerp, Belgium, and.,the Department of Biomedical Sciences, University of Antwerp, AN-2610 Antwerp, Belgium
| | - Stuart Maudsley
- the Receptor Pharmacology Unit, NIA, National Institutes of Health, Baltimore, Maryland 21224, .,the Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp, AN-2610 Antwerp, Belgium, and.,the Department of Biomedical Sciences, University of Antwerp, AN-2610 Antwerp, Belgium
| |
Collapse
|
31
|
Vogel Ciernia A, Pride MC, Durbin-Johnson B, Noronha A, Chang A, Yasui DH, Crawley JN, LaSalle JM. Early motor phenotype detection in a female mouse model of Rett syndrome is improved by cross-fostering. Hum Mol Genet 2017; 26:1839-1854. [PMID: 28334953 PMCID: PMC6075042 DOI: 10.1093/hmg/ddx087] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 12/14/2022] Open
Abstract
Rett syndrome (RTT) is an X-linked neurodevelopmental disorder caused by mutations in the gene encoding methyl CpG binding protein 2 (MeCP2) that occur sporadically in 1:10,000 female births. RTT is characterized by a period of largely normal development followed by regression in language and motor skills at 6-18 months of age. Mecp2 mutant mice recapitulate many of the clinical features of RTT, but the majority of behavioral assessments have been conducted in male Mecp2 hemizygous null mice as offspring of heterozygous dams. Given that RTT patients are predominantly female, we conducted a systematic analysis of developmental milestones, sensory abilities, and motor deficits, following the longitudinal decline of function from early postnatal to adult ages in female Mecp2 heterozygotes of the conventional Bird line (Mecp2tm1.1bird-/+), as compared to their female wildtype littermate controls. Further, we assessed the impact of postnatal maternal environment on developmental milestones and behavioral phenotypes. Cross-fostering to CD1 dams accelerated several developmental milestones independent of genotype, and induced earlier onset of weight gain in adult female Mecp2tm1.1bird-/+ mice. Cross-fostering improved the sensitivity of a number of motor behaviors that resulted in observable deficits in Mecp2tm1.1bird-/+ mice at much earlier (6-7 weeks) ages than were previously reported (6-9 months). Our findings indicate that female Mecp2tm1.1bird-/+ mice recapitulate many of the motor aspects of RTT syndrome earlier than previously appreciated. In addition, rearing conditions may impact the phenotypic severity and improve the ability to detect genotype differences in female Mecp2 mutant mice.
Collapse
Affiliation(s)
| | | | | | - Adriana Noronha
- Medical Microbiology and Immunology
- Genome Center
- MIND Institute
| | - Alene Chang
- Medical Microbiology and Immunology
- Genome Center
- MIND Institute
| | - Dag H. Yasui
- Medical Microbiology and Immunology
- Genome Center
- MIND Institute
- Department of Psychiatry and Behavioral Sciences
| | | | - Janine M. LaSalle
- Medical Microbiology and Immunology
- Genome Center
- MIND Institute
- Department of Psychiatry and Behavioral Sciences
- Center for Children's Environmental Health, University of California, Davis, UC Davis, CA, USA
| |
Collapse
|
32
|
Bains RS, Wells S, Sillito RR, Armstrong JD, Cater HL, Banks G, Nolan PM. Assessing mouse behaviour throughout the light/dark cycle using automated in-cage analysis tools. J Neurosci Methods 2017; 300:37-47. [PMID: 28456660 PMCID: PMC5909039 DOI: 10.1016/j.jneumeth.2017.04.014] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 12/15/2022]
Abstract
Automated assessment of mouse home-cage behaviour is robust and reliable. Analysis over multiple light/dark cycles improves ability to classify behaviours. Combined RFID and video analysis enables home-cage analysis in group housed animals.
An important factor in reducing variability in mouse test outcomes has been to develop assays that can be used for continuous automated home cage assessment. Our experience has shown that this has been most evidenced in long-term assessment of wheel-running activity in mice. Historically, wheel-running in mice and other rodents have been used as a robust assay to determine, with precision, the inherent period of circadian rhythms in mice. Furthermore, this assay has been instrumental in dissecting the molecular genetic basis of mammalian circadian rhythms. In teasing out the elements of this test that have determined its robustness – automated assessment of an unforced behaviour in the home cage over long time intervals – we and others have been investigating whether similar test apparatus could be used to accurately discriminate differences in distinct behavioural parameters in mice. Firstly, using these systems, we explored behaviours in a number of mouse inbred strains to determine whether we could extract biologically meaningful differences. Secondly, we tested a number of relevant mutant lines to determine how discriminative these parameters were. Our findings show that, when compared to conventional out-of-cage phenotyping, a far deeper understanding of mouse mutant phenotype can be established by monitoring behaviour in the home cage over one or more light:dark cycles.
