1
|
Xue R, Liu K, Hu X, Ma X, Li S, Deng Z, Zhong K, Yang J, Zhu X, Liu S, Shi Z, Zhou M, Tang Y. Longitudinal observation of radiation-induced cognitive impairment and emotional dysfunction: Based on animal model and clinical cohort. Neuroscience 2025; 572:171-181. [PMID: 39921023 DOI: 10.1016/j.neuroscience.2025.02.010] [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: 10/24/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Radiation-induced cognitive impairment (RICI) and emotional disorder (RIED) are comorbidities that seriously affect the quality of life in patients with radiation-induced brain injury (RIBI). A longitudinal study was conducted to observe the occurrence and development of RICI and RIED in an RIBI animal model and a clinical cohort following intensity modulated radiotherapy (IMRT). RIBI mice and sham controls were subjected to three cycles of behavior tests for cognitive and emotional function at post-irradiation 1 week (1w), 5 weeks (5w), and 9 weeks (9w) corresponding to early, middle and late stage after radiotherapy. Additionally, 139 patients who underwent IMRT after nasopharyngeal carcinoma and were firstly diagnosed with radiation-induced brain injury were enrolled. Pre-treatment and follow-up neuropsychological assessments of cognition, anxiety and depression were completed. Compared with control, significant declines in working memory, object recognition memory and social memory were observed in RIBI mice at post-irradiation 5w and 9w. Longitudinal observations revealed that memory impairment predominantly occurred in the middle stage and persisted into the late stage. Anxiety-like behaviors were only observed at post-irradiation 9w. In the clinical cohort, RICI exhibited a parallel cumulative incidence curve and a similar median onset to RIBI. RICI predominantly occurred 2-6 years post-IMRT 2-6 and progressively deteriorated beyond 6 years while RIED gradually increased beyond 6 years after IMRT. During two-year follow-up visits, half of the patients with RICI combined with RIBI benefited from drug treatment, achieving stable or improved cognition, while the other half showed no response or experienced cognition aggravation. In summary, RICI predominantly occurred in the middle stage post-irradiation and progressed to the late stage while RIED mostly emerged in the late stage in RIBI. Consistency in the development process of RICI and RIED was observed in the animal model and the clinical cohort.
Collapse
Affiliation(s)
- Ruiqi Xue
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kejia Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xia Hu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xueying Ma
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shaojian Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhenhong Deng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ke Zhong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingwen Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoqiu Zhu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Brain Research Center, Department of Neurology, Vice President, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
2
|
Ruiz-Sobremazas D, Coca M, Morales-Navas M, Rodulfo-Cardenas R, Lopez-Granero C, Colomina MT, Perez-Fernandez C, Sanchez-Santed F. The effects of oral gestational particulate matter 10 exposure: Insights into neurodevelopmental milestones, inhibitory control, adult sociability, and object recognition. Neurotoxicology 2025; 108:231-245. [PMID: 40252736 DOI: 10.1016/j.neuro.2025.04.007] [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: 01/17/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Air pollutants have been associated with various neurodevelopmental disorders, with several studies specifically linking Particulate Matter (PM) exposure to attentional and social deficits. This link is even more pronounced when exposure occurs during the prenatal period, as it can disrupt normal brain development. However, while social deficits have been extensively studied during adolescence, their impact on adult social behaviors remains largely unexplored. To investigate these effects, pregnant Wistar rats were exposed throughout gestation (GD1-GD21) to PM10 at a dosage of 200 μg/Kg/day diluted in PBS that was freely drunk. After birth, the pups were evaluated on developmental milestones such as weight progression, ocular opening, and muscular strength. In adulthood, inhibitory control was assessed using the Five Choice Serial Reaction Time Task (5-CSRTT), social behavior using the Three-Chambered Crawley's Test (3-CT), and object recognition using the Novelty Object Recognition test (NOR). The results indicated that prenatal PM10 exposure is associated with higher birth weight and poorer performance in neuromuscular tests. However, no significant differences were observed in inhibitory control (5-CSRTT) or social behavior (3-CT). Interestingly, prenatally exposed rodents showed heightened novelty responses in the NOR test. In conclusion, gestational exposure to PM10 is related to differences in neurodevelopmental milestones, including weight and muscular strength. While it does not impact adult inhibitory control or social behavior, it influences novelty recognition in later life.
Collapse
Affiliation(s)
- Diego Ruiz-Sobremazas
- Department of Psychology and Research Center for Well-Being and Social Inclusion (CIBIS), University of Almeria, Almeria 04120, Spain; Department of Psychology and Sociology, University of Zaragoza, Teruel, Aragón 44003, Spain
| | - Mario Coca
- Department of Psychology and Research Center for Well-Being and Social Inclusion (CIBIS), University of Almeria, Almeria 04120, Spain
| | - Miguel Morales-Navas
- Department of Health Sciences, Universidad de Burgos, Paseo de los Comendadores, Burgos 09001, Spain
| | - Rocío Rodulfo-Cardenas
- Universitat Rovira I Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Cataluña, Spain; Universitat Rovira I Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Cataluña, Spain; Universitat Rovira I Virgili, Center of Environmental, Food and Toxicological Technology (TECNATOX), Reus, Cataluña, Spain
| | - Caridad Lopez-Granero
- Department of Psychology and Sociology, University of Zaragoza, Teruel, Aragón 44003, Spain
| | - Maria-Teresa Colomina
- Universitat Rovira I Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Cataluña, Spain; Universitat Rovira I Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Cataluña, Spain; Universitat Rovira I Virgili, Center of Environmental, Food and Toxicological Technology (TECNATOX), Reus, Cataluña, Spain
| | - Cristian Perez-Fernandez
- Department of Health Sciences, Universidad de Burgos, Paseo de los Comendadores, Burgos 09001, Spain
| | - Fernando Sanchez-Santed
- Department of Psychology and Research Center for Well-Being and Social Inclusion (CIBIS), University of Almeria, Almeria 04120, Spain.
| |
Collapse
|
3
|
Frye CA, Cleveland DM, Sadarangani A, Torgersen JK. Progesterone Promotes Anti-Anxiety/Depressant-like Behavior and Trophic Actions of BDNF in the Hippocampus of Female Nuclear Progesterone Receptor, but Not 5α-Reductase, Knockout Mice. Int J Mol Sci 2025; 26:1173. [PMID: 39940941 PMCID: PMC11818940 DOI: 10.3390/ijms26031173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Progestogens' anti-anxiety and anti-depressive effects and mechanisms are not well-understood. Progestogens are hypothesized to have anti-anxiety and anti-depressive effects on behavior, independent of actions at nuclear progestin receptors (NPRs) and dependent on allopregnanolone (5α-pregnan-3α-ol-20-one; 3α,5α-THP), a 5α-reduced, neuroactive metabolite of progesterone (P4). Adult c57 mice in behavioral estrus (proestrus; pro) showed more anti-anxiety-like and anti-depressant-like behavior and higher levels of estradiol (E2), P4, and allopregnanolone in the hippocampus/amygdala complex. Proestrus c57 > 5α-reductase knockout (5αRKO) mice made more central entries in an open field than diestrus c57 and 5αRKO mice that were not different. Ovariectomized (OVX) c57 mice administered 1, 2, or 4 mg/kg P4 SC showed dosage-dependent increases in central entries in an open field (more anti-anxiety-like behavior); 5αRKO mice had maximal increases at 1-2 mg/kg P4. OVX c57 and 5αRKO mice showed maximum increases in central entries with SC 3α,5α-THP (4 mg/kg), and c57s showed a similar maximal response to P4 (4 mg/kg), but 5αRKOs response was half at that dosage. P4 (4 mg/kg SC to OVX c57 or progestin receptor knockout (PRKO) mice decreased immobility (depression-like behavior) in the forced swim task. Effects of E2 and veh were similar in both groups. Levels of 3α,5α-THP in the hippocampus/amygdala were consistent with effects on central entries in the open field. Levels of brain-derived neurotrophic factor (BDNF) in the hippocampus/amygdala were greater among E2-primed (0.09 mg/kg, SC) vs vehicle-administered mice. In sum, adult female mice can be responsive to P4 for anti-anxiety/anti-depressant-like behavior; such effects may be independent of NPRs but require 5α-reduction and E2's priming actions at BDNF in the hippocampus/amygdala complex.
Collapse
Affiliation(s)
- Cheryl A. Frye
- Comprehensive Neuropsychological Services, 490 Western Avenue, Albany, NY 12203, USA; (D.M.C.); (A.S.); (J.K.T.)
- Department of Psychology, The University at Albany, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Daina M. Cleveland
- Comprehensive Neuropsychological Services, 490 Western Avenue, Albany, NY 12203, USA; (D.M.C.); (A.S.); (J.K.T.)
| | - Anjali Sadarangani
- Comprehensive Neuropsychological Services, 490 Western Avenue, Albany, NY 12203, USA; (D.M.C.); (A.S.); (J.K.T.)
| | - Jennifer K. Torgersen
- Comprehensive Neuropsychological Services, 490 Western Avenue, Albany, NY 12203, USA; (D.M.C.); (A.S.); (J.K.T.)
- Department of Psychology, The University at Albany, 1400 Washington Avenue, Albany, NY 12222, USA
| |
Collapse
|
4
|
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
|
5
|
Wahl L, Karim A, Hassett AR, van der Doe M, Dijkhuizen S, Badura A. Multiparametric Assays Capture Sex- and Environment-Dependent Modifiers of Behavioral Phenotypes in Autism Mouse Models. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100366. [PMID: 39262819 PMCID: PMC11387692 DOI: 10.1016/j.bpsgos.2024.100366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 09/13/2024] Open
Abstract
Background Current phenotyping approaches for murine autism models often focus on one selected behavioral feature, making the translation onto a spectrum of autistic characteristics in humans challenging. Furthermore, sex and environmental factors are rarely considered. Here, we aimed to capture the full spectrum of behavioral manifestations in 3 autism mouse models to develop a "behavioral fingerprint" that takes environmental and sex influences under consideration. Methods To this end, we employed a wide range of classical standardized behavioral tests and 2 multiparametric behavioral assays-the Live Mouse Tracker and Motion Sequencing-on male and female Shank2, Tsc1, and Purkinje cell-specific Tsc1 mutant mice raised in standard or enriched environments. Our aim was to integrate our high dimensional data into one single platform to classify differences in all experimental groups along dimensions with maximum discriminative power. Results Multiparametric behavioral assays enabled a more accurate classification of experimental groups than classical tests, and dimensionality reduction analysis demonstrated significant additional gains in classification accuracy, highlighting the presence of sex, environmental, and genotype differences in our experimental groups. Conclusions Together, our results provide a complete phenotypic description of all tested groups, suggesting that multiparametric assays can capture the entire spectrum of the heterogeneous phenotype in autism mouse models.
Collapse
Affiliation(s)
- Lucas Wahl
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Arun Karim
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Amy R Hassett
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Max van der Doe
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | | |
Collapse
|
6
|
Martínez-Drudis L, Bérard M, Musiol D, Rivest S, Oueslati A. Pharmacological inhibition of PLK2 kinase activity mitigates cognitive decline but aggravates APP pathology in a sex-dependent manner in APP/PS1 mouse model of Alzheimer's disease. Heliyon 2024; 10:e39571. [PMID: 39498012 PMCID: PMC11532864 DOI: 10.1016/j.heliyon.2024.e39571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Converging evidence from clinical and experimental studies suggest the potential significance of Polo-like kinase 2 (PLK2) in regulating the phosphorylation and toxicity of the Alzheimer's disease (AD)-related protein, amyloid precursor protein (APP). These findings have prompted various experimental trials aimed at inhibiting PLK2 kinase activity in different transgenic mouse models of AD. While positive impacts on cognitive decline were reported in these studies, the cellular effects remained controversial. In the present study, we sought to assess the cognitive and cellular consequences of chronic PLK2 inhibitor treatment in the APP/PS1 transgenic mouse model of AD. First, we confirmed that inhibiting PLK2 prevented cognitive decline in a sex-dependent manner, particularly by enhancing working memory in male APP/PS1 mice. Surprisingly, cellular analysis revealed that treatment with PLK2 inhibitor increased the load of amyloid plaques and elevated levels of soluble amyloid β (Aβ) 40 and Aβ42 in the cortex, as well as insoluble Aβ42 in the hippocampus of female mice, without affecting APP pathology in males. These results underscore the potential of PLK2 inhibition to mitigate cognitive symptoms in males. However, paradoxically, it intensifies amyloid pathology in females by enhancing APP amyloidogenic processing, creating a controversial aspect to its therapeutic impact. Overall, these data highlight the sex-dependent nature of the effects of PLK2 inhibition, which may also be influenced by the genetic background of the transgenic mouse model utilized.
Collapse
Affiliation(s)
- Laura Martínez-Drudis
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Morgan Bérard
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Dylan Musiol
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Serge Rivest
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Abid Oueslati
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
7
|
Ornoy A, Echefu B, Becker M. Animal Models of Autistic-like Behavior in Rodents: A Scoping Review and Call for a Comprehensive Scoring System. Int J Mol Sci 2024; 25:10469. [PMID: 39408797 PMCID: PMC11477392 DOI: 10.3390/ijms251910469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Appropriate animal models of human diseases are a cornerstone in the advancement of science and medicine. To create animal models of neuropsychiatric and neurobehavioral diseases such as autism spectrum disorder (ASD) necessitates the development of sufficient neurobehavioral measuring tools to translate human behavior to expected measurable behavioral features in animals. If possible, the severity of the symptoms should also be assessed. Indeed, at least in rodents, adequate neurobehavioral and neurological tests have been developed. Since ASD is characterized by a number of specific behavioral trends with significant severity, animal models of autistic-like behavior have to demonstrate the specific characteristic features, namely impaired social interactions, communication deficits, and restricted, repetitive behavioral patterns, with association to several additional impairments such as somatosensory, motor, and memory impairments. Thus, an appropriate model must show behavioral impairment of a minimal number of neurobehavioral characteristics using an adequate number of behavioral tests. The proper animal models enable the study of ASD-like-behavior from the etiologic, pathogenetic, and therapeutic aspects. From the etiologic aspects, models have been developed by the use of immunogenic substances like polyinosinic-polycytidylic acid (PolyIC), lipopolysaccharide (LPS), and propionic acid, or other well-documented immunogens or pathogens, like Mycobacterium tuberculosis. Another approach is the use of chemicals like valproic acid, polychlorinated biphenyls (PCBs), organophosphate pesticides like chlorpyrifos (CPF), and others. These substances were administered either prenatally, generally after the period of major organogenesis, or, especially in rodents, during early postnatal life. In addition, using modern genetic manipulation methods, genetic models have been created of almost all human genetic diseases that are manifested by autistic-like behavior (i.e., fragile X, Rett syndrome, SHANK gene mutation, neuroligin genes, and others). Ideally, we should not only evaluate the different behavioral modes affected by the ASD-like behavior, but also assess the severity of the behavioral deviations by an appropriate scoring system, as applied to humans. We therefore propose a scoring system for improved assessment of ASD-like behavior in animal models.
Collapse
Affiliation(s)
- Asher Ornoy
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
- Hadassah Academic College, Jerusalem 9101001, Israel
- Hadassah Medical School, Hebrew University, Jerusalem 9112102, Israel
| | - Boniface Echefu
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| | - Maria Becker
- Department of Morphological Sciences and Teratology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (B.E.); (M.B.)
| |
Collapse
|
8
|
Zilkha N, Chuartzman SG, Fishman R, Ben-Dor S, Kimchi T. Maternal high-fat or low-protein diets promote autism-related behavior and altered social behavior within groups in offspring male mice. Sci Rep 2024; 14:19227. [PMID: 39164365 PMCID: PMC11336096 DOI: 10.1038/s41598-024-70062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
Maternal malnutrition has been associated with neurodevelopmental deficits and long-term implications on the offspring's health and behavior. Here, we investigated the effects of maternal low-protein diet (LPD) or obesity-inducing maternal high-fat diet (HFD) on dyadic social interactions, group organization and autism-related behaviors in mice. We found that maternal HFD induced an autism-related behavioral phenotype in the male offspring, including a robust decrease in sociability, increased aggression, cognitive rigidity and repetitive behaviors. Maternal LPD led to a milder yet significant effect on autism-related symptoms, with no effects on olfactory-mediated social behavior. Under naturalistic conditions in a group setting, this manifested in altered behavioral repertoires, increased magnitude in dominance relations, and reduced interactions with novel social stimuli in the HFD male offspring, but not in the LPD offspring. Finally, we found HFD-induced transcriptomic changes in the olfactory bulbs of the male offspring. Together, our findings show that maternal malnutrition induces long-lasting effects on aggression and autism-related behaviors in male offspring, and potential impairments in brain regions processing chemosensory signals.
Collapse
Affiliation(s)
- Noga Zilkha
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | | | - Ruth Fishman
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Shifra Ben-Dor
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Tali Kimchi
- Department of Brain Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
9
|
Varga A, Kedves R, Sághy K, Garab D, Zádor F, Lendvai B, Lévay G, Román V. R-Baclofen Treatment Corrects Autistic-like Behavioral Deficits in the RjIbm(m):FH Fawn-Hooded Rat Strain. Pharmaceuticals (Basel) 2024; 17:939. [PMID: 39065788 PMCID: PMC11279403 DOI: 10.3390/ph17070939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
The Fawn-hooded rat has long been used as a model for various peripheral and central disorders and the data available indicate that the social behavior of this strain may be compromised. However, a thorough description of the Fawn-hooded rat is unavailable in this regard. The objective of the present study was to investigate various aspects of the Fawn-hooded rat's social behavior in depth. Our results show that several facets of socio-communicational behavior are impaired in the RjIbm(m):FH strain, including defective ultrasonic vocalizations in pups upon maternal deprivation, reduced social play in adolescence and impaired social novelty discrimination in adulthood. In addition, Fawn-hooded rats exhibited heightened tactile sensitivity and hyperactivity. The defects observed were comparable to those induced by prenatal valproate exposure, a widely utilized model of autism spectrum disorder. Further on, the pro-social drug R-baclofen (0.25-1 mg/kg) reversed the autistic-like defects observed in Fawn-hooded rats, specifically the deficiency in ultrasonic vocalization, tactile sensitivity and social novelty discrimination endpoints. In conclusion, the asocial, hypersensitive and hyperactive phenotype as well as the responsivity to R-baclofen indicate this variant of the Fawn-hooded rat strain may serve as a model of autism spectrum disorder and could be useful in the identification of novel drug candidates.
Collapse
Affiliation(s)
- Anita Varga
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
- Doctoral School of Biology and Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/A, 1117 Budapest, Hungary
| | - Rita Kedves
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
| | - Katalin Sághy
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
| | - Dénes Garab
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
| | - Ferenc Zádor
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
| | - Balázs Lendvai
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
- Richter Department, Semmelweis University, Gyömrői út 19-21, 1103 Budapest, Hungary
| | - György Lévay
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Vas utca 17, 1088 Budapest, Hungary
| | - Viktor Román
- Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, 1103 Budapest, Hungary
- Richter Department, Semmelweis University, Gyömrői út 19-21, 1103 Budapest, Hungary
| |
Collapse
|
10
|
Arake M, Ohta H, Nozawa T, Satoh Y, Fujita M, Nakata T, Meredith AL, Shinomiya N, Ishizuka T, Morimoto Y. BK channel dysfunction disrupts attention-controlled behaviors and altered perseverative responses in murine instrumental learning. Behav Brain Res 2024; 468:115015. [PMID: 38670533 DOI: 10.1016/j.bbr.2024.115015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
This study examined the effect of knockout of KCNMA1 gene, coding for the BK channel, on cognitive and attentional functions in mice, with an aim to better understand its implications for human neurodevelopmental disorders. The study used the 3-choice serial reaction time task (3-CSRTT) to assess the learning performance, attentional abilities, and repetitive behaviors in mice lacking the KCNMA1 gene (KCNMA1-/-) compared to wild-type (WT) controls. Results showed no significant differences in learning accuracy between the two groups. However, KCNMA1-/- mice were more prone to omitting responses to stimuli. In addition, when the timing of cue presentation was randomized, the KCNMA1-/- showed premature responses. Notably, these mice also demonstrated a marked reduction in perseverative responses, which include repeated nose-poke behaviors following decisions. These findings highlight the involvement of the KCNMA1 gene in managing attention, impulsivity, and potentially moderating repetitive actions.
Collapse
Affiliation(s)
- Masashi Arake
- Department of Physiology, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama 359-8513, Japan
| | - Hiroyuki Ohta
- Department of Pharmacology, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama 359-8513, Japan.
| | - Takashi Nozawa
- Department of Psychology, Mejiro University, Nakaochiai 4-31-1, Shinjuku-ku, Tokyo 161-8539, Japan
| | - Yasushi Satoh
- Department of Biochemistry, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama 359-8513, Japan
| | - Masanori Fujita
- Division of Environmental Medicine, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama 359-8513, Japan
| | - Takahiro Nakata
- Department of Molecular and Cellular Anatomy, Faculty of Health Promotional Sciences, Tokoha University, Hamamatsu, Shizuoka, Japan
| | - Andrea L Meredith
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama 359-8513, Japan
| | - Toshiaki Ishizuka
- Department of Pharmacology, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama 359-8513, Japan
| | - Yuji Morimoto
- Department of Physiology, National Defense Medical College, Namiki 3-2, Tokorozawa, Saitama 359-8513, Japan
| |
Collapse
|
11
|
Camilleri MPJ, Bains RS, Williams CKI. Of Mice and Mates: Automated Classification and Modelling of Mouse Behaviour in Groups Using a Single Model Across Cages. Int J Comput Vis 2024; 132:5491-5513. [PMID: 39554493 PMCID: PMC11568001 DOI: 10.1007/s11263-024-02118-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 05/07/2024] [Indexed: 11/19/2024]
Abstract
Behavioural experiments often happen in specialised arenas, but this may confound the analysis. To address this issue, we provide tools to study mice in the home-cage environment, equipping biologists with the possibility to capture the temporal aspect of the individual's behaviour and model the interaction and interdependence between cage-mates with minimal human intervention. Our main contribution is the novel Global Behaviour Model (GBM) which summarises the joint behaviour of groups of mice across cages, using a permutation matrix to match the mouse identities in each cage to the model. In support of the above, we also (a) developed the Activity Labelling Module (ALM) to automatically classify mouse behaviour from video, and (b) released two datasets, ABODe for training behaviour classifiers and IMADGE for modelling behaviour. Supplementary Information The online version contains supplementary material available at 10.1007/s11263-024-02118-3.
Collapse
|
12
|
Li X, Yennawar M, Wiest A, O'Brien WT, Babrowicz B, White RS, Talos DM, Jensen FE. Cannabidiol attenuates seizure susceptibility and behavioural deficits in adult CDKL5 R59X knock-in mice. Eur J Neurosci 2024; 59:3337-3352. [PMID: 38654472 DOI: 10.1111/ejn.16350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 02/15/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is caused by a loss-of-function mutation in CDKL5 gene, encoding a serine-threonine kinase highly expressed in the brain. CDD manifests with early-onset epilepsy, autism, motor impairment and severe intellectual disability. While there are no known treatments for CDD, the use of cannabidiol has recently been introduced into clinical practice for neurodevelopmental disorders. Given the increased clinical utilization of cannabidiol, we examined its efficacy in the CDKL5R59X knock-in (R59X) mice, a CDD model based on a human mutation that exhibits both lifelong seizure susceptibility and behavioural deficits. We found that cannabidiol pre-treatment rescued the increased seizure susceptibility in response to the chemoconvulsant pentylenetetrazol (PTZ), attenuated working memory and long-term memory impairments, and rescued social deficits in adult R59X mice. To elucidate a potential mechanism, we compared the developmental hippocampal and cortical expression of common endocannabinoid (eCB) targets in R59X mice and their wild-type littermates, including cannabinoid type 1 receptor (CB1R), transient receptor potential vanilloid type 1 (TRPV1) and 2 (TRPV2), G-coupled protein receptor 55 (GPR55) and adenosine receptor 1 (A1R). Many of these eCB targets were developmentally regulated in both R59X and wild-type mice. In addition, adult R59X mice demonstrated significantly decreased expression of CB1R and TRPV1 in the hippocampus, and TRPV2 in the cortex, while TRPV1 was increased in the cortex. These findings support the potential for dysregulation of eCB signalling as a plausible mechanism and therapeutic target in CDD, given the efficacy of cannabidiol to attenuate hyperexcitability and behavioural deficits in this disorder.
Collapse
Affiliation(s)
- Xiaofan Li
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Madhumita Yennawar
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alyssa Wiest
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William T O'Brien
- Neurobehavior Testing Core, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bergan Babrowicz
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rachel S White
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Delia M Talos
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Frances E Jensen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Chen C, Altafi M, Corbu MA, Trenk A, van den Munkhof H, Weineck K, Bender F, Carus-Cadavieco M, Bakhareva A, Korotkova T, Ponomarenko A. The dynamic state of a prefrontal-hypothalamic-midbrain circuit commands behavioral transitions. Nat Neurosci 2024; 27:952-963. [PMID: 38499854 PMCID: PMC11089001 DOI: 10.1038/s41593-024-01598-3] [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: 08/26/2022] [Accepted: 02/12/2024] [Indexed: 03/20/2024]
Abstract
Innate behaviors meet multiple needs adaptively and in a serial order, suggesting the existence of a hitherto elusive brain dynamics that brings together representations of upcoming behaviors during their selection. Here we show that during behavioral transitions, possible upcoming behaviors are encoded by specific signatures of neuronal populations in the lateral hypothalamus (LH) that are active near beta oscillation peaks. Optogenetic recruitment of intrahypothalamic inhibition at this phase eliminates behavioral transitions. We show that transitions are elicited by beta-rhythmic inputs from the prefrontal cortex that spontaneously synchronize with LH 'transition cells' encoding multiple behaviors. Downstream of the LH, dopamine neurons increase firing during beta oscillations and also encode behavioral transitions. Thus, a hypothalamic transition state signals alternative future behaviors, encodes the one most likely to be selected and enables rapid coordination with cognitive and reward-processing circuitries, commanding adaptive social contact and eating behaviors.
Collapse
Affiliation(s)
- Changwan Chen
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne/University Clinic Cologne, Cologne, Germany
| | - Mahsa Altafi
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mihaela-Anca Corbu
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne/University Clinic Cologne, Cologne, Germany
| | - Aleksandra Trenk
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Hanna van den Munkhof
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne/University Clinic Cologne, Cologne, Germany
| | - Kristin Weineck
- Behavioural Neurodynamics Group, Leibniz Institute for Molecular Pharmacology (FMP)/NeuroCure Cluster of Excellence, Berlin, Germany
| | - Franziska Bender
- Behavioural Neurodynamics Group, Leibniz Institute for Molecular Pharmacology (FMP)/NeuroCure Cluster of Excellence, Berlin, Germany
| | - Marta Carus-Cadavieco
- Max Planck Institute for Metabolism Research, Cologne, Germany
- Behavioural Neurodynamics Group, Leibniz Institute for Molecular Pharmacology (FMP)/NeuroCure Cluster of Excellence, Berlin, Germany
| | - Alisa Bakhareva
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne/University Clinic Cologne, Cologne, Germany
| | - Tatiana Korotkova
- Max Planck Institute for Metabolism Research, Cologne, Germany.
- Institute for Systems Physiology, Faculty of Medicine, University of Cologne/University Clinic Cologne, Cologne, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| | - Alexey Ponomarenko
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
- Behavioural Neurodynamics Group, Leibniz Institute for Molecular Pharmacology (FMP)/NeuroCure Cluster of Excellence, Berlin, Germany.
| |
Collapse
|
14
|
Matsumoto Y, Miwa H, Katayama KI, Watanabe A, Yamada K, Ito T, Nakagawa S, Aruga J. Slitrk4 is required for the development of inhibitory neurons in the fear memory circuit of the lateral amygdala. Front Mol Neurosci 2024; 17:1386924. [PMID: 38736483 PMCID: PMC11082273 DOI: 10.3389/fnmol.2024.1386924] [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: 02/16/2024] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
The Slitrk family consists of six synaptic adhesion molecules, some of which are associated with neuropsychiatric disorders. In this study, we aimed to investigate the physiological role of Slitrk4 by analyzing Slitrk4 knockout (KO) mice. The Slitrk4 protein was widely detected in the brain and was abundant in the olfactory bulb and amygdala. In a systematic behavioral analysis, male Slitrk4 KO mice exhibited an enhanced fear memory acquisition in a cued test for classical fear conditioning, and social behavior deficits in reciprocal social interaction tests. In an electrophysiological analysis using amygdala slices, Slitrk4 KO mice showed enhanced long-term potentiation in the thalamo-amygdala afferents and reduced feedback inhibition. In the molecular marker analysis of Slitrk4 KO brains, the number of calretinin (CR)-positive interneurons was decreased in the anterior part of the lateral amygdala nuclei at the adult stage. In in vitro experiments for neuronal differentiation, Slitrk4-deficient embryonic stem cells were defective in inducing GABAergic interneurons with an altered response to sonic hedgehog signaling activation that was involved in the generation of GABAergic interneuron subsets. These results indicate that Slitrk4 function is related to the development of inhibitory neurons in the fear memory circuit and would contribute to a better understanding of osttraumatic stress disorder, in which an altered expression of Slitrk4 has been reported.
Collapse
Affiliation(s)
- Yoshifumi Matsumoto
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute, Wako-shi, Japan
| | - Hideki Miwa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kei-ichi Katayama
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute, Wako-shi, Japan
| | - Arata Watanabe
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Kazuyuki Yamada
- Support Unit for Animal Experiments, RIKEN Brain Science Institute, Wako-shi, Japan
| | - Takashi Ito
- Department of Biochemistry, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Shinsuke Nakagawa
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Jun Aruga
- Laboratory for Behavioral and Developmental Disorders, RIKEN Brain Science Institute, Wako-shi, Japan
- Department of Medical Pharmacology, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
15
|
Gencturk S, Unal G. Rodent tests of depression and anxiety: Construct validity and translational relevance. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2024; 24:191-224. [PMID: 38413466 PMCID: PMC11039509 DOI: 10.3758/s13415-024-01171-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/03/2024] [Indexed: 02/29/2024]
Abstract
Behavioral testing constitutes the primary method to measure the emotional states of nonhuman animals in preclinical research. Emerging as the characteristic tool of the behaviorist school of psychology, behavioral testing of animals, particularly rodents, is employed to understand the complex cognitive and affective symptoms of neuropsychiatric disorders. Following the symptom-based diagnosis model of the DSM, rodent models and tests of depression and anxiety focus on behavioral patterns that resemble the superficial symptoms of these disorders. While these practices provided researchers with a platform to screen novel antidepressant and anxiolytic drug candidates, their construct validity-involving relevant underlying mechanisms-has been questioned. In this review, we present the laboratory procedures used to assess depressive- and anxiety-like behaviors in rats and mice. These include constructs that rely on stress-triggered responses, such as behavioral despair, and those that emerge with nonaversive training, such as cognitive bias. We describe the specific behavioral tests that are used to assess these constructs and discuss the criticisms on their theoretical background. We review specific concerns about the construct validity and translational relevance of individual behavioral tests, outline the limitations of the traditional, symptom-based interpretation, and introduce novel, ethologically relevant frameworks that emphasize simple behavioral patterns. Finally, we explore behavioral monitoring and morphological analysis methods that can be integrated into behavioral testing and discuss how they can enhance the construct validity of these tests.
Collapse
Affiliation(s)
- Sinem Gencturk
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey
| | - Gunes Unal
- Behavioral Neuroscience Laboratory, Department of Psychology, Boğaziçi University, 34342, Istanbul, Turkey.
| |
Collapse
|
16
|
Vorhees CV, Williams MT. Tests for learning and memory in rodent regulatory studies. Curr Res Toxicol 2024; 6:100151. [PMID: 38304257 PMCID: PMC10832385 DOI: 10.1016/j.crtox.2024.100151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
For decades, regulatory guidelines for safety assessment in rodents for drugs, chemicals, pesticides, and food additives with developmental neurotoxic potential have recommended a single test of learning and memory (L&M). In recent years some agencies have requested two such tests. Given the importance of higher cognitive function to health, and the fact that different types of L&M are mediated by different brain regions assessing higher functions represents a step forward in providing better evidence-based protection against adverse brain effects. Given the myriad of tests available for assessing L&M in rodents this leads to the question of which tests best fit regulatory guidelines. To address this question, we begin by describing the central role of two types of L&M essential to all mammalian species and the regions/networks that mediate them. We suggest that the tests recommended possess characteristics that make them well suited to the needs in regulatory safety studies. By brain region, these are (1) the hippocampus and entorhinal cortex for spatial navigation, which assesses explicit L&M for reference and episodic memory and (2) the striatum and related structures for egocentric navigation, which assesses implicit or procedural memory and path integration. Of the tests available, we suggest that in this context, the evidence supports the use of water mazes, specifically, the Morris water maze (MWM) for spatial L&M and the Cincinnati water maze (CWM) for egocentric/procedural L&M. We review the evidentiary basis for these tests, describe their use, and explain procedures that optimize their sensitivity.
Collapse
Affiliation(s)
- Charles V. Vorhees
- Corresponding author at: Div. of Neurology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA.
| | | |
Collapse
|
17
|
Ross MM, Hernandez-Espinosa DR, Aizenman E. Neurodevelopmental Consequences of Dietary Zinc Deficiency: A Status Report. Biol Trace Elem Res 2023; 201:5616-5639. [PMID: 36964812 DOI: 10.1007/s12011-023-03630-2] [Citation(s) in RCA: 5] [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: 01/25/2023] [Accepted: 03/11/2023] [Indexed: 03/26/2023]
Abstract
Zinc is a tightly regulated trace mineral element playing critical roles in growth, immunity, neurodevelopment, and synaptic and hormonal signaling. Although severe dietary zinc deficiency is relatively uncommon in the United States, dietary zinc deficiency is a substantial public health concern in low- and middle-income countries. Zinc status may be a key determinant of neurodevelopmental processes. Indeed, limited cohort studies have shown that serum zinc is lower in people diagnosed with autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), and depression. These observations have sparked multiple studies investigating the mechanisms underlying zinc status and neurodevelopmental outcomes. Animal models of perinatal and adult dietary zinc restriction yield distinct behavioral phenotypes reminiscent of features of ASD, ADHD, and depression, including increased anxiety and immobility, repetitive behaviors, and altered social behaviors. At the cellular and molecular level, zinc has demonstrated roles in neurogenesis, regulation of cellular redox status, transcription factor trafficking, synaptogenesis, and the regulation of synaptic architecture via the Shank family of scaffolding proteins. Although mechanistic questions remain, the current evidence suggests that zinc status is important for adequate neuronal development and may be a yet overlooked factor in the pathogenesis of several psychiatric conditions. This review aims to summarize current knowledge of the role of zinc in the neurophysiology of the perinatal period, the many cellular targets of zinc in the developing brain, and the potential consequences of alterations in zinc homeostasis in early life.
Collapse
Affiliation(s)
- Madeline M Ross
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Diego R Hernandez-Espinosa
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
18
|
Mosconi MW, Stevens CJ, Unruh KE, Shafer R, Elison JT. Endophenotype trait domains for advancing gene discovery in autism spectrum disorder. J Neurodev Disord 2023; 15:41. [PMID: 37993779 PMCID: PMC10664534 DOI: 10.1186/s11689-023-09511-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/09/2023] [Indexed: 11/24/2023] Open
Abstract
Autism spectrum disorder (ASD) is associated with a diverse range of etiological processes, including both genetic and non-genetic causes. For a plurality of individuals with ASD, it is likely that the primary causes involve multiple common inherited variants that individually account for only small levels of variation in phenotypic outcomes. This genetic landscape creates a major challenge for detecting small but important pathogenic effects associated with ASD. To address similar challenges, separate fields of medicine have identified endophenotypes, or discrete, quantitative traits that reflect genetic likelihood for a particular clinical condition and leveraged the study of these traits to map polygenic mechanisms and advance more personalized therapeutic strategies for complex diseases. Endophenotypes represent a distinct class of biomarkers useful for understanding genetic contributions to psychiatric and developmental disorders because they are embedded within the causal chain between genotype and clinical phenotype, and they are more proximal to the action of the gene(s) than behavioral traits. Despite their demonstrated power for guiding new understanding of complex genetic structures of clinical conditions, few endophenotypes associated with ASD have been identified and integrated into family genetic studies. In this review, we argue that advancing knowledge of the complex pathogenic processes that contribute to ASD can be accelerated by refocusing attention toward identifying endophenotypic traits reflective of inherited mechanisms. This pivot requires renewed emphasis on study designs with measurement of familial co-variation including infant sibling studies, family trio and quad designs, and analysis of monozygotic and dizygotic twin concordance for select trait dimensions. We also emphasize that clarification of endophenotypic traits necessarily will involve integration of transdiagnostic approaches as candidate traits likely reflect liability for multiple clinical conditions and often are agnostic to diagnostic boundaries. Multiple candidate endophenotypes associated with ASD likelihood are described, and we propose a new focus on the analysis of "endophenotype trait domains" (ETDs), or traits measured across multiple levels (e.g., molecular, cellular, neural system, neuropsychological) along the causal pathway from genes to behavior. To inform our central argument for research efforts toward ETD discovery, we first provide a brief review of the concept of endophenotypes and their application to psychiatry. Next, we highlight key criteria for determining the value of candidate endophenotypes, including unique considerations for the study of ASD. Descriptions of different study designs for assessing endophenotypes in ASD research then are offered, including analysis of how select patterns of results may help prioritize candidate traits in future research. We also present multiple candidate ETDs that collectively cover a breadth of clinical phenomena associated with ASD, including social, language/communication, cognitive control, and sensorimotor processes. These ETDs are described because they represent promising targets for gene discovery related to clinical autistic traits, and they serve as models for analysis of separate candidate domains that may inform understanding of inherited etiological processes associated with ASD as well as overlapping neurodevelopmental disorders.
Collapse
Affiliation(s)
- Matthew W Mosconi
- Schiefelbusch Institute for Life Span Studies and Kansas Center for Autism Research and Training (K-CART), University of Kansas, Lawrence, KS, USA.
- Clinical Child Psychology Program, University of Kansas, Lawrence, KS, USA.
| | - Cassandra J Stevens
- Schiefelbusch Institute for Life Span Studies and Kansas Center for Autism Research and Training (K-CART), University of Kansas, Lawrence, KS, USA
- Clinical Child Psychology Program, University of Kansas, Lawrence, KS, USA
| | - Kathryn E Unruh
- Schiefelbusch Institute for Life Span Studies and Kansas Center for Autism Research and Training (K-CART), University of Kansas, Lawrence, KS, USA
| | - Robin Shafer
- Schiefelbusch Institute for Life Span Studies and Kansas Center for Autism Research and Training (K-CART), University of Kansas, Lawrence, KS, USA
| | - Jed T Elison
- Institute of Child Development, University of Minnesota, Minneapolis, MN, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
19
|
Gal A, Raykin E, Giladi S, Lederman D, Kofman O, Golan HM. Temporal dynamics of isolation calls emitted by pups in environmental and genetic mouse models of autism spectrum disorder. Front Neurosci 2023; 17:1274039. [PMID: 37942134 PMCID: PMC10629105 DOI: 10.3389/fnins.2023.1274039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction Environmental and genetic factors contribute to the increased risk for neurodevelopmental disorders, including deficits in the development of social communication. In the mouse, ultrasonic vocalizations emitted by the pup stimulate maternal retrieval and potentiate maternal care. Therefore, isolation induced ultrasonic vocalization emitted by pups provides a means to evaluate deficits in communication during early development, before other ways of communication are apparent. Previous studies in our labs showed that gestational exposure to the pesticide chlorpyrifos (CPF) and the Methylenetetrahydrofolate (Mthfr)-knock-out mice are associated with impaired social preference, restricted or repetitive behavior and altered spectral properties of pups' ultrasonic vocalization. In this study, we explore the temporal dynamics of pups' vocalization in these Autism spectrum disorder (ASD) models. Methods We utilized the maternal potentiation protocol and analyzed the time course of pup vocalizations following isolation from the nest. Two models of ASD were studied: gestational exposure to the pesticide CPF and the Mthfr-knock-out mice. Results Vocalization emitted by pups of both ASD models were dynamically modified in quantity and spectral structure within each session and between the two isolation sessions. The first isolation session was characterized by a buildup of call quantity and significant effects of USV spectral structure variables, and the second isolation session was characterized by enhanced calls and vocalization time, but minute effect on USV properties. Moreover, in both models we described an increased usage of harmonic calls with time during the isolation sessions. Discussion Communication between two or more individuals requires an interplay between the two sides and depends on the response and the time since the stimulus. As such, the presence of dynamic changes in vocalization structure in the control pups, and the alteration observed in the pups of the ASD models, suggest impaired regulation of vocalization associated with the environmental and genetic factors. Last, we propose that temporal dynamics of ultrasonic vocalization communication should be considered in future analysis in rodent models of ASD to maximize the sensitivity of the study of vocalizations.
Collapse
Affiliation(s)
- Ayelet Gal
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Eynav Raykin
- Psychology Department, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Shaked Giladi
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Dror Lederman
- Faculty of Engineering, Holon Institute of Technology Holon, Holon, Israel
| | - Ora Kofman
- Psychology Department, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Hava M. Golan
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Center for Autism Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
20
|
László K, Vörös D, Correia P, Fazekas CL, Török B, Plangár I, Zelena D. Vasopressin as Possible Treatment Option in Autism Spectrum Disorder. Biomedicines 2023; 11:2603. [PMID: 37892977 PMCID: PMC10603886 DOI: 10.3390/biomedicines11102603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is rather common, presenting with prevalent early problems in social communication and accompanied by repetitive behavior. As vasopressin was implicated not only in salt-water homeostasis and stress-axis regulation, but also in social behavior, its role in the development of ASD might be suggested. In this review, we summarized a wide range of problems associated with ASD to which vasopressin might contribute, from social skills to communication, motor function problems, autonomous nervous system alterations as well as sleep disturbances, and altered sensory information processing. Beside functional connections between vasopressin and ASD, we draw attention to the anatomical background, highlighting several brain areas, including the paraventricular nucleus of the hypothalamus, medial preoptic area, lateral septum, bed nucleus of stria terminalis, amygdala, hippocampus, olfactory bulb and even the cerebellum, either producing vasopressin or containing vasopressinergic receptors (presumably V1a). Sex differences in the vasopressinergic system might underline the male prevalence of ASD. Moreover, vasopressin might contribute to the effectiveness of available off-label therapies as well as serve as a possible target for intervention. In this sense, vasopressin, but paradoxically also V1a receptor antagonist, were found to be effective in some clinical trials. We concluded that although vasopressin might be an effective candidate for ASD treatment, we might assume that only a subgroup (e.g., with stress-axis disturbances), a certain sex (most probably males) and a certain brain area (targeting by means of virus vectors) would benefit from this therapy.
Collapse
Affiliation(s)
- Kristóf László
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Dávid Vörös
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Pedro Correia
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Bibiána Török
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| | - Imola Plangár
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
| | - Dóra Zelena
- Institute of Physiology, Medical School, University of Pécs, 7624 Pecs, Hungary; (K.L.); (D.V.); (P.C.); (C.L.F.); (B.T.); (I.P.)
- Center of Neuroscience, University of Pécs, 7624 Pecs, Hungary
- Szentágothai Research Center, University of Pécs, 7624 Pecs, Hungary
- Hungarian Research Network, Institute of Experimental Medicine, 1083 Budapest, Hungary
| |
Collapse
|
21
|
Woo T, King C, Ahmed NI, Cordes M, Nistala S, Will MJ, Bloomer C, Kibiryeva N, Rivera RM, Talebizadeh Z, Beversdorf DQ. microRNA as a Maternal Marker for Prenatal Stress-Associated ASD, Evidence from a Murine Model. J Pers Med 2023; 13:1412. [PMID: 37763179 PMCID: PMC10533003 DOI: 10.3390/jpm13091412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Autism Spectrum Disorder (ASD) has been associated with a complex interplay between genetic and environmental factors. Prenatal stress exposure has been identified as a possible risk factor, although most stress-exposed pregnancies do not result in ASD. The serotonin transporter (SERT) gene has been linked to stress reactivity, and the presence of the SERT short (S)-allele has been shown to mediate the association between maternal stress exposure and ASD. In a mouse model, we investigated the effects of prenatal stress exposure and maternal SERT genotype on offspring behavior and explored its association with maternal microRNA (miRNA) expression during pregnancy. Pregnant female mice were divided into four groups based on genotype (wildtype or SERT heterozygous knockout (Sert-het)) and the presence or absence of chronic variable stress (CVS) during pregnancy. Offspring behavior was assessed at 60 days old (PD60) using the three-chamber test, open field test, elevated plus-maze test, and marble-burying test. We found that the social preference index (SPI) of SERT-het/stress offspring was significantly lower than that of wildtype control offspring, indicating a reduced preference for social interaction on social approach, specifically for males. SERT-het/stress offspring also showed significantly more frequent grooming behavior compared to wildtype controls, specifically for males, suggesting elevated repetitive behavior. We profiled miRNA expression in maternal blood samples collected at embryonic day 21 (E21) and identified three miRNAs (mmu-miR-7684-3p, mmu-miR-5622-3p, mmu-miR-6900-3p) that were differentially expressed in the SERT-het/stress group compared to all other groups. These findings suggest that maternal SERT genotype and prenatal stress exposure interact to influence offspring behavior, and that maternal miRNA expression late in pregnancy may serve as a potential marker of a particular subtype of ASD pathogenesis.
Collapse
Affiliation(s)
- Taeseon Woo
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO 65211, USA;
| | - Candice King
- Department of Biological Science, University of Missouri, Columbia, MO 65211, USA; (C.K.); (M.C.)
| | - Nick I. Ahmed
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA; (N.I.A.); (M.J.W.)
| | - Madison Cordes
- Department of Biological Science, University of Missouri, Columbia, MO 65211, USA; (C.K.); (M.C.)
| | | | - Matthew J. Will
- Department of Psychological Sciences, University of Missouri, Columbia, MO 65211, USA; (N.I.A.); (M.J.W.)
| | - Clark Bloomer
- Genomics Core, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Nataliya Kibiryeva
- College of Bioscience, Kansas City University, Kansas City, MO 64106, USA;
| | - Rocio M. Rivera
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA;
| | - Zohreh Talebizadeh
- American College of Medical Genetics and Genomics, Bethesda, MD 20814, USA;
| | - David Q. Beversdorf
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO 65211, USA;
- Department of Radiology, Neurology, and Psychological Science, William and Nancy Thompson Endowed Chair in Radiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
22
|
Leung BK, Merlin S, Walker AK, Lawther AJ, Paxinos G, Eapen V, Clarke R, Balleine BW, Furlong TM. Immp2l knockdown in male mice increases stimulus-driven instrumental behaviour but does not alter goal-directed learning or neuron density in cortico-striatal circuits in a model of Tourette syndrome and autism spectrum disorder. Behav Brain Res 2023; 452:114610. [PMID: 37541448 DOI: 10.1016/j.bbr.2023.114610] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 07/22/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Cortico-striatal neurocircuits mediate goal-directed and habitual actions which are necessary for adaptive behaviour. It has recently been proposed that some of the core symptoms of autism spectrum disorder (ASD) and Gilles de la Tourette syndrome (GTS), such as tics and other repetitive behaviours, may emerge because of imbalances in these neurocircuits. We have recently developed a model of ASD and GTS by knocking down Immp2l, a mitochondrial gene frequently associated with these disorders. The current study sought to determine whether Immp2l knockdown (KD) in male mice alters flexible, goal- or cue- driven behaviour using procedures specifically designed to examine response-outcome and stimulus-response associations, which underlie goal-directed and habitual behaviour, respectively. Whether Immp2l KD alters neuron density in cortico-striatal neurocircuits known to regulate these behaviours was also examined. Immp2l KD mice and wild type-like mice (WT) were trained on Pavlovian and instrumental learning procedures where auditory cues predicted food delivery and lever-press responses earned a food outcome. It was demonstrated that goal-directed learning was not changed for Immp2l KD mice compared to WT mice, as lever-press responses were sensitive to changes in the value of the food outcome, and to contingency reversal and degradation. There was also no difference in the capacity of KD mice to form habitual behaviours compared to WT mice following extending training of the instrumental action. However, Immp2l KD mice were more responsive to auditory stimuli paired with food as indicated by a non-specific increase in lever response rates during Pavlovian-to-instrumental transfer. Finally, there were no alterations to neuron density in striatum or any prefrontal cortex or limbic brain structures examined. Thus, the current study suggests that Immp2l is not necessary for learned maladaptive goal or stimulus driven behaviours in ASD or GTS, but that it may contribute to increased capacity for external stimuli to drive behaviour. Alterations to stimulus-driven behaviour could potentially influence the expression of tics and repetitive behaviours, suggesting that genetic alterations to Immp2l may contribute to these core symptoms in ASD and GTS. Given that this is the first application of this battery of instrumental learning procedures to a mouse model of ASD or GTS, it is an important initial step in determining the contribution of known risk-genes to goal-directed versus habitual behaviours, which should be more broadly applied to other rodent models of ASD and GTS in the future.
Collapse
Affiliation(s)
- Beatrice K Leung
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - Sam Merlin
- School of Science, Western Sydney University, Campbelltown, Sydney, NSW, Australia
| | - Adam K Walker
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, NSW, Australia; Discipline of Psychiatry and Mental Health, University of New South Wales, NSW, Australia
| | - Adam J Lawther
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, NSW, Australia
| | - George Paxinos
- Neuroscience Research Australia, Randwick, NSW, Australia; School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Valsamma Eapen
- Discipline of Psychiatry and Mental Health, University of New South Wales, NSW, Australia; Mental Health Research Unit, South Western Sydney Local Health District, Liverpool, Australia
| | - Raymond Clarke
- Ingham Institute, Discipline of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Bernard W Balleine
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - Teri M Furlong
- Neuroscience Research Australia, Randwick, NSW, Australia; School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
23
|
Bagosi Z, Megyesi K, Ayman J, Rudersdorf H, Ayaz MK, Csabafi K. The Role of Corticotropin-Releasing Factor (CRF) and CRF-Related Peptides in the Social Behavior of Rodents. Biomedicines 2023; 11:2217. [PMID: 37626714 PMCID: PMC10452353 DOI: 10.3390/biomedicines11082217] [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: 07/13/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Since the corticotropin-releasing factor (CRF) was isolated from an ovine brain, a growing family of CRF-related peptides has been discovered. Today, the mammalian CRF system consists of four ligands (CRF, urocortin 1 (Ucn1), urocortin 2 (Ucn2), and urocortin 3 (Ucn3)); two receptors (CRF receptor type 1 (CRF1) and CRF receptor type 2 (CRF2)); and a CRF-binding protein (CRF-BP). Besides the regulation of the neuroendocrine, autonomic, and behavioral responses to stress, CRF and CRF-related peptides are also involved in different aspects of social behavior. In the present study, we review the experiments that investigated the role of CRF and the urocortins involved in the social behavior of rats, mice, and voles, with a special focus on sociability and preference for social novelty, as well as the ability for social recognition, discrimination, and memory. In general, these experiments demonstrate that CRF, Ucn1, Ucn2, and Ucn3 play important, but distinct roles in the social behavior of rodents, and that they are mediated by CRF1 and/or CRF2. In addition, we suggest the possible brain regions and pathways that express CRF and CRF-related peptides and that might be involved in social interactions. Furthermore, we also emphasize the differences between the species, strains, and sexes that make translation of these roles from rodents to humans difficult.
Collapse
Affiliation(s)
- Zsolt Bagosi
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| | - Kíra Megyesi
- Interdisciplinary Center for Excellence, Clinical Research Competence Center, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Jázmin Ayman
- Department of Obstetrics and Gynecology, Albert Szent-Györgyi Albert School of Medicine, University of Szeged, 6720 Szeged, Hungary;
| | - Hanna Rudersdorf
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| | - Maieda Khan Ayaz
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| | - Krisztina Csabafi
- Department of Pathophysiology, Albert Szent-Györgyi School of Medicine, University of Szeged, 6720 Szeged, Hungary; (H.R.); (M.K.A.); (K.C.)
| |
Collapse
|
24
|
Guo M, Sun L. From rodents to humans: Rodent behavioral paradigms for social behavioral disorders. Brain Circ 2023; 9:154-161. [PMID: 38020957 PMCID: PMC10679632 DOI: 10.4103/bc.bc_48_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 12/01/2023] Open
Abstract
Social cognition guides social behavior. Subjects with proper social cognition should be able to: (1) have reasonable social motivation, (2) recognize other people and infer their intentions, and (3) weigh social hierarchies and other values. The choice of appropriate behavioral paradigms enables the use of rodents to study social behavior disorders in humans, thus enabling research to go deeper into neural mechanisms. This paper reviews commonly used rodent behavioral paradigms in studies of social behavior disorders. We focused specifically on sorting out ways to transfer the study of human social behavior to rodents through behavioral paradigms.
Collapse
Affiliation(s)
- Mingyue Guo
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Capital Medical University, Beijing, China
| | - Le Sun
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Mann B, Crawford JC, Reddy K, Lott J, Youn YH, Gao G, Guy C, Chou CH, Darnell D, Trivedi S, Bomme P, Loughran AJ, Thomas PG, Han YG, Tuomanen EI. Bacterial TLR2/6 Ligands Block Ciliogenesis, Derepress Hedgehog Signaling, and Expand the Neocortex. mBio 2023; 14:e0051023. [PMID: 37052506 PMCID: PMC10294647 DOI: 10.1128/mbio.00510-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Microbial components have a range of direct effects on the fetal brain. However, little is known about the cellular targets and molecular mechanisms that mediate these effects. Neural progenitor cells (NPCs) control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. We identify ventricular radial glia (vRG), the primary NPC, as the target of bacterial cell wall (BCW) generated during the antibiotic treatment of maternal pneumonia. BCW enhanced proliferative potential of vRGs by shortening the cell cycle and increasing self-renewal. Expanded vRGs propagated to increase neuronal output in all cortical layers. Remarkably, Toll-like receptor 2 (TLR2), which recognizes BCW, localized at the base of primary cilia in vRGs and the BCW-TLR2 interaction suppressed ciliogenesis leading to derepression of Hedgehog (HH) signaling and expansion of vRGs. We also show that TLR6 is an essential partner of TLR2 in this process. Surprisingly, TLR6 alone was required to set the number of cortical neurons under healthy conditions. These findings suggest that an endogenous signal from TLRs suppresses cortical expansion during normal development of the neocortex and that BCW antagonizes that signal through the TLR2/cilia/HH signaling axis changing brain structure and function. IMPORTANCE Fetal brain development in early gestation can be impacted by transplacental infection, altered metabolites from the maternal microbiome, or maternal immune activation. It is less well understood how maternal microbial subcomponents that cross the placenta, such as bacterial cell wall (BCW), directly interact with fetal neural progenitors and neurons and affect development. This scenario plays out in the clinic when BCW debris released during antibiotic therapy of maternal infection traffics to the fetal brain. This study identifies the direct interaction of BCW with TLR2/6 present on the primary cilium, the signaling hub on fetal neural progenitor cells (NPCs). NPCs control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. Within a window of vulnerability before the appearance of fetal immune cells, the BCW-TLR2/6 interaction results in the inhibition of ciliogenesis, derepression of Sonic Hedgehog signaling, excess proliferation of neural progenitors, and abnormal cortical architecture. In the first example of TLR signaling linked to Sonic Hedgehog, BCW/TLR2/6 appears to act during fetal brain morphogenesis to play a role in setting the total cell number in the neocortex.
Collapse
Affiliation(s)
- Beth Mann
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kavya Reddy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Josi Lott
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yong Ha Youn
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Geli Gao
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ching-Heng Chou
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Daniel Darnell
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sanchit Trivedi
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Perrine Bomme
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Allister J. Loughran
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Young-Goo Han
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Elaine I. Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
26
|
Ravache TT, Batistuzzo A, Nunes GG, Gomez TGB, Lorena FB, Do Nascimento BPP, Bernardi MM, Lima ERR, Martins DO, Campos ACP, Pagano RL, Ribeiro MO. Multisensory Stimulation Reverses Memory Impairment in Adrβ 3KO Male Mice. Int J Mol Sci 2023; 24:10522. [PMID: 37445699 DOI: 10.3390/ijms241310522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Norepinephrine plays an important role in modulating memory through its beta-adrenergic receptors (Adrβ: β1, β2 and β3). Here, we hypothesized that multisensory stimulation would reverse memory impairment caused by the inactivation of Adrβ3 (Adrβ3KO) with consequent inhibition of sustained glial-mediated inflammation. To test this, 21- and 86-day-old Adrβ3KO mice were exposed to an 8-week multisensory stimulation (MS) protocol that comprised gustatory and olfactory stimuli of positive and negative valence; intellectual challenges to reach food; the use of hidden objects; and the presentation of food in ways that prompted foraging, which was followed by analysis of GFAP, Iba-1 and EAAT2 protein expression in the hippocampus (HC) and amygdala (AMY). The MS protocol reduced GFAP and Iba-1 expression in the HC of young mice but not in older mice. While this protocol restored memory impairment when applied to Adrβ3KO animals immediately after weaning, it had no effect when applied to adult animals. In fact, we observed that aging worsened the memory of Adrβ3KO mice. In the AMY of Adrβ3KO older mice, we observed an increase in GFAP and EAAT2 expression when compared to wild-type (WT) mice that MS was unable to reduce. These results suggest that a richer and more diverse environment helps to correct memory impairment when applied immediately after weaning in Adrβ3KO animals and indicates that the control of neuroinflammation mediates this response.
Collapse
Affiliation(s)
- Thaís T Ravache
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Alice Batistuzzo
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Gabriela G Nunes
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Thiago G B Gomez
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| | - Fernanda B Lorena
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
- Departamento de Medicina Translacional, Universidade Federal de São Paulo 04023-062, SP, Brazil
| | - Bruna P P Do Nascimento
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
- Departamento de Medicina Translacional, Universidade Federal de São Paulo 04023-062, SP, Brazil
| | - Maria Martha Bernardi
- Graduate Program in Environmental and Experimental Pathology, Paulista University, São Paulo 04026-002, SP, Brazil
| | - Eduarda R R Lima
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
| | - Daniel O Martins
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
| | - Ana Carolina P Campos
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Rosana L Pagano
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-050, SP, Brazil
| | - Miriam O Ribeiro
- Programa de Pós-Graduação em Distúrbios do Desenvolvimento, Centro de Ciências Biológicas e da Saúde Universidade Presbiteriana Mackenzie, São Paulo 01302-907, SP, Brazil
| |
Collapse
|
27
|
Xi K, Cai SQ, Yan HF, Tian Y, Cai J, Yang XM, Wang JM, Xing GG. CSMD3 Deficiency Leads to Motor Impairments and Autism-Like Behaviors via Dysfunction of Cerebellar Purkinje Cells in Mice. J Neurosci 2023; 43:3949-3969. [PMID: 37037606 PMCID: PMC10219040 DOI: 10.1523/jneurosci.1835-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/18/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with highly heritable heterogeneity. Mutations of CUB and sushi multiple domains 3 (CSMD3) gene have been reported in individuals with ASD. However, the underlying mechanisms of CSMD3 for the onset of ASD remain unexplored. Here, using male CSMD3 knock-out (CSMD3 -/-) mice, we found that genetic deletion of CSMD3 produced core autistic-like symptoms (social interaction deficits, restricted interests, and repetitive and stereotyped behaviors) and motor dysfunction in mice, indicating that the CSMD3 gene can be considered as a candidate for ASD. Moreover, we discovered that the ablation of CSMD3 in mice led to abnormal cerebellar Purkinje cell (PC) morphology in Crus I/II lobules, including aberrant developmental dendritogenesis and spinogenesis of PCs. Furthermore, combining in vivo fiber photometry calcium imaging and ex vivo electrophysiological recordings, we showed that the CSMD3 -/- mice exhibited an increased neuronal activity (calcium fluorescence signals) in PCs of Crus I/II lobules in response to movement activity, as well as an enhanced intrinsic excitability of PCs and an increase of excitatory rather than inhibitory synaptic input to the PCs, and an impaired long-term depression at the parallel fiber-PC synapse. These results suggest that CSMD3 plays an important role in the development of cerebellar PCs. Loss of CSMD3 causes abnormal PC morphology and dysfunction in the cerebellum, which may underlie the pathogenesis of motor deficits and core autistic-like symptoms in CSMD3 -/- mice. Our findings provide novel insight into the pathophysiological mechanisms by which CSMD3 mutations cause impairments in cerebellar function that may contribute to ASD.SIGNIFICANCE STATEMENT Autism spectrum disorder (ASD) is a neurodevelopmental disorder with highly heritable heterogeneity. Advances in genomic analysis have contributed to numerous candidate genes for the risk of ASD. Recently, a novel giant gene CSMD3 encoding a protein with CUB and sushi multiple domains (CSMDs) has been identified as a candidate gene for ASD. However, the underlying mechanisms of CSMD3 for the onset of ASD remain largely unknown. Here, we unravel that loss of CSMD3 results in abnormal morphology, increased intrinsic excitabilities, and impaired synaptic plasticity in cerebellar PCs, subsequently leading to motor deficits and ASD-like behaviors in mice. These results provide novel insight into the pathophysiological mechanisms by which CSMD3 mutations cause impairments in cerebellar function that may contribute to ASD.
Collapse
Affiliation(s)
- Ke Xi
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Si-Qing Cai
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Hui-Fang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Yue Tian
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Jie Cai
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Xiao-Mei Yang
- Department of Human Anatomy and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Jing-Min Wang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Guo-Gang Xing
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
- Second Affiliated Hospital of Xinxiang Medical University, Henan 453002, People's Republic of China
| |
Collapse
|
28
|
Schwabenland M, Mossad O, Sievert A, Peres AG, Ringel E, Baasch S, Kolter J, Cascone G, Dokalis N, Vlachos A, Ruzsics Z, Henneke P, Prinz M, Blank T. Neonatal immune challenge poses a sex-specific risk for epigenetic microglial reprogramming and behavioral impairment. Nat Commun 2023; 14:2721. [PMID: 37169749 PMCID: PMC10175500 DOI: 10.1038/s41467-023-38373-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/27/2023] [Indexed: 05/13/2023] Open
Abstract
While the precise processes underlying a sex bias in the development of central nervous system (CNS) disorders are unknown, there is growing evidence that an early life immune activation can contribute to the disease pathogenesis. When we mimicked an early systemic viral infection or applied murine cytomegalovirus (MCMV) systemically in neonatal female and male mice, only male adolescent mice presented behavioral deficits, including reduced social behavior and cognition. This was paralleled by an increased amount of infiltrating T cells in the brain parenchyma, enhanced interferon-γ (IFNγ) signaling, and epigenetic reprogramming of microglial cells. These microglial cells showed increased phagocytic activity, which resulted in abnormal loss of excitatory synapses within the hippocampal brain region. None of these alterations were seen in female adolescent mice. Our findings underscore the early postnatal period's susceptibility to cause sex-dependent long-term CNS deficiencies following infections.
Collapse
Affiliation(s)
- Marius Schwabenland
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Omar Mossad
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Annika Sievert
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Adam G Peres
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elena Ringel
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Baasch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Giulia Cascone
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nikolaos Dokalis
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Zsolt Ruzsics
- Institute for Virology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Pediatrics and Adolescent Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
29
|
Guerra M, Medici V, Weatheritt R, Corvino V, Palacios D, Geloso MC, Farini D, Sette C. Fetal exposure to valproic acid dysregulates the expression of autism-linked genes in the developing cerebellum. Transl Psychiatry 2023; 13:114. [PMID: 37019889 PMCID: PMC10076313 DOI: 10.1038/s41398-023-02391-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 04/07/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a set of highly heritable neurodevelopmental syndromes characterized by social and communication impairment, repetitive behaviour, and intellectual disability. Although mutations in multiple genes have been associated to ASD, most patients lack detectable genetic alterations. For this reason, environmental factors are commonly thought to also contribute to ASD aetiology. Transcriptome analyses have revealed that autistic brains possess distinct gene expression signatures, whose elucidation can provide insights about the mechanisms underlying the effects of ASD-causing genetic and environmental factors. Herein, we have identified a coordinated and temporally regulated programme of gene expression in the post-natal development of cerebellum, a brain area whose defects are strongly associated with ASD. Notably, this cerebellar developmental programme is significantly enriched in ASD-linked genes. Clustering analyses highlighted six different patterns of gene expression modulated during cerebellar development, with most of them being enriched in functional processes that are frequently dysregulated in ASD. By using the valproic acid mouse model of ASD, we found that ASD-linked genes are dysregulated in the developing cerebellum of ASD-like mice, a defect that correlates with impaired social behaviour and altered cerebellar cortical morphology. Moreover, changes in transcript levels were reflected in aberrant protein expression, indicating the functional relevance of these alterations. Thus, our work uncovers a complex ASD-related transcriptional programme regulated during cerebellar development and highlight genes whose expression is dysregulated in this brain area of an ASD mouse model.
Collapse
Affiliation(s)
- Marika Guerra
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Vanessa Medici
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
| | - Robert Weatheritt
- Garvan Institute of Medical Research, EMBL Australia, Darlinghurst, NSW, Australia
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
| | - Daniela Palacios
- Department of Life Science and Public Health, Section of Biology, Catholic University of the Sacred Hearth, Rome, Italy
| | - Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Donatella Farini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Hearth, Rome, Italy.
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy.
| |
Collapse
|
30
|
de Oliveira Ferreira E, Pessoa Gomes JM, Neves KRT, Lima FAV, de Barros Viana GS, de Andrade GM. Maternal treatment with aripiprazole prevents the development of a valproic acid-induced autism-like phenotype in juvenile male mice. Behav Pharmacol 2023; 34:154-168. [PMID: 36853856 DOI: 10.1097/fbp.0000000000000718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Autism spectrum disorder (ASD) describes a heterogeneous group of neurodevelopmental conditions characterized by deficits in social communication and repetitive behaviors. Aripiprazole (APZ) is an atypical antipsychotic that can safeguard mice against autism-like behavior induced by valproic acid (VPA). In the present study, we examined the effects of maternal treatment with APZ (10 mg/kg) in juvenile mice prenatally exposed to VPA on neurodevelopmental behaviors, social interactions, communication, and working memory, as well as synaptophysin (SYP), synaptosomal-associated protein, 25 kDa (SNAP-25) and microtubule-associated protein 2 (MAP-2) expression in the medial prefrontal cortex (mPFC) and cell viability in the hippocampus. In addition, to evaluate possible APZ interference with the anticonvulsant properties of VPA on pentylenetetrazole (PTZ)-induced seizures were evaluated. Maternal treatment with APZ significantly prevented body weight loss, self-righting, eye-opening, social interactions, social communication, and working memory deficits in mice prenatally exposed to VPA. Additionally, the decrease in the SYP, SNAP-25, and MAP-2 expressions in the mPFC and cell death in the hippocampus was prevented by APZ. Furthermore, APZ (10 mg/kg) did not interfere with the anticonvulsant effect of VPA (15 mg/kg) in animals with PTZ-induced seizures. These findings indicate that maternal treatment with APZ in pregnant mice exposed to VPA protects animals against the ASD-like behavioral phenotype, and this effect may be related, at least in part, to synaptic plasticity and neuronal protection in the PFC and hippocampus. APZ may serve as an effective pharmacological therapeutic target against autistic behaviors in the VPA animal model of ASD, which should be further investigated to verify its clinical relevance.
Collapse
Affiliation(s)
| | | | | | | | - Glauce Socorro de Barros Viana
- Department of Physiology and Pharmacology
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Brazil
| | - Geanne Matos de Andrade
- Department of Clinical Medicine
- Department of Physiology and Pharmacology
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza, Brazil
| |
Collapse
|
31
|
Liu B, Qian Y, Wang J. EDDSN-MRT: multiple rodent tracking based on ear detection and dual siamese network for rodent social behavior analysis. BMC Neurosci 2023; 24:23. [PMID: 36973649 PMCID: PMC10044788 DOI: 10.1186/s12868-023-00787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Rodent social behavior is a commonly used preclinical model to interrogate the mechanisms underpinning various human neurological conditions. To investigate the interplay between neural systems and social behaviors, neuroscientists need a precise quantitative measure for multi-rodent tracking and behavior assessment in laboratory settings. However, identifying individual differences across multiple rodents due to visual occlusion precludes the generation of stable individual tracks across time. METHODS To overcome the present limitations of multi-rodent tracking, we have developed an Ear Detection and Dual Siamese Network for Multiple Rodent Tracking (EDDSN-MRT). The aim of this study is to validate the EDDSN-MRT system in mice using a publicly available dataset and compare it with several current state-of-the-art methods for behavioral assessment. To demonstrate its application and effectiveness in the assessment of multi-rodent social behavior, we implemented an intermittent fasting intervention experiment on 4 groups of mice (each group is with different ages and fasting status and contains 8 individuals). We used the EDDSN-MRT system to track multiple mice simultaneously and for the identification and analysis of individual differences in rodent social behavior and compared our proposed method with Toxtrac and idtracker.ai. RESULTS The locomotion behavior of up to 4 mice can be tracked simultaneously using the EDDSN-MRT system. Unexpectedly, we found intermittent fasting led to a decrease in the spatial distribution of the mice, contrasting with previous findings. Furthermore, we show that the EDDSN-MRT system can be used to analyze the social behavior of multiple mice of different ages and fasting status and provide data on locomotion behavior across multiple mice simultaneously. CONCLUSIONS Compared with several state-of-the-art methods, the EDDSN-MRT system provided better tracking performance according to Multiple Object Tracking Accuracy (MOTA) and ID Correct Rate (ICR). External experimental validation suggests that the EDDSN-MRT system has sensitivity to distinguish the behaviors of mice on different intermittent fasting regimens. The EDDSN-MRT system code is freely available here: https://github.com/fliessen/EDDSN-MRT .
Collapse
Affiliation(s)
- Bingbin Liu
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Yuxuan Qian
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China.
| |
Collapse
|
32
|
Haddad FL, De Oliveira C, Schmid S. Investigating behavioral phenotypes related to autism spectrum disorder in a gene-environment interaction model of Cntnap2 deficiency and Poly I:C maternal immune activation. Front Neurosci 2023; 17:1160243. [PMID: 36998729 PMCID: PMC10043204 DOI: 10.3389/fnins.2023.1160243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
IntroductionAutism Spectrum Disorder (ASD) has been associated with a wide variety of genetic and environmental risk factors in both human and preclinical studies. Together, findings support a gene-environment interaction hypothesis whereby different risk factors independently and synergistically impair neurodevelopment and lead to the core symptoms of ASD. To date, this hypothesis has not been commonly investigated in preclinical ASD models. Mutations in the Contactin-associated protein-like 2 (Cntnap2) gene and exposure to maternal immune activation (MIA) during pregnancy have both been linked to ASD in humans, and preclinical rodent models have shown that both MIA and Cntnap2 deficiency lead to similar behavioral deficits.MethodsIn this study, we tested the interaction between these two risk factors by exposing Wildtype, Cntnap2+/–, and Cntnap2–/– rats to Polyinosinic: Polycytidylic acid (Poly I:C) MIA at gestation day 9.5.ResultsOur findings showed that Cntnap2 deficiency and Poly I:C MIA independently and synergistically altered ASD-related behaviors like open field exploration, social behavior, and sensory processing as measured through reactivity, sensitization, and pre-pulse inhibition (PPI) of the acoustic startle response. In support of the double-hit hypothesis, Poly I:C MIA acted synergistically with the Cntnap2–/– genotype to decrease PPI in adolescent offspring. In addition, Poly I:C MIA also interacted with the Cntnap2+/– genotype to produce subtle changes in locomotor hyperactivity and social behavior. On the other hand, Cntnap2 knockout and Poly I:C MIA showed independent effects on acoustic startle reactivity and sensitization.DiscussionTogether, our findings support the gene-environment interaction hypothesis of ASD by showing that different genetic and environmental risk factors could act synergistically to exacerbate behavioral changes. In addition, by showing the independent effects of each risk factor, our findings suggest that ASD phenotypes could be caused by different underlying mechanisms.
Collapse
Affiliation(s)
- Faraj L. Haddad
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Cleusa De Oliveira
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Psychology, The University of Western Ontario, London, ON, Canada
- *Correspondence: Susanne Schmid,
| |
Collapse
|
33
|
Anderson JM, Boardman AA, Bates R, Zou X, Huang W, Cao L. Hypothalamic TrkB.FL overexpression improves metabolic outcomes in the BTBR mouse model of autism. PLoS One 2023; 18:e0282566. [PMID: 36893171 PMCID: PMC9997972 DOI: 10.1371/journal.pone.0282566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
BTBR T+ Itpr3tf/J (BTBR) mice are used as a model of autism spectrum disorder (ASD), displaying similar behavioral and physiological deficits observed in patients with ASD. Our recent study found that implementation of an enriched environment (EE) in BTBR mice improved metabolic and behavioral outcomes. Brain-derived neurotrophic factor (Bdnf) and its receptor tropomyosin kinase receptor B (Ntrk2) were upregulated in the hypothalamus, hippocampus, and amygdala by implementing EE in BTBR mice, suggesting that BDNF-TrkB signaling plays a role in the EE-BTBR phenotype. Here, we used an adeno-associated virus (AAV) vector to overexpress the TrkB full-length (TrkB.FL) BDNF receptor in the BTBR mouse hypothalamus in order to assess whether hypothalamic BDNF-TrkB signaling is responsible for the improved metabolic and behavioral phenotypes associated with EE. Normal chow diet (NCD)-fed and high fat diet (HFD)-fed BTBR mice were randomized to receive either bilateral injections of AAV-TrkB.FL or AAV-YFP as control, and were subjected to metabolic and behavioral assessments up to 24 weeks post-injection. Both NCD and HFD TrkB.FL overexpressing mice displayed improved metabolic outcomes, characterized as reduced percent weight gain and increased energy expenditure. NCD TrkB.FL mice showed improved glycemic control, reduced adiposity, and increased lean mass. In NCD mice, TrkB.FL overexpression altered the ratio of TrkB.FL/TrkB.T1 protein expression and increased phosphorylation of PLCγ in the hypothalamus. TrkB.FL overexpression also upregulated expression of hypothalamic genes involved in energy regulation and altered expression of genes involved in thermogenesis, lipolysis, and energy expenditure in white adipose tissue and brown adipose tissue. In HFD mice, TrkB.FL overexpression increased phosphorylation of PLCγ. TrkB.FL overexpression in the hypothalamus did not improve behavioral deficits in either NCD or HFD mice. Together, these results suggest that enhancing hypothalamic TrkB.FL signaling improves metabolic health in BTBR mice.
Collapse
Affiliation(s)
- Jacqueline M. Anderson
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Amber A. Boardman
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Rhiannon Bates
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Xunchang Zou
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Wei Huang
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| | - Lei Cao
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States of America
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
34
|
Cabana-Domínguez J, Antón-Galindo E, Fernàndez-Castillo N, Singgih EL, O'Leary A, Norton WH, Strekalova T, Schenck A, Reif A, Lesch KP, Slattery D, Cormand B. The translational genetics of ADHD and related phenotypes in model organisms. Neurosci Biobehav Rev 2023; 144:104949. [PMID: 36368527 DOI: 10.1016/j.neubiorev.2022.104949] [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: 07/01/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly prevalent neurodevelopmental disorder resulting from the interaction between genetic and environmental risk factors. It is well known that ADHD co-occurs frequently with other psychiatric disorders due, in part, to shared genetics factors. Although many studies have contributed to delineate the genetic landscape of psychiatric disorders, their specific molecular underpinnings are still not fully understood. The use of animal models can help us to understand the role of specific genes and environmental stimuli-induced epigenetic modifications in the pathogenesis of ADHD and its comorbidities. The aim of this review is to provide an overview on the functional work performed in rodents, zebrafish and fruit fly and highlight the generated insights into the biology of ADHD, with a special focus on genetics and epigenetics. We also describe the behavioral tests that are available to study ADHD-relevant phenotypes and comorbid traits in these models. Furthermore, we have searched for new models to study ADHD and its comorbidities, which can be useful to test potential pharmacological treatments.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| | - Ester Antón-Galindo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Noèlia Fernàndez-Castillo
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain
| | - Euginia L Singgih
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia
| | - William Hg Norton
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany, and Department of Neuropsychology and Psychiatry, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - David Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Bru Cormand
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Catalonia, Spain.
| |
Collapse
|
35
|
Basilico B, Ferrucci L, Khan A, Di Angelantonio S, Ragozzino D, Reverte I. What microglia depletion approaches tell us about the role of microglia on synaptic function and behavior. Front Cell Neurosci 2022; 16:1022431. [PMID: 36406752 PMCID: PMC9673171 DOI: 10.3389/fncel.2022.1022431] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Microglia are dynamic cells, constantly surveying their surroundings and interacting with neurons and synapses. Indeed, a wealth of knowledge has revealed a critical role of microglia in modulating synaptic transmission and plasticity in the developing brain. In the past decade, novel pharmacological and genetic strategies have allowed the acute removal of microglia, opening the possibility to explore and understand the role of microglia also in the adult brain. In this review, we summarized and discussed the contribution of microglia depletion strategies to the current understanding of the role of microglia on synaptic function, learning and memory, and behavior both in physiological and pathological conditions. We first described the available microglia depletion methods highlighting their main strengths and weaknesses. We then reviewed the impact of microglia depletion on structural and functional synaptic plasticity. Next, we focused our analysis on the effects of microglia depletion on behavior, including general locomotor activity, sensory perception, motor function, sociability, learning and memory both in healthy animals and animal models of disease. Finally, we integrated the findings from the reviewed studies and discussed the emerging roles of microglia on the maintenance of synaptic function, learning, memory strength and forgetfulness, and the implications of microglia depletion in models of brain disease.
Collapse
Affiliation(s)
| | - Laura Ferrucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Azka Khan
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- and Neuro-Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Davide Ragozzino
- Laboratory Affiliated to Institute Pasteur Italia – Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
- *Correspondence: Davide Ragozzino,
| | - Ingrid Reverte
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), Rome, Italy
- Ingrid Reverte,
| |
Collapse
|
36
|
Corredor K, Duran J, Herrera-Isaza L, Forero S, Quintanilla J, Gomez A, Martínez GS, Cardenas FP. Behavioral effects of environmental enrichment on male and female wistar rats with early life stress experiences. Front Physiol 2022; 13:837661. [PMID: 36225294 PMCID: PMC9548697 DOI: 10.3389/fphys.2022.837661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure to adverse childhood experiences or early life stress experiences (ELSs) increase the risk of non-adaptive behaviors and psychopathology in adulthood. Environmental enrichment (EE) has been proposed to minimize these effects. The vast number of methodological variations in animal studies underscores the lack of systematicity in the studies and the need for a detailed understanding of how enrichment interacts with other variables. Here we evaluate the effects of environmental enrichment in male and female Wistar rats exposed to adverse early life experiences (prenatal, postnatal, and combined) on emotional (elevated plus maze), social (social interaction chamber), memory (Morris water maze) and flexibility tasks. Our results—collected from PND 51 to 64—confirmed: 1) the positive effect of environmental enrichment (PND 28–49) on anxiety-like behaviors in animals submitted to ELSs. These effects depended on type of experience and type of enrichment: foraging enrichment reduced anxiety-like behaviors in animals with prenatal and postnatal stress but increased them in animals without ELSs. This effect was sex-dependent: females showed lower anxiety compared to males. Our data also indicated that females exposed to prenatal and postnatal stress had lower anxious responses than males in the same conditions; 2) no differences were found for social interactions; 3) concerning memory, there was a significant interaction between the three factors: A significant interaction for males with prenatal stress was observed for foraging enrichment, while physical enrichment was positive for males with postnatal stress; d) regarding cognitive flexibility, a positive effect of EE was found in animals exposed to adverse ELSs: animals with combined stress and exposed to physical enrichment showed a higher index of cognitive flexibility than those not exposed to enrichment. Yet, within animals with no EE, those exposed to combined stress showed lower flexibility than those exposed to both prenatal stress and no stress. On the other hand, animals with prenatal stress and exposed to foraging-type enrichment showed lower cognitive flexibility than those with no EE. The prenatal stress-inducing conditions used here 5) did not induced fetal or maternal problems and 6) did not induced changes in the volume of the dentate gyrus of the hippocampus.
Collapse
Affiliation(s)
- K. Corredor
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
- Centro de Investigación en Biomodelos, Bogotá, Colombia
| | - J.M. Duran
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - L. Herrera-Isaza
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - S. Forero
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - J.P. Quintanilla
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | - A. Gomez
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
| | | | - F. P. Cardenas
- Laboratory of Neuroscience and Behavior, Universidad de los Andes, Bogotá, Colombia
- *Correspondence: F. P. Cardenas,
| |
Collapse
|
37
|
Punt AM, Judson MC, Sidorov MS, Williams BN, Johnson NS, Belder S, den Hertog D, Davis CR, Feygin MS, Lang PF, Jolfaei MA, Curran PJ, van IJcken WF, Elgersma Y, Philpot BD. Molecular and behavioral consequences of Ube3a gene overdosage in mice. JCI Insight 2022; 7:e158953. [PMID: 36134658 PMCID: PMC9675564 DOI: 10.1172/jci.insight.158953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/17/2022] [Indexed: 12/01/2022] Open
Abstract
Chromosome 15q11.2-q13.1 duplication syndrome (Dup15q syndrome) is a severe neurodevelopmental disorder characterized by intellectual disability, impaired motor coordination, and autism spectrum disorder. Chromosomal multiplication of the UBE3A gene is presumed to be the primary driver of Dup15q pathophysiology, given that UBE3A exhibits maternal monoallelic expression in neurons and that maternal duplications typically yield far more severe neurodevelopmental outcomes than paternal duplications. However, studies into the pathogenic effects of UBE3A overexpression in mice have yielded conflicting results. Here, we investigated the neurodevelopmental impact of Ube3a gene overdosage using bacterial artificial chromosome-based transgenic mouse models (Ube3aOE) that recapitulate the increases in Ube3a copy number most often observed in Dup15q. In contrast to previously published Ube3a overexpression models, Ube3aOE mice were indistinguishable from wild-type controls on a number of molecular and behavioral measures, despite suffering increased mortality when challenged with seizures, a phenotype reminiscent of sudden unexpected death in epilepsy. Collectively, our data support a model wherein pathogenic synergy between UBE3A and other overexpressed 15q11.2-q13.1 genes is required for full penetrance of Dup15q syndrome phenotypes.
Collapse
Affiliation(s)
- A. Mattijs Punt
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Matthew C. Judson
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Michael S. Sidorov
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Brittany N. Williams
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Naomi S. Johnson
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Sabine Belder
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Dion den Hertog
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Courtney R. Davis
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Maximillian S. Feygin
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Patrick F. Lang
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| | - Mehrnoush Aghadavoud Jolfaei
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Patrick J. Curran
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Ype Elgersma
- Department of Clinical Genetics and Department of Neuroscience and
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, Netherlands
| | - Benjamin D. Philpot
- Neuroscience Center, Department of Cell Biology and Physiology, and the Carolina Institute for Developmental Disabilities and
| |
Collapse
|
38
|
Biosca-Brull J, Guardia-Escote L, Blanco J, Basaure P, Cabré M, Sánchez-Santed F, Domingo JL, Colomina MT. Prenatal, but not postnatal exposure to chlorpyrifos affects social behavior of mice and the excitatory-inhibitory balance in a sex-dependent manner. Food Chem Toxicol 2022; 169:113423. [PMID: 36113784 DOI: 10.1016/j.fct.2022.113423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022]
Abstract
The balance between excitatory and inhibitory neurotransmitters is essential for proper brain development. An imbalance between these two systems has been associated with neurodevelopmental disorders. On the other hand, literature also associates the massive use of pesticides with the increase of these disorders, with a particular focus on chlorpyrifos (CPF) a world-wide used organophosphate pesticide. This study was aimed at assessing social autistic-like behaviors on mice pre or postnatally exposed to CPF (0 or 1 mg/kg/day), in both sexes. In prenatal exposure, C57BL/6J pregnant mice were exposed to CPF through the diet, between gestational days (GD) 12 and 18, while a positive control group for some autistic behaviors was exposed to valproic acid (VPA) on GD 12 and 13. To assess postnatal exposure, C57BL/6J mice were orally exposed to the vehicle (corn oil) or CPF, from postnatal days (PND) 10-15. Social behavior and gene expression analysis were assessed on PND 45. Results showed social alterations only in males prenatally treated. GABA system was upregulated in CPF-treated females, whereas an increase in both systems was observed in both treated males. These findings suggest that males are more sensitive to prenatal CPF exposure, favoring the sex bias observed in ASD.
Collapse
Affiliation(s)
- Judit Biosca-Brull
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain.
| | - Laia Guardia-Escote
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain
| | - Jordi Blanco
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain; Universitat Rovira i Virgili, Department of Basic Medical Sciences, Reus, Spain
| | - Pia Basaure
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain
| | - Maria Cabré
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Biochemistry and Biotechnology, Tarragona, Spain
| | - Fernando Sánchez-Santed
- Department of Psychology, Health Research Center (CEINSA), Almeria University, 04120, Almeria, Spain
| | - José L Domingo
- Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain
| | - Maria Teresa Colomina
- Universitat Rovira i Virgili, Research Group in Neurobehavior and Health (NEUROLAB), Tarragona, Spain; Universitat Rovira i Virgili, Department of Psychology and Research Center for Behavior Assessment (CRAMC), Tarragona, Spain; Universitat Rovira i Virgili, Laboratory of Toxicology and Environmental Health (TECNATOX), Reus, Spain.
| |
Collapse
|
39
|
Rigney N, Zbib A, de Vries GJ, Petrulis A. Knockdown of sexually differentiated vasopressin expression in the bed nucleus of the stria terminalis reduces social and sexual behaviour in male, but not female, mice. J Neuroendocrinol 2022; 34:e13083. [PMID: 34978098 PMCID: PMC9213575 DOI: 10.1111/jne.13083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022]
Abstract
The neuropeptide arginine-vasopressin (AVP) has long been implicated in the regulation of social behaviour and communication, but the sources of AVP release relevant for behaviour have not been precisely determined. Ablations of the sexually dimorphic AVP cells within the bed nucleus of the stria terminalis (BNST), which are more numerous in males, affect social behaviour differently in males and females. However, it is unknown whether these behavioural effects are caused by a reduction of AVP or of other factors associated with these cells. To test the role of AVP specifically, we used an shRNA viral construct to knock down AVP gene expression within the BNST of wild-type male and female mice, using scrambled sequence virus as a control, and evaluated subsequent changes in social behaviours (social investigation, ultrasonic vocalization (USV), scent marking, copulation, and aggression), or anxiety-like behaviours (elevated plus maze). We observed that, in males, knockdown of AVP expression in the BNST strongly reduced investigation of novel males, aggressive signalling towards other males (tail rattling, USV), and copulatory behaviour, but did not alter attack initiation, other measures of social communication, or anxiety-like behaviours. In females, however, BNST AVP knockdown did not alter any of these behaviours. These results point to differential involvement of AVP derived from the BNST in social behaviour.
Collapse
Affiliation(s)
- Nicole Rigney
- Center for Behavioral NeuroscienceNeuroscience InstituteGeorgia State UniversityAtlantaGeorgiaUSA
| | - Adam Zbib
- Center for Behavioral NeuroscienceNeuroscience InstituteGeorgia State UniversityAtlantaGeorgiaUSA
| | - Geert J. de Vries
- Center for Behavioral NeuroscienceNeuroscience InstituteGeorgia State UniversityAtlantaGeorgiaUSA
| | - Aras Petrulis
- Center for Behavioral NeuroscienceNeuroscience InstituteGeorgia State UniversityAtlantaGeorgiaUSA
| |
Collapse
|
40
|
Haddad FL, Patel SV, Doornaert EE, De Oliveira C, Allman BL, Baines KJ, Renaud SJ, Schmid S. Interleukin 15 modulates the effects of poly I:C maternal immune activation on offspring behaviour. Brain Behav Immun Health 2022; 23:100473. [PMID: 35668725 PMCID: PMC9166394 DOI: 10.1016/j.bbih.2022.100473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 01/21/2023] Open
Abstract
Maternal infections during pregnancy are linked with an increased risk for disorders like Autism Spectrum Disorder and schizophrenia in the offspring. Although precise mechanisms are still unclear, clinical and preclinical evidence suggest a strong role for maternal immune activation (MIA) in the neurodevelopmental disruptions caused by maternal infection. Previously, studies using the Polyinosinic:Polycytidylic (Poly I:C) MIA preclinical model showed that cytokines like Interleukin 6 (Il6) are important mediators of MIA's effects. In this study, we hypothesized that Il15 may similarly act as a mediator of Poly I:C MIA, given its role in the antiviral immune response. To test this hypothesis, we induced Poly I:C MIA at gestational day 9.5 in wildtype (WT) and Il15−/− rat dams and tested their offspring in adolescence and adulthood. Poly I:C MIA and Il15 knockout produced both independent and synergistic effects on offspring behaviour. Poly I:C MIA decreased startle reactivity in adult WT offspring but resulted in increased adolescent anxiety and decreased adult locomotor activity in Il15−/− offspring. In addition, Poly I:C MIA led to genotype-independent effects on locomotor activity and prepulse inhibition. Finally, we showed that Il15−/− offspring exhibit distinct phenotypes that were unrelated to Poly I:C MIA including altered startle reactivity, locomotion and signal transduction in the auditory brainstem. Overall, our findings indicate that the lack of Il15 can leave offspring either more or less susceptible to Poly I:C MIA, depending on the phenotype in question. Future studies should examine the contribution of fetal versus maternal Il15 in MIA to determine the precise developmental mechanisms underlying these changes. Poly I:C MIA decreases startle reactivity in adult WT but not Il15−/− offspring. Il15−/− offspring exposed to Poly I:C MIA show altered PPI and open field exploration. Il15−/− rats exhibit distinct behavioural phenotypes independent from MIA.
Collapse
|
41
|
Huo Y, Lu W, Tian Y, Hou Q, Man HY. Prkn knockout mice show autistic-like behaviors and aberrant synapse formation. iScience 2022; 25:104573. [PMID: 35789851 PMCID: PMC9249611 DOI: 10.1016/j.isci.2022.104573] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/26/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with high genetic heterogeneity, affecting one in 44 children in the United States. Recent genomic sequencing studies from autistic human individuals indicate that PARK2, a gene that has long been considered in the pathogenesis of Parkinson's disease, is involved in ASD. Here, we report that Prkn knockout (KO) mice demonstrate autistic-like behaviors including impaired social interaction, elevated repetitive behaviors, and deficits in communication. In addition, Prkn KO mice show reduced neuronal activity in the context of sociability in the prelimbic cortex. Cell morphological examination of layer 5 prelimbic cortical neurons shows a reduction in dendritic arborization and spine number. Furthermore, biochemistry and immunocytochemistry analyses reveal alterations in synapse density and the molecular composition of synapses. These findings indicate that Prkn is implicated in brain development and suggest the potential use of the Prkn KO mouse as a model for autism research.
Collapse
Affiliation(s)
- Yuda Huo
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Wen Lu
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Yuan Tian
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Qingming Hou
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA
| |
Collapse
|
42
|
Establishment of a social conditioned place preference paradigm for the study of social reward in female mice. Sci Rep 2022; 12:11271. [PMID: 35789188 PMCID: PMC9253334 DOI: 10.1038/s41598-022-15427-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/23/2022] [Indexed: 11/12/2022] Open
Abstract
Social interactions can be and often are rewarding. The effect of social contact strongly depends on circumstances, and the reward may be driven by varied motivational processes, ranging from parental or affiliative behaviors to investigation or aggression. Reward associated with nonreproductive interactions in rodents is measured using the social conditioned place preference (sCPP) paradigm, where a change in preference for an initially neutral context confirms reinforcing effects of social contact. Here, we revised the sCPP method and reexamined social reward in adult female mice. Contrary to earlier studies, we found that robust rewarding effects of social contact could be detected in adult (14-week-old) female C57BL/6 mice when the sCPP task was refined to remove confounding factors. Strikingly, the rewarding effects of social interaction were only observed among female siblings who remained together from birth. Contact with same-age nonsiblings was not rewarding even after 8 weeks of cohousing. Other factors critical for the social reward effect in the sCPP paradigm included the number of conditioning sessions and the inherent preference for contextual cues. Thus, we show that social interaction is rewarding in adult female mice, but this effect strictly depends on the familiarity of the interaction partners. Furthermore, by identifying confounding factors, we provide a behavioral model to study the mechanisms underlying the rewarding effects of nonreproductive social interaction in adult mice.
Collapse
|
43
|
Chaudry S, Vasudevan N. mTOR-Dependent Spine Dynamics in Autism. Front Mol Neurosci 2022; 15:877609. [PMID: 35782388 PMCID: PMC9241970 DOI: 10.3389/fnmol.2022.877609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Conditions (ASC) are a group of neurodevelopmental disorders characterized by deficits in social communication and interaction as well as repetitive behaviors and restricted range of interests. ASC are complex genetic disorders with moderate to high heritability, and associated with atypical patterns of neural connectivity. Many of the genes implicated in ASC are involved in dendritic spine pruning and spine development, both of which can be mediated by the mammalian target of rapamycin (mTOR) signaling pathway. Consistent with this idea, human postmortem studies have shown increased spine density in ASC compared to controls suggesting that the balance between autophagy and spinogenesis is altered in ASC. However, murine models of ASC have shown inconsistent results for spine morphology, which may underlie functional connectivity. This review seeks to establish the relevance of changes in dendritic spines in ASC using data gathered from rodent models. Using a literature survey, we identify 20 genes that are linked to dendritic spine pruning or development in rodents that are also strongly implicated in ASC in humans. Furthermore, we show that all 20 genes are linked to the mTOR pathway and propose that the mTOR pathway regulating spine dynamics is a potential mechanism underlying the ASC signaling pathway in ASC. We show here that the direction of change in spine density was mostly correlated to the upstream positive or negative regulation of the mTOR pathway and most rodent models of mutant mTOR regulators show increases in immature spines, based on morphological analyses. We further explore the idea that these mutations in these genes result in aberrant social behavior in rodent models that is due to these altered spine dynamics. This review should therefore pave the way for further research on the specific genes outlined, their effect on spine morphology or density with an emphasis on understanding the functional role of these changes in ASC.
Collapse
|
44
|
Keysers C, Knapska E, Moita MA, Gazzola V. Emotional contagion and prosocial behavior in rodents. Trends Cogn Sci 2022; 26:688-706. [PMID: 35667978 DOI: 10.1016/j.tics.2022.05.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 01/09/2023]
Abstract
Empathy is critical to adjusting our behavior to the state of others. The past decade dramatically deepened our understanding of the biological origin of this capacity. We now understand that rodents robustly show emotional contagion for the distress of others via neural structures homologous to those involved in human empathy. Their propensity to approach others in distress strengthens this effect. Although rodents can also learn to favor behaviors that benefit others via structures overlapping with those of emotional contagion, they do so less reliably and more selectively. Together, this suggests evolution selected mechanisms for emotional contagion to prepare animals for dangers by using others as sentinels. Such shared emotions additionally can, under certain circumstances, promote prosocial behavior.
Collapse
Affiliation(s)
- Christian Keysers
- Social Brain Lab, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Art and Sciences, Amsterdam, the Netherlands; Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands.
| | - Ewelina Knapska
- Laboratory of Emotions' Neurobiology, Center of Excellence for Neural Plasticity and Brain Disorders BRAINCITY, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Marta A Moita
- Champalimaud Neuroscience Progamme, Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Valeria Gazzola
- Social Brain Lab, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Art and Sciences, Amsterdam, the Netherlands; Department of Psychology, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
45
|
Fabregat M, Niño-Rivero S, Pose S, Cárdenas-Rodríguez M, Bresque M, Hernández K, Prieto-Echagüe V, Schlapp G, Crispo M, Lagos P, Lago N, Escande C, Irigoín F, Badano JL. Generation and characterization of Ccdc28b mutant mice links the Bardet-Biedl associated gene with mild social behavioral phenotypes. PLoS Genet 2022; 18:e1009896. [PMID: 35653384 PMCID: PMC9197067 DOI: 10.1371/journal.pgen.1009896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 06/14/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
CCDC28B (coiled-coil domain-containing protein 28B) was identified as a modifier in the ciliopathy Bardet-Biedl syndrome (BBS). Our previous work in cells and zebrafish showed that CCDC28B plays a role regulating cilia length in a mechanism that is not completely understood. Here we report the generation of a Ccdc28b mutant mouse using CRISPR/Cas9 (Ccdc28b mut). Depletion of CCDC28B resulted in a mild phenotype. Ccdc28b mut animals i) do not present clear structural cilia affectation, although we did observe mild defects in cilia density and cilia length in some tissues, ii) reproduce normally, and iii) do not develop retinal degeneration or obesity, two hallmark features of reported BBS murine models. In contrast, Ccdc28b mut mice did show clear social interaction defects as well as stereotypical behaviors. This finding is indeed relevant regarding CCDC28B as a modifier of BBS since behavioral phenotypes have been documented in BBS. Overall, this work reports a novel mouse model that will be key to continue evaluating genetic interactions in BBS, deciphering the contribution of CCDC28B to modulate the presentation of BBS phenotypes. In addition, our data underscores a novel link between CCDC28B and behavioral defects, providing a novel opportunity to further our understanding of the genetic, cellular, and molecular basis of these complex phenotypes. BBS is caused by mutations in any one of 22 genes known to date. In some families, BBS can be inherited as an oligogenic trait whereby mutations in more than one BBS gene collaborate in the presentation of the syndrome. In addition, CCDC28B was originally identified as a modifier of BBS, whereby a reduction in CCDC28B levels was associated with a more severe presentation of the syndrome. Different mechanisms, all relying on functional redundancy, have been proposed to explain these genetic interactions. The characterization of BBS proteins supported this functional redundancy hypothesis: BBS proteins play a role in cilia maintenance/function and subsets of BBS proteins can even interact directly in multiprotein complexes. We have previously shown that CCDC28B also participates in cilia biology regulating the length of the organelle: knockdown of CCDC28B in cells results in cilia shortening and targeting ccdc28b in zebrafish also results in early embryonic phenotypes characteristic of other cilia mutants. In this work, we generated a Ccdc28b mutant mouse to determine whether abrogating Ccdc28b function would be sufficient to cause a ciliopathy phenotype in mammals, and to generate a tool to continue dissecting its modifying role in the context of BBS. Overall, Ccdc28b mutant mice presented a mild phenotype, a finding fully compatible with its role as a modifier, rather than a causal BBS gene. In addition, we found that Ccdc28b mutants showed behavioral phenotypes, similar to the deficits observed in rodent autism spectrum disorder (ASD) models. Thus, our results underscore a novel causal link between CCDC28B and behavioral phenotypes in mice.
Collapse
Affiliation(s)
- Matías Fabregat
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Sofía Niño-Rivero
- Departamento de Fisiología, Universidad de la República, Montevideo, Uruguay
| | - Sabrina Pose
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Magdalena Cárdenas-Rodríguez
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Mariana Bresque
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Metabolic Diseases and Aging Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Karina Hernández
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Victoria Prieto-Echagüe
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Geraldine Schlapp
- Laboratory Animal Biotechnology Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Martina Crispo
- Laboratory Animal Biotechnology Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Patricia Lagos
- Departamento de Fisiología, Universidad de la República, Montevideo, Uruguay
| | - Natalia Lago
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Carlos Escande
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Metabolic Diseases and Aging Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Florencia Irigoín
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- * E-mail: (FI); (JLB)
| | - Jose L. Badano
- Human Molecular Genetics Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
- INDICyO Institutional Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
- * E-mail: (FI); (JLB)
| |
Collapse
|
46
|
Sauer AK, Hagmeyer S, Grabrucker AM. Prenatal Zinc Deficient Mice as a Model for Autism Spectrum Disorders. Int J Mol Sci 2022; 23:ijms23116082. [PMID: 35682762 PMCID: PMC9181257 DOI: 10.3390/ijms23116082] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies have shown a clear association between early life zinc deficiency and Autism Spectrum Disorders (ASD). In line with this, mouse models have revealed prenatal zinc deficiency as a profound risk factor for neurobiological and behavioral abnormalities in the offspring reminiscent of ASD behavior. From these studies, a complex pathology emerges, with alterations in the gastrointestinal and immune system and synaptic signaling in the brain, as a major consequence of prenatal zinc deficiency. The features represent a critical link in a causal chain that leads to various neuronal dysfunctions and behavioral phenotypes observed in prenatal zinc deficient (PZD) mice and probably other mouse models for ASD. Given that the complete phenotype of PZD mice may be key to understanding how non-genetic factors can modify the clinical features and severity of autistic patients and explain the observed heterogeneity, here, we summarize published data on PZD mice. We critically review the emerging evidence that prenatal zinc deficiency is at the core of several environmental risk factors associated with ASD, being mechanistically linked to ASD-associated genetic factors. In addition, we highlight future directions and outstanding questions, including potential symptomatic, disease-modifying, and preventive treatment strategies.
Collapse
Affiliation(s)
- Ann Katrin Sauer
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
- Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
| | - Simone Hagmeyer
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
| | - Andreas M. Grabrucker
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland; (A.K.S.); (S.H.)
- Bernal Institute, University of Limerick, V94 T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94 T9PX Limerick, Ireland
- Correspondence: ; Tel.: +353-61-237756
| |
Collapse
|
47
|
Galizio A, Odum AL. Reinforced behavioral variability in the valproate rat model of autism spectrum disorder. J Exp Anal Behav 2022; 117:576-596. [PMID: 35467762 DOI: 10.1002/jeab.760] [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: 10/16/2021] [Revised: 02/28/2022] [Accepted: 03/29/2022] [Indexed: 11/05/2022]
Abstract
Individuals diagnosed with autism spectrum disorder (ASD) tend to display restricted, repetitive behaviors and deficits in social interaction. Rats exposed to valproate (VPA) in utero have been shown to model symptoms of ASD. In previous research, VPA rats engaged in less social interaction and more repetitive responding than controls. The purpose of the present study was to further investigate behavioral variability in the VPA rat model of ASD by testing VPA and control rats in a reinforced-behavioral-variability operant task. In this procedure, rats emitted sequences of lever presses, some of which produced food. During baseline, food was delivered probabilistically, and variability was not required. Next, rats were exposed either to a variability contingency, in which food was only delivered following sequences that differed sufficiently from previous sequences (i.e., variability required), or to a yoked contingency, in which variability was not required. We hypothesized that VPA rats would behave less variably than controls in this task. However, VPA and control rats responded similarly variably when variability was required. Furthermore, VPA rats behaved slightly more variably than controls during baseline and yoked conditions, when variability was not required. These findings contribute to the complex literature surrounding the VPA rat model of ASD.
Collapse
Affiliation(s)
- Ann Galizio
- Utah State University, Department of Psychology
| | - Amy L Odum
- Utah State University, Department of Psychology
| |
Collapse
|
48
|
Silverman JL, Thurm A, Ethridge SB, Soller MM, Petkova SP, Abel T, Bauman MD, Brodkin ES, Harony‐Nicolas H, Wöhr M, Halladay A. Reconsidering animal models used to study autism spectrum disorder: Current state and optimizing future. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12803. [PMID: 35285132 PMCID: PMC9189007 DOI: 10.1111/gbb.12803] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/15/2022]
Abstract
Neurodevelopmental disorders (NDDs), including autism spectrum disorder (ASD) and intellectual disability (ID), are pervasive, often lifelong disorders, lacking evidence-based interventions for core symptoms. With no established biological markers, diagnoses are defined by behavioral criteria. Thus, preclinical in vivo animal models of NDDs must be optimally utilized. For this reason, experts in the field of behavioral neuroscience convened a workshop with the goals of reviewing current behavioral studies, reports, and assessments in rodent models. Goals included: (a) identifying the maximal utility and limitations of behavior in animal models with construct validity; (b) providing recommendations for phenotyping animal models; and (c) guidelines on how in vivo models should be used and reported reliably and rigorously while acknowledging their limitations. We concluded by recommending minimal criteria for reporting in manuscripts going forward. The workshop elucidated a consensus of potential solutions to several problems, including revisiting claims made about animal model links to ASD (and related conditions). Specific conclusions included: mice (or other rodent or preclinical models) are models of the neurodevelopmental insult, not specifically any disorder (e.g., ASD); a model that perfectly recapitulates a disorder such as ASD is untenable; and greater attention needs be given to validation of behavioral testing methods, data analysis, and critical interpretation.
Collapse
Affiliation(s)
- Jill L. Silverman
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping ServiceNational Institute of Mental HealthBethesdaMarylandUSA
| | - Sarah B. Ethridge
- Neurodevelopmental and Behavioral Phenotyping ServiceNational Institute of Mental HealthBethesdaMarylandUSA
| | - Makayla M. Soller
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Stela P. Petkova
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Ted Abel
- Department of Neuroscience and PharmacologyIowa Neuroscience Institute, University of IowaIowa CityIowaUSA
| | - Melissa D. Bauman
- MIND Institute, Department of Psychiatry and Behavioral SciencesUniversity of California Davis School of MedicineSacramentoCaliforniaUSA
| | - Edward S. Brodkin
- Department of PsychiatryPerelman School of Medicine at the University of Pennsylvania, Translational Research LaboratoryPhiladelphiaPennsylvaniaUSA
| | - Hala Harony‐Nicolas
- Seaver Autism Center for Research and TreatmentIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Markus Wöhr
- Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological PsychologySocial and Affective Neuroscience Research Group, KU LeuvenLeuvenBelgium,Leuven Brain InstituteKU LeuvenLeuvenBelgium,Faculty of Psychology, Experimental and Biological Psychology, Behavioral NeurosciencePhilipps‐University of MarburgMarburgGermany,Center for Mind, Brain, and BehaviorPhilipps‐University of MarburgMarburgGermany
| | - Alycia Halladay
- Autism Science FoundationUSA,Department of Pharmacology and ToxicologyRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
49
|
Interspecific differences in sociability, social novelty preference, anxiety- and depression-like behaviors between Brandt's voles and C57BL/6J mice. Behav Processes 2022; 197:104624. [DOI: 10.1016/j.beproc.2022.104624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 11/24/2022]
|
50
|
Riet F, Mittelhaeuser C, Lux A, Bour R, Selloum M, Sorg T, Herault Y, Meziane H. Behavioral Testing Design for Evaluation of Cognitive Disabilities. Curr Protoc 2022; 2:e382. [PMID: 35195951 DOI: 10.1002/cpz1.382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intellectual disabilities (ID) constitute a class of human neurodevelopmental diseases and are a major medical and socioeconomic problem owing to their high incidence and enormous burden to the families of those affected. In the past three decades, mutant mouse technologies have provided powerful tools for elucidating the pathophysiological mechanisms underlying behavioral and developmental alterations related to IDs and for addressing new therapeutic strategies, and major progress has been made revealing previously unidentified genes involved in ID. However, the pathological hallmarks of IDs are very heterogeneous in regard to both the functional deficits observed and the severity of the phenotype, even within the same mutation types. For this reason, an appropriate experimental design is required to reduce the risk of false negatives and positives in animal functional genomic studies. This experimental design should address functions important to evaluate, tests, and the appropriate workflow. Here, we propose an extensive behavioral screen with detailed protocols, which was successfully used in a systematic mouse functional genomic approach to gain pathway-based insights into mechanisms leading to cognitive dysfunction in humans. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Assessment of circadian activity and ingestive behavior Basic Protocol 2: Assessment of neurological reflexes and motor abilities using the grip and rotarod tests Basic Protocol 3: Evaluation of anxiety-related behavior using the elevated plus maze Basic Protocol 4: Evaluation of recognition memory using the object recognition task Basic Protocol 5: Evaluation of social behavior using the social recognition test Basic Protocol 6: Evaluation of working memory using the Y-maze spontaneous alternation test Basic Protocol 7: Evaluation of emotional learning and memory using the fear conditioning test.
Collapse
Affiliation(s)
- Fabrice Riet
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Christophe Mittelhaeuser
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Aline Lux
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Raphael Bour
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Mohammed Selloum
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Tania Sorg
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France.,Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire, IGBMC, Illkirch, France
| | - Hamid Meziane
- Université de Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, Institut Clinique de la Souris (ICS), Illkirch, France
| |
Collapse
|