1
|
Ortiz A, Ayhan F, Harper M, Konopka G. Cell type specific roles of FOXP1 during early neocortical murine development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.08.598089. [PMID: 38895440 PMCID: PMC11185780 DOI: 10.1101/2024.06.08.598089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Cortical development is a tightly controlled process and any deviation during development may increase the susceptibility to neurodevelopmental disorders, such as autism spectrum disorders (ASD). Numerous studies identified mutations in FOXP1, a transcription factor enriched in the neocortex, as causal for ASD and FOXP1 syndrome. Our group has shown that Foxp1 deletion in the mouse cortex leads to overall reduced cortex thickness, alterations in cortical lamination, and changes in the relative thickness of cortical layers. However, the developmental and cell type-specific mechanisms underlying these changes remained unclear. This work characterizes the developmental requirement of neocortical Foxp1 at key embryonic and perinatal ages using a conditional knock-out of Foxp1. We find that Foxp1 deletion results in accelerated pseudo-age during early neurogenesis, increased cell cycle exit during late neurogenesis, altered gene expression and chromatin accessibility, and selective migration deficits in a subset of upper-layer neurons. These data explain the postnatal differences observed in cortical layers and relative cortical thickness. We also highlight genes regulated by FOXP1 and their enrichment with high-confidence ASD or synaptic genes. Together, these results underscore a network of neurodevelopmental disorder-related genes that may serve as potential modulatory targets for postnatal modification relevant to ASD and FOXP1 syndrome.
Collapse
Affiliation(s)
- Ana Ortiz
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Fatma Ayhan
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Matthew Harper
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Genevieve Konopka
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Ahmed NI, Khandelwal N, Anderson AG, Oh E, Vollmer RM, Kulkarni A, Gibson JR, Konopka G. Compensation between FOXP transcription factors maintains proper striatal function. Cell Rep 2024; 43:114257. [PMID: 38761373 PMCID: PMC11234887 DOI: 10.1016/j.celrep.2024.114257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 02/05/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024] Open
Abstract
Spiny projection neurons (SPNs) of the striatum are critical in integrating neurochemical information to coordinate motor and reward-based behavior. Mutations in the regulatory transcription factors expressed in SPNs can result in neurodevelopmental disorders (NDDs). Paralogous transcription factors Foxp1 and Foxp2, which are both expressed in the dopamine receptor 1 (D1) expressing SPNs, are known to have variants implicated in NDDs. Utilizing mice with a D1-SPN-specific loss of Foxp1, Foxp2, or both and a combination of behavior, electrophysiology, and cell-type-specific genomic analysis, loss of both genes results in impaired motor and social behavior as well as increased firing of the D1-SPNs. Differential gene expression analysis implicates genes involved in autism risk, electrophysiological properties, and neuronal development and function. Viral-mediated re-expression of Foxp1 into the double knockouts is sufficient to restore electrophysiological and behavioral deficits. These data indicate complementary roles between Foxp1 and Foxp2 in the D1-SPNs.
Collapse
Affiliation(s)
- Newaz I Ahmed
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Nitin Khandelwal
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ashley G Anderson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Emily Oh
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Rachael M Vollmer
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Jay R Gibson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
3
|
Heim F, Scharff C, Fisher SE, Riebel K, Ten Cate C. Auditory discrimination learning and acoustic cue weighing in female zebra finches with localized FoxP1 knockdowns. J Neurophysiol 2024; 131:950-963. [PMID: 38629163 DOI: 10.1152/jn.00228.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 05/21/2024] Open
Abstract
Rare disruptions of the transcription factor FOXP1 are implicated in a human neurodevelopmental disorder characterized by autism and/or intellectual disability with prominent problems in speech and language abilities. Avian orthologues of this transcription factor are evolutionarily conserved and highly expressed in specific regions of songbird brains, including areas associated with vocal production learning and auditory perception. Here, we investigated possible contributions of FoxP1 to song discrimination and auditory perception in juvenile and adult female zebra finches. They received lentiviral knockdowns of FoxP1 in one of two brain areas involved in auditory stimulus processing, HVC (proper name) or CMM (caudomedial mesopallium). Ninety-six females, distributed over different experimental and control groups were trained to discriminate between two stimulus songs in an operant Go/Nogo paradigm and subsequently tested with an array of stimuli. This made it possible to assess how well they recognized and categorized altered versions of training stimuli and whether localized FoxP1 knockdowns affected the role of different features during discrimination and categorization of song. Although FoxP1 expression was significantly reduced by the knockdowns, neither discrimination of the stimulus songs nor categorization of songs modified in pitch, sequential order of syllables or by reversed playback were affected. Subsequently, we analyzed the full dataset to assess the impact of the different stimulus manipulations for cue weighing in song discrimination. Our findings show that zebra finches rely on multiple parameters for song discrimination, but with relatively more prominent roles for spectral parameters and syllable sequencing as cues for song discrimination.NEW & NOTEWORTHY In humans, mutations of the transcription factor FoxP1 are implicated in speech and language problems. In songbirds, FoxP1 has been linked to male song learning and female preference strength. We found that FoxP1 knockdowns in female HVC and caudomedial mesopallium (CMM) did not alter song discrimination or categorization based on spectral and temporal information. However, this large dataset allowed to validate different cue weights for spectral over temporal information for song recognition.
Collapse
Affiliation(s)
- Fabian Heim
- Institute of Biology, Leiden University, Leiden, The Netherlands
- Language and Genetics Department, Max Planck Institute for Psycholinguistics,Nijmegen, The Netherlands
- Institute of Biology, Freie Universität Berlin, Berlin, Germany
| | | | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics,Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Katharina Riebel
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Carel Ten Cate
- Institute of Biology, Leiden University, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| |
Collapse
|
4
|
Tyson HR, Harrison DJ, Higgs MJ, Isles AR, John RM. Deficiency of the paternally-expressed imprinted Peg3 gene in mice has sexually dimorphic consequences for offspring communication and social behaviour. Front Neurosci 2024; 18:1374781. [PMID: 38595977 PMCID: PMC11002209 DOI: 10.3389/fnins.2024.1374781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Imprinted genes are expressed from one parental allele as a consequence of epigenetic processes initiated in the germline. Consequently, their ability to influence phenotype depends on their parent-of-origin. Recent research suggests that the sex of the individual expressing the imprinted gene is also important. We have previously reported that genetically wildtype (WT) dams carrying and caring for pups mutant for PEG3 exhibit anxiety-like behaviours and their mutant pups show a reduction in ultrasonic vocalisation when separated from their mothers. Sex-specificity was not examined. Methods WT female mice were mated with WT, heterozygous Peg3-/+ or homozygous Peg3-/- studs to generate all WT (control), 50:50 mixed or 100% mutant litters, respectively, followed by behavioural assessment of both dams and their pups. Results We reproduced our original finding that WT dams carrying and caring for 100% mutant litters exhibit postpartum anxiety-like symptoms and delayed pup retrieval. Additionally, these WT dams were found to allocate less time to pup-directed care behaviours relative to controls. Male Peg3-deficient pups demonstrated significantly reduced vocalisation with a more subtle communication deficit in females. Postweaning, male mutants exhibited deficits across a number of key social behaviours as did WT males sharing their environment with mutants. Only modest variations in social behaviour were detected in experimental females. Discussion We have experimentally demonstrated that Peg3 deficiency confined to the offspring causes anxiety in mouse mothers and atypical behaviour including deficits in communication in their male offspring. A male-specific reduction in expression PEG3 in the fetally-derived placenta has previously been associated with maternal depression in human pregnancy. Maternal mood disorders such as depression and anxiety are associated with delays in language development and neuroatypical behaviour more common in sons. Peg3 deficiency could drive the association of maternal and offspring behavioural disorders reported in humans.
