1
|
Wagner L, Cakar ME, Banchik M, Chiem E, Glynn SS, Than AH, Green SA, Dapretto M. Beyond motor learning: Insights from infant magnetic resonance imaging on the critical role of the cerebellum in behavioral development. Dev Cogn Neurosci 2025; 72:101514. [PMID: 39919679 PMCID: PMC11848473 DOI: 10.1016/j.dcn.2025.101514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 02/09/2025] Open
Abstract
Although the cerebellum is now recognized for its crucial role in non-motor functions such as language, perceptual processes, social communication, and executive function in adults, it is often overlooked in studies of non-motor behavioral development in infancy. Recent magnetic resonance imaging (MRI) research increasingly shows the cerebellum is key to understanding the emergence of complex human behaviors and neurodevelopmental conditions. This review summarizes studies from diverse MRI modalities that link early cerebellar development from birth to age two with emerging non-motor behaviors and psychiatric symptomatology. Our focus centered on both term and preterm infants, excluding studies of perinatal injury and cerebellar pathology. We conclude that the cerebellum is implicated in many non-motor behaviors and implicit learning mechanisms in infancy. The field's current limitations include inconsistencies in study design, a paucity of gold-standard infant neuroimaging tools, and treatment of the cerebellum as a uniform structure. Moving forward, the cerebellum should be considered a structure of greater interest to the developmental neuroimaging community. Studies should test developmental hypotheses about the behavioral roles of specific cerebro-cerebellar circuits, and theoretical frameworks such as Olson's "model switch" hypothesis of cerebellar learning. Large-scale, longitudinal, well-powered neuroimaging studies of typical and preterm development will be key.
Collapse
Affiliation(s)
- Lauren Wagner
- Neuroscience Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Melis E Cakar
- Neuroscience Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Megan Banchik
- Department of Psychiatry and Biobehavioral Science, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Emily Chiem
- Molecular, Cellular, Integrative Physiology Program, University of California Los Angeles, Los Angeles, CA, 90095, United States
| | - Siobhan Sive Glynn
- Department of Psychology, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Amy H Than
- Neuroscience Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Shulamite A Green
- Department of Psychiatry and Biobehavioral Science, University of California Los Angeles, Los Angeles, CA 90095, United States; Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, United States
| | - Mirella Dapretto
- Department of Psychiatry and Biobehavioral Science, University of California Los Angeles, Los Angeles, CA 90095, United States; Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, United States.
| |
Collapse
|
2
|
Boonstra JT. The cerebellar connectome. Behav Brain Res 2025; 482:115457. [PMID: 39884319 DOI: 10.1016/j.bbr.2025.115457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The cerebellum, once primarily associated with motor functions, has emerged as a critical component in higher cognitive processes and emotional regulation. This paradigm shift frames the cerebellum as an essential focal point for elucidating sophisticated functional brain circuitry. Network neuroscience often maintains a cortical-centric viewpoint, potentially overlooking the significant contributions of the cerebellum in connectome organization. Enhanced recognition and integration of cerebellar aspects in connectomic analyses hold significant potential for elucidating cerebellar circuitry within comprehensive brain networks and in neuropsychiatric conditions where cerebellar involvement is evident. This review explores the intricate anatomy, connectivity, and functional organization of the cerebellum within the broader context of large-scale brain networks. Cerebellar-specific networks are examined, emphasizing their role in supporting diverse cognitive functions via the cerebellum's hierarchical functional organization. The clinical significance of cerebellar connectomics is then addressed, highlighting the interplay between cerebellar circuitry and neurological and psychiatric conditions. The paper concludes by considering neurostimulation treatments and future directions in the field. This comprehensive review underscores the cerebellum's integral role in the human connectome.
Collapse
Affiliation(s)
- Jackson Tyler Boonstra
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands; Department of Neurology, Amsterdam University Medical Centers, Amsterdam Neuroscience, University of Amsterdam, Amsterdam 1105 AZ, The Netherlands.
| |
Collapse
|
3
|
Atterton C, Pelenyi A, Jones J, Currey L, Al-Khalily M, Wright L, Doonan M, Knight D, Kurniawan ND, Walters S, Thor S, Piper M. The Hippo effector TEAD1 regulates postnatal murine cerebellar development. Brain Struct Funct 2025; 230:42. [PMID: 40064689 PMCID: PMC11893647 DOI: 10.1007/s00429-025-02903-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
The Hippo signalling cascade is an evolutionarily conserved pathway critical for the development of numerous organ systems and is required for the development of many parts of the mammalian nervous system, including the cerebellum. The Hippo pathway converges, via the nuclear YAP/TAZ co-transcription factors, on transcription factors of the TEA Domain (TEAD) family (TEAD1-4) and promotes the expression of pro-proliferative genes. Despite the importance of TEAD function, our understanding of spatial and temporal expression of this family is limited, as is our understanding of which TEAD family members regulate Hippo-dependent organ development. Here, we focus on TEAD1 and how this factor contributes to postnatal murine cerebellar development. We find expression of TEAD1 within cerebellar progenitor cells and glial cells, including astrocytes and Bergmann glia, as well as by some interneurons within the granular layer. The importance of TEAD1 expression for cerebellar development was investigated using a conditional ablation approach, which revealed a range of developmental deficits in Tead1 mutants, including an underdeveloped cerebellum, morphological defects in Bergmann Glia and Purkinje Neurons, as well as granule neuron migration defects. Collectively, these findings suggest a major role for TEAD1 as an effector of the Hippo pathway during cerebellar development.
Collapse
Affiliation(s)
- Cooper Atterton
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Alexandra Pelenyi
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Justin Jones
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Laura Currey
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Majd Al-Khalily
- The Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Lucinda Wright
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Mikki Doonan
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - David Knight
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Nyoman D Kurniawan
- The Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Shaun Walters
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Stefan Thor
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michael Piper
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia.
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
4
|
Yin H, Wang Z, Wang W, Liu J, Xue Y, Liu L, Shen J, Duan L. Dysregulated Pathways During Pregnancy Predict Drug Candidates in Neurodevelopmental Disorders. Neurosci Bull 2025:10.1007/s12264-025-01360-0. [PMID: 39913063 DOI: 10.1007/s12264-025-01360-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/06/2024] [Indexed: 02/07/2025] Open
Abstract
Maternal health during pregnancy has a direct impact on the risk and severity of neurodevelopmental disorders (NDDs) in the offspring, especially in the case of drug exposure. However, little progress has been made to assess the risk of drug exposure during pregnancy due to ethical constraints and drug use factors. We collected and manually curated sub-pathways and pathways (sub-/pathways) and drug information to propose an analytical framework for predicting drug candidates. This framework linked sub-/pathway activity and drug response scores derived from gene transcription data and was applied to human fetal brain development and six NDDs. Further, specific and pleiotropic sub-/pathways/drugs were identified using entropy, and sex bias was analyzed in conjunction with logistic regression and random forest models. We identified 19 disorder-associated and 256 regionally pleiotropic and specific candidate drugs that targeted risk sub-/pathways in NDDs, showing temporal or spatial changes across fetal development. Moreover, 5443 differential drug-sub-/pathways exhibited sex-biased differences after filling in the gender labels. A user-friendly NDDP visualization website ( https://ndd-lab.shinyapps.io/NDDP ) was developed to allow researchers and clinicians to access and retrieve data easily. Our framework overcame data gaps and identified numerous pleiotropic and specific candidates across six disorders and fetal developmental trajectories. This could significantly contribute to drug discovery during pregnancy and can be applied to a wide range of traits.
Collapse
Affiliation(s)
- Huamin Yin
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Zhendong Wang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenhang Wang
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxin Liu
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Yirui Xue
- Wenzhou Medical University, Wenzhou, 325035, China
| | - Li Liu
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Jingling Shen
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China.
| | - Lian Duan
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
5
|
Ajongbolo AO, Langhans SA. YAP/TAZ-associated cell signaling - at the crossroads of cancer and neurodevelopmental disorders. Front Cell Dev Biol 2025; 13:1522705. [PMID: 39936032 PMCID: PMC11810912 DOI: 10.3389/fcell.2025.1522705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
YAP/TAZ (Yes-associated protein/paralog transcriptional co-activator with PDZ-binding domain) are transcriptional cofactors that are the key and major downstream effectors of the Hippo signaling pathway. Both are known to play a crucial role in defining cellular outcomes, including cell differentiation, cell proliferation, and apoptosis. Aside from the canonical Hippo signaling cascade with the key components MST1/2 (mammalian STE20-like kinase 1/2), SAV1 (Salvador homologue 1), MOB1A/B (Mps one binder kinase activator 1A/B) and LATS1/2 (large tumor suppressor kinase 1/2) upstream of YAP/TAZ, YAP/TAZ activation is also influenced by numerous other signaling pathways. Such non-canonical regulation of YAP/TAZ includes well-known growth factor signaling pathways such as the epidermal growth factor receptor (EGFR)/ErbB family, Notch, and Wnt signaling as well as cell-cell adhesion, cell-matrix interactions and mechanical cues from a cell's microenvironment. This puts YAP/TAZ at the center of a complex signaling network capable of regulating developmental processes and tissue regeneration. On the other hand, dysregulation of YAP/TAZ signaling has been implicated in numerous diseases including various cancers and neurodevelopmental disorders. Indeed, in recent years, parallels between cancer development and neurodevelopmental disorders have become apparent with YAP/TAZ signaling being one of these pathways. This review discusses the role of YAP/TAZ in brain development, cancer and neurodevelopmental disorders with a special focus on the interconnection in the role of YAP/TAZ in these different conditions.
Collapse
Affiliation(s)
- Aderonke O. Ajongbolo
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
- Biological Sciences Graduate Program, University of Delaware, Newark, DE, United States
| | - Sigrid A. Langhans
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
| |
Collapse
|
6
|
O’Neill E, Curham L, Ní Chasaide C, O’Brien S, McManus G, Moran B, Rubin K, Glazer S, Lynch MA, Mills KH. Neonatal infection with Bordetella pertussis promotes autism-like phenotypes in mice. iScience 2025; 28:111548. [PMID: 39897939 PMCID: PMC11784780 DOI: 10.1016/j.isci.2024.111548] [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: 04/11/2024] [Revised: 09/09/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Autism spectrum disorder (ASD) has been linked with infections early in life. Here we demonstrate that the infection of neonatal mice with the respiratory pathogen Bordetella pertussis leads to neuroinflammation, neurodevelopmental defects, and ASD-like behaviors. Following the respiratory challenge of neonatal mice with B. pertussis, multiple atypical CNS findings were observed, including blood-brain barrier disruption, dissemination of live B. pertussis bacteria to the brain with the concomitant infiltration of inflammatory monocytes, neutrophils, and activated IL-17A- and IFN-γ-producing CD4 T cells. Microglia from infected mice were activated, with impaired phagocytic function, resulting in defective synaptic pruning and disrupted neuronal circuit formation. Impaired neurodevelopment in B. pertussis-infected post-natal mice was associated with ASD-like behavioral abnormalities in young adulthood. Our data indicate that infection with virulent B. pertussis during infancy increases the risk of autism-like behavior in young adult mice. A study into the potential role of B. pertussis in human ASD is warranted.
Collapse
Affiliation(s)
- Eoin O’Neill
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Lucy Curham
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Caitlín Ní Chasaide
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Síofra O’Brien
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Gavin McManus
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Barry Moran
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Keith Rubin
- ILiAD Biotechnologies, Weston, FL 33331, USA
| | | | - Marina A. Lynch
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Kingston H.G. Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| |
Collapse
|
7
|
Butera C, Delafield-Butt J, Lu SC, Sobota K, McGowan T, Harrison L, Kilroy E, Jayashankar A, Aziz-Zadeh L. Motor Signature Differences Between Autism Spectrum Disorder and Developmental Coordination Disorder, and Their Neural Mechanisms. J Autism Dev Disord 2025; 55:353-368. [PMID: 38062243 PMCID: PMC11802596 DOI: 10.1007/s10803-023-06171-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 02/07/2025]
Abstract
Autism spectrum disorder (ASD) and Developmental Coordination Disorder (DCD) are distinct clinical groups with overlapping motor features. We attempted to (1) differentiate children with ASD from those with DCD, and from those typically developing (TD) (ages 8-17; 18 ASD, 16 DCD, 20 TD) using a 5-min coloring game on a smart tablet and (2) identify neural correlates of these differences. We utilized standardized behavioral motor assessments (e.g. fine motor, gross motor, and balance skills) and video recordings of a smart tablet task to capture any visible motor, behavioral, posture, or engagement differences. We employed machine learning analytics of motor kinematics during a 5-min coloring game on a smart tablet. Imaging data was captured using functional magnetic resonance imaging (fMRI) during action production tasks. While subject-rated motor assessments could not differentiate the two clinical groups, machine learning computational analysis provided good predictive discrimination: between TD and ASD (76% accuracy), TD and DCD (78% accuracy), and ASD and DCD (71% accuracy). Two kinematic markers which strongly drove categorization were significantly correlated with cerebellar activity. Findings demonstrate unique neuromotor patterns between ASD and DCD relate to cerebellar function and present a promising route for computational techniques in early identification. These are promising preliminary results that warrant replication with larger samples.
Collapse
Affiliation(s)
- Christiana Butera
- USC Mrs. T.H. Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, USA.
- Brain and Creativity Institute, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA.
| | - Jonathan Delafield-Butt
- Laboratory for Innovation in Autism, University of Strathclyde, Glasgow, Scotland, UK
- Faculty of Humanities and Social Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Szu-Ching Lu
- Laboratory for Innovation in Autism, University of Strathclyde, Glasgow, Scotland, UK
- Faculty of Humanities and Social Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | | | - Timothy McGowan
- Laboratory for Innovation in Autism, University of Strathclyde, Glasgow, Scotland, UK
- Faculty of Humanities and Social Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Laura Harrison
- USC Mrs. T.H. Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, USA
- Brain and Creativity Institute, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Emily Kilroy
- USC Mrs. T.H. Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, USA
- Brain and Creativity Institute, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Aditya Jayashankar
- USC Mrs. T.H. Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, USA
- Brain and Creativity Institute, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Lisa Aziz-Zadeh
- USC Mrs. T.H. Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, USA
- Brain and Creativity Institute, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
8
|
Alnakhli AM, Saleh A, Kabel AM, Estfanous RS, Borg HM, Alsufyani KM, Sabry NM, Gomaa FAM, Abd Elmaaboud MA. Perindopril Ameliorates Sodium Valproate-Induced Rat Model of Autism: Involvement of Sirtuin-1, JAK2/STAT3 Axis, PI3K/Akt/GSK-3β Pathway, and PPAR-Gamma Signaling. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1802. [PMID: 39596986 PMCID: PMC11596946 DOI: 10.3390/medicina60111802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: Autism is a developmental disability characterized by impairment of motor functions and social communication together with the development of repetitive or stereotyped behaviors. Neither the exact etiology or the curative treatment of autism are yet completely explored. The goals of this study were to evaluate the possible effects of perindopril on a rat model of autism and to elucidate the possible molecular mechanisms that may contribute to these effects. Materials and Methods: In a rat model of sodium valproate (VPA)-induced autism, the effect of postnatal administration of different doses of perindopril on growth and motor development, social and repetitive behaviors, sirtuin-1, oxidative stress and inflammatory markers, PI3K/Akt/GSK-3β pathway, JAK2/STAT3 axis, and PPAR-gamma signaling in the hippocampal tissues were investigated. The histopathological and electron microscopic changes elicited by administration of the different treatments were also investigated. Results: Perindopril dose-dependently combatted the effects of prenatal exposure to VPA on growth and maturation, motor development, and social and repetitive behaviors. In addition, the different doses of perindopril ameliorated the effects of prenatal exposure to VPA on sirtuin-1, oxidative stress and inflammatory markers, PI3K/Akt/GSK-3β pathway, JAK2/STAT3 axis, and PPAR-gamma signaling. These effects had a mitigating impact on VPA-induced histopathological and electron microscopic changes in the hippocampal tissues. Conclusions: Perindopril may emerge as a promising agent for amelioration of the pathologic changes of autism spectrum disorders.
Collapse
Affiliation(s)
- Anwar M. Alnakhli
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (A.M.A.); (A.S.)
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (A.M.A.); (A.S.)
| | - Ahmed M. Kabel
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Remon S. Estfanous
- Anatomy and Embryology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Hany M. Borg
- Physiology Department, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt
| | | | - Nesreen M. Sabry
- Clinical Oncology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Fatma Alzahraa M. Gomaa
- Pharamcognosy and Medicinal Herbs Department, Faculty of Pharmacy, Al-Baha University, AlBaha 65779, Saudi Arabia;
| | | |
Collapse
|
9
|
Christensen ZP, Freedman EG, Foxe JJ. Autism is associated with in vivo changes in gray matter neurite architecture. Autism Res 2024; 17:2261-2277. [PMID: 39324563 DOI: 10.1002/aur.3239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Postmortem investigations in autism have identified anomalies in neural cytoarchitecture across limbic, cerebellar, and neocortical networks. These anomalies include narrow cell mini-columns and variable neuron density. However, difficulty obtaining sufficient post-mortem samples has often prevented investigations from converging on reproducible measures. Recent advances in processing magnetic resonance diffusion weighted images (DWI) make in vivo characterization of neuronal cytoarchitecture a potential alternative to post-mortem studies. Using extensive DWI data from the Adolescent Brain Cognitive Developmentsm (ABCD®) study 142 individuals with an autism diagnosis were compared with 8971 controls using a restriction spectrum imaging (RSI) framework that characterized total neurite density (TND), its component restricted normalized directional diffusion (RND), and restricted normalized isotropic diffusion (RNI). A significant decrease in TND was observed in autism in the right cerebellar cortex (β = -0.005, SE =0.0015, p = 0.0267), with significant decreases in RNI and significant increases in RND found diffusely throughout posterior and anterior aspects of the brain, respectively. Furthermore, these regions remained significant in post-hoc analysis when the autism sample was compared against a subset of 1404 individuals with other psychiatric conditions (pulled from the original 8971). These findings highlight the importance of characterizing neuron cytoarchitecture in autism and the significance of their incorporation as physiological covariates in future studies.
Collapse
Affiliation(s)
- Zachary P Christensen
- Frederick J. and Marion A. Schindler Cognitive Neurophysiology Laboratory, The Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Edward G Freedman
- Frederick J. and Marion A. Schindler Cognitive Neurophysiology Laboratory, The Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - John J Foxe
- Frederick J. and Marion A. Schindler Cognitive Neurophysiology Laboratory, The Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
10
|
Mundorf A, Merklein SA, Rice LC, Desmond JE, Peterburs J. Early Adversity Affects Cerebellar Structure and Function-A Systematic Review of Human and Animal Studies. Dev Psychobiol 2024; 66:e22556. [PMID: 39378310 DOI: 10.1002/dev.22556] [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: 01/12/2024] [Revised: 07/23/2024] [Accepted: 09/18/2024] [Indexed: 10/10/2024]
Abstract
Recent research has highlighted cerebellar involvement in cognition and several psychiatric conditions such as mood and anxiety disorders and schizophrenia. Attention-deficit/hyperactivity disorder and autism spectrum disorder have been linked to reduced cerebellar volume as well. Cerebellar alterations are frequently present after early adversity in humans and animals, but a systematic integration of results is lacking. To this end, a systematic literature search was conducted in PubMed, Web of Science, and EBSCO databases using the keywords "early adversity OR early life stress" AND "cerebellum OR cerebellar." A total of 45 publications met the inclusion criteria: 25 studies investigated human subjects and 20 reported results from animal models. Findings in healthy subjects show bilateral volume reduction and decreased functional connectivity within the cerebellum and between the cerebellum and frontal regions after adversity throughout life, especially when adversity was assessed with the Childhood Trauma Questionnaire. In clinical populations, adults demonstrate increased cerebellar volume and functional connectivity after adversity, whereas pediatric patients show reduced cerebellar volume. Animal findings reveal cerebellar alterations without necessarily co-occurring pathological behavior, highlighting alterations in stress hormone receptor levels, cell density, and neuroinflammation markers. Cerebellar alterations after early adversity are robust findings across human and animal studies and occur independent of clinical symptoms.
Collapse
Affiliation(s)
- Annakarina Mundorf
- Department of Neurology, Division of Cognitive Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Sarah A Merklein
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Department of Psychology, Medical School Hamburg, Hamburg, Germany
| | - Laura C Rice
- Center for Neurodevelopmental and Imaging Research, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - John E Desmond
- Department of Neurology, Division of Cognitive Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jutta Peterburs
- Department of Neurology, Division of Cognitive Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
11
|
van der Heijden ME. Converging and Diverging Cerebellar Pathways for Motor and Social Behaviors in Mice. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1754-1767. [PMID: 38780757 PMCID: PMC11489171 DOI: 10.1007/s12311-024-01706-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Evidence from clinical and preclinical studies has shown that the cerebellum contributes to cognitive functions, including social behaviors. Now that the cerebellum's role in a wider range of behaviors has been confirmed, the question arises whether the cerebellum contributes to social behaviors via the same mechanisms with which it modulates movements. This review seeks to answer whether the cerebellum guides motor and social behaviors through identical pathways. It focuses on studies in which cerebellar cells, synapses, or genes are manipulated in a cell-type specific manner followed by testing of the effects on social and motor behaviors. These studies show that both anatomically restricted and cerebellar cortex-wide manipulations can lead to social impairments without abnormal motor control, and vice versa. These studies suggest that the cerebellum employs different cellular, synaptic, and molecular pathways for social and motor behaviors. Future studies warrant a focus on the diverging mechanisms by which the cerebellum contributes to a wide range of neural functions.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, USA.
- Center for Neurobiology Research, Virginia Tech Carilion, Roanoke, VA, USA.
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
12
|
Hosaka S, Hosokawa M, Hibi M, Shimizu T. The Zebrafish Cerebellar Neural Circuits Are Involved in Orienting Behavior. eNeuro 2024; 11:ENEURO.0141-24.2024. [PMID: 39406478 PMCID: PMC11521796 DOI: 10.1523/eneuro.0141-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/22/2024] [Accepted: 08/13/2024] [Indexed: 11/01/2024] Open
Abstract
Deficits in social behavior are found in neurodevelopmental disorders, including autism spectrum disorders (ASDs). Since abnormalities in cerebellar morphology and function are observed in ASD patients, the cerebellum is thought to play a role in social behavior. However, it remains unknown whether the cerebellum is involved in social behavior in other animals and how cerebellar circuits control social behavior. To address this issue, we employed zebrafish stereotyped orienting behavior as a model of social behaviors, in which a pair of adult zebrafish in two separate tanks approach each other, with one swimming at synchronized angles (orienting angles) with the other. We harnessed transgenic zebrafish that express botulinum toxin, which inhibits the release of neurotransmitters, in either granule cells or Purkinje cells (PCs), and zebrafish mutants of reelin, which is involved in the positioning of cerebellar neurons, including PCs. These zebrafish, deficient in the function or formation of cerebellar neural circuits, showed a significantly shorter period of orienting behavior compared with their control siblings. We found an increase in c-fos and egr1 expression in the cerebellum after the orienting behavior. These results suggest that zebrafish cerebellar circuits play an important role in social orienting behavior.
Collapse
Affiliation(s)
- Shiori Hosaka
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Miu Hosokawa
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Masahiko Hibi
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Takashi Shimizu
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Aichi 464-8602, Japan
| |
Collapse
|
13
|
King C, Maze T, Plakke B. Altered prefrontal and cerebellar parvalbumin neuron counts are associated with cognitive changes in male rats. Exp Brain Res 2024; 242:2295-2308. [PMID: 39085433 DOI: 10.1007/s00221-024-06902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Exposure to valproic acid (VPA), a common anti-seizure medication, in utero is a risk factor for autism spectrum disorder (ASD). People with ASD often display changes in the cerebellum, including volume changes, altered circuitry, and changes in Purkinje cell populations. ASD is also characterized by changes in the medial prefrontal cortex (mPFC), where excitatory/inhibitory balance is often altered. This study exposed rats to a high dose of VPA during gestation and assessed cognition and anxiety-like behaviors during young adulthood using a set-shifting task and the elevated plus maze. Inhibitory parvalbumin-expressing (PV +) neuron counts were assessed in the mPFC and cerebellar lobules VI and VII (Purkinje cell layers), which are known to modulate cognition. VPA males had increased PV + counts in crus I and II of lobule VII. VPA males also had decreased parvalbumin-expressing neuron counts in the mPFC. It was also found that VPA-exposed rats, regardless of sex, had increased parvalbumin-expressing Purkinje cell counts in lobule VI. In males, this was associated with impaired intra-dimensional shifting on a set-shifting task. Purkinje cell over proliferation may be contributing to the previously observed increase in volume of Lobule VI. These findings suggest that altered inhibitory signaling in cerebellar-frontal circuits may contribute to the cognitive deficits that occur within ASD.
Collapse
Affiliation(s)
- Cole King
- Psychological Sciences, Kansas State University, 1114 Mid-Campus Dr., Manhattan, KS, 66506, USA
| | - Tessa Maze
- Psychological Sciences, Kansas State University, 1114 Mid-Campus Dr., Manhattan, KS, 66506, USA
| | - Bethany Plakke
- Psychological Sciences, Kansas State University, 1114 Mid-Campus Dr., Manhattan, KS, 66506, USA.
| |
Collapse
|
14
|
Antonioni A, Raho EM, Straudi S, Granieri E, Koch G, Fadiga L. The cerebellum and the Mirror Neuron System: A matter of inhibition? From neurophysiological evidence to neuromodulatory implications. A narrative review. Neurosci Biobehav Rev 2024; 164:105830. [PMID: 39069236 DOI: 10.1016/j.neubiorev.2024.105830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Mirror neurons show activity during both the execution (AE) and observation of actions (AO). The Mirror Neuron System (MNS) could be involved during motor imagery (MI) as well. Extensive research suggests that the cerebellum is interