1
|
Wang Y, Ma L, Wang J, Liu N, Men W, Tan S, Gao JH, Qin S, He Y, Dong Q, Tao S. Emotional and behavioral problems accelerate hypothalamic development from childhood to adolescence: Findings from a longitudinal cohort study. J Affect Disord 2025; 371:124-133. [PMID: 39542114 DOI: 10.1016/j.jad.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Despite the pivotal role of the hypothalamus in regulating various physiological processes, our understanding of its developmental trajectory and subregional organization during childhood and adolescence remains limited, as well as how emotional and behavioral problems can impact hypothalamic development, potentially leading to neurodevelopmental disorders. METHODS This population-based longitudinal cohort study utilized data from a representative sample of 702 children, who were followed two to five times. Emotional and behavioral problems were assessed using the Strengths and Difficulties Questionnaire (SDQ). Linear mixed models were employed to delineate developmental trajectories and behavioral regulation. RESULTS Using an automated segmentation technique, we quantified the volumes and asymmetries of the hypothalamus and its subregions in a large longitudinal sample of 702 subjects aged 6-15 years with 1371 MRI scans, and mapped their developmental trajectories. Our findings indicate that while the anterior and posterior regions of the hypothalamus exhibit a tendency toward decline, the tubular region demonstrates a linear increase which is influenced by lateralization, sex, and intracranial volume. Furthermore, emotional and behavioral problems - particularly emotional symptoms and peer relationship problems - accelerate development in superior tubular and anterior-superior regions. CONCLUSIONS In this study, we initially delineated the developmental trajectories of the hypothalamus and its subregions from childhood to adolescence based on a longitudinal cohort study. Our findings revealed that the development of hypothalamus followed the pattern of "lateral early to medial late, and dorsomedial early to ventromedial late", and the emotional and behavioral problems accelerate hypothalamic development. This study provides preliminary evidence regarding the impact of emotional and behavioral problems on the dynamic development of the hypothalamus, offering a crucial foundation for future prevention and intervention strategies targeting cognitive and emotional behavioral problems.
Collapse
Affiliation(s)
- Yanpei Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Leilei Ma
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Jiali Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Ningyu Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Weiwei Men
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuping Tan
- Psychiatry Research Center, Beijing HuiLongGuan Hospital, Peking University, Beijing 100096, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shaozheng Qin
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Sha Tao
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China; IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
2
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: implications for brain development and autism. Transl Psychiatry 2024; 14:482. [PMID: 39632793 PMCID: PMC11618798 DOI: 10.1038/s41398-024-03179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood-stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNA-seq data obtained from E12.5 fetal mouse brains 3 hours after VPA administration to the pregnant dam revealed that VPA rapidly and significantly increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of 399 autism risk genes was significantly altered by VPA as was expression of 258 genes that have been reported to modulate fetal brain development but are not otherwise linked to autism. Expression of genes associated with intracellular signaling pathways, neurogenesis, and excitation-inhibition balance as well as synaptogenesis, neuronal fate determination, axon and dendritic development, neuroinflammation, circadian rhythms, and epigenetic modulation of gene expression was dysregulated by VPA. Notably, at least 40 genes that are known to regulate embryonic neurogenesis were dysregulated by VPA. The goal of this study was to identify mouse genes that are: (a) significantly up- or downregulated by VPA in the fetal brain and (b) associated with autism and/or known to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity and, consequently behavior, in the adult. The genes meeting these criteria provide potential targets for future hypothesis-driven studies to elucidate the proximal causes of errors in brain connectivity underlying neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Susan G Dorsey
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
| | - Evelina Mocci
- Department of Pain and Translational Symptom Science University of Maryland School of Nursing, Baltimore, MD, 21201, USA
- Institute for Genome Sciences University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Malcolm V Lane
- Translational Toxicology/Department of Epidemiology and Public Health University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Bruce K Krueger
- Departments of Physiology and Psychiatry University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
3
|
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
|
4
|
Lai W, Zhao Y, Chen Y, Dai Z, Chen R, Niu Y, Chen X, Chen S, Huang G, Shan Z, Zheng J, Hu Y, Chen Q, Gong S, Kang S, Guo H, Ma X, Song Y, Xia K, Wang J, Zhou L, So KF, Wang K, Qiu S, Zhang L, Chen J, Shi L. Autism patient-derived SHANK2B Y29X mutation affects the development of ALDH1A1 negative dopamine neuron. Mol Psychiatry 2024; 29:3180-3194. [PMID: 38704506 PMCID: PMC11449796 DOI: 10.1038/s41380-024-02578-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Autism spectrum disorder (ASD) encompasses a range of neurodevelopmental conditions. Different mutations on a single ASD gene contribute to heterogeneity of disease phenotypes, possibly due to functional diversity of generated isoforms. SHANK2, a causative gene in ASD, demonstrates this phenomenon, but there is a scarcity of tools for studying endogenous SHANK2 proteins in an isoform-specific manner. Here, we report a point mutation on SHANK2, which is found in a patient with autism, located on exon of the SHANK2B transcript variant (NM_133266.5), hereby SHANK2BY29X. This mutation results in an early stop codon and an aberrant splicing event that impacts SHANK2 transcript variants distinctly. Induced pluripotent stem cells (iPSCs) carrying this mutation, from the patient or isogenic editing, fail to differentiate into functional dopamine (DA) neurons, which can be rescued by genetic correction. Available SMART-Seq single-cell data from human midbrain reveals the abundance of SHANK2B transcript in the ALDH1A1 negative DA neurons. We then show that SHANK2BY29X mutation primarily affects SHANK2B expression and ALDH1A1 negative DA neurons in vitro during early neuronal developmental stage. Mice knocked in with the identical mutation exhibit autistic-like behavior, decreased occupancy of ALDH1A1 negative DA neurons and decreased dopamine release in ventral tegmental area (VTA). Our study provides novel insights on a SHANK2 mutation derived from autism patient and highlights SHANK2B significance in ALDH1A1 negative DA neuron.
Collapse
Affiliation(s)
- Wanjing Lai
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Yingying Zhao
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, 999077, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yalan Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Zhenzhu Dai
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Ruhai Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Yimei Niu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Xiaoxia Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Shuting Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Guanqun Huang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Ziyun Shan
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiajun Zheng
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Yu Hu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Qingpei Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Siyi Gong
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Sai Kang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Hui Guo
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Xiaokuang Ma
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 850004, USA
| | - Youqiang Song
- School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Kun Xia
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Jie Wang
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Shenfeng Qiu
- Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ, 850004, USA
| | - Li Zhang
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China.
| | - Jiekai Chen
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, 999077, China.
| | - Lingling Shi
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, China.
- Department of Psychiatry, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510632, China.
- Co-innovation Center of Neuro-regeneration, Nantong University, Nantong, Jiangsu, 226019, China.
- Department of Neurology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China.
| |
Collapse
|
5
|
Kreutzmann JC, Kahl E, Fendt M. Sex-specific modulation of safety learning in Shank2-deficient mice. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110973. [PMID: 38369099 DOI: 10.1016/j.pnpbp.2024.110973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterized by impaired perceptual processing and social communication, intellectual disabilities, and repetitive behaviors. Interestingly, while not a core symptom, anxiety disorders frequently co-occur in individuals with ASD and deficits in safety learning have been described in patients with anxiety-related disorders. Because genetic factors, such as SHANK deficiency (loss-of-function mutations), have been linked to ASD, the aim of the present study was to investigate whether Shank2 deficiency interferes with associative fear and safety signal learning. To first investigate trait anxiety, male and female Shank2-deficient mice were exposed to a light-dark box test. Mice were then submitted to a combination of contextual fear conditioning and single-cue safety conditioning. The results show that Shank2 deficiency increases trait anxiety but reduces contextual fear learning. In male but not female Shank2-deficient mice, reduced single-cued safety learning was observed. This safety learning deficit was not caused by altered anxiety levels, increased locomotor activity, or reduced contextual fear since these changes were also observed in female Shank2-deficient mice. Concluding, our data indicate that the observed safety learning deficits in Shank2-deficient male mice could contribute to the emotional symptoms observed in ASD and the high comorbidity with anxiety-related disorders.
Collapse
Affiliation(s)
- Judith C Kreutzmann
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany.; Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Evelyn Kahl
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Germany.; Center of Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Germany.
| |
Collapse
|
6
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: Implications for brain development and autism. RESEARCH SQUARE 2024:rs.3.rs-3684653. [PMID: 38260618 PMCID: PMC10802704 DOI: 10.21203/rs.3.rs-3684653/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood-stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNA-seq data obtained from E12.5 fetal mouse brains 3 hours after VPA administration to the pregnant dam revealed that VPA rapidly and significantly increased or decreased the expression of approximately 7,300 genes. No significant sex differences in VPA-induced gene expression were observed. Expression of 399 autism risk genes was significantly altered by VPA as was expression of 255 genes that have been reported to play fundamental roles in fetal brain development but are not otherwise linked to autism. Expression of genes associated with intracellular signaling pathways, neurogenesis, and excitation-inhibition balance as well as synaptogenesis, neuronal fate determination, axon and dendritic development, neuroinflammation, circadian rhythms, and epigenetic modulation of gene expression was dysregulated by VPA. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity and, consequently behavior, in the adult. The set of genes meeting these criteria provides potential targets for future hypothesis-driven studies to elucidate the proximal causes of errors in brain connectivity underlying neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Susan G. Dorsey
- Department of Pain and Translational Symptom Sciences, University of Maryland School of Nursing, Baltimore, MD 21201
| | - Evelina Mocci
- Department of Pain and Translational Symptom Sciences, University of Maryland School of Nursing, Baltimore, MD 21201
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Malcolm V. Lane
- Translational Toxicology/Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Bruce K. Krueger
- Departments of Physiology and Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
7
|
Bannerman DM, Barkus C, Eltokhi A. Behavioral Analysis of NMDAR Function in Rodents: Tests of Long-Term Spatial Memory. Methods Mol Biol 2024; 2799:107-138. [PMID: 38727905 DOI: 10.1007/978-1-0716-3830-9_7] [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] [Indexed: 07/03/2024]
Abstract
NMDAR-dependent forms of synaptic plasticity in brain regions like the hippocampus are widely believed to provide the neural substrate for long-term associative memory formation. However, the experimental data are equivocal at best and may suggest a more nuanced role for NMDARs and synaptic plasticity in memory. Much of the experimental data available comes from studies in genetically modified mice in which NMDAR subunits have been deleted or mutated in order to disrupt NMDAR function. Behavioral assessment of long-term memory in these mice has involved tests like the Morris watermaze and the radial arm maze. Here we describe these behavioral tests and some of the different testing protocols that can be used to assess memory performance. We discuss the importance of distinguishing selective effects on learning and memory processes from nonspecific effects on sensorimotor or motivational aspects of performance.