Collapse
Affiliation(s)
- Hannah R. Tyson
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - David J. Harrison
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Mathew J. Higgs
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Anthony R. Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Rosalind M. John
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
5
|
Sterling ML, Teunisse R, Englitz B. Rodent ultrasonic vocal interaction resolved with millimeter precision using hybrid beamforming. eLife 2023; 12:e86126. [PMID: 37493217 PMCID: PMC10522333 DOI: 10.7554/elife.86126] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/25/2023] [Indexed: 07/27/2023] Open
Abstract
Ultrasonic vocalizations (USVs) fulfill an important role in communication and navigation in many species. Because of their social and affective significance, rodent USVs are increasingly used as a behavioral measure in neurodevelopmental and neurolinguistic research. Reliably attributing USVs to their emitter during close interactions has emerged as a difficult, key challenge. If addressed, all subsequent analyses gain substantial confidence. We present a hybrid ultrasonic tracking system, Hybrid Vocalization Localizer (HyVL), that synergistically integrates a high-resolution acoustic camera with high-quality ultrasonic microphones. HyVL is the first to achieve millimeter precision (~3.4-4.8 mm, 91% assigned) in localizing USVs, ~3× better than other systems, approaching the physical limits (mouse snout ~10 mm). We analyze mouse courtship interactions and demonstrate that males and females vocalize in starkly different relative spatial positions, and that the fraction of female vocalizations has likely been overestimated previously due to imprecise localization. Further, we find that when two male mice interact with one female, one of the males takes a dominant role in the interaction both in terms of the vocalization rate and the location relative to the female. HyVL substantially improves the precision with which social communication between rodents can be studied. It is also affordable, open-source, easy to set up, can be integrated with existing setups, and reduces the required number of experiments and animals.
Collapse
Affiliation(s)
- Max L Sterling
- Computational Neuroscience Lab, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
- Visual Neuroscience Lab, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Ruben Teunisse
- Computational Neuroscience Lab, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Bernhard Englitz
- Computational Neuroscience Lab, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| |
Collapse
|
6
|
Ahmed NI, Khandelwal N, Anderson AG, Kulkarni A, Gibson J, Konopka G. Compensation between FOXP transcription factors maintains proper striatal function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546567. [PMID: 37425820 PMCID: PMC10327074 DOI: 10.1101/2023.06.26.546567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Spiny projection neurons (SPNs) of the striatum are critical in integrating neurochemical information to coordinate motor and reward-based behavior. Mutations in the regulatory transcription factors expressed in SPNs can result in neurodevelopmental disorders (NDDs). Paralogous transcription factors Foxp1 and Foxp2, which are both expressed in the dopamine receptor 1 (D1) expressing SPNs, are known to have variants implicated in NDDs. Utilizing mice with a D1-SPN specific loss of Foxp1, Foxp2, or both and a combination of behavior, electrophysiology, and cell-type specific genomic analysis, loss of both genes results in impaired motor and social behavior as well as increased firing of the D1-SPNs. Differential gene expression analysis implicates genes involved in autism risk, electrophysiological properties, and neuronal development and function. Viral mediated re-expression of Foxp1 into the double knockouts was sufficient to restore electrophysiological and behavioral deficits. These data indicate complementary roles between Foxp1 and Foxp2 in the D1-SPNs.
Collapse
Affiliation(s)
- Newaz I. Ahmed
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Nitin Khandelwal
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ashley G. Anderson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, 77030, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Jay Gibson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
- Lead Contact
| |
Collapse
|
7
|
Loan A, Leung JWH, Cook DP, Ko C, Vanderhyden BC, Wang J, Chan HM. Prenatal low-dose methylmercury exposure causes premature neuronal differentiation and autism-like behaviors in a rodent model. iScience 2023; 26:106093. [PMID: 36843845 PMCID: PMC9947313 DOI: 10.1016/j.isci.2023.106093] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/10/2022] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Aberrant neurodevelopment is a core deficit of autism spectrum disorder (ASD). Here we ask whether a non-genetic factor, prenatal exposure to the environmental pollutant methylmercury (MeHg), is a contributing factor in ASD onset. We showed that adult mice prenatally exposed to non-apoptotic MeHg exhibited key ASD characteristics, including impaired communication, reduced sociability, and increased restrictive repetitive behaviors, whereas in the embryonic cortex, prenatal MeHg exposure caused premature neuronal differentiation. Further single-cell RNA sequencing (scRNA-seq) analysis disclosed that prenatal exposure to MeHg resulted in cortical radial glial precursors (RGPs) favoring asymmetric differentiation to directly generate cortical neurons, omitting the intermediate progenitor stage. In addition, MeHg exposure in cultured RGPs increased CREB phosphorylation and enhanced the interaction between CREB and CREB binding protein (CBP). Intriguingly, metformin, an FDA-approved drug, can reverse MeHg-induced premature neuronal differentiation via CREB/CBP repulsion. These findings provide insights into ASD etiology, its underlying mechanism, and a potential therapeutic strategy.
Collapse
Affiliation(s)
- Allison Loan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Joseph Wai-Hin Leung
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - David P. Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Chelsea Ko
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Barbara C. Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jing Wang
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Hing Man Chan
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
8
|
Qi H, Luo L, Lu C, Chen R, Zhou X, Zhang X, Jia Y. TCF7L2 acts as a molecular switch in midbrain to control mammal vocalization through its DNA binding domain but not transcription activation domain. Mol Psychiatry 2023; 28:1703-1717. [PMID: 36782064 DOI: 10.1038/s41380-023-01993-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 01/15/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023]
Abstract
Vocalization is an essential medium for social signaling in birds and mammals. Periaqueductal gray (PAG) a conserved midbrain structure is believed to be responsible for innate vocalizations, but its molecular regulation remains largely unknown. Here, through a mouse forward genetic screening we identified one of the key Wnt/β-catenin effectors TCF7L2/TCF4 controls ultrasonic vocalization (USV) production and syllable complexity during maternal deprivation and sexual encounter. Early developmental expression of TCF7L2 in PAG excitatory neurons is necessary for the complex trait, while TCF7L2 loss reduces neuronal gene expressions and synaptic transmission in PAG. TCF7L2-mediated vocal control is independent of its β-catenin-binding domain but dependent of its DNA binding ability. Patient mutations associated with developmental disorders, including autism spectrum disorders, disrupt the transcriptional repression effect of TCF7L2, while mice carrying those mutations display severe USV impairments. Therefore, we conclude that TCF7L2 orchestrates gene expression in midbrain to control vocal production through its DNA binding but not transcription activation domain.
Collapse
Affiliation(s)
- Huihui Qi
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,School of Medicine, Tsinghua University, Beijing, 100084, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Li Luo
- Tsinghua Laboratory of Brain and Intelligence (THBI), Tsinghua University, Beijing, 100084, China
| | - Caijing Lu
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Runze Chen
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Xianyao Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu, China
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Science, Beijing Normal University, Beijing, 100875, China
| | - Yichang Jia
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, 100084, China. .,School of Medicine, Tsinghua University, Beijing, 100084, China. .,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China. .,Tsinghua Laboratory of Brain and Intelligence (THBI), Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
9
|
Shekel I, Giladi S, Raykin E, Weiner M, Chalifa-Caspi V, Lederman D, Kofman O, Golan HM. Isolation-Induced Ultrasonic Vocalization in Environmental and Genetic Mice Models of Autism. Front Neurosci 2021; 15:769670. [PMID: 34880723 PMCID: PMC8645772 DOI: 10.3389/fnins.2021.769670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/18/2021] [Indexed: 11/20/2022] Open
Abstract
Studies in rodent models suggest that calls emitted by isolated pups serve as an early behavioral manifestation of communication deficits and autistic like behavior. Previous studies in our labs showed that gestational exposure to the pesticide chlorpyrifos (CPF) and the Mthfr-knock-out mice are associated with impaired social preference and restricted or repetitive behavior. To extend these studies, we examine how pup communication via ultrasonic vocalizations is altered in these ASD models. We implemented an unsupervised hierarchical clustering method based on the spectral properties of the syllables in order to exploit syllable classification to homogeneous categories while avoiding over-categorization. Comparative exploration of the spectral and temporal aspects of syllables emitted by pups in two ASD models point to the following: (1) Most clusters showed a significant effect of the ASD factor on the start and end frequencies and bandwidth and (2) The highest percent change due to the ASD factor was on the bandwidth and duration. In addition, we found sex differences in the spectral and temporal properties of the calls in both control groups as well as an interaction between sex and the gene/environment factor. Considering the basal differences in the characteristics of syllables emitted by pups of the C57Bl/6 and Balb/c strains used as a background in the two models, we suggest that the above spectral-temporal parameters start frequency, bandwidth, and duration are the most sensitive USV features that may represent developmental changes in ASD models.