Collapse
Affiliation(s)
- Rasneer S Bains
- Mary Lyon Centre, MRC Harwell Institute, Harwell Science Campus, Oxfordshire, UK
| | - Sara Wells
- Mary Lyon Centre, MRC Harwell Institute, Harwell Science Campus, Oxfordshire, UK
| | | | - J Douglas Armstrong
- Actual Analytics Ltd., Edinburgh, UK; School of Informatics, University of Edinburgh, Edinburgh, UK
| | - Heather L Cater
- Mary Lyon Centre, MRC Harwell Institute, Harwell Science Campus, Oxfordshire, UK
| | - Gareth Banks
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Science Campus, Oxfordshire, UK
| | - Patrick M Nolan
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Science Campus, Oxfordshire, UK.
| |
Collapse
|
33
|
McCarty R. Cross-fostering: Elucidating the effects of gene×environment interactions on phenotypic development. Neurosci Biobehav Rev 2016; 73:219-254. [PMID: 28034661 DOI: 10.1016/j.neubiorev.2016.12.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/15/2016] [Accepted: 12/17/2016] [Indexed: 02/03/2023]
Abstract
Cross-fostering of litters from soon after birth until weaning is a valuable tool to study the ways in which gene×environment interactions program the development of neural, physiological and behavioral characteristics of mammalian species. In laboratory mice and rats, the primary focus of this review, cross-fostering of litters between mothers of different strains or treatment groups (intraspecific) or between mothers of different species (interspecific) has been conducted over the past 9 decades. Areas of particular interest have included maternal effects on emotionality, social preferences, responses to stressful stimulation, nutrition and growth, blood pressure regulation, and epigenetic effects on brain development and behavior. Results from these areas of research highlight the critical role of the postnatal maternal environment in programming the development of offspring phenotypic characteristics. In addition, experimental paradigms that have included cross-fostering have permitted investigators to tease apart prenatal versus postnatal effects of various treatments on offspring development and behavior.
Collapse
Affiliation(s)
- Richard McCarty
- Department of Psychology, Vanderbilt University, Nashville, TN 37240 USA.
| |
Collapse
|
34
|
Mychasiuk R, Rho JM. Genetic modifications associated with ketogenic diet treatment in the BTBR T+Tf/J mouse model of autism spectrum disorder. Autism Res 2016; 10:456-471. [PMID: 27529337 DOI: 10.1002/aur.1682] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/08/2016] [Accepted: 07/12/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a prevalent and heterogeneous neurodevelopmental disorder characterized by hallmark behavioral features. The spectrum of disorders that fall within the ASD umbrella encompass a distinct but overlapping symptom complex that likely results from an array of molecular and genetic aberrations rather than a single genetic mutation. The ketogenic diet (KD) is a high-fat low-carbohydrate anti-seizure and neuroprotective diet that has demonstrated efficacy in the treatment of ASD-like behaviors in animal and human studies. METHODS We investigated changes in mRNA and gene expression in the BTBR mouse model of ASD that may contribute to the behavioral phenotype. In addition, we sought to examine changes in gene expression following KD treatment in BTBR mice. RESULTS Despite significant behavioral abnormalities, expression changes in BTBR mice did not differ substantially from controls; only 33 genes were differentially expressed in the temporal cortex, and 48 in the hippocampus. Examination of these differentially expressed genes suggested deficits in the stress response and in neuronal signaling/communication. After treatment with the KD, both brain regions demonstrated improvements in ASD deficits associated with myelin formation and white matter development. CONCLUSIONS Although our study supports many of the previously known impairments associated with ASD, such as excessive myelin formation and impaired GABAergic transmission, the RNAseq data and pathway analysis utilized here identified new therapeutic targets for analysis, such as Vitamin D pathways and cAMP signaling. Autism Res 2017, 10: 456-471. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Richelle Mychasiuk
- Departments of Psychology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jong M Rho
- Departments of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
35
|
Kosaki Y, Watanabe S. Impaired Pavlovian predictive learning between temporally phasic but not static events in autism-model strain mice. Neurobiol Learn Mem 2016; 134 Pt B:304-16. [PMID: 27521755 DOI: 10.1016/j.nlm.2016.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 08/01/2016] [Accepted: 08/08/2016] [Indexed: 11/24/2022]
Abstract
Autism-spectrum disorder (ASD) is a multi-aspect developmental disorder characterised by various social and non-social behavioural abnormalities. Using BTBR T+ tf mouse strain (BTBR), a promising animal model displaying a number of behavioural and neural characteristics associated with ASD, we tested the hypothesis that at the core of various symptoms of ASD lies a fundamental deficit in predictive learning between events. In five experiments, we conducted a variety of Pavlovian conditioning tasks, some requiring the establishment of associations between temporally phasic events and others involving static events. BTBR mice were impaired in the acquisition of conditioned magazine approach responses with an appetitive unconditioned stimulus (US) (Experiment 1) and conditioned freezing with an electric shock US (Experiment 2). Both of these tasks had temporally phasic conditioned stimuli (CSs). Conversely, these mice showed normal acquisition of conditioned place preference (CPP), whether the US was a systemic injection of methamphetamine (Experiment 3A) or the presence of food (Experiment 3B). Experiment 4 showed normal acquisition of conditioned taste aversion (CTA) to a flavour-taste compound CS, although BTBR mice still exhibited an abnormal stimulus selection when learning for each element of the compound CS was assessed separately. Experiment 5 revealed a weaker latent inhibition of CTA in BTBR mice. The BTBR mouse's impaired predictive learning between phasic events and intact associations between static events are discussed in terms of dysfunctional contingency-based, but not contiguity-based learning, which may accompany abnormal selective attention to relevant cues. We propose that such dysfunctional contingency learning mechanisms may underlie the development of various social and non-social symptoms of ASD.