Collapse
Affiliation(s)
- David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford, UK.
| | - Chris Barkus
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Ahmed Eltokhi
- Department of Biomedical Sciences, School of Medicine, Mercer University, Columbus, GA, USA
| |
Collapse
|
8
|
Zhong F, Li W, Zhao C, Jin L, Lu X, Zhao Y, Pu J, Ge H. Basigin Deficiency Induces Spontaneous Polycystic Kidney in Mice. Hypertension 2024; 81:114-125. [PMID: 37955149 DOI: 10.1161/hypertensionaha.123.21486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Polycystic kidney disease is the most common hereditary kidney disorder with early and frequent hypertension symptoms. The mechanisms of cyst progression in polycystic kidney disease remain incompletely understood. METHODS Bsg (basigin) heterozygous and homozygous knockout mice were generated using cas9 system, and Bsg overexpression was achieved by adeno-associated virus serotype 9 injection. Renal morphology was investigated through histological and imaging analysis. Molecular analysis was performed through transcriptomic profiling and biochemical approaches. RESULTS Bsg-deficient mice exhibited significantly elevated arterial blood pressure. Further investigation demonstrated that Bsg deficiency triggers spontaneous cystic formation in mouse kidneys, which shares similar cyst pathological features and common transcriptional regulatory pathways with human polycystic kidney disease. Moreover, Bsg disruption promoted polycystin-1 ubiquitination and degradation, leading to activation of polycystic kidney disease associated cAMP and AMPK signaling pathways in Bsg knockout mouse kidneys. Finally, adeno-associated virus serotype 9 mediated Bsg reexpression reversed cystic progression in Bsg knockout mice in vivo, and Bsg overexpression inhibited the expansion of Madin-Darby canine kidney cysts in vitro. CONCLUSIONS Our findings show that Bsg deficiency leads to an early-onset spontaneous polycystic kidney phenotype, suggesting that dysregulated Bsg signaling may be a contributing factor in cystogenesis.
Collapse
Affiliation(s)
- Fangyuan Zhong
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Wenli Li
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Chenxu Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Lixing Jin
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xiyuan Lu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yichao Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Heng Ge
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|
9
|
Martinez-Lopez S, Angel-Gomis E, Sanchez-Ardid E, Pastor-Campos A, Picó J, Gomez-Hurtado I. The 3Rs in Experimental Liver Disease. Animals (Basel) 2023; 13:2357. [PMID: 37508134 PMCID: PMC10376896 DOI: 10.3390/ani13142357] [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: 06/14/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with cirrhosis present multiple physiological and immunological alterations that play a very important role in the development of clinically relevant secondary complications to the disease. Experimentation in animal models is essential to understand the pathogenesis of human diseases and, considering the high prevalence of liver disease worldwide, to understand the pathophysiology of disease progression and the molecular pathways involved, due to the complexity of the liver as an organ and its relationship with the rest of the organism. However, today there is a growing awareness about the sensitivity and suffering of animals, causing opposition to animal research among a minority in society and some scientists, but also about the attention to the welfare of laboratory animals since this has been built into regulations in most nations that conduct animal research. In 1959, Russell and Burch published the book "The Principles of Humane Experimental Technique", proposing that in those experiments where animals were necessary, everything possible should be done to try to replace them with non-sentient alternatives, to reduce to a minimum their number, and to refine experiments that are essential so that they caused the least amount of pain and distress. In this review, a comprehensive summary of the most widely used techniques to replace, reduce, and refine in experimental liver research is offered, to assess the advantages and weaknesses of available experimental liver disease models for researchers who are planning to perform animal studies in the near future.
Collapse
Affiliation(s)
- Sebastian Martinez-Lopez
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Enrique Angel-Gomis
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Elisabet Sanchez-Ardid
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Servicio de Patología Digestiva, Institut de Recerca IIB-Sant Pau, Hospital de Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Alberto Pastor-Campos
- Oficina de Investigación Responsable, Universidad Miguel Hernández, 03202 Elche, Spain
| | - Joanna Picó
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
| | - Isabel Gomez-Hurtado
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
| |
Collapse
|
10
|
Dorsey SG, Mocci E, Lane MV, Krueger BK. Rapid effects of valproic acid on the fetal brain transcriptome: Implications for brain development and autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538959. [PMID: 37205520 PMCID: PMC10187231 DOI: 10.1101/2023.05.01.538959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
There is an increased incidence of autism among the children of women who take the anti-epileptic, mood stabilizing drug, valproic acid (VPA) during pregnancy; moreover, exposure to VPA in utero causes autistic-like symptoms in rodents and non-human primates. Analysis of RNAseq data ob-tained from E12.5 fetal mouse brains 3 hours after VPA administration revealed that VPA significant-ly increased or decreased the expression of approximately 7,300 genes. No significant sex differ-ences in VPA-induced gene expression were observed. Expression of genes associated with neu-rodevelopmental disorders (NDDs) such as autism as well as neurogenesis, axon growth and syn-aptogenesis, GABAergic, glutaminergic and dopaminergic synaptic transmission, perineuronal nets, and circadian rhythms was dysregulated by VPA. Moreover, expression of 399 autism risk genes was significantly altered by VPA as was expression of 252 genes that have been reported to play fundamental roles in the development of the nervous system but are not otherwise linked to autism. The goal of this study was to identify mouse genes that are: (a) significantly up- or down-regulated by VPA in the fetal brain and (b) known to be associated with autism and/or to play a role in embryonic neurodevelopmental processes, perturbation of which has the potential to alter brain connectivity in the postnatal and adult brain. The set of genes meeting these criteria pro-vides potential targets for future hypothesis-driven approaches to elucidating the proximal underly-ing causes of defective brain connectivity in NDDs such as autism.
Collapse
|
11
|
Verderio P, Lecchi M, Ciniselli CM, Shishmani B, Apolone G, Manenti G. 3Rs Principle and Legislative Decrees to Achieve High Standard of Animal Research. Animals (Basel) 2023; 13:ani13020277. [PMID: 36670818 PMCID: PMC9854901 DOI: 10.3390/ani13020277] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Animal experimentation is a vast ecosystem that tries to make different issues such as legislative, ethical and scientific coexist. Research in animal experimentation has made many strides thanks to the 3Rs principle and the attached legislative decrees, but for this very reason, it needs to be evenly implemented both among the countries that have adhered to the decrees and among the team members who design and execute the experimental practice. In this article, we emphasize the importance of the 3Rs principle's application, with a particular focus on the concept of Reduction and related key aspects that can best be handled with the contribution of experts from different fields.
Collapse
Affiliation(s)
- Paolo Verderio
- Unit of Bioinformatics and Biostatistics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
- Correspondence: ; Tel.: +39-02-2390-3201
| | - Mara Lecchi
- Unit of Bioinformatics and Biostatistics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Chiara Maura Ciniselli
- Unit of Bioinformatics and Biostatistics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Bjorn Shishmani
- Unit of Bioinformatics and Biostatistics, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Giacomo Manenti
- Unit of Animal Health and Welfare, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| |
Collapse
|
12
|
Kim Y, Ko TH, Jin C, Zhang Y, Kang HR, Ma R, Li H, Choi JI, Han K. The emerging roles of Shank3 in cardiac function and dysfunction. Front Cell Dev Biol 2023; 11:1191369. [PMID: 37187620 PMCID: PMC10175600 DOI: 10.3389/fcell.2023.1191369] [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: 03/22/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Shank3 is a member of the Shank family proteins (Shank1-3), which are abundantly present in the postsynaptic density (PSD) of neuronal excitatory synapses. As a core scaffold in the PSD, Shank3 plays a critical role in organizing the macromolecular complex, ensuring proper synaptic development and function. Clinically, various mutations of the SHANK3 gene are causally associated with brain disorders such as autism spectrum disorders and schizophrenia. However, recent in vitro and in vivo functional studies and expression profiling in various tissues and cell types suggest that Shank3 also plays a role in cardiac function and dysfunction. For example, Shank3 interacts with phospholipase Cβ1b (PLCβ1b) in cardiomyocytes, regulating its localization to the sarcolemma and its role in mediating Gq-induced signaling. In addition, changes in cardiac morphology and function associated with myocardial infarction and aging have been investigated in a few Shank3 mutant mouse models. This review highlights these results and potential underlying mechanisms, and predicts additional molecular functions of Shank3 based on its protein interactors in the PSD, which are also highly expressed and function in the heart. Finally, we provide perspectives and possible directions for future studies to better understand the roles of Shank3 in the heart.
Collapse
Affiliation(s)
- Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Republic of Korea
| | - Chunmei Jin
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Republic of Korea
| | - Yinhua Zhang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyae Rim Kang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Huiling Li
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jong-Il Choi, ; Kihoon Han,
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- *Correspondence: Jong-Il Choi, ; Kihoon Han,
| |
Collapse
|
13
|
Yoo YE, Yoo T, Kang H, Kim E. Brain region and gene dosage-differential transcriptomic changes in Shank2-mutant mice. Front Mol Neurosci 2022; 15:977305. [PMID: 36311025 PMCID: PMC9612946 DOI: 10.3389/fnmol.2022.977305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/16/2022] [Indexed: 12/20/2022] Open
Abstract
Shank2 is an abundant excitatory postsynaptic scaffolding protein that has been implicated in various neurodevelopmental and psychiatric disorders, including autism spectrum disorder (ASD), intellectual disability, attention-deficit/hyperactivity disorder, and schizophrenia. Shank2-mutant mice show ASD-like behavioral deficits and altered synaptic and neuronal functions, but little is known about how different brain regions and gene dosages affect the transcriptomic phenotypes of these mice. Here, we performed RNA-Seq-based transcriptomic analyses of the prefrontal cortex, hippocampus, and striatum in adult Shank2 heterozygous (HT)- and homozygous (HM)-mutant mice lacking exons 6–7. The prefrontal cortical, hippocampal, and striatal regions showed distinct transcriptomic patterns associated with synapse, ribosome, mitochondria, spliceosome, and extracellular matrix (ECM). The three brain regions were also distinct in the expression of ASD-related and ASD-risk genes. These differential patterns were stronger in the prefrontal cortex where the HT transcriptome displayed increased synaptic gene expression and reverse-ASD patterns whereas the HM transcriptome showed decreased synaptic gene expression and ASD-like patterns. These results suggest brain region- and gene dosage-differential transcriptomic changes in Shank2-mutant mice.