Collapse
Affiliation(s)
- Itay Shekel
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Shaked Giladi
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Eynav Raykin
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Department of Psychology, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - May Weiner
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Vered Chalifa-Caspi
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Dror Lederman
- Faculty of Engineering, Holon Institute of Technology, Holon, Israel
| | - Ora Kofman
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Department of Psychology, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Hava M Golan
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,National Center for Autism Research, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|
10
|
Sanjuán J, Castro-Martínez XH, García-Martí G, González-Fernández J, Sanz-Requena R, Haro JM, Meana JJ, Martí-Bonmatí L, Nacher J, Sebastiá-Ortega N, Gilabert-Juan J, Moltó MD. FOXP2 expression and gray matter density in the male brains of patients with schizophrenia. Brain Imaging Behav 2021; 15:1403-1411. [PMID: 32734433 PMCID: PMC8286223 DOI: 10.1007/s11682-020-00339-x] [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] [Indexed: 02/08/2023]
Abstract
Common genetic variants of FOXP2 may contribute to schizophrenia vulnerability, but controversial results have been reported for this proposal. Here we evaluated the potential impact of the common FOXP2 rs2396753 polymorphism in schizophrenia. It was previously reported to be part of a risk haplotype for this disease and to have significant effects on gray matter concentration in the patients. We undertook the first examination into whether rs2396753 affects the brain expression of FOXP2 and a replication study of earlier neuroimaging findings of the influence of this genetic variant on brain structure. FOXP2 expression levels were measured in postmortem prefrontal cortex samples of 84 male subjects (48 patients and 36 controls) from the CIBERSAM Brain and the Stanley Foundation Array Collections. High-resolution anatomical magnetic resonance imaging was performed on 79 male subjects (61 patients, 18 controls) using optimized voxel-based morphometry. We found differences in FOXP2 expression and brain morphometry depending on the rs2396753, relating low FOXP2 mRNA levels with reduction of gray matter density. We detected an interaction between rs2396753 and the clinical groups, showing that heterozygous patients for this polymorphism have gray matter density decrease and low FOXP2 expression comparing with the heterozygous controls. This study shows the importance of independent replication of neuroimaging genetic studies of FOXP2 as a candidate gene in schizophrenia. Furthermore, our results suggest that the FOXP2 rs2396753 affects mRNA levels, thus providing new knowledge about its significance as a potential susceptibility polymorphism in schizophrenia.
Collapse
Affiliation(s)
- Julio Sanjuán
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain.,Unit of Psychiatry, University of Valencia, Valencia, Spain.,INCLIVA Biomedical Research Institute, Fundación Investigación Hospital Clínico de Valencia, Valencia, Spain
| | - Xochitl Helga Castro-Martínez
- Department of Genetics, University of Valencia, Valencia, Spain.,Laboratorio de Genómica de Enfermedades Psiquiátricas y Neurodegenerativas, INMEGEN, Ciudad de México, México
| | - Gracián García-Martí
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain.,Biomedical Engineering Unit / Radiology Department, Quirónsalud Hospital, Valencia, Spain
| | | | - Roberto Sanz-Requena
- Biomedical Engineering Unit / Radiology Department, Quirónsalud Hospital, Valencia, Spain
| | - Josep María Haro
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain.,Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Deu, Barcelona, Spain
| | - J Javier Meana
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain.,Department of Pharmacology, Universidad del País Vasco/Euskal Herriko Unibertsitatea UPV/EHU, Leioa, Spain
| | - Luis Martí-Bonmatí
- Biomedical Engineering Unit / Radiology Department, Quirónsalud Hospital, Valencia, Spain
| | - Juan Nacher
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain.,INCLIVA Biomedical Research Institute, Fundación Investigación Hospital Clínico de Valencia, Valencia, Spain.,Neurobiology Unit, Cell Biology Department, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, Spain
| | - Noelia Sebastiá-Ortega
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain.,Department of Genetics, University of Valencia, Valencia, Spain
| | - Javier Gilabert-Juan
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain. .,INCLIVA Biomedical Research Institute, Fundación Investigación Hospital Clínico de Valencia, Valencia, Spain. .,Department of Genetics, University of Valencia, Valencia, Spain. .,Neurobiology Unit, Cell Biology Department, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, Spain. .,Department of Genetics, Universitat de València, Dr. Moliner 50, 46100, Burjassot, Valencia, Spain.
| | - María Dolores Moltó
- Spanish National Network for Research in Mental Health CIBERSAM, Valencia, Spain. .,INCLIVA Biomedical Research Institute, Fundación Investigación Hospital Clínico de Valencia, Valencia, Spain. .,Department of Genetics, University of Valencia, Valencia, Spain. .,Department of Genetics, Universitat de València, Dr. Moliner 50, 46100, Burjassot, Valencia, Spain.
| |
Collapse
|
11
|
Santos-Terra J, Deckmann I, Fontes-Dutra M, Schwingel GB, Bambini-Junior V, Gottfried C. Transcription factors in neurodevelopmental and associated psychiatric disorders: A potential convergence for genetic and environmental risk factors. Int J Dev Neurosci 2021; 81:545-578. [PMID: 34240460 DOI: 10.1002/jdn.10141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/23/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a heterogeneous and highly prevalent group of psychiatric conditions marked by impairments in the nervous system. Their onset occurs during gestation, and the alterations are observed throughout the postnatal life. Although many genetic and environmental risk factors have been described in this context, the interactions between them challenge the understanding of the pathways associated with NDDs. Transcription factors (TFs)-a group of over 1,600 proteins that can interact with DNA, regulating gene expression through modulation of RNA synthesis-represent a point of convergence for different risk factors. In addition, TFs organize critical processes like angiogenesis, blood-brain barrier formation, myelination, neuronal migration, immune activation, and many others in a time and location-dependent way. In this review, we summarize important TF alterations in NDD and associated disorders, along with specific impairments observed in animal models, and, finally, establish hypotheses to explain how these proteins may be critical mediators in the context of genome-environment interactions.
Collapse
Affiliation(s)
- Júlio Santos-Terra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Iohanna Deckmann
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Mellanie Fontes-Dutra
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Gustavo Brum Schwingel
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| | - Victorio Bambini-Junior
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Preston, UK
| | - Carmem Gottfried
- Translational Research Group in Autism Spectrum Disorders (GETTEA), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Autism Wellbeing And Research Development (AWARD) Institute, BR-UK-CA, Preston, UK
| |
Collapse
|
12
|
Lenell C, Broadfoot CK, Schaen-Heacock NE, Ciucci MR. Biological and Acoustic Sex Differences in Rat Ultrasonic Vocalization. Brain Sci 2021; 11:459. [PMID: 33916537 PMCID: PMC8067311 DOI: 10.3390/brainsci11040459] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/26/2021] [Accepted: 04/01/2021] [Indexed: 11/30/2022] Open
Abstract
The rat model is a useful tool for understanding peripheral and central mechanisms of laryngeal biology. Rats produce ultrasonic vocalizations (USVs) that have communicative intent and are altered by experimental conditions such as social environment, stress, diet, drugs, age, and neurological diseases, validating the rat model's utility for studying communication and related deficits. Sex differences are apparent in both the rat larynx and USV acoustics and are differentially affected by experimental conditions. Therefore, the purpose of this review paper is to highlight the known sex differences in rat USV production, acoustics, and laryngeal biology detailed in the literature across the lifespan.
Collapse
Affiliation(s)
- Charles Lenell
- Department of Surgery, University of Wisconsin Madison, Madison, WI 53792, USA; (C.L.); (C.K.B.); (N.E.S.-H.)
- Communicative Sciences and Disorders, New York University, New York, NY 10001, USA
| | - Courtney K. Broadfoot
- Department of Surgery, University of Wisconsin Madison, Madison, WI 53792, USA; (C.L.); (C.K.B.); (N.E.S.-H.)
- Department of Communication Sciences and Disorders, University of Wisconsin Madison, Madison, WI 53706, USA
| | - Nicole E. Schaen-Heacock
- Department of Surgery, University of Wisconsin Madison, Madison, WI 53792, USA; (C.L.); (C.K.B.); (N.E.S.-H.)
- Department of Communication Sciences and Disorders, University of Wisconsin Madison, Madison, WI 53706, USA
| | - Michelle R. Ciucci
- Department of Surgery, University of Wisconsin Madison, Madison, WI 53792, USA; (C.L.); (C.K.B.); (N.E.S.-H.)
- Department of Communication Sciences and Disorders, University of Wisconsin Madison, Madison, WI 53706, USA
| |
Collapse
|
13
|
Age and Sex-Dependent ADNP Regulation of Muscle Gene Expression Is Correlated with Motor Behavior: Possible Feedback Mechanism with PACAP. Int J Mol Sci 2020; 21:ijms21186715. [PMID: 32937737 PMCID: PMC7555576 DOI: 10.3390/ijms21186715] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022] Open
Abstract
The activity-dependent neuroprotective protein (ADNP), a double-edged sword, sex-dependently regulates multiple genes and was previously associated with the control of early muscle development and aging. Here we aimed to decipher the involvement of ADNP in versatile muscle gene expression patterns in correlation with motor function throughout life. Using quantitative RT-PCR we showed that Adnp+/− heterozygous deficiency in mice resulted in aberrant gastrocnemius (GC) muscle, tongue and bladder gene expression, which was corrected by the Adnp snippet, drug candidate, NAP (CP201). A significant sexual dichotomy was discovered, coupled to muscle and age-specific gene regulation. As such, Adnp was shown to regulate myosin light chain (Myl) in the gastrocnemius (GC) muscle, the language acquisition gene forkhead box protein P2 (Foxp2) in the tongue and the pituitary-adenylate cyclase activating polypeptide (PACAP) receptor PAC1 mRNA (Adcyap1r1) in the bladder, with PACAP linked to bladder function. A tight age regulation was observed, coupled to an extensive correlation to muscle function (gait analysis), placing ADNP as a muscle-regulating gene/protein.
Collapse
|
14
|
Co M, Anderson AG, Konopka G. FOXP transcription factors in vertebrate brain development, function, and disorders. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e375. [PMID: 31999079 PMCID: PMC8286808 DOI: 10.1002/wdev.375] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/17/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
FOXP transcription factors are an evolutionarily ancient protein subfamily coordinating the development of several organ systems in the vertebrate body. Association of their genes with neurodevelopmental disorders has sparked particular interest in their expression patterns and functions in the brain. Here, FOXP1, FOXP2, and FOXP4 are expressed in distinct cell type-specific spatiotemporal patterns in multiple regions, including the cortex, hippocampus, amygdala, basal ganglia, thalamus, and cerebellum. These varied sites and timepoints of expression have complicated efforts to link FOXP1 and FOXP2 mutations to their respective developmental disorders, the former affecting global neural functions and the latter specifically affecting speech and language. However, the use of animal models, particularly those with brain region- and cell type-specific manipulations, has greatly advanced our understanding of how FOXP expression patterns could underlie disorder-related phenotypes. While many questions remain regarding FOXP expression and function in the brain, studies to date have illuminated the roles of these transcription factors in vertebrate brain development and have greatly informed our understanding of human development and disorders. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Marissa Co
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Ashley G Anderson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
15
|
Caruso A, Ricceri L, Scattoni ML. Ultrasonic vocalizations as a fundamental tool for early and adult behavioral phenotyping of Autism Spectrum Disorder rodent models. Neurosci Biobehav Rev 2020; 116:31-43. [DOI: 10.1016/j.neubiorev.2020.06.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 05/08/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
|
16
|
Riede T, Coyne M, Tafoya B, Baab KL. Postnatal Development of the Mouse Larynx: Negative Allometry, Age-Dependent Shape Changes, Morphological Integration, and a Size-Dependent Spectral Feature. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2020; 63:2680-2694. [PMID: 32762490 DOI: 10.1044/2020_jslhr-20-00070] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Purpose The larynx plays a role in swallowing, respiration, and voice production. All three functions change during ontogeny. We investigated ontogenetic shape changes using a mouse model to inform our understanding of how laryngeal form and function are integrated. We understand the characterization of developmental changes to larynx anatomy as a critical step toward using rodent models to study human vocal communication disorders. Method Contrast-enhanced micro-computed tomography image stacks were used to generate three-dimensional reconstructions of the CD-1 mouse (Mus musculus) laryngeal cartilaginous framework. Then, we quantified size and shape in four age groups: pups, weanlings, young, and old adults using a combination of landmark and linear morphometrics. We analyzed postnatal patterns of growth and shape in the laryngeal skeleton, as well as morphological integration among four laryngeal cartilages using geometric morphometric methods. Acoustic analysis of vocal patterns was employed to investigate morphological and functional integration. Results Four cartilages scaled with negative allometry on body mass. Additionally, thyroid, arytenoid, and epiglottic cartilages, but not the cricoid cartilage, showed shape change associated with developmental age. A test for modularity between the four cartilages suggests greater independence of thyroid cartilage shape, hinting at the importance of embryological origin during postnatal development. Finally, mean fundamental frequency, but not fundamental frequency range, varied predictably with size. Conclusion In a mouse model, the four main laryngeal cartilages do not develop uniformly throughout the first 12 months of life. High-dimensional shape analysis effectively quantified variation in shape across development and in relation to size, as well as clarifying patterns of covariation in shape among cartilages and possibly the ventral pouch. Supplemental Material https://doi.org/10.23641/asha.12735917.
Collapse
Affiliation(s)
- Tobias Riede
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, AZ
- College of Veterinary Medicine, Midwestern University, Glendale, AZ
| | - Megan Coyne
- College of Veterinary Medicine, Midwestern University, Glendale, AZ
| | - Blake Tafoya
- College of Veterinary Medicine, Midwestern University, Glendale, AZ
| | - Karen L Baab
- Department of Anatomy, College of Graduate Studies, Midwestern University, Glendale, AZ
| |
Collapse
|
17
|
Taylor MR, Roby CR, Elziny S, Duricy E, Taylor TM, Bowers JM. Age, but Not Sex, Modulates Foxp3 Expression in the Rat Brain across Development. Neuroscience 2020; 442:87-99. [PMID: 32599120 DOI: 10.1016/j.neuroscience.2020.06.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 06/17/2020] [Accepted: 06/20/2020] [Indexed: 12/18/2022]
Abstract
The interconnectivity between brain development and the immune system has become an area of interest for many neuroscientists. However, to date, a limited number of known immune mediators of the peripheral nervous system (PNS) have been found to influence the development of the central nervous system (CNS). FOXP3 is a well-established mediator of regulatory T-cells in the PNS. However, the expression pattern of FOXP3 in the CNS and the PNS throughout development is unknown. To fill this void, we have characterized, in several brain regions, the developmental profile of Foxp3 for both sexes using rats. We found different patterns of Foxp3 in the CNS and PNS. In the CNS, we found Foxp3 was ubiquitously expressed, with the levels of Foxp3 varying by brain region. We also found both Foxp3 mRNA and protein levels peak during embryonic development and then steadily decrease with a peak increase during adulthood. In adulthood, the protein but not mRNA increases to the equivalent levels found at the embryonic stage of life. In the PNS, Foxp3 protein levels were low embryonically and increased steadily over the life of the animal with maximal levels reached in adulthood. Patterns observed for both the PNS and CNS were similar in males and females across all developmental timepoints. Our novel findings have implications for understanding how the neural immune system impacts neurodevelopmental disorders such as autism and schizophrenia.
Collapse
Affiliation(s)
- Makenzlie R Taylor
- School of Neuroscience, 1981 Kraft Drive, ILSB, Virginia Tech, Blacksburg, VA 24061-0913, USA
| | - Clinton R Roby
- School of Neuroscience, 1981 Kraft Drive, ILSB, Virginia Tech, Blacksburg, VA 24061-0913, USA
| | - Soad Elziny
- School of Neuroscience, 1981 Kraft Drive, ILSB, Virginia Tech, Blacksburg, VA 24061-0913, USA
| | - Erin Duricy
- School of Neuroscience, 1981 Kraft Drive, ILSB, Virginia Tech, Blacksburg, VA 24061-0913, USA
| | - Tina M Taylor
- School of Neuroscience, 1981 Kraft Drive, ILSB, Virginia Tech, Blacksburg, VA 24061-0913, USA
| | - J Michael Bowers
- School of Neuroscience, 1981 Kraft Drive, ILSB, Virginia Tech, Blacksburg, VA 24061-0913, USA.
| |
Collapse
|
18
|
Xiang D, Lu J, Wei C, Cai X, Wang Y, Liang Y, Xu M, Wang Z, Liu M, Wang M, Liang X, Li L, Yao P. Berberine Ameliorates Prenatal Dihydrotestosterone Exposure-Induced Autism-Like Behavior by Suppression of Androgen Receptor. Front Cell Neurosci 2020; 14:87. [PMID: 32327976 PMCID: PMC7161090 DOI: 10.3389/fncel.2020.00087] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/23/2020] [Indexed: 01/03/2023] Open
Abstract
Many epidemiology studies have shown that maternal polycystic ovary syndrome (PCOS) results in a greater risk of autism spectrum disorders (ASD) development, although the detailed mechanism remains unclear. In this study, we aimed to investigate the potential mechanism and provide a possible treatment for PCOS-mediated ASD through three experiments: Experiment 1: real-time PCR and western blots were employed to measure gene expression in human neurons, and the luciferase reporter assay and chromatin immunoprecipitation (ChIP) was used to map the responsive elements on related gene promoters. Experiment 2: pregnant dams were prenatally exposed to dihydrotestosterone (DHT), androgen receptor (AR) knockdown (shAR) in the amygdala, or berberine (BBR), and the subsequent male offspring were used for autism-like behavior (ALB) assay followed by biomedical analysis, including gene expression, oxidative stress, and mitochondrial function. Experiment 3: the male offspring from prenatal DHT exposed dams were postnatally treated by either shAR or BBR, and the offspring were used for ALB assay followed by biomedical analysis. Our findings showed that DHT treatment suppresses the expression of estrogen receptor β (ERβ) and superoxide dismutase 2 (SOD2) through AR-mediated hypermethylation on the ERβ promoter, and BBR treatment suppresses AR expression through hypermethylation on the AR promoter. Prenatal DHT treatment induces ERβ suppression, oxidative stress and mitochondria dysfunction in the amygdala with subsequent ALB behavior in male offspring, and AR knockdown partly diminishes this effect. Furthermore, both prenatal and postnatal treatment of BBR partly restores prenatal DHT exposure-mediated ALB. In conclusion, DHT suppresses ERβ expression through the AR signaling pathway by hypermethylation on the ERβ promoter, and BBR restores this effect through AR suppression. Prenatal DHT exposure induces ALB in offspring through AR-mediated ERβ suppression, and both prenatal and postnatal treatment of BBR ameliorates this effect. We conclude that BBR ameliorates prenatal DHT exposure-induced ALB through AR suppression, this study may help elucidate the potential mechanism and identify a potential treatment through using BBR for PCOS-mediated ASD.
Collapse
Affiliation(s)
- Dongfang Xiang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianping Lu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Chongxia Wei
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Xiaofan Cai
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Yongxia Wang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujie Liang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Mingtao Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zichen Wang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Min Liu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Wang
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Xuefang Liang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Li
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Paul Yao
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China.,Hainan Maternal and Child Health Hospital, Haikou, China
| |
Collapse
|
19
|
Alteration in the time and/or mode of delivery differentially modulates early development in mice. Mol Brain 2020; 13:34. [PMID: 32151280 PMCID: PMC7063737 DOI: 10.1186/s13041-020-00578-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 03/02/2020] [Indexed: 12/27/2022] Open
Abstract
Delivery is a complex biological process involving hormonal and mechanical stimuli that together condition the survival and development of the fetus out of the womb. Accordingly, changes in the time or way of being born are associated with an alteration of fundamental biological functions and hypothesized to promote the emergence of neurodevelopmental disorders. Hence, the steadily rise in preterm birth and cesarean section (CS) delivery rates over the past years has become a worldwide health concern. In our previous work, we reported that even though no long-term autistic-like deficits were observed, mice born preterm by CS presented early transient neuronal and communicative defects. However, understanding if these alterations were due to an early birth combined with CS delivery, or if prematurity solely could lead to a similar outcome remained to be evaluated. Using mice born either at term or preterm by vaginal or CS delivery, we assessed early life ultrasonic vocalizations and the onset of eye opening. We report that alterations in communicative behaviors are finely attuned and specifically affected either by preterm birth or by the association between CS delivery and preterm birth in mice, while delayed onset of eye opening is due to prematurity. Moreover, our work further underlies a gender-dependent vulnerability to changes in the time and/or way of being born with distinct outcomes observed in males and females. Thus, our results shed light on the intricacy of birth alterations and might further explain the disparities reported in epidemiological studies.
Collapse
|
20
|
Liu P, Huang S, Ling S, Xu S, Wang F, Zhang W, Zhou R, He L, Xia X, Yao Z, Fan Y, Wang N, Hu C, Zhao X, Tucker HO, Wang J, Guo X. Foxp1 controls brown/beige adipocyte differentiation and thermogenesis through regulating β3-AR desensitization. Nat Commun 2019; 10:5070. [PMID: 31699980 PMCID: PMC6838312 DOI: 10.1038/s41467-019-12988-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/02/2019] [Indexed: 01/08/2023] Open
Abstract
β-Adrenergic receptor (β-AR) signaling is a pathway controlling adaptive thermogenesis in brown or beige adipocytes. Here we investigate the biological roles of the transcription factor Foxp1 in brown/beige adipocyte differentiation and thermogenesis. Adipose-specific deletion of Foxp1 leads to an increase of brown adipose activity and browning program of white adipose tissues. The Foxp1-deficient mice show an augmented energy expenditure and are protected from diet-induced obesity and insulin resistance. Consistently, overexpression of Foxp1 in adipocytes impairs adaptive thermogenesis and promotes diet-induced obesity. A robust change in abundance of the β3-adrenergic receptor (β3-AR) is observed in brown/beige adipocytes from both lines of mice. Molecularly, Foxp1 directly represses β3-AR transcription and regulates its desensitization behavior. Taken together, our findings reveal Foxp1 as a master transcriptional repressor of brown/beige adipocyte differentiation and thermogenesis, and provide an important clue for its targeting and treatment of obesity. Beta3-adrenergic receptor (b3-AR) signaling in response to cold activates adipose tissue thermogenesis. Here the authors identify the transcription factor FoxP1 as a direct negative regulator of b3-AR expression and show that loss of FoxP1 leads to enhanced development of thermogenic adipose tissue.
Collapse
Affiliation(s)
- Pei Liu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Sixia Huang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shifeng Ling
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuqin Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fuhua Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rujiang Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuechun Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhengju Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Fan
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Congxia Hu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaodong Zhao
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haley O Tucker
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xizhi Guo
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
21
|
O'Connor AM, Burton TJ, Mansuri H, Hand GR, Leamey CA, Sawatari A. Environmental Enrichment From Birth Impacts Parvalbumin Expressing Cells and Wisteria Floribunda Agglutinin Labelled Peri-Neuronal Nets Within the Developing Murine Striatum. Front Neuroanat 2019; 13:90. [PMID: 31708753 PMCID: PMC6821641 DOI: 10.3389/fnana.2019.00090] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 10/01/2019] [Indexed: 11/13/2022] Open
Abstract
Environmental enrichment can dramatically affect both the development and function of neural circuits. This is accomplished, at least in part, by the regulation of inhibitory cellular networks and related extracellular matrix glycoprotein structures known as perineuronal nets. The degree to which enhanced housing can influence brain areas involved in the planning and execution of actions is not well known. We examined the effect of enriching mice from birth on parvalbumin expression and perineuronal net formation in developing and adult striatum. This input nucleus of the basal ganglia consists of topographically discernible regions that serve different functions, providing a means of simultaneously examining the influence of environmental factors on discrete, but related networks. Greater densities of striatal parvalbumin positive cells and wisteria floribunda agglutinin labelled perineuronal nets were present in enriched pups during the second postnatal week, primarily within the lateral portion of the nucleus. Housing conditions continued to have an impact into adulthood, with enriched mice exhibiting higher parvalbumin positive cell densities in both medial and lateral striatum. Curiously, no differences due to housing conditions were detected in striatal perineuronal net densities of mature animals. The degree of overlap between striatal parvalbumin expression and perineuronal net formation was also increased, suggesting that heightened neural activity associated with enrichment may have contributed to greater engagement of networks affiliated with cells that express the calcium binding protein. Brain derived neurotrophic factor, an important regulator of inhibitory network maturation, is also subtly, but significantly affected within the striatum of enriched cohorts. Together, these findings suggest that environmental enrichment can exert cell specific effects within different divisions of an area vital for the regulation of action.
Collapse
Affiliation(s)
- Angela May O'Connor
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Thomas Joseph Burton
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Hannan Mansuri
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Gabriel Rhys Hand
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Catherine Anne Leamey
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Atomu Sawatari
- Systems Neuroscience Laboratory, Discipline of Physiology, School of Medical Sciences and the Bosch Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Sullivan JM, De Rubeis S, Schaefer A. Convergence of spectrums: neuronal gene network states in autism spectrum disorder. Curr Opin Neurobiol 2019; 59:102-111. [PMID: 31220745 DOI: 10.1016/j.conb.2019.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/20/2019] [Accepted: 04/24/2019] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by social deficits and restrictive and/or repetitive behaviors. The breadth of ASD symptoms is paralleled by the multiplicity of genes that have been implicated in its etiology. Initial findings revealed numerous ASD risk genes that contribute to synaptic function. More recently, genomic and gene expression studies point to altered chromatin function and impaired transcriptional control as additional risk factors for ASD. The consequences of impaired transcriptional alterations in ASD involve consistent changes in synaptic gene expression and cortical neuron specification during brain development. The multiplicity of genetic and environmental factors associated with ASD risk and their convergence onto common molecular pathways in neurons point to ASD as a disorder of gene regulatory networks.
Collapse
Affiliation(s)
- Josefa M Sullivan
- Nash Family Department of Neuroscience, New York, NY, USA; Department of Psychiatry, New York, NY, USA; Friedman Brain Institute, New York, NY, USA; Seaver Autism Center for Research and Treatment, New York, NY, USA
| | - Silvia De Rubeis
- Department of Psychiatry, New York, NY, USA; Seaver Autism Center for Research and Treatment, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anne Schaefer
- Nash Family Department of Neuroscience, New York, NY, USA; Department of Psychiatry, New York, NY, USA; Friedman Brain Institute, New York, NY, USA; Seaver Autism Center for Research and Treatment, New York, NY, USA.
| |
Collapse
|
23
|
McNamara GI, Creeth HDJ, Harrison DJ, Tansey KE, Andrews RM, Isles AR, John RM. Loss of offspring Peg3 reduces neonatal ultrasonic vocalizations and increases maternal anxiety in wild-type mothers. Hum Mol Genet 2019; 27:440-450. [PMID: 29186532 PMCID: PMC5886183 DOI: 10.1093/hmg/ddx412] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
Depression and anxiety are the most common mental health conditions during pregnancy and can impair the normal development of mother-infant interactions. These adversities are associated with low birth weight and increased risk of behavioural disorders in children. We recently reported reduced expression of the imprinted gene PATERNALLY EXPRESSED GENE 3 (PEG3) in placenta of human infants born to depressed mothers. Expression of Peg3 in the brain has previously been linked maternal behaviour in rodents, at least in some studies, with mutant dams neglecting their pups. However, in our human study decreased expression was in the placenta derived from the fetus. Here, we examined maternal behaviour in response to reduced expression of Peg3 in the feto-placental unit. Prenatally we found novelty reactivity was altered in wild-type females carrying litters with a null mutation in Peg3. This behavioural alteration was short-lived and there were no significant differences the transcriptomes of either the maternal hypothalamus or hippocampus at E16.5. In contrast, while maternal gross maternal care was intact postnatally, the exposed dams were significantly slower to retrieve their pups and displayed a marked increase in anxiety. We also observed a significant reduction in the isolation-induced ultrasonic vocalizations (USVs) emitted by mutant pups separated from their mothers. USVs are a form of communication known to elicit maternal care suggesting Peg3 mutant pups drive the deficit in maternal behaviour. These data support the hypothesis that reduced placental PEG3 in human pregnancies occurs as a consequence of prenatal depression but leaves scope for feto-placental Peg3 dosage, during gestation, influencing aspects of maternal behaviour.
Collapse
Affiliation(s)
- G I McNamara
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - H D J Creeth
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - D J Harrison
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - K E Tansey
- Core Bioinformatics and Statistics Team, College of Biomedical & Life Sciences
| | - R M Andrews
- Systems Immunity University Research Institute, Cardiff University, Cardiff CF10 3XQ, UK
| | - A R Isles
- MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - R M John
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| |
Collapse
|
24
|
Castellucci GA, Calbick D, McCormick D. The temporal organization of mouse ultrasonic vocalizations. PLoS One 2018; 13:e0199929. [PMID: 30376572 PMCID: PMC6207298 DOI: 10.1371/journal.pone.0199929] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/15/2018] [Indexed: 12/30/2022] Open
Abstract
House mice, like many tetrapods, produce multielement calls consisting of individual vocalizations repeated in rhythmic series. In this study, we examine the multielement ultrasonic vocalizations (USVs) of adult male C57Bl/6J mice and specifically assess their temporal properties and organization. We found that male mice produce two classes of USVs which display unique temporal features and arise from discrete respiratory patterns. We also observed that nearly all USVs were produced in repetitive series exhibiting a hierarchical organization and a stereotyped rhythmic structure. Furthermore, series rhythmicity alone was determined to be sufficient for the mathematical discrimination of USVs produced by adult males, adult females, and pups, underscoring the known importance of call timing in USV perception. Finally, the gross spectrotemporal features of male USVs were found to develop continuously from birth and stabilize by P50, suggesting that USV production in infants and adults relies on common biological mechanisms. In conclusion, we demonstrate that the temporal organization of multielement mouse USVs is both stable and informative, and we propose that call timing be explicitly assessed when examining mouse USV production. Furthermore, this is the first report of putative USV classes arising from distinct articulatory patterns in mice, and is the first to empirically define multielement USV series and provide a detailed description of their temporal structure and development. This study therefore represents an important point of reference for the analysis of mouse USVs, a commonly used metric of social behavior in mouse models of human disease, and furthers the understanding of vocalization production in an accessible mammalian species.
Collapse
Affiliation(s)
- Gregg A. Castellucci
- Neuroscience Institute, New York University School of Medicine, New York, NY, United States of America
- Haskins Laboratories, New Haven, CT, United States of America
- Department of Genetics, Yale University of Medicine, New Haven, CT, United States of America
| | - Daniel Calbick
- Department of Genetics, Yale University of Medicine, New Haven, CT, United States of America
| | - David McCormick
- Institute of Neuroscience, University of Oregon, Eugene, OR, United States of America
- Department of Biology, University of Oregon, Eugene, OR, United States of America
| |
Collapse
|
25
|
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: 4.2] [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
|
26
|
Foxp2 regulates anatomical features that may be relevant for vocal behaviors and bipedal locomotion. Proc Natl Acad Sci U S A 2018; 115:8799-8804. [PMID: 30104377 DOI: 10.1073/pnas.1721820115] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Fundamental human traits, such as language and bipedalism, are associated with a range of anatomical adaptations in craniofacial shaping and skeletal remodeling. However, it is unclear how such morphological features arose during hominin evolution. FOXP2 is a brain-expressed transcription factor implicated in a rare disorder involving speech apraxia and language impairments. Analysis of its evolutionary history suggests that this gene may have contributed to the emergence of proficient spoken language. In the present study, through analyses of skeleton-specific knockout mice, we identified roles of Foxp2 in skull shaping and bone remodeling. Selective ablation of Foxp2 in cartilage disrupted pup vocalizations in a similar way to that of global Foxp2 mutants, which may be due to pleiotropic effects on craniofacial morphogenesis. Our findings also indicate that Foxp2 helps to regulate strength and length of hind limbs and maintenance of joint cartilage and intervertebral discs, which are all anatomical features that are susceptible to adaptations for bipedal locomotion. In light of the known roles of Foxp2 in brain circuits that are important for motor skills and spoken language, we suggest that this gene may have been well placed to contribute to coevolution of neural and anatomical adaptations related to speech and bipedal locomotion.
Collapse
|
27
|
Braccioli L, Vervoort SJ, Adolfs Y, Heijnen CJ, Basak O, Pasterkamp RJ, Nijboer CH, Coffer PJ. FOXP1 Promotes Embryonic Neural Stem Cell Differentiation by Repressing Jagged1 Expression. Stem Cell Reports 2018; 9:1530-1545. [PMID: 29141232 PMCID: PMC5688236 DOI: 10.1016/j.stemcr.2017.10.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 01/11/2023] Open
Abstract
Mutations in FOXP1 have been linked to neurodevelopmental disorders including intellectual disability and autism; however, the underlying molecular mechanisms remain ill-defined. Here, we demonstrate with RNA and chromatin immunoprecipitation sequencing that FOXP1 directly regulates genes controlling neurogenesis. We show that FOXP1 is expressed in embryonic neural stem cells (NSCs), and modulation of FOXP1 expression affects both neuron and astrocyte differentiation. Using a murine model of cortical development, FOXP1-knockdown in utero was found to reduce NSC differentiation and migration during corticogenesis. Furthermore, transplantation of FOXP1-knockdown NSCs in neonatal mice after hypoxia-ischemia challenge demonstrated that FOXP1 is also required for neuronal differentiation and functionality in vivo. FOXP1 was found to repress the expression of Notch pathway genes including the Notch-ligand Jagged1, resulting in inhibition of Notch signaling. Finally, blockade of Jagged1 in FOXP1-knockdown NSCs rescued neuronal differentiation in vitro. Together, these data support a role for FOXP1 in regulating embryonic NSC differentiation by modulating Notch signaling. FOXP1 promotes astrocyte and neuronal differentiation of NSCs in vitro FOXP1 promotes neuronal differentiation of NSCs in vivo FOXP1 transcriptionally regulates pro-neural genes and represses Notch pathway genes FOXP1 promotes neuronal differentiation by limiting Jagged1 expression
Collapse
Affiliation(s)
- Luca Braccioli
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands; Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Stephin J Vervoort
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Onur Basak
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, 3584 CT Utrecht, the Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht 3584 CX, the Netherlands
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease (NIDOD), University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands.
| | - Paul J Coffer
- Center for Molecular Medicine and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584 CT, the Netherlands.
| |
Collapse
|
28
|
Bell MR. Comparing Postnatal Development of Gonadal Hormones and Associated Social Behaviors in Rats, Mice, and Humans. Endocrinology 2018; 159:2596-2613. [PMID: 29767714 PMCID: PMC6692888 DOI: 10.1210/en.2018-00220] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/08/2018] [Indexed: 12/20/2022]
Abstract
Postnatal development includes dramatic changes in gonadal hormones and the many social behaviors they help regulate, both in rodents and humans. Parental care-seeking is the most salient social interaction in neonates and infants, play and prosocial behaviors are commonly studied in juveniles, and the development of aggression and sexual behavior begins in peripubertal stages but continues through late adolescence into adulthood. Although parental behaviors are shown after reproductive success in adulthood, alloparenting behaviors are actually high in juveniles as well. These behaviors are sensitive to both early-life organizational effects of gonadal hormones and later-life activational regulation. However, changes in circulating gonadal hormones and the display of the previous behaviors over development differ between rats, mice, and humans. These endpoints are of interest to endocrinologist, toxicologists, and neuroscientists because of their relevance to mental health disorders and their vulnerability to effects of endocrine-disrupting chemical exposure. As such, the goal of this mini-review is to succinctly describe and relate the postnatal development of gonadal hormones and social behaviors to each other, over time, and across animal models. Ideally, this will help identify appropriate animal models and age ranges for continued study of both normative development and in contexts of environmental disruption.
Collapse
Affiliation(s)
- Margaret R Bell
- Department of Biological Sciences, DePaul University, Chicago, Illinois
- Department of Health Sciences, DePaul University, Chicago, Illinois
| |
Collapse
|
29
|
Usui N, Araujo DJ, Kulkarni A, Co M, Ellegood J, Harper M, Toriumi K, Lerch JP, Konopka G. Foxp1 regulation of neonatal vocalizations via cortical development. Genes Dev 2017; 31:2039-2055. [PMID: 29138280 PMCID: PMC5733496 DOI: 10.1101/gad.305037.117] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022]
Abstract
Usui et al. show that deletion of Foxp1 in the developing forebrain leads to impairments in neonatal vocalizations as well as neocortical cytoarchitectonic alterations via neuronal positioning and migration. Sumoylation of Foxp1 affects neuronal differentiation and migration in the developing neocortex. The molecular mechanisms driving brain development at risk in autism spectrum disorders (ASDs) remain mostly unknown. Previous studies have implicated the transcription factor FOXP1 in both brain development and ASD pathophysiology. However, the specific molecular pathways both upstream of and downstream from FOXP1 are not fully understood. To elucidate the contribution of FOXP1-mediated signaling to brain development and, in particular, neocortical development, we generated forebrain-specific Foxp1 conditional knockout mice. We show that deletion of Foxp1 in the developing forebrain leads to impairments in neonatal vocalizations as well as neocortical cytoarchitectonic alterations via neuronal positioning and migration. Using a genomics approach, we identified the transcriptional networks regulated by Foxp1 in the developing neocortex and found that such networks are enriched for downstream targets involved in neurogenesis and neuronal migration. We also uncovered mechanistic insight into Foxp1 function by demonstrating that sumoylation of Foxp1 during embryonic brain development is necessary for mediating proper interactions between Foxp1 and the NuRD complex. Furthermore, we demonstrated that sumoylation of Foxp1 affects neuronal differentiation and migration in the developing neocortex. Together, these data provide critical mechanistic insights into the function of FOXP1 in the developing neocortex and may reveal molecular pathways at risk in ASD.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Division of Development of Mental Functions, Research Center for Child Mental Development, University of Fukui, Fukui 910-1193, Japan.,Division of Developmental Higher Brain Functions, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Osaka 565-0871, Japan
| | - Daniel J Araujo
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Marissa Co
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario M5S 1A1, Canada
| | - Matthew Harper
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kazuya Toriumi
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Project for Schizophrenia Research, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Jason P Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Ontario M5S 1A1, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| | - Genevieve Konopka
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
30
|
Siper PM, De Rubeis S, Trelles MDP, Durkin A, Di Marino D, Muratet F, Frank Y, Lozano R, Eichler EE, Kelly M, Beighley J, Gerdts J, Wallace AS, Mefford HC, Bernier RA, Kolevzon A, Buxbaum JD. Prospective investigation of FOXP1 syndrome. Mol Autism 2017; 8:57. [PMID: 29090079 PMCID: PMC5655854 DOI: 10.1186/s13229-017-0172-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
Background Haploinsufficiency of the forkhead-box protein P1 (FOXP1) gene leads to a neurodevelopmental disorder termed FOXP1 syndrome. Previous studies in individuals carrying FOXP1 mutations and deletions have described the presence of autism spectrum disorder (ASD) traits, intellectual disability, language impairment, and psychiatric features. The goal of the present study was to comprehensively characterize the genetic and clinical spectrum of FOXP1 syndrome. This is the first study to prospectively examine the genotype-phenotype relationship in multiple individuals with FOXP1 syndrome, using a battery of standardized clinical assessments. Methods Genetic and clinical data was obtained and analyzed from nine children and adolescents between the ages of 5–17 with mutations in FOXP1. Phenotypic characterization included gold standard ASD testing and norm-referenced measures of cognition, adaptive behavior, language, motor, and visual-motor integration skills. In addition, psychiatric, medical, neurological, and dysmorphology examinations were completed by a multidisciplinary team of clinicians. A comprehensive review of reported cases was also performed. All missense and in-frame mutations were mapped onto the three-dimensional structure of DNA-bound FOXP1. Results We have identified nine de novo mutations, including three frameshift, one nonsense, one mutation in an essential splice site resulting in frameshift and insertion of a premature stop codon, three missense, and one in-frame deletion. Reviewing prior literature, we found seven instances of recurrent mutations and another 34 private mutations. The majority of pathogenic missense and in-frame mutations, including all four missense mutations in our cohort, lie in the DNA-binding domain. Through structural analyses, we show that the mutations perturb amino acids necessary for binding to the DNA or interfere with the domain swapping that mediates FOXP1 dimerization. Individuals with FOXP1 syndrome presented with delays in early motor and language milestones, language impairment (expressive language > receptive language), ASD symptoms, visual-motor integration deficits, and complex psychiatric presentations characterized by anxiety, obsessive-compulsive traits, attention deficits, and externalizing symptoms. Medical features included non-specific structural brain abnormalities and dysmorphic features, endocrine and gastrointestinal problems, sleep disturbances, and sinopulmonary infections. Conclusions This study identifies novel FOXP1 mutations associated with FOXP1 syndrome, identifies recurrent mutations, and demonstrates significant clustering of missense mutations in the DNA-binding domain. Clinical findings confirm the role FOXP1 plays in development across multiple domains of functioning. The genetic findings can be incorporated into clinical genetics practice to improve accurate genetic diagnosis of FOXP1 syndrome and the clinical findings can inform monitoring and treatment of individuals with FOXP1 syndrome. Electronic supplementary material The online version of this article (10.1186/s13229-017-0172-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paige M Siper
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Silvia De Rubeis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Maria Del Pilar Trelles
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Allison Durkin
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Daniele Di Marino
- Department of Informatics, Institute of Computational Science, Università della Svizzera Italiana, Lugano, Switzerland
| | - François Muratet
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Yitzchak Frank
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Reymundo Lozano
- Department of Genetics and Genomic Sciences, Seaver Autism Center for Research and Treatment, Department of Psychiatry, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington, Seattle, WA USA
| | - Morgan Kelly
- Department of Psychiatry, University of Washington, Seattle, WA USA
| | | | - Jennifer Gerdts
- Department of Psychiatry, University of Washington, Seattle, WA USA
| | | | | | | | - Alexander Kolevzon
- Department of Psychiatry, Department of Pediatrics, Friedman Brain Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Joseph D Buxbaum
- Department of Psychiatry, Department of Genetics and Genomic Sciences, Department of Neuroscience, Friedman Brain Institute, Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA.,Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
31
|
Mendoza E, Scharff C. Protein-Protein Interaction Among the FoxP Family Members and their Regulation of Two Target Genes, VLDLR and CNTNAP2 in the Zebra Finch Song System. Front Mol Neurosci 2017; 10:112. [PMID: 28507505 PMCID: PMC5410569 DOI: 10.3389/fnmol.2017.00112] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/05/2017] [Indexed: 12/18/2022] Open
Abstract
The Forkhead transcription factor FOXP2 is implicated in speech perception and production. The avian homolog, FoxP21 contributes to song learning and production in birds. In human cell lines, transcriptional activity of FOXP2 requires homo-dimerization or dimerization with paralogs FOXP1 or FOXP4. Whether FoxP dimerization occurs in the brain is unknown. We recently showed that FoxP1, FoxP2 and FoxP4 (FoxP1/2/4) proteins are co-expressed in neurons of Area X, a song control region in zebra finches. We now report on dimer- and oligomerization of zebra finch FoxPs and how this affects transcription. In cell lines and in the brain we identify homo- and hetero-dimers, and an oligomer composed of FoxP1/2/4. We further show that FoxP1/2 but not FoxP4 bind to the regulatory region of the target gene Contactin-associated protein-like 2 (CNTNAP2). In addition, we demonstrate that FoxP1/4 bind to the regulatory region of very low density lipoprotein receptor (VLDLR), as has been shown for FoxP2 previously. Interestingly, FoxP1/2/4 individually or in combinations regulate the promoters for SV40, zebra finch VLDLR and CNTNAP2 differentially. These data exemplify the potential for complex transcriptional regulation of FoxP1/2/4, highlighting the need for future functional studies dissecting their differential regulation in the brain.
Collapse
Affiliation(s)
- Ezequiel Mendoza
- Institut für Verhaltensbiologie, Freie Universität BerlinBerlin, Germany
| | - Constance Scharff
- Institut für Verhaltensbiologie, Freie Universität BerlinBerlin, Germany
| |
Collapse
|