Collapse
|
36
|
Guo YP, Commons KG. Serotonin neuron abnormalities in the BTBR mouse model of autism. Autism Res 2016; 10:66-77. [PMID: 27478061 DOI: 10.1002/aur.1665] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/08/2016] [Accepted: 06/13/2016] [Indexed: 12/21/2022]
Abstract
The inbred mouse strain BTBR T+ Itpr3tf /J (BTBR) is studied as a model of idiopathic autism because they are less social and more resistant to change than other strains. Forebrain serotonin receptors and the response to serotonin drugs are altered in BTBR mice, yet it remains unknown if serotonin neurons themselves are abnormal. In this study, we found that serotonin tissue content and the density of serotonin axons is reduced in the hippocampus of BTBR mice in comparison to C57BL/6J (C57) mice. This was accompanied by possible compensatory changes in serotonin neurons that were most pronounced in regions known to provide innervation to the hippocampus: the caudal dorsal raphe (B6) and the median raphe. These changes included increased numbers of serotonin neurons and hyperactivation of Fos expression. Metrics of serotonin neurons in the rostral 2/3 of the dorsal raphe and serotonin content of the prefrontal cortex were less impacted. Thus, serotonin neurons exhibit region-dependent abnormalities in the BTBR mouse that may contribute to their altered behavioral profile. Autism Res 2017, 10: 66-77. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yue-Ping Guo
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, Harbin, China.,Department of Anesthesiology, Perioperative, and Pain Medicine, Boston Children's Hospital; Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Kathryn G Commons
- Department of Anesthesiology, Perioperative, and Pain Medicine, Boston Children's Hospital; Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
37
|
Ergaz Z, Weinstein-Fudim L, Ornoy A. Genetic and non-genetic animal models for autism spectrum disorders (ASD). Reprod Toxicol 2016; 64:116-40. [PMID: 27142188 DOI: 10.1016/j.reprotox.2016.04.024] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) is associated, in addition to complex genetic factors, with a variety of prenatal, perinatal and postnatal etiologies. We discuss the known animal models, mostly in mice and rats, of ASD that helps us to understand the etiology, pathogenesis and treatment of human ASD. We describe only models where behavioral testing has shown autistic like behaviors. Some genetic models mimic known human syndromes like fragile X where ASD is part of the clinical picture, and others are without defined human syndromes. Among the environmentally induced ASD models in rodents, the most common model is the one induced by valproic acid (VPA) either prenatally or early postnatally. VPA induces autism-like behaviors following single exposure during different phases of brain development, implying that the mechanism of action is via a general biological mechanism like epigenetic changes. Maternal infection and inflammation are also associated with ASD in man and animal models.
Collapse
Affiliation(s)
- Zivanit Ergaz
- Laboratory of Teratology, Department of Medical Neurobiology Hebrew University Hadassah Medical School and Hadassah Hospital, Jerusalem, Israel
| | - Liza Weinstein-Fudim
- Laboratory of Teratology, Department of Medical Neurobiology Hebrew University Hadassah Medical School and Hadassah Hospital, Jerusalem, Israel
| | - Asher Ornoy
- Laboratory of Teratology, Department of Medical Neurobiology Hebrew University Hadassah Medical School and Hadassah Hospital, Jerusalem, Israel.
| |
Collapse
|
38
|
Wang Y, Billon C, Walker JK, Burris TP. Therapeutic Effect of a Synthetic RORα/γ Agonist in an Animal Model of Autism. ACS Chem Neurosci 2016; 7:143-8. [PMID: 26625251 PMCID: PMC4759619 DOI: 10.1021/acschemneuro.5b00159] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
![]()
Autism is a developmental disorder
of the nervous system associated
with impaired social communication and interactions as well excessive
repetitive behaviors. There are no drug therapies that directly target
the pathology of this disease. The retinoic acid receptor-related
orphan receptor α (RORα) is a nuclear receptor that has
been demonstrated to have reduced expression in many individuals with
autism spectrum disorder (ASD). Several genes that have been shown
to be downregulated in individuals with ASD have also been identified
as putative RORα target genes. Utilizing a synthetic RORα/γ
agonist, SR1078, that we identified previously, we demonstrate that
treatment of BTBR mice (a model of autism) with SR1078 results in
reduced repetitive behavior. Furthermore, these mice display increased
expression of ASD-associated RORα target genes in both the brains
of the BTBR mice and in a human neuroblastoma cell line treated with
SR1078. These data suggest that pharmacological activation of RORα
may be a method for treatment of autism.
Collapse
Affiliation(s)
- Yongjun Wang
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Cyrielle Billon
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - John K. Walker
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Thomas P. Burris
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| |
Collapse
|
39
|
Kazdoba TM, Hagerman RJ, Zolkowska D, Rogawski MA, Crawley JN. Evaluation of the neuroactive steroid ganaxolone on social and repetitive behaviors in the BTBR mouse model of autism. Psychopharmacology (Berl) 2016; 233:309-23. [PMID: 26525567 PMCID: PMC4703522 DOI: 10.1007/s00213-015-4115-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 10/06/2015] [Indexed: 10/22/2022]
Abstract
RATIONALE Abnormalities in excitatory/inhibitory neurotransmission are hypothesized to contribute to autism spectrum disorder (ASD) etiology. BTBR T (+) Itpr3 (tf) /J (BTBR), an inbred mouse strain, displays social deficits and repetitive self-grooming, offering face validity to ASD diagnostic symptoms. Reduced GABAergic neurotransmission in BTBR suggests that GABAA receptor positive allosteric modulators (PAMs) could improve ASD-relevant BTBR phenotypes. The neuroactive steroid ganaxolone acts as a PAM, displaying anticonvulsant properties in rodent epilepsy models and an anxiolytic-like profile in the elevated plus-maze. OBJECTIVES We evaluated ganaxolone in BTBR and C57BL/6J mice in standardized assays for sociability and repetitive behaviors. Open field and anxiety-related behaviors were tested as internal controls and for comparison with the existing neuroactive steroid literature. RESULTS Ganaxolone improved aspects of social approach and reciprocal social interactions in BTBR, with no effect on repetitive self-grooming, and no detrimental effects in C57BL/6J. Ganaxolone increased overall exploratory activity in BTBR and C57BL/6J in the open field, social approach, and elevated plus-maze, introducing a confound for the interpretation of social improvements. Allopregnanolone and diazepam similarly increased total entries in the elevated plus-maze, indicating that behavioral activation may be a general property of GABAA receptor PAMs in these strains. CONCLUSIONS Ganaxolone shows promise for improving sociability. In addition, ganaxolone, as well as other GABAA receptor PAMs, enhanced overall BTBR activity. The translational implications of specific sociability improvements and nonspecific behavioral activation by ganaxolone in the BTBR model remain to be determined. Future studies to explore whether PAMs provide a novel profile with unique benefits for ASD treatment will be worthwhile.
Collapse
Affiliation(s)
- Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
| | - Randi J Hagerman
- MIND Institute, Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Dorota Zolkowska
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Michael A Rogawski
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
| |
Collapse
|
40
|
GABAB Receptor Agonist R-Baclofen Reverses Social Deficits and Reduces Repetitive Behavior in Two Mouse Models of Autism. Neuropsychopharmacology 2015; 40:2228-39. [PMID: 25754761 PMCID: PMC4613612 DOI: 10.1038/npp.2015.66] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 02/28/2015] [Accepted: 03/03/2015] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is diagnosed by two core behavioral criteria, unusual reciprocal social interactions and communication, and stereotyped, repetitive behaviors with restricted interests. Excitatory/inhibitory imbalance is a prominent hypothesis for the etiology of autism. The selective GABAB receptor agonist R-baclofen previously reversed social deficits and reduced repetitive behaviors in a mouse model of Fragile X syndrome, and Arbaclofen improved some clinical symptoms in some Fragile X and ASD patients. To evaluate R-baclofen in a broader range of mouse models of ASD, we tested both the R-baclofen enantiomer and the less potent S-baclofen enantiomer in two inbred strains of mice that display low sociability and/or high repetitive or stereotyped behaviors. R-baclofen treatment reversed social approach deficits in BTBR T+ Itpr3tf/J (BTBR), reduced repetitive self-grooming and high marble burying scores in BTBR, and reduced stereotyped jumping in C58/J (C58), at nonsedating doses. S-baclofen produced minimal effects at the same doses. These findings encourage investigations of R-baclofen in other preclinical model systems. Additional clinical studies may be warranted to further evaluate the hypothesis that the GABAB receptor represents a promising pharmacological target for treating appropriately stratified subsets of individuals with ASD.
Collapse
|
41
|
Burket JA, Benson AD, Tang AH, Deutsch SI. NMDA receptor activation regulates sociability by its effect on mTOR signaling activity. Prog Neuropsychopharmacol Biol Psychiatry 2015; 60:60-5. [PMID: 25703582 PMCID: PMC5549784 DOI: 10.1016/j.pnpbp.2015.02.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/22/2015] [Accepted: 02/15/2015] [Indexed: 11/20/2022]
Abstract
Tuberous Sclerosis Complex is one example of a syndromic form of autism spectrum disorder associated with disinhibited activity of mTORC1 in neurons (e.g., cerebellar Purkinje cells). mTORC1 is a complex protein possessing serine/threonine kinase activity and a key downstream molecule in a signaling cascade beginning at the cell surface with the transduction of neurotransmitters (e.g., glutamate and acetylcholine) and nerve growth factors (e.g., Brain-Derived Neurotrophic Factor). Interestingly, the severity of the intellectual disability in Tuberous Sclerosis Complex may relate more to this metabolic disturbance (i.e., overactivity of mTOR signaling) than the density of cortical tubers. Several recent reports showed that rapamycin, an inhibitor of mTORC1, improved sociability and other symptoms in mouse models of Tuberous Sclerosis Complex and autism spectrum disorder, consistent with mTORC1 overactivity playing an important pathogenic role. NMDA receptor activation may also dampen mTORC1 activity by at least two possible mechanisms: regulating intraneuronal accumulation of arginine and the phosphorylation status of a specific extracellular signal regulating kinase (i.e., ERK1/2), both of which are "drivers" of mTORC1 activity. Conceivably, the prosocial effects of targeting the NMDA receptor with agonists in mouse models of autism spectrum disorders result from their ability to dampen mTORC1 activity in neurons. Strategies for dampening mTORC1 overactivity by NMDA receptor activation may be preferred to its direct inhibition in chronic neurodevelopmental disorders, such as autism spectrum disorders.
Collapse
Affiliation(s)
- Jessica A Burket
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Andrew D Benson
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Amy H Tang
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States.
| |
Collapse
|
42
|
Kalueff AV, Stewart AM, Song C, Gottesman II. Targeting dynamic interplay among disordered domains or endophenotypes to understand complex neuropsychiatric disorders: Translational lessons from preclinical models. Neurosci Biobehav Rev 2015; 53:25-36. [PMID: 25813308 DOI: 10.1016/j.neubiorev.2015.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 03/12/2015] [Accepted: 03/13/2015] [Indexed: 12/15/2022]
Abstract
Contemporary biological psychiatry uses clinical and experimental (animal) models to increase our understanding of brain pathogenesis. Modeling psychiatric disorders is currently performed by targeting various key neurobehavioral clusters of phenotypic traits (domains), including affective, cognitive, social, motor and reward. Analyses of such domains and their 'smaller units' - individual endophenotypes - are critical for the study of complex brain disorders and their neural underpinnings. The spectrum nature of brain disorders and the importance of pathogenetic linkage among various disordered domains or endophenotypes have also been recognized as an important strategic direction of translational research. Here, we discuss cross-domain analyses of animal models, and focus on their value for mimicking the clinical overlap between disordered neurobehavioral domains in humans. Based on recent experimental evidence, we argue that understanding of brain pathogenesis requires modeling the clinically relevant inter-relationships between various individual endophenotypes (or their domains).
Collapse
Affiliation(s)
- Allan V Kalueff
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524025, Guangdong, China; ZENEREI Institute, 309 Palmer Court, Slidell, LA 70458, USA.
| | - Adam Michael Stewart
- ZENEREI Institute, 309 Palmer Court, Slidell, LA 70458, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524025, Guangdong, China; Department of Psychology and Neuroscience, Dalhousie University, 1355 Oxford St, Halifax, NS B3H 4R2, Canada
| | - Irving I Gottesman
- Department of Psychology, University of Minnesota, Elliot Hall, Minneapolis, MN 55455, USA
| |
Collapse
|
43
|
De Felice A, Scattoni ML, Ricceri L, Calamandrei G. Prenatal exposure to a common organophosphate insecticide delays motor development in a mouse model of idiopathic autism. PLoS One 2015; 10:e0121663. [PMID: 25803479 PMCID: PMC4372449 DOI: 10.1371/journal.pone.0121663] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 01/27/2015] [Indexed: 11/18/2022] Open
Abstract
Autism spectrum disorders are characterized by impaired social and communicative skills and repetitive behaviors. Emerging evidence supported the hypothesis that these neurodevelopmental disorders may result from a combination of genetic susceptibility and exposure to environmental toxins in early developmental phases. This study assessed the effects of prenatal exposure to chlorpyrifos (CPF), a widely diffused organophosphate insecticide endowed with developmental neurotoxicity at sub-toxic doses, in the BTBR T+tf/J mouse strain, a validated model of idiopathic autism that displays several behavioral traits relevant to the autism spectrum. To this aim, pregnant BTBR mice were administered from gestational day 14 to 17 with either vehicle or CPF at a dose of 6 mg/kg/bw by oral gavages. Offspring of both sexes underwent assessment of early developmental milestones, including somatic growth, motor behavior and ultrasound vocalization. To evaluate the potential long-term effects of CPF, two different social behavior patterns typically altered in the BTBR strain (free social interaction with a same-sex companion in females, or interaction with a sexually receptive female in males) were also examined in the two sexes at adulthood. Our findings indicate significant effects of CPF on somatic growth and neonatal motor patterns. CPF treated pups showed reduced weight gain, delayed motor maturation (i.e., persistency of immature patterns such as pivoting at the expenses of coordinated locomotion) and a trend to enhanced ultrasound vocalization. At adulthood, CPF associated alterations were found in males only: the altered pattern of investigation of a sexual partner, previously described in BTBR mice, was enhanced in CPF males, and associated to increased ultrasonic vocalization rate. These findings strengthen the need of future studies to evaluate the role of environmental chemicals in the etiology of neurodevelopment disorders.
Collapse
Affiliation(s)
- Alessia De Felice
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Maria Luisa Scattoni
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Ricceri
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Gemma Calamandrei
- Section of Neurotoxicology and Neuroendocrinology, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
- * E-mail:
| |
Collapse
|
44
|
Wöhr M. Effect of social odor context on the emission of isolation-induced ultrasonic vocalizations in the BTBR T+tf/J mouse model for autism. Front Neurosci 2015; 9:73. [PMID: 25852455 PMCID: PMC4364166 DOI: 10.3389/fnins.2015.00073] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/19/2015] [Indexed: 11/24/2022] Open
Abstract
An important diagnostic criterion for social communication deficits in autism spectrum disorders (ASD) are difficulties in adjusting behavior to suit different social contexts. While the BTBR T+tf/J (BTBR) inbred strain of mice is one of the most commonly used mouse models for ASD, little is known about whether BTBR mice display deficits in detecting changes in social context and their ability to adjust to them. Here, it was tested therefore whether the emission of isolation-induced ultrasonic vocalizations (USV) in BTBR mouse pups is affected by the social odor context, in comparison to the standard control strain with high sociability, C57BL/6J (B6). It is known that the presence of odors from mothers and littermates leads to a calming of the isolated mouse pup, and hence to a reduction in isolation-induced USV emission. In accordance with their behavioral phenotypes with relevance to all diagnostic core symptoms of ASD, it was predicted that BTBR mouse pups would not display a calming response when tested under soiled bedding conditions with home cage bedding material containing maternal odors, and that similar isolation-induced USV emission rates would be seen in BTBR mice tested under clean and soiled bedding conditions. Unexpectedly, however, the present findings show that BTBR mouse pups display such a calming response and emit fewer isolation-induced USV when tested under soiled as compared to clean bedding conditions, similar to B6 mouse pups. Yet, in contrast to B6 mouse pups, which emitted isolation-induced USV with shorter call durations and lower levels of frequency modulation under soiled bedding conditions, social odor context had no effect on acoustic call features in BTBR mouse pups. This indicates that the BTBR mouse model for ASD does not display deficits in detecting changes in social context, but has a limited ability and/or reduced motivation to adjust to them.
Collapse
Affiliation(s)
- Markus Wöhr
- Behavioral Neuroscience, Experimental and Physiological Psychology, Philipps-University of Marburg Marburg, Germany
| |
Collapse
|
45
|
Careaga M, Schwartzer J, Ashwood P. Inflammatory profiles in the BTBR mouse: how relevant are they to autism spectrum disorders? Brain Behav Immun 2015; 43:11-6. [PMID: 24937468 PMCID: PMC4776653 DOI: 10.1016/j.bbi.2014.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/23/2014] [Accepted: 06/06/2014] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of disorders characterized by core behavioral features including stereotyped interests, repetitive behaviors and impairments in communication and social interaction. In addition, widespread changes in the immune systems of individuals with ASD have been identified, in particular increased evidence of inflammation in the periphery and central nervous system. While the etiology of these disorders remains unclear, it appears that multiple gene and environmental factors are involved. The need for animal models paralleling the behavioral and immunological features of ASD is paramount to better understand the link between immune system dysregulation and behavioral deficits observed in these disorders. As such, the asocial BTBR mouse strain displays both ASD relevant behaviors and persistent immune dysregulation, providing a model system that has and continues to be instructive in understanding the complex nature of ASD.
Collapse
Affiliation(s)
- Milo Careaga
- Department of Medical Microbiology and Immunology, UC Davis, United States,The M.I.N.D. Institute, University of California at Davis, CA, United States
| | - Jared Schwartzer
- Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, United States
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, United States; The M.I.N.D. Institute, University of California at Davis, CA, United States.
| |
Collapse
|
46
|
Long-term exposure to intranasal oxytocin in a mouse autism model. Transl Psychiatry 2014; 4:e480. [PMID: 25386957 PMCID: PMC4259989 DOI: 10.1038/tp.2014.117] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 08/28/2014] [Accepted: 09/17/2014] [Indexed: 11/09/2022] Open
Abstract
Oxytocin (OT) is a neuropeptide involved in mammalian social behavior. It is currently in clinical trials for the treatment of autism spectrum disorder (ASD). Previous studies in healthy rodents (prairie voles and C57BL/6J mice) have shown that there may be detrimental effects of long-term intranasal administration, raising the questions about safety and efficacy. To investigate the effects of OT on the aspects of ASD phenotype, we conducted the first study of chronic intranasal OT in a well-validated mouse model of autism, the BTBR T+ Itpr3tf/J inbred strain (BTBR), which displays low sociability and high repetitive behaviors. BTBR and C57BL/6J (B6) mice (N=94) were administered 0.8 IU/kg of OT intranasally, daily for 30 days, starting on day 21. We ran a well-characterized set of behavioral tasks relevant to diagnostic and associated symptoms of autism, including juvenile reciprocal social interactions, three-chambered social approach, open-field exploratory activity, repetitive self-grooming and fear-conditioned learning and memory, some during and some post treatment. Intranasal OT did not improve autism-relevant behaviors in BTBR, except for female sniffing in the three-chambered social interaction test. Male saline-treated BTBR mice showed increased interest in a novel mouse, both in chamber time and sniffing time, whereas OT-treated male BTBR mice showed a preference for the novel mouse in sniffing time only. No deleterious effects of OT were detected in either B6 or BTBR mice, except possibly for the lack of a preference for the novel mouse's chamber in OT-treated male BTBR mice. These results highlight the complexity inherent in understanding the effects of OT on behavior. Future investigations of chronic intranasal OT should include a wider dose range and early developmental time points in both healthy rodents and ASD models to affirm the efficacy and safety of OT.
Collapse
|
47
|
Subchronic treatment of donepezil rescues impaired social, hyperactive, and stereotypic behavior in valproic acid-induced animal model of autism. PLoS One 2014; 9:e104927. [PMID: 25133713 PMCID: PMC4136791 DOI: 10.1371/journal.pone.0104927] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 07/16/2014] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of pervasive developmental disorders with core symptoms such as sociability deficit, language impairment, and repetitive/restricted behaviors. Although worldwide prevalence of ASD has been increased continuously, therapeutic agents to ameliorate the core symptoms especially social deficits, are very limited. In this study, we investigated therapeutic potential of donepezil for ASD using valproic acid-induced autistic animal model (VPA animal model). We found that prenatal exposure of valproic acid (VPA) induced dysregulation of cholinergic neuronal development, most notably the up-regulation of acetylcholinesterase (AChE) in the prefrontal cortex of affected rat and mouse offspring. Similarly, differentiating cortical neural progenitor cell in culture treated with VPA showed increased expression of AChE in vitro. Chromatin precipitation experiments revealed that acetylation of histone H3 bound to AChE promoter region was increased by VPA. In addition, other histone deacetyalse inhibitors (HDACIs) such as trichostatin A and sodium butyrate also increased the expression of AChE in differentiating neural progenitor cells suggesting the essential role of HDACIs in the regulation of AChE expression. For behavioral analysis, we injected PBS or donepezil (0.3 mg/kg) intraperitoneally to control and VPA mice once daily from postnatal day 14 all throughout the experiment. Subchronic treatment of donepezil improved sociability and prevented repetitive behavior and hyperactivity of VPA-treated mice offspring. Taken together, these results provide evidence that dysregulation of ACh system represented by the up-regulation of AChE may serve as an effective pharmacological therapeutic target against autistic behaviors in VPA animal model of ASD, which should be subjected for further investigation to verify the clinical relevance.
Collapse
|
48
|
Amodeo DA, Yi J, Sweeney JA, Ragozzino ME. Oxotremorine treatment reduces repetitive behaviors in BTBR T+ tf/J mice. Front Synaptic Neurosci 2014; 6:17. [PMID: 25165445 PMCID: PMC4131251 DOI: 10.3389/fnsyn.2014.00017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/20/2014] [Indexed: 11/18/2022] Open
Abstract
Repetitive behaviors with restricted interests is one of the core criteria for the diagnosis of autism spectrum disorder (ASD). Current pharmacotherapies that target the dopaminergic or serotonergic systems have limited effectiveness in treating repetitive behaviors. Previous research has demonstrated that administration of muscarinic cholinergic receptor (mAChR) antagonists can exacerbate motor stereotypies while mAChR agonists reduce stereotypies. The present study determined whether the mAChR agonist, oxotremorine affected repetitive behaviors in the BTBR T+ tf/J (BTBR) mouse model of autism. To test the effects of oxotremorine on repetitive behaviors, marble burying and grooming behavior were measured in BTBR mice and compared to that in C57BL/6J (B6) mice. The effects of oxotremorine on locomotor activity was also measured. Thirty minutes before each test, mice received an intraperitoneal (ip) injection of saline, 0.001 mg or 0.01 mg of oxotremorine methiodide. Saline- treated BTBR mice exhibited increased marble burying and self-grooming behavior compared to that of saline-treated B6 mice. Oxotremorine significantly reduced marble burying and self-grooming behavior in BTBR mice, but had no significant effect in B6 mice. In addition, oxotremorine did not affect locomotor activity in BTBR mice, but significantly reduced locomotor activity in B6 mice at the 0.01 mg dose. These findings demonstrate that activation of mAChRs reduces repetitive behavior in the BTBR mouse and suggest that treatment with a mAChR agonist may be effective in reducing repetitive behaviors in ASD.
Collapse
Affiliation(s)
- Dionisio A Amodeo
- Laboratory of Integrative Neuroscience, Department of Psychology, University of Illinois at Chicago Chicago, IL, USA
| | - Julia Yi
- Laboratory of Integrative Neuroscience, Department of Psychology, University of Illinois at Chicago Chicago, IL, USA
| | - John A Sweeney
- Department of Psychiatry, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - Michael E Ragozzino
- Laboratory of Integrative Neuroscience, Department of Psychology, University of Illinois at Chicago Chicago, IL, USA
| |
Collapse
|
49
|
Kulesskaya N, Karpova NN, Ma L, Tian L, Voikar V. Mixed housing with DBA/2 mice induces stress in C57BL/6 mice: implications for interventions based on social enrichment. Front Behav Neurosci 2014; 8:257. [PMID: 25147512 PMCID: PMC4123727 DOI: 10.3389/fnbeh.2014.00257] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 07/13/2014] [Indexed: 12/11/2022] Open
Abstract
Several behavioral interventions, based on social enrichment and observational learning are applied in treatment of neuropsychiatric disorders. However, the mechanism of such modulatory effect and the safety of applied methods on individuals involved in social support need further investigation. We took advantage of known differences between inbred mouse strains to reveal the effect of social enrichment on behavior and neurobiology of animals with different behavioral phenotypes. C57BL/6 and DBA/2 female mice displaying multiple differences in cognitive, social, and emotional behavior were group-housed either in same-strain or in mixed-strain conditions. Comprehensive behavioral phenotyping and analysis of expression of several plasticity- and stress-related genes were done to measure the reciprocal effects of social interaction between the strains. Contrary to our expectation, mixed housing did not change the behavior of DBA/2 mice. Nevertheless, the level of serum corticosterone and the expression of glucocorticoid receptor Nr3c1 in the brain were increased in mixed housed DBA/2 as compared with those of separately housed DBA/2 mice. In contrast, socially active C57BL/6 animals were more sensitive to the mixed housing, displaying several signs of stress: alterations in learning, social, and anxiety-like behavior and anhedonia. These behavioral impairments were accompanied by the elevated serum corticosterone and the reduced expression of Nr3c1, as well as the elevated Bdnf levels in the cortex and hippocampus. Our results demonstrate the importance of social factors in modulation of both behavior and the underlying neurobiological mechanisms in stress response, and draw attention to the potential negative impact of social interventions for individuals involved in social support.
Collapse
Affiliation(s)
| | - Nina N Karpova
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Li Ma
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Li Tian
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Vootele Voikar
- Neuroscience Center, University of Helsinki Helsinki, Finland
| |
Collapse
|
50
|
Squillace M, Dodero L, Federici M, Migliarini S, Errico F, Napolitano F, Krashia P, Di Maio A, Galbusera A, Bifone A, Scattoni ML, Pasqualetti M, Mercuri NB, Usiello A, Gozzi A. Dysfunctional dopaminergic neurotransmission in asocial BTBR mice. Transl Psychiatry 2014; 4:e427. [PMID: 25136890 PMCID: PMC4150243 DOI: 10.1038/tp.2014.69] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/28/2014] [Accepted: 06/23/2014] [Indexed: 01/05/2023] Open
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental conditions characterized by pronounced social and communication deficits and stereotyped behaviours. Recent psychosocial and neuroimaging studies have highlighted reward-processing deficits and reduced dopamine (DA) mesolimbic circuit reactivity in ASD patients. However, the neurobiological and molecular determinants of these deficits remain undetermined. Mouse models recapitulating ASD-like phenotypes could help generate hypotheses about the origin and neurophysiological underpinnings of clinically relevant traits. Here we used functional magnetic resonance imaging (fMRI), behavioural and molecular readouts to probe dopamine neurotransmission responsivity in BTBR T(+) Itpr3(tf)/J mice (BTBR), an inbred mouse line widely used to model ASD-like symptoms owing to its robust social and communication deficits, and high level of repetitive stereotyped behaviours. C57BL/6J (B6) mice were used as normosocial reference comparators. DA reuptake inhibition with GBR 12909 produced significant striatal DA release in both strains, but failed to elicit fMRI activation in widespread forebrain areas of BTBR mice, including mesolimbic reward and striatal terminals. In addition, BTBR mice exhibited no appreciable motor responses to GBR 12909. DA D1 receptor-dependent behavioural and signalling responses were found to be unaltered in BTBR mice, whereas dramatic reductions in pre- and postsynaptic DA D2 and adenosine A2A receptor function was observed in these animals. Overall these results document profoundly compromised DA D2-mediated neurotransmission in BTBR mice, a finding that is likely to have a role in the distinctive social and behavioural deficits exhibited by these mice. Our results call for a deeper investigation of the role of dopaminergic dysfunction in mouse lines exhibiting ASD-like phenotypes, and possibly in ASD patient populations.
Collapse
Affiliation(s)
- M Squillace
- Ceinge Biotecnologie Avanzate, Naples, Italy
| | - L Dodero
- Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy,Istituto Italiano di Tecnologia, Pavis, Genoa, Italy
| | - M Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy,Laboratorio di Neurologia Sperimentale, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - S Migliarini
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - F Errico
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - F Napolitano
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - P Krashia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Di Maio
- Ceinge Biotecnologie Avanzate, Naples, Italy
| | - A Galbusera
- Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - A Bifone
- Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - M L Scattoni
- Neurotoxicology and Neuroendocrinology Section, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - M Pasqualetti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - N B Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy,Laboratorio di Neurologia Sperimentale, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - A Usiello
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples (SUN), Caserta, Italy, Dr , Ceinge Biotecnologie Avanzate, Naples, Italy E-mail:
| | - A Gozzi
- Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy,Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Sciences@uniTn, Corso Bettini 31, 38068 Rovereto, Italy. E-mail:
| |
Collapse
|