Collapse
Affiliation(s)
- Ye-Eun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- *Correspondence: Eunjoon Kim,
| |
Collapse
|
14
|
Yoo T, Yoo YE, Kang H, Kim E. Age, brain region, and gene dosage-differential transcriptomic changes in Shank3-mutant mice. Front Mol Neurosci 2022; 15:1017512. [PMID: 36311023 PMCID: PMC9597470 DOI: 10.3389/fnmol.2022.1017512] [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: 08/12/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Shank3 is an abundant excitatory postsynaptic scaffolding protein implicated in various neurodevelopmental disorders, including autism spectrum disorder (ASD), Phelan-McDermid syndrome, intellectual disability, and schizophrenia. Shank3-mutant mice show various molecular, synaptic, and behavioral deficits, but little is known about how transcriptomic phenotypes vary across different ages, brain regions, and gene dosages. Here, we report transcriptomic patterns in the forebrains of juvenile and adult homozygous Shank3-mutant mice that lack exons 14-16 and also the prefrontal, hippocampal, and striatal transcriptomes in adult heterozygous and homozygous Shank3-mutant mice. The juvenile and adult mutant transcriptomes show patterns opposite from and similar to those observed in ASD (termed reverse-ASD and ASD-like patterns), respectively. The juvenile transcriptomic changes accompany synaptic upregulations and ribosomal and mitochondrial downregulations, whereas the adult transcriptome show opposite changes. The prefrontal, hippocampal, and striatal transcriptomes show differential changes in ASD-related gene expressions and biological functions associated with synapse, ribosome, mitochondria, and spliceosome. These patterns also differ across heterozygous and homozygous Shank3-mutant mice. These results suggest age, brain region, and gene dosage-differential transcriptomic changes in Shank3-mutant mice.
Collapse
Affiliation(s)
- Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Ye-Eun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| |
Collapse
|
15
|
Yun M, Kim E, Jung MW. Enhanced fear limits behavioral flexibility in Shank2-deficient mice. Mol Autism 2022; 13:40. [PMID: 36192805 PMCID: PMC9531513 DOI: 10.1186/s13229-022-00518-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background A core symptom of autism spectrum disorder (ASD) is repetitive and restrictive patterns of behavior. Cognitive inflexibility has been proposed as a potential basis for these symptoms of ASD. More generally, behavioral inflexibility has been proposed to underlie repetitive and restrictive behavior in ASD. Here, we investigated whether and how behavioral flexibility is compromised in a widely used animal model of ASD.
Methods We compared the behavioral performance of Shank2-knockout mice and wild-type littermates in reversal learning employing a probabilistic classical trace conditioning paradigm. A conditioned stimulus (odor) was paired with an unconditioned appetitive (water, 6 µl) or aversive (air puff) stimulus in a probabilistic manner. We also compared air puff-induced eye closure responses of Shank2-knockout and wild-type mice. Results Male, but not female, Shank2-knockout mice showed impaired reversal learning when the expected outcomes consisted of a water reward and a strong air puff. Moreover, male, but not female, Shank2-knockout mice showed stronger anticipatory eye closure responses to the air puff compared to wild-type littermates, raising the possibility that the impairment might reflect enhanced fear. In support of this contention, male Shank2-knockout mice showed intact reversal learning when the strong air puff was replaced with a mild air puff and when the expected outcomes consisted of only rewards. Limitations We examined behavioral flexibility in one behavioral task (reversal learning in a probabilistic classical trace conditioning paradigm) using one ASD mouse model (Shank2-knockout mice). Thus, future work is needed to clarify the extent to which our findings (that enhanced fear limits behavioral flexibility in ASD) can explain the behavioral inflexibility associated with ASD. Also, we examined only the relationship between fear and behavioral flexibility, leaving open the question of whether abnormalities in processes other than fear contribute to behavioral inflexibility in ASD. Finally, the neurobiological mechanisms linking Shank2-knockout and enhanced fear remain to be elucidated. Conclusions Our results indicate that enhanced fear suppresses reversal learning in the presence of an intact capability to learn cue-outcome contingency changes in Shank2-knockout mice. Our findings suggest that behavioral flexibility might be seriously limited by abnormal emotional responses in ASD. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-022-00518-1.
Collapse
Affiliation(s)
- Miru Yun
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, 34141, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea. .,Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, 34141, Korea.
| | - Min Whan Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea. .,Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, 34141, Korea.
| |
Collapse
|
16
|
Purushotham SS, Reddy NMN, D'Souza MN, Choudhury NR, Ganguly A, Gopalakrishna N, Muddashetty R, Clement JP. A perspective on molecular signalling dysfunction, its clinical relevance and therapeutics in autism spectrum disorder. Exp Brain Res 2022; 240:2525-2567. [PMID: 36063192 DOI: 10.1007/s00221-022-06448-x] [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: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022]
Abstract
Intellectual disability (ID) and autism spectrum disorder (ASD) are neurodevelopmental disorders that have become a primary clinical and social concern, with a prevalence of 2-3% in the population. Neuronal function and behaviour undergo significant malleability during the critical period of development that is found to be impaired in ID/ASD. Human genome sequencing studies have revealed many genetic variations associated with ASD/ID that are further verified by many approaches, including many mouse and other models. These models have facilitated the identification of fundamental mechanisms underlying the pathogenesis of ASD/ID, and several studies have proposed converging molecular pathways in ASD/ID. However, linking the mechanisms of the pathogenic genes and their molecular characteristics that lead to ID/ASD has progressed slowly, hampering the development of potential therapeutic strategies. This review discusses the possibility of recognising the common molecular causes for most ASD/ID based on studies from the available models that may enable a better therapeutic strategy to treat ID/ASD. We also reviewed the potential biomarkers to detect ASD/ID at early stages that may aid in diagnosis and initiating medical treatment, the concerns with drug failure in clinical trials, and developing therapeutic strategies that can be applied beyond a particular mutation associated with ASD/ID.
Collapse
Affiliation(s)
- Sushmitha S Purushotham
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Neeharika M N Reddy
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Michelle Ninochka D'Souza
- Centre for Brain Research, Indian Institute of Science Campus, CV Raman Avenue, Bangalore, 560 012, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - Nilpawan Roy Choudhury
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Anusa Ganguly
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Niharika Gopalakrishna
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India
| | - Ravi Muddashetty
- Centre for Brain Research, Indian Institute of Science Campus, CV Raman Avenue, Bangalore, 560 012, India.,The University of Trans-Disciplinary Health Sciences and Technology (TDU), Bangalore, 560064, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, 560064, India.
| |
Collapse
|
17
|
Eltokhi A, Sommer IE. A Reciprocal Link Between Gut Microbiota, Inflammation and Depression: A Place for Probiotics? Front Neurosci 2022; 16:852506. [PMID: 35546876 PMCID: PMC9081810 DOI: 10.3389/fnins.2022.852506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
Depression is a severe mental disorder that places a significant economic burden on public health. The reciprocal link between the trillions of bacteria in the gut, the microbiota, and depression is a controversial topic in neuroscience research and has drawn the attention of public interest and press coverage in recent years. Mounting pieces of evidence shed light on the role of the gut microbiota in depression, which is suggested to involve immune, endocrine, and neural pathways that are the main components of the microbiota-gut-brain axis. The gut microbiota play major roles in brain development and physiology and ultimately behavior. The bidirectional communication between the gut microbiota and brain function has been extensively explored in animal models of depression and clinical research in humans. Certain gut microbiota strains have been associated with the pathophysiology of depression. Therefore, oral intake of probiotics, the beneficial living bacteria and yeast, may represent a therapeutic approach for depression treatment. In this review, we summarize the findings describing the possible links between the gut microbiota and depression, focusing mainly on the inflammatory markers and sex hormones. By discussing preclinical and clinical studies on probiotics as a supplementary therapy for depression, we suggest that probiotics may be beneficial in alleviating depressive symptoms, possibly through immune modulation. Still, further comprehensive studies are required to draw a more solid conclusion regarding the efficacy of probiotics and their mechanisms of action.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Iris E Sommer
- Department of Biomedical Sciences of Cells & Systems, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
18
|
Pitzer C, Kurpiers B, Eltokhi A. Sex Differences in Depression-Like Behaviors in Adult Mice Depend on Endophenotype and Strain. Front Behav Neurosci 2022; 16:838122. [PMID: 35368297 PMCID: PMC8969904 DOI: 10.3389/fnbeh.2022.838122] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/14/2022] [Indexed: 12/27/2022] Open
Abstract
Depression affects women nearly twice as frequently as men. In contrast, rodent models of depression have shown inconsistent results regarding sex bias, often reporting more depression-like behaviors in males. This sex discrepancy in rodents modeling depression may rely on differences in the baseline activity of males and females in depression-related behavioral tests. We previously showed that the baseline despair and anhedonia behaviors, major endophenotypes of depression, are not sex biased in young adolescent wild-type mice of C57BL/6N, DBA/2, and FVB/N strains. Since the prevalence of depression in women peaks in their reproductive years, we here investigated sex differences of the baseline depression-like behaviors in adult mice using these three strains. Similar to the results in young mice, no difference was found between adult male and female mice in behavioral tests measuring despair in both tail suspension and forced swim tests, and anhedonia in the sucrose preference test. We then extended our study and tested apathy, another endophenotype of depression, using the splash test. Adult male and female mice showed significantly different results in the baseline apathy-like behaviors depending on the investigated strain. This study dissects the complex sex effects of different depression endophenotypes, stresses the importance of considering strain, and puts forward a hypothesis of the inconsistency of results between different laboratories investigating rodent models of depression.
Collapse
Affiliation(s)
- Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
- *Correspondence: Claudia Pitzer,
| | - Barbara Kurpiers
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, WA, United States
- Ahmed Eltokhi,
| |
Collapse
|
19
|
A Novel Automated Approach for Improving Standardization of the Marble Burying Test Enables Quantification of Burying Bouts and Activity Characteristics. eNeuro 2022; 9:ENEURO.0446-21.2022. [PMID: 35288451 PMCID: PMC8982638 DOI: 10.1523/eneuro.0446-21.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/10/2022] [Accepted: 02/22/2022] [Indexed: 11/21/2022] Open
Abstract
The marble burying test is a commonly used paradigm to describe phenotypes in mouse models of neurodevelopmental and psychiatric disorders. The current methodological approach relies predominantly on reporting the number of buried marbles at the end of the test. By measuring the proxy of the behavior (buried marbles), many important characteristics regarding the temporal aspect of this assay are lost. Here, we introduce a novel, automated method to quantify mouse behavior during the marble burying test with the focus on the burying bouts and movement dynamics. Using open-source software packages, we trained a supervised machine learning algorithm (the "classifier") to distinguish burying behavior in freely moving mice. In order to confirm the classifier's accuracy and characterize burying events in high detail, we performed the marble burying test in three mouse models: Ube3am-/p+ [Angelman syndrome (AS) model], Shank2 -/- (autism model), and Sapap3 -/- [obsessive-compulsive disorder (OCD) model] mice. The classifier scored burying behavior accurately and consistent with the previously reported phenotype of the Ube3am-/p+ mice, which showed decreased levels of burying compared with controls. Shank2 -/- mice showed a similar pattern of decreased burying behavior, which was not found in Sapap3 -/- mice. Tracking mouse behavior throughout the test revealed hypoactivity in Ube3am-/p+ and hyperactivity in the Shank2 -/- mice, indicating that mouse activity is unrelated to burying behavior. Reducing activity with midazolam in Shank2 -/- mice did not alter the burying behavior. Together, we demonstrate that our classifier is an accurate method for the analysis of the marble burying test, providing more information than currently used methods.
Collapse
|
20
|
Al Dera H. Cellular and molecular mechanisms underlying autism spectrum disorders and associated comorbidities: A pathophysiological review. Biomed Pharmacother 2022; 148:112688. [PMID: 35149383 DOI: 10.1016/j.biopha.2022.112688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders that develop in early life due to interaction between several genetic and environmental factors and lead to alterations in brain function and structure. During the last decades, several mechanisms have been placed to explain the pathogenesis of autism. Unfortunately, these are reported in several studies and reviews which make it difficult to follow by the reader. In addition, some recent molecular mechanisms related to ASD have been unrevealed. This paper revises and highlights the major common molecular mechanisms responsible for the clinical symptoms seen in people with ASD, including the roles of common genetic factors and disorders, neuroinflammation, GABAergic signaling, and alterations in Ca+2 signaling. Besides, it covers the major molecular mechanisms and signaling pathways involved in initiating the epileptic seizure, including the alterations in the GABAergic and glutamate signaling, vitamin and mineral deficiency, disorders of metabolism, and autoimmunity. Finally, this review also discusses sleep disorder patterns and the molecular mechanisms underlying them.
Collapse
Affiliation(s)
- Hussain Al Dera
- Department of Basic Medical Sciences, College of Medicine at King Saud, Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia; King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia.
| |
Collapse
|
21
|
Martin LJ, Poulson SJ, Mannan E, Sivaselvachandran S, Cho M, Setak F, Chan C. Altered nociceptive behavior and emotional contagion of pain in mouse models of autism. GENES, BRAIN, AND BEHAVIOR 2021; 21:e12778. [PMID: 34812576 PMCID: PMC9744566 DOI: 10.1111/gbb.12778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/26/2021] [Accepted: 11/04/2021] [Indexed: 12/17/2022]
Abstract
Individuals with autism spectrum disorder (ASD) have altered sensory processing but may ineffectively communicate their experiences. Here, we used a battery of nociceptive behavioral tests to assess sensory alterations in two commonly used mouse models of ASD, BTBR T+ Itpr3tf /J (BTBR), and fragile-X mental retardation-1 knockout (Fmr1-KO) mice. We also asked whether emotional contagion, a primitive form of empathy, was altered in BTBR and Fmr1 KO mice when experiencing pain with a social partner. BTBR mice demonstrated mixed nociceptive responses with hyporesponsivity to mechanical/thermal stimuli and intraplantar injections of formalin and capsaicin while displaying hypersensitivity on the acetic acid test. Fmr1-KO mice were hyposensitive to mechanical stimuli and intraplantar injections of capsaicin and formalin. BTBR and Fmr1-KO mice developed significantly less mechanical allodynia following intraplantar injections of complete Freund's adjuvant, while BTBR mice developed slightly more thermal hyperalgesia. Finally, as measured by the formalin and acetic acid writhing tests, BTBR and Fmr1-KO mice did not show emotional contagion of pain. In sum, our findings indicate that depending on the sensation, pain responses may be mixed, which reflects findings in ASD individuals.
Collapse
Affiliation(s)
- Loren J. Martin
- Department of PsychologyUniversity of Toronto MississaugaMississaugaOntarioCanada,Cell and Systems BiologyUniversity of TorontoTorontoOntarioCanada
| | - Sandra J. Poulson
- Department of PsychologyUniversity of Toronto MississaugaMississaugaOntarioCanada
| | - Emma Mannan
- Cell and Systems BiologyUniversity of TorontoTorontoOntarioCanada
| | | | - Moonjeong Cho
- Department of PsychologyUniversity of Toronto MississaugaMississaugaOntarioCanada
| | - Fatima Setak
- Department of PsychologyUniversity of Toronto MississaugaMississaugaOntarioCanada
| | - Claire Chan
- Cell and Systems BiologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
22
|
Abdelrahman AH, Eid OM, Ibrahim MH, Abd El-Fattah SN, Eid MM, Meguid NA. Evaluation of circulating miRNAs and mRNAs expression patterns in autism spectrum disorder. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00202-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Autism spectrum disorder is a condition related to brain development that affects a person’s perception and socialization, resulting in problems in social interaction and communication. It has no single known cause, yet several different genes appear to be involved in autism. As a genetically complex disease, dysregulation of miRNA expression and miRNA–mRNA interactions might be a feature of autism spectrum disorder. The aim of the current study was to investigate the expression profile of circulating miRNA-128, miRNA-7 and SHANK gene family in ASD patients and to assess the possible influence of miRNA-128 and miRNA-7 on SHANK genes, which might provide an insight into the pathogenic mechanisms of ASD and introduce noninvasive molecular biomarkers for the disease diagnosis and prognosis. Quantitative real-time PCR technique was employed to determine expression levels of miRNA-128, miRNA-7 and SHANK gene family in blood samples of 40 autistic cases along with 30 age- and sex-matched normal volunteer subjects.
Results
Our study revealed a statistical significant upregulation of miRNA-128 expression levels in ASD cases compared to controls (p value < 0.001). A statistical significant difference in SHANK-3 expression was encountered on comparing cases to controls (p value < 0.001). However, miRNA-7 expression showed no significant difference between the studied groups.
Conclusions
MiRNA-128 and SHANK-3 gene are emerging players in the field of ASD. They are promising candidates as noninvasive biomarkers in autism. Future studies are needed to emphasize their pivotal role.
Collapse
|
23
|
Eltokhi A, Kurpiers B, Pitzer C. Baseline Depression-Like Behaviors in Wild-Type Adolescent Mice Are Strain and Age but Not Sex Dependent. Front Behav Neurosci 2021; 15:759574. [PMID: 34690714 PMCID: PMC8529326 DOI: 10.3389/fnbeh.2021.759574] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Depression is a major neuropsychiatric disorder, decreasing the ability of hundreds of millions of individuals worldwide to function in social, academic, and employment settings. Beyond the alarming public health problem, depression leads to morbidity across the entire age including adolescence and adulthood. Modeling depression in rodents has been used to understand the pathophysiological mechanisms behind this disorder and create new therapeutics. Although women are two times more likely to be diagnosed with depression compared to men, behavioral experiments on rodent models of depression are mainly performed in males based on the assumption that the estrous cycles in females may affect the behavioral outcome and cause an increase in the intrinsic variability compared to males. Still, the inclusion of female rodents in the behavioral analysis is mandatory to establish the origin of sex bias in depression. Here, we investigated the baseline depression-like behaviors in male and female mice of three adolescent wild-type inbred strains, C57BL/6N, DBA/2, and FVB/N, that are typically used as background strains for mouse models of neuropsychiatric disorders. Our experiments, performed at two different developmental stages during adolescence (P22-P26 and P32-P36), revealed strain but no sex differences in a set of depression-related tests, including tail suspension, sucrose preference and forced swim tests. Additionally, the 10-day interval during this sensitive period uncovered a strong impact on the behavioral outcome of C57BL/6N and FVB/N mice, highlighting a significant effect of maturation on behavioral patterns. Since anxiety-related behavioral tests are often performed together with depression tests in mouse models of neuropsychiatric disorders, we extended our study and included hyponeophagia as an anxiety test. Consistent with a previous study revealing sex differences in other anxiety tests in adolescent mice, male and females mice behaved differently in the hyponeophagia test at P27. Our study gives insight into the behavioral experiments assessing depression and stresses the importance of considering strain, age and sex when evaluating neuropsychiatric-like traits in rodent models.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Barbara Kurpiers
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
24
|
Vallés AS, Barrantes FJ. Dendritic spine membrane proteome and its alterations in autistic spectrum disorder. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:435-474. [PMID: 35034726 DOI: 10.1016/bs.apcsb.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dendritic spines are small protrusions stemming from the dendritic shaft that constitute the primary specialization for receiving and processing excitatory neurotransmission in brain synapses. The disruption of dendritic spine function in several neurological and neuropsychiatric diseases leads to severe information-processing deficits with impairments in neuronal connectivity and plasticity. Spine dysregulation is usually accompanied by morphological alterations to spine shape, size and/or number that may occur at early pathophysiological stages and not necessarily be reflected in clinical manifestations. Autism spectrum disorder (ASD) is one such group of diseases involving changes in neuronal connectivity and abnormal morphology of dendritic spines on postsynaptic neurons. These alterations at the subcellular level correlate with molecular changes in the spine proteome, with alterations in the copy number, topography, or in severe cases in the phenotype of the molecular components, predominantly of those proteins involved in spine recognition and adhesion, reflected in abnormally short lifetimes of the synapse and compensatory increases in synaptic connections. Since cholinergic neurotransmission participates in the regulation of cognitive function (attention, memory, learning processes, cognitive flexibility, social interactions) brain acetylcholine receptors are likely to play an important role in the dysfunctional synapses in ASD, either directly or indirectly via the modulatory functions exerted on other neurotransmitter receptor proteins and spine-resident proteins.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca, Argentina
| | - Francisco J Barrantes
- Instituto de Investigaciones Biomédicas (BIOMED), UCA-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
25
|
Li S, DeLisi LE, McDonough SI. Rare germline variants in individuals diagnosed with schizophrenia within multiplex families. Psychiatry Res 2021; 303:114038. [PMID: 34174581 DOI: 10.1016/j.psychres.2021.114038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/27/2021] [Indexed: 12/30/2022]
Abstract
An extensive catalog of common and rare genetic variants contributes to overall risk for schizophrenia and related disorders. As a complement to population genetics efforts, here we present whole genome sequences of multiple affected probands within individual families to search for possible high penetrance driver variants. From a total of 15 families diagnostically evaluated by a single research psychiatrist, we performed whole genome sequencing of a total of 61 affected individuals, called SNPs, indels, and copy number variants, and compared to reference genomes. In fourteen out of fifteen families, the schizophrenia polygenic risk score for each proband was within the control range defined by the Thousand Genomes cohort. In six families, each affected member carried a very rare or private, predicted-damaging, variant in at least one gene. Among these genes, variants in LRP1 and TENM2 suggest these are candidate disease-related genes when taken into context with existing population genetic studies and biological information. Results add to the number of pedigree sequences reported, suggest pathways for the investigation of biological mechanisms, and are consistent with the overall accumulating evidence that very rare damaging variants contribute to the heritability of schizophrenia.
Collapse
Affiliation(s)
| | - Lynn E DeLisi
- Cambridge Health Alliance, Cambridge, MA, United States; Harvard Medical School, Boston, MA, United States
| | | |
Collapse
|
26
|
Dalal JS, Winden KD, Salussolia CL, Sundberg M, Singh A, Pham TT, Zhou P, Pu WT, Miller MT, Sahin M. Loss of Tsc1 in cerebellar Purkinje cells induces transcriptional and translation changes in FMRP target transcripts. eLife 2021; 10:e67399. [PMID: 34259631 PMCID: PMC8279760 DOI: 10.7554/elife.67399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is a genetic disorder that is associated with multiple neurological manifestations. Previously, we demonstrated that Tsc1 loss in cerebellar Purkinje cells (PCs) can cause altered social behavior in mice. Here, we performed detailed transcriptional and translational analyses of Tsc1-deficient PCs to understand the molecular alterations in these cells. We found that target transcripts of the Fragile X Mental Retardation Protein (FMRP) are reduced in mutant PCs with evidence of increased degradation. Surprisingly, we observed unchanged ribosomal binding for many of these genes using translating ribosome affinity purification. Finally, we found that multiple FMRP targets, including SHANK2, were reduced, suggesting that compensatory increases in ribosomal binding efficiency may be unable to overcome reduced transcript levels. These data further implicate dysfunction of FMRP and its targets in TSC and suggest that treatments aimed at restoring the function of these pathways may be beneficial.
Collapse
Affiliation(s)
- Jasbir Singh Dalal
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Kirby Neurobiology Center, Boston Children’s HospitalBostonUnited States
| | - Kellen Diamond Winden
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Kirby Neurobiology Center, Boston Children’s HospitalBostonUnited States
| | - Catherine Lourdes Salussolia
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Kirby Neurobiology Center, Boston Children’s HospitalBostonUnited States
| | - Maria Sundberg
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Kirby Neurobiology Center, Boston Children’s HospitalBostonUnited States
| | - Achint Singh
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Kirby Neurobiology Center, Boston Children’s HospitalBostonUnited States
| | - Truc Thanh Pham
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Kirby Neurobiology Center, Boston Children’s HospitalBostonUnited States
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children’s HospitalBostonUnited States
| | - William T Pu
- Department of Cardiology, Boston Children’s HospitalBostonUnited States
- Harvard Medical SchoolBostonUnited States
| | - Meghan T Miller
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center BaselBaselSwitzerland
| | - Mustafa Sahin
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Kirby Neurobiology Center, Boston Children’s HospitalBostonUnited States
- Harvard Medical SchoolBostonUnited States
| |
Collapse
|
27
|
Saadatmand F, Gurdziel K, Jackson L, Kwabi-Addo B, Ruden DM. DNA methylation and exposure to violence among African American young adult males. Brain Behav Immun Health 2021; 14:100247. [PMID: 34589758 PMCID: PMC8474503 DOI: 10.1016/j.bbih.2021.100247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/18/2021] [Accepted: 03/22/2021] [Indexed: 02/02/2023] Open
Abstract
Exposure to violence (ETV) has been linked to epigenomics mechanisms such as DNA methylation (DNAm). We used epigenetic profiling of blood collected from 32 African American young adult males who lived in Washington DC to determine if changes in DNAm at CpG sites affiliated with nervous and immune system were associated with exposure to violence. Pathway analysis of differentially methylated regions comparing high and low ETV groups revealed an enrichment of gene sets annotated to nervous system and immune ontologies. Many of these genes are known to interact with each other which suggests DNAm alters gene function in the nervous and immune system in response to ETV. Using data from a unique age group, young African American adult males, we provide evidence that lifetime ETV could impact DNA methylation in genes impacted at Central Nervous System and Immune Function sites. METHOD Methylation analysis was performed on DNA collected from the blood of participants classified with either high or low lifetime ETV. Illumina®MethylationEPIC Beadchips (~850k CpG sites) were processed on the iScan System to examine whole-genome methylation differences. Differentially methylated CpG-sites between high (n = 19) and low (n = 13) groups were identified using linear regression with violence and substance abuse as model covariates. Gene ontology analysis was used to identify enrichment categories from probes annotated to the nearest gene. RESULTS A total of 595 probes (279 hypermethylated; 316 hypomethylated) annotated to 383 genes were considered differentially methylated in association with ETV. Males with high ETV showed elevated methylation in several signaling pathways but were most impacted at Central Nervous System and Immune Function affiliated sites. Eight candidate genes were identified that play important biological roles in stress response to violence with HDAC4 (10%), NR4A3 (11%), NR4A2 (12%), DSCAML1(12%), and ELAVL3 (13%) exhibiting higher levels in the low ETV group and DLGAP1 (10%), SHANK2 (10%), and NRG1(11%) having increased methylation in the high ETV group. These findings suggest that individuals subjected to high ETV may be at risk for poor health outcomes that have not been reported previously.
Collapse
Affiliation(s)
- Forough Saadatmand
- Department of Pediatrics, College of Medicine, Howard University, Washington, DC, USA
| | - Katherine Gurdziel
- Office of the Vice President of Research, Wayne State University, Detroit, MI, USA
| | - Latifa Jackson
- Department of Pediatrics, College of Medicine, Howard University, Washington, DC, USA
- W. Montague Cobb Research Laboratory, College of Arts and Sciences, Howard University, Washington, DC, USA
| | - Bernard Kwabi-Addo
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, Washington, DC, USA
| | - Douglas M. Ruden
- Department of Ob/Gyn, CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
- Institutes for Environmental Health Science, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
28
|
Steubler V, Erdinger S, Back MK, Ludewig S, Fässler D, Richter M, Han K, Slomianka L, Amrein I, von Engelhardt J, Wolfer DP, Korte M, Müller UC. Loss of all three APP family members during development impairs synaptic function and plasticity, disrupts learning, and causes an autism-like phenotype. EMBO J 2021; 40:e107471. [PMID: 34008862 PMCID: PMC8204861 DOI: 10.15252/embj.2020107471] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
The key role of APP for Alzheimer pathogenesis is well established. However, perinatal lethality of germline knockout mice lacking the entire APP family has so far precluded the analysis of its physiological functions for the developing and adult brain. Here, we generated conditional APP/APLP1/APLP2 triple KO (cTKO) mice lacking the APP family in excitatory forebrain neurons from embryonic day 11.5 onwards. NexCre cTKO mice showed altered brain morphology with agenesis of the corpus callosum and disrupted hippocampal lamination. Further, NexCre cTKOs revealed reduced basal synaptic transmission and drastically reduced long-term potentiation that was associated with reduced dendritic length and reduced spine density of pyramidal cells. With regard to behavior, lack of the APP family leads not only to severe impairments in a panel of tests for learning and memory, but also to an autism-like phenotype including repetitive rearing and climbing, impaired social communication, and deficits in social interaction. Together, our study identifies essential functions of the APP family during development, for normal hippocampal function and circuits important for learning and social behavior.
Collapse
Affiliation(s)
- Vicky Steubler
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Susanne Erdinger
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Michaela K Back
- Institute of PathophysiologyFocus Program Translational Neuroscience (FTN)University Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Susann Ludewig
- Division of Cellular NeurobiologyZoological Institute, TU BraunschweigBraunschweigGermany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration GroupBraunschweigGermany
| | - Dominique Fässler
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Max Richter
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Kang Han
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Lutz Slomianka
- Institute of Anatomy and Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Irmgard Amrein
- Institute of Anatomy and Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Jakob von Engelhardt
- Institute of PathophysiologyFocus Program Translational Neuroscience (FTN)University Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - David P Wolfer
- Institute of Anatomy and Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
- Institute of Human Movement SciencesETH ZurichZurichSwitzerland
| | - Martin Korte
- Division of Cellular NeurobiologyZoological Institute, TU BraunschweigBraunschweigGermany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration GroupBraunschweigGermany
| | - Ulrike C Müller
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| |
Collapse
|
29
|
Lee S, Kang H, Jung H, Kim E, Lee E. Gene Dosage- and Age-Dependent Differential Transcriptomic Changes in the Prefrontal Cortex of Shank2-Mutant Mice. Front Mol Neurosci 2021; 14:683196. [PMID: 34177464 PMCID: PMC8226033 DOI: 10.3389/fnmol.2021.683196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/18/2021] [Indexed: 11/20/2022] Open
Abstract
Shank2 is an abundant postsynaptic scaffolding protein that is known to regulate excitatory synapse assembly and synaptic transmission and has been implicated in various neurodevelopmental disorders, including autism spectrum disorders (ASD). Previous studies on Shank2-mutant mice provided mechanistic insights into their autistic-like phenotypes, but it remains unclear how transcriptomic patterns are changed in brain regions of the mutant mice in age- and gene dosage-dependent manners. To this end, we performed RNA-Seq analyses of the transcripts from the prefrontal cortex (PFC) of heterozygous and homozygous Shank2-mutant mice lacking exons 6 and 7 at juvenile (week 3) and adult (week 12) stages. Juvenile heterozygous Shank2-mutant mice showed upregulation of glutamate synapse-related genes, downregulation of ribosomal and mitochondrial genes, and transcriptomic changes that are opposite to those observed in ASD (anti-ASD) such as upregulation of ASD_down (downregulated in ASD), GABA neuron-related, and oligodendrocyte-related genes. Juvenile homozygous Shank2 mice showed upregulation of chromatin-related genes and transcriptomic changes that are in line with those occurring in ASD (pro-ASD) such as downregulation of ASD_down, GABA neuron-related, and oligodendrocyte-related genes. Adult heterozygous and homozygous Shank2-mutant mice both exhibited downregulation of ribosomal and mitochondrial genes and pro-ASD transcriptomic changes. Therefore, the gene dosage- and age-dependent effects of Shank2 deletions in mice include differential transcriptomic changes across distinct functional contexts, including synapses, chromatin, ribosomes, mitochondria, GABA neurons, and oligodendrocytes.
Collapse
Affiliation(s)
- Seungjoon Lee
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, KISTI, Daejeon, South Korea
| | - Hwajin Jung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, South Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Eunee Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea.,Department of Anatomy, School of Medicine, Yonsei University, Seoul, South Korea
| |
Collapse
|
30
|
Premoli M, Memo M, Bonini SA. Ultrasonic vocalizations in mice: relevance for ethologic and neurodevelopmental disorders studies. Neural Regen Res 2021; 16:1158-1167. [PMID: 33269765 PMCID: PMC8224126 DOI: 10.4103/1673-5374.300340] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/09/2020] [Accepted: 09/23/2020] [Indexed: 12/21/2022] Open
Abstract
Mice use ultrasonic vocalizations (USVs) to communicate each other and to convey their emotional state. USVs have been greatly characterized in specific life phases and contexts, such as mother isolation-induced USVs for pups or female-induced USVs for male mice during courtship. USVs can be acquired by means of specific tools and later analyzed on the base of both quantitative and qualitative parameters. Indeed, different ultrasonic call categories exist and have already been defined. The understanding of different calls meaning is still missing, and it will represent an essential step forward in the field of USVs. They have long been studied in the ethological context, but recently they emerged as a precious instrument to study pathologies characterized by deficits in communication, in particular neurodevelopmental disorders (NDDs), such as autism spectrum disorders. This review covers the topics of USVs characteristics in mice, contexts for USVs emission and factors that modulate their expression. A particular focus will be devoted to mouse USVs in the context of NDDs. Indeed, several NDDs murine models exist and an intense study of USVs is currently in progress, with the aim of both performing an early diagnosis and to find a pharmacological/behavioral intervention to improve patients' quality of life.
Collapse
Affiliation(s)
- Marika Premoli
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Sara Anna Bonini
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, Italy
| |
Collapse
|
31
|
Prashanth G, Vastrad B, Tengli A, Vastrad C, Kotturshetti I. Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules. BMC Endocr Disord 2021; 21:80. [PMID: 33902539 PMCID: PMC8074411 DOI: 10.1186/s12902-021-00718-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. METHODS To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. RESULTS A total of 820 DEGs were identified between healthy obese and metabolically unhealthy obese, among 409 up regulated and 411 down regulated genes. The GO enrichment analysis results showed that these DEGs were significantly enriched in ion transmembrane transport, intrinsic component of plasma membrane, transferase activity, transferring phosphorus-containing groups, cell adhesion, integral component of plasma membrane and signaling receptor binding, whereas, the REACTOME pathway enrichment analysis results showed that these DEGs were significantly enriched in integration of energy metabolism and extracellular matrix organization. The hub genes CEBPD, TP73, ESR2, TAB1, MAP 3K5, FN1, UBD, RUNX1, PIK3R2 and TNF, which might play an essential role in obesity associated type 2 diabetes mellitus was further screened. CONCLUSIONS The present study could deepen the understanding of the molecular mechanism of obesity associated type 2 diabetes mellitus, which could be useful in developing therapeutic targets for obesity associated type 2 diabetes mellitus.
Collapse
Affiliation(s)
- G Prashanth
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, Karnataka, 577501, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka, 582103, India
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India.
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka, 582209, India
| |
Collapse
|
32
|
Grabrucker S, Pagano J, Schweizer J, Urrutia-Ruiz C, Schön M, Thome K, Ehret G, Grabrucker AM, Zhang R, Hengerer B, Bockmann J, Verpelli C, Sala C, Boeckers TM. Activation of the medial preoptic area (MPOA) ameliorates loss of maternal behavior in a Shank2 mouse model for autism. EMBO J 2021; 40:e104267. [PMID: 33491217 PMCID: PMC7917557 DOI: 10.15252/embj.2019104267] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 11/20/2022] Open
Abstract
Impairments in social relationships and awareness are features observed in autism spectrum disorders (ASDs). However, the underlying mechanisms remain poorly understood. Shank2 is a high‐confidence ASD candidate gene and localizes primarily to postsynaptic densities (PSDs) of excitatory synapses in the central nervous system (CNS). We show here that loss of Shank2 in mice leads to a lack of social attachment and bonding behavior towards pubs independent of hormonal, cognitive, or sensitive deficits. Shank2−/− mice display functional changes in nuclei of the social attachment circuit that were most prominent in the medial preoptic area (MPOA) of the hypothalamus. Selective enhancement of MPOA activity by DREADD technology re‐established social bonding behavior in Shank2−/− mice, providing evidence that the identified circuit might be crucial for explaining how social deficits in ASD can arise.
Collapse
Affiliation(s)
- Stefanie Grabrucker
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Jessica Pagano
- CNR Neuroscience Institute, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Johanna Schweizer
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Kevin Thome
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Günter Ehret
- Institute of Neurobiology, Ulm University, Ulm, Germany
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Rong Zhang
- Neuroscience Research Institute, Peking University, Beijing, China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, China.,Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | | | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Carlo Sala
- CNR Neuroscience Institute, Milan, Italy
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,DZNE, Ulm Site, Ulm, Germany
| |
Collapse
|
33
|
Unsicker C, Cristian FB, von Hahn M, Eckstein V, Rappold GA, Berkel S. SHANK2 mutations impair apoptosis, proliferation and neurite outgrowth during early neuronal differentiation in SH-SY5Y cells. Sci Rep 2021; 11:2128. [PMID: 33483523 PMCID: PMC7822837 DOI: 10.1038/s41598-021-81241-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/30/2020] [Indexed: 12/25/2022] Open
Abstract
SHANK2 mutations have been identified in individuals with neurodevelopmental disorders, including intellectual disability and autism spectrum disorders (ASD). Using CRISPR/Cas9 genome editing, we obtained SH-SY5Y cell lines with frameshift mutations on one or both SHANK2 alleles. We investigated the effects of the different SHANK2 mutations on cell morphology, cell proliferation and differentiation potential during early neuronal differentiation. All mutant cell lines showed impaired neuronal differentiation marker expression. Cells with bi-allelic SHANK2 mutations revealed diminished apoptosis and increased proliferation, as well as decreased neurite outgrowth during early neuronal differentiation. Bi-allelic SHANK2 mutations resulted in an increase in p-AKT levels, suggesting that SHANK2 mutations impair downstream signaling of tyrosine kinase receptors. Additionally, cells with bi-allelic SHANK2 mutations had lower amyloid precursor protein (APP) expression compared to controls, suggesting a molecular link between SHANK2 and APP. Together, we can show that frameshift mutations on one or both SHANK2 alleles lead to an alteration of neuronal differentiation in SH-SY5Y cells, characterized by changes in cell growth and pre- and postsynaptic protein expression. We also provide first evidence that downstream signaling of tyrosine kinase receptors and amyloid precursor protein expression are affected.
Collapse
Affiliation(s)
- Christine Unsicker
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Flavia-Bianca Cristian
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Manja von Hahn
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Volker Eckstein
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Simone Berkel
- Department of Human Molecular Genetics, Institute of Human Genetics, University Hospital Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
34
|
Jin C, Kang H, Yoo T, Ryu JR, Yoo YE, Ma R, Zhang Y, Kang HR, Kim Y, Seong H, Bang G, Park S, Kwon SK, Sun W, Kim H, Kim JY, Kim E, Han K. The Neomycin Resistance Cassette in the Targeted Allele of Shank3B Knock-Out Mice Has Potential Off-Target Effects to Produce an Unusual Shank3 Isoform. Front Mol Neurosci 2021; 13:614435. [PMID: 33505245 PMCID: PMC7831789 DOI: 10.3389/fnmol.2020.614435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/02/2020] [Indexed: 01/20/2023] Open
Abstract
Variants of the SH3 and multiple ankyrin repeat domains 3 (SHANK3), which encodes postsynaptic scaffolds, are associated with brain disorders. The targeted alleles in a few Shank3 knock-out (KO) lines contain a neomycin resistance (Neo) cassette, which may perturb the normal expression of neighboring genes; however, this has not been investigated in detail. We previously reported an unexpected increase in the mRNA expression of Shank3 exons 1–12 in the brains of Shank3B KO mice generated by replacing Shank3 exons 13–16 with the Neo cassette. In this study, we confirmed that the increased Shank3 mRNA in Shank3B KO brains produced an unusual ∼60 kDa Shank3 isoform (Shank3-N), which did not properly localize to the synaptic compartment. Functionally, Shank3-N overexpression altered the dendritic spine morphology in cultured neurons. Importantly, Shank3-N expression in Shank3B KO mice was not a compensatory response to a reduction of full-length Shank3 because expression was still detected in the brain after normalizing the level of full-length Shank3. Moreover, in another Shank3 KO line (Shank3 gKO) with a similar Shank3 exonal deletion as that in Shank3B KO mice but without a Neo cassette, the mRNA expression levels of Shank3 exons 1–12 were lower than those of wild-type mice and Shank3-N was not detected in the brain. In addition, the expression levels of genes neighboring Shank3 on chromosome 15 were altered in the striatum of Shank3B KO but not Shank3 gKO mice. These results suggest that the Neo cassette has potential off-target effects in Shank3B KO mice.
Collapse
Affiliation(s)
- Chunmei Jin
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information, Daejeon, South Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Jae Ryun Ryu
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Ye-Eun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Ruiying Ma
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yinhua Zhang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Hyae Rim Kang
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| | - Yoonhee Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - Hyunyoung Seong
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea.,College of Pharmacy, Korea University, Sejong, South Korea
| | - Sangwoo Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea
| | - Seok-Kyu Kwon
- Center for Functional Connectomics, Korea Institute of Science and Technology, Brain Science Institute, Seoul, South Korea
| | - Woong Sun
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.,Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hyunkyung Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea.,Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul, South Korea
| | - Jin Young Kim
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Ochang, South Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Kihoon Han
- Department of Neuroscience, College of Medicine, Korea University, Seoul, South Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
35
|
Imbalanced post- and extrasynaptic SHANK2A functions during development affect social behavior in SHANK2-mediated neuropsychiatric disorders. Mol Psychiatry 2021; 26:6482-6504. [PMID: 34021263 PMCID: PMC8760046 DOI: 10.1038/s41380-021-01140-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Mutations in SHANK genes play an undisputed role in neuropsychiatric disorders. Until now, research has focused on the postsynaptic function of SHANKs, and prominent postsynaptic alterations in glutamatergic signal transmission have been reported in Shank KO mouse models. Recent studies have also suggested a possible presynaptic function of SHANK proteins, but these remain poorly defined. In this study, we examined how SHANK2 can mediate electrophysiological, molecular, and behavioral effects by conditionally overexpressing either wild-type SHANK2A or the extrasynaptic SHANK2A(R462X) variant. SHANK2A overexpression affected pre- and postsynaptic targets and revealed a reversible, development-dependent autism spectrum disorder-like behavior. SHANK2A also mediated redistribution of Ca2+-permeable AMPA receptors between apical and basal hippocampal CA1 dendrites, leading to impaired synaptic plasticity in the basal dendrites. Moreover, SHANK2A overexpression reduced social interaction and increased the excitatory noise in the olfactory cortex during odor processing. In contrast, overexpression of the extrasynaptic SHANK2A(R462X) variant did not impair hippocampal synaptic plasticity, but still altered the expression of presynaptic/axonal signaling proteins. We also observed an attention-deficit/hyperactivity-like behavior and improved social interaction along with enhanced signal-to-noise ratio in cortical odor processing. Our results suggest that the disruption of pre- and postsynaptic SHANK2 functions caused by SHANK2 mutations has a strong impact on social behavior. These findings indicate that pre- and postsynaptic SHANK2 actions cooperate for normal neuronal function, and that an imbalance between these functions may lead to different neuropsychiatric disorders.
Collapse
|
36
|
Glutamatergic Dysfunction and Synaptic Ultrastructural Alterations in Schizophrenia and Autism Spectrum Disorder: Evidence from Human and Rodent Studies. Int J Mol Sci 2020; 22:ijms22010059. [PMID: 33374598 PMCID: PMC7793137 DOI: 10.3390/ijms22010059] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The correlation between dysfunction in the glutamatergic system and neuropsychiatric disorders, including schizophrenia and autism spectrum disorder, is undisputed. Both disorders are associated with molecular and ultrastructural alterations that affect synaptic plasticity and thus the molecular and physiological basis of learning and memory. Altered synaptic plasticity, accompanied by changes in protein synthesis and trafficking of postsynaptic proteins, as well as structural modifications of excitatory synapses, are critically involved in the postnatal development of the mammalian nervous system. In this review, we summarize glutamatergic alterations and ultrastructural changes in synapses in schizophrenia and autism spectrum disorder of genetic or drug-related origin, and briefly comment on the possible reversibility of these neuropsychiatric disorders in the light of findings in regular synaptic physiology.
Collapse
|
37
|
Chen ST, Lai WJ, Zhang WJ, Chen QP, Zhou LB, So KF, Shi LL. Insulin-like growth factor 1 partially rescues early developmental defects caused by SHANK2 knockdown in human neurons. Neural Regen Res 2020; 15:2335-2343. [PMID: 32594058 PMCID: PMC7749486 DOI: 10.4103/1673-5374.285002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 02/22/2020] [Accepted: 04/01/2020] [Indexed: 11/16/2022] Open
Abstract
SHANK2 is a scaffold protein that serves as a protein anchor at the postsynaptic density in neurons. Genetic variants of SHANK2 are strongly associated with synaptic dysfunction and the pathophysiology of autism spectrum disorder. Recent studies indicate that early neuronal developmental defects play a role in the pathogenesis of autism spectrum disorder, and that insulin-like growth factor 1 has a positive effect on neurite development. To investigate the effects of SHANK2 knockdown on early neuronal development, we generated a sparse culture system using human induced pluripotent stem cells, which then differentiated into neural progenitor cells after 3-14 days in culture, and which were dissociated into single neurons. Neurons in the experimental group were infected with shSHANK2 lentivirus carrying a red fluorescent protein reporter (shSHANK2 group). Control neurons were infected with scrambled shControl lentivirus carrying a red fluorescent protein reporter (shControl group). Neuronal somata and neurites were reconstructed based on the lentiviral red fluorescent protein signal. Developmental dendritic and motility changes in VGLUT1+ glutamatergic neurons and TH+ dopaminergic neurons were then evaluated in both groups. Compared with shControl VGLUT1+ neurons, the dendritic length and arborizations of shSHANK2 VGLUT1+ neurons were shorter and fewer, while cell soma speed was higher. Furthermore, dendritic length and arborization were significantly increased after insulin-like growth factor 1 treatment of shSHANK2 neurons, while cell soma speed remained unaffected. These results suggest that insulin-like growth factor 1 can rescue morphological defects, but not the change in neuronal motility. Collectively, our findings demonstrate that SHANK2 deficiency perturbs early neuronal development, and that IGF1 can partially rescue the neuronal defects caused by SHANK2 knockdown. All experimental procedures and protocols were approved by the Laboratory Animal Ethics Committee of Jinan University, China (approval No. 20170228010) on February 28, 2017.
Collapse
Affiliation(s)
- Shu-Ting Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Wan-Jing Lai
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
- Clinical Medicine, Jinan University, Guangzhou, Guangdong Province, China
| | - Wei-Jia Zhang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Qing-Pei Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Li-Bing Zhou
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Ling-Ling Shi
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
- Department of Psychiatry, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
38
|
Zhang H, Wang D, Chen J, Li X, Yi Q, Shi Y. Identification of SHANK2 Pathogenic Variants in a Chinese Uygur Population with Schizophrenia. J Mol Neurosci 2020; 71:1-8. [PMID: 32897530 DOI: 10.1007/s12031-020-01606-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/19/2020] [Indexed: 11/25/2022]
Abstract
Genomic studies on schizophrenia (SCZ) have revealed several candidate genes involved in excitatory synapse function and plasticity associated with its etiology. SHANK2 is a postsynaptic scaffolding protein, which anchors a protein complex connecting NMDAR, AMPAR, and mGluR receptors at excitatory neuronal synapses. Mutations in the SHANK2 gene have been reported to be associated with human autism spectrum disorders (ASDs) and SCZ. To identify variants in the SHANK2 gene and determine the association of SHANK2 with SCZ in the Chinese Uygur population, we conducted targeted sequencing of whole exon regions and exon-intron boundaries of SHANK2 in 1574 SCZ patients and 1481 healthy controls. A total of 149 variants were identified, including six common variants and 143 rare variants. For common variants, rs62622853 and rs3924047 showed allelic significance with SCZ before correction, but the association was eliminated after Bonferroni correction. Seven rare nonsynonymous variants, p.Arg739Trp, p.Pro807Leu, p.Ile854Phe, p.Thr1322Ser, p.Leu1434Arg, p.Val1486Ile, and p.Thr1674Met, occurred only in the patients but not in any of the healthy controls. In silico analysis predicted that p.Arg739Trp, p.Leu1434Arg, and p.Val1486Ile variants are likely to be damaging. The present study suggests that individuals with two novel rare nonsynonymous variants (p.Arg739Trp, p.Leu1434Arg) and p.Val1486Ile variants of SHANK2 might increase the susceptibility to developing SCZ disorder.
Collapse
Affiliation(s)
- Han Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Dong Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhua Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuli Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qizhong Yi
- Psychological Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China.
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China. .,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Psychological Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China. .,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
39
|
Mikkelsen EK, Weir JT. The genome of the Xingu scale-backed antbird (Willisornis vidua nigrigula) reveals lineage-specific adaptations. Genomics 2020; 112:4552-4560. [PMID: 32771623 DOI: 10.1016/j.ygeno.2020.07.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/08/2020] [Accepted: 07/30/2020] [Indexed: 12/23/2022]
Abstract
Antbirds (Thamnophilidae) are a large neotropical family of passerine bird renowned for the ant-following foraging strategies of several members of this clade. The high diversity of antbirds provides ample opportunity for speciation studies, however these studies can be hindered by the lack of an annotated antbird reference genome. In this study, we produced a high-quality annotated reference genome for the Xingu Scale-backed Antbird (Willisornis vidua nigrigula) using 10X Genomics Chromium linked-reads technology. The assembly is 1.09 Gb, with a scaffold N50 of 12.1 Mb and 17,475 annotated protein coding genes. We compare the proteome of W. v. nigrigula to several other passerines, and produce annotations for two additional antbird genomes in order to identify genes under lineage-specific positive selection and gene families with evidence for significant expansions in antbirds. Several of these genes have functions potentially related to the lineage-specific traits of antbirds, including adaptations for thermoregulation in a humid tropical environment.
Collapse
Affiliation(s)
- Else K Mikkelsen
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto M5S 3B2, ON, Canada.
| | - Jason T Weir
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto M5S 3B2, ON, Canada; Department of Biological Sciences, University of Toronto Scarborough, Toronto M1C 1A4, ON, Canada; Department of Ornithology, Royal Ontario Museum, Toronto, Canada
| |
Collapse
|
40
|
Eltokhi A, Kurpiers B, Pitzer C. Behavioral tests assessing neuropsychiatric phenotypes in adolescent mice reveal strain- and sex-specific effects. Sci Rep 2020; 10:11263. [PMID: 32647155 PMCID: PMC7347854 DOI: 10.1038/s41598-020-67758-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/10/2020] [Indexed: 12/29/2022] Open
Abstract
In humans, infancy and adolescence are associated with major changes in synaptic functions and ongoing maturation of neural networks, which underlie the major behavioral changes during these periods. Among adult cases with neuropsychiatric disorders including autism spectrum disorder, schizophrenia, attention deficit hyperactivity, and bipolar disorders, 50% have developed behavioral symptoms and received a diagnosis before 15 years of age. However, most of the behavioral studies in mice modeling neuropsychiatric phenotypes are performed in adult animals, missing valuable phenotypic information related to the effect of synaptic maturation during development. Here, we explored which behavioral experiments assessing neuropsychiatric phenotypes can be performed during a specific window of development in adolescent male and female C57BL/6N, DBA/2, and FVB/N mice that are typically used as background strains for generating genetically-modified mouse models. The three wild-type strains were evaluated across anxiety, social behaviors, and cognitive functions in order to cover the main behavioral impairments that occur in neuropsychiatric disorders. During adolescence, the three strains displayed significant differences under certain behavioral paradigms. In addition, C57BL/6N and FVB/N, but not DBA/2 mice revealed some sex-related differences. Our results provide new insights into discrete behaviors during development and emphasize the crucial importance of the genetic background, sex, and experimental settings in the age-dependent regulation of different behaviors.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany. .,Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany.
| | - Barbara Kurpiers
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
41
|
Culotta L, Penzes P. Exploring the mechanisms underlying excitation/inhibition imbalance in human iPSC-derived models of ASD. Mol Autism 2020; 11:32. [PMID: 32393347 PMCID: PMC7216514 DOI: 10.1186/s13229-020-00339-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is a range of neurodevelopmental disorders characterized by impaired social interaction and communication, and repetitive or restricted behaviors. ASD subjects exhibit complex genetic and clinical heterogeneity, thus hindering the discovery of pathophysiological mechanisms. Considering that several ASD-risk genes encode proteins involved in the regulation of synaptic plasticity, neuronal excitability, and neuronal connectivity, one hypothesis that has emerged is that ASD arises from a disruption of the neuronal network activity due to perturbation of the synaptic excitation and inhibition (E/I) balance. The development of induced pluripotent stem cell (iPSC) technology and recent advances in neuronal differentiation techniques provide a unique opportunity to model complex neuronal connectivity and to test the E/I hypothesis of ASD in human-based models. Here, we aim to review the latest advances in studying the different cellular and molecular mechanisms contributing to E/I balance using iPSC-based in vitro models of ASD.
Collapse
Affiliation(s)
- Lorenza Culotta
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
42
|
CaMKIIα phosphorylation of Shank3 modulates ABI1-Shank3 interaction. Biochem Biophys Res Commun 2020; 524:262-267. [PMID: 31983435 DOI: 10.1016/j.bbrc.2020.01.089] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 01/15/2020] [Indexed: 12/22/2022]
Abstract
Protein-protein interactions can be modulated by phosphorylation of either binding partner, thereby altering subcellular localization and/or physiological function. Shank3, a master postsynaptic scaffolding protein that controls the developmental maturation of excitatory synapses, was recently shown to be phosphorylated by Protein Kinase A (PKA) at Ser685 in vivo. Mutation of Shank3 Ser685 was shown to modulate the binding of Abelson interactor 1 (ABI1), a component of the WAVE regulatory complex for actin remodeling, but a direct effect of Ser685 phosphorylation on ABI1 binding was not investigated. Here, we demonstrate that Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα) also phosphorylates Shank3 at Ser685. Mutation of Ser685 to phospho-null alanine (S685A) prevented both CaMKIIα and PKA phosphorylation of a GST-Shank3 fusion protein. The co-immunoprecipitation of ABI1 with Shank3 from HEK293 cell extracts is reduced by mutation of Ser685 to either Ala or Asp. However, pre-phosphorylation of GST-Shank3 by purified CaMKIIα significantly increased binding of ABI1, and this effect was abrogated by Ser685 to Ala mutation in GST-Shank3. Taken together, our data suggest that neuronal ABI1-Shank3 interactions may be convergently regulated by Shank3 Ser685 phosphorylation in response to both Ca2+ and cAMP signaling, potentially modulating dendritic spine morphology.
Collapse
|
43
|
Peleh T, Eltokhi A, Pitzer C. Longitudinal analysis of ultrasonic vocalizations in mice from infancy to adolescence: Insights into the vocal repertoire of three wild-type strains in two different social contexts. PLoS One 2019; 14:e0220238. [PMID: 31365551 PMCID: PMC6668806 DOI: 10.1371/journal.pone.0220238] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 07/11/2019] [Indexed: 11/19/2022] Open
Abstract
Ultrasonic vocalizations (USV) are emitted by mice under certain developmental, social and behavioral conditions. The analysis of USV can be used as a reliable measure of the general affective state, for testing the efficacy of pharmacological compounds and for investigating communication in mutant mice with predicted social or communication deficits. Social and communication studies in mice have focused mainly on the investigation of USV emitted by neonatal pups after separation from the dam and during social interaction between adult males and females. Longitudinal USV analysis among the different developmental states remained uninvestigated. In our study, we first recorded USV from three inbred mouse strains C57BL/6N, DBA/2 and FVB/N during the neonatal stages after separation from the littermates and then during a reunion with one littermate. Our results revealed significant strain-specific differences in the numbers and categories of USV calls. In addition, the USV profiles seemed to be sensitive to small developmental progress during infancy. By following these mice to the adolescent stage and measuring USV in the three-chamber social test, we found that USV profiles still showed significant differences between these strains in the different trials of the test. To study the effects of social context on USV characteristics, we measured USV emitted by another cohort of adolescent mice during the direct social interaction test. To this end, this study provides a strategy for evaluating novel mouse mutants in behavioral questions relevant to disorders with deficits in communication and sociability and emphasizes the important contribution of genetics and experimental contexts on the behavioral outcome.
Collapse
Affiliation(s)
- Tatiana Peleh
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Ahmed Eltokhi
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
- Research Group of the Max Planck Institute for Medical Research at the Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- * E-mail:
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
44
|
SHANK2 mutations associated with autism spectrum disorder cause hyperconnectivity of human neurons. Nat Neurosci 2019; 22:556-564. [PMID: 30911184 PMCID: PMC6475597 DOI: 10.1038/s41593-019-0365-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Heterozygous loss-of-function mutations in SHANK2 are associated with autism spectrum disorder (ASD). We generated cortical neurons from induced pluripotent stem cells (iPSC) derived from neurotypic and ASD-affected donors. We developed Sparse coculture for Connectivity (SparCon) assays where SHANK2 and control neurons were differentially labeled and sparsely seeded together on a lawn of unlabeled control neurons. We observed increases in dendrite length, dendrite complexity, synapse number, and frequency of spontaneous excitatory postsynaptic currents. These findings were phenocopied in gene-edited homozygous SHANK2 knockout cells and rescued by gene correction of an ASD SHANK2 mutation. Dendrite length increases were exacerbated by IGF1, TG003, or BDNF, and suppressed by DHPG treatment. The transcriptome in isogenic SHANK2 neurons was perturbed in synapse, plasticity, and neuronal morphogenesis gene sets and ASD gene modules, and activity-dependent dendrite extension was impaired. Our findings provide evidence for hyperconnectivity and altered transcriptome in SHANK2 neurons derived from ASD subjects.
Collapse
|
45
|
Ey E, Torquet N, de Chaumont F, Lévi-Strauss J, Ferhat AT, Le Sourd AM, Boeckers TM, Bourgeron T. Shank2 Mutant Mice Display Hyperactivity Insensitive to Methylphenidate and Reduced Flexibility in Social Motivation, but Normal Social Recognition. Front Mol Neurosci 2018; 11:365. [PMID: 30337855 PMCID: PMC6180161 DOI: 10.3389/fnmol.2018.00365] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/13/2018] [Indexed: 12/17/2022] Open
Abstract
Mouse models of autism can be used to study evolutionarily conserved mechanisms underlying behavioral abnormalities in social communication and repetitive behaviors. SHANK genes code for synaptic scaffolding proteins at excitatory synapses and mutations in all SHANK genes have been associated with autism. Here, we present three behavioral aspects of the mutant mice deleted for exon 16 in Shank2. First, we treated Shank2 mutant mice with methylphenidate to rescue the hyperactivity. Our failure to do so suggests that the hyperactivity displayed by Shank2 mutant mice is not related to the one displayed by the typical mouse models of hyperactivity, and might be more closely related to manic-like behaviors. Second, by testing the effect of group housing and social isolation on social interest, we highlighted that Shank2 mutant mice lack the typical flexibility to modulate social interest, in comparison with wild-type littermates. Finally, we established a new protocol to test for social recognition in a social context. We used this protocol to show that Shank2 mutant mice were able to discriminate familiar and unknown conspecifics in free interactions. Altogether, these studies shed some light on specific aspects of the behavioral defects displayed by the Shank2 mouse model. Such information could be used to orient therapeutic strategies and to design more specific tests to characterize the complex behavior of mouse models of autism.
Collapse
Affiliation(s)
- Elodie Ey
- CNRS UMR 3571, Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
| | - Nicolas Torquet
- Sorbonne Université, UPMC Univ Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Paris, France
| | | | - Julie Lévi-Strauss
- CNRS UMR 3571, Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
| | | | - Anne-Marie Le Sourd
- CNRS UMR 3571, Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Thomas Bourgeron
- CNRS UMR 3571, Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| |
Collapse
|
46
|
Berkel S, Eltokhi A, Fröhlich H, Porras-Gonzalez D, Rafiullah R, Sprengel R, Rappold GA. Sex Hormones Regulate SHANK Expression. Front Mol Neurosci 2018; 11:337. [PMID: 30319350 PMCID: PMC6167484 DOI: 10.3389/fnmol.2018.00337] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/28/2018] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorders (ASD) have a higher prevalence in male individuals compared to females, with a ratio of affected boys compared to girls of 4:1 for ASD and 11:1 for Asperger syndrome. Mutations in the SHANK genes (comprising SHANK1, SHANK2 and SHANK3) coding for postsynaptic scaffolding proteins have been tightly associated with ASD. As early brain development is strongly influenced by sex hormones, we investigated the effect of dihydrotestosterone (DHT) and 17β-estradiol on SHANK expression in a human neuroblastoma cell model. Both sex hormones had a significant impact on the expression of all three SHANK genes, which could be effectively blocked by androgen and estrogen receptor antagonists. In neuron-specific androgen receptor knock-out mice (ArNesCre), we found a nominal significant reduction of all Shank genes at postnatal day 7.5 in the cortex. In the developing cortex of wild-type (WT) CD1 mice, a sex-differential protein expression was identified for all Shanks at embryonic day 17.5 and postnatal day 7.5 with significantly higher protein levels in male compared to female mice. Together, we could show that SHANK expression is influenced by sex hormones leading to a sex-differential expression, thus providing novel insights into the sex bias in ASD.
Collapse
Affiliation(s)
- Simone Berkel
- Department of Human Molecular Genetics, Institute of Human Genetics, Ruprecht-Karls-University, Heidelberg, Germany
| | - Ahmed Eltokhi
- Department of Human Molecular Genetics, Institute of Human Genetics, Ruprecht-Karls-University, Heidelberg, Germany.,Research Group of the Max Planck Institute for Medical Research at the Institute of Anatomy and Cell Biology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Henning Fröhlich
- Department of Human Molecular Genetics, Institute of Human Genetics, Ruprecht-Karls-University, Heidelberg, Germany
| | - Diana Porras-Gonzalez
- Department of Human Molecular Genetics, Institute of Human Genetics, Ruprecht-Karls-University, Heidelberg, Germany
| | - Rafiullah Rafiullah
- Department of Human Molecular Genetics, Institute of Human Genetics, Ruprecht-Karls-University, Heidelberg, Germany
| | - Rolf Sprengel
- Research Group of the Max Planck Institute for Medical Research at the Institute of Anatomy and Cell Biology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Gudrun A Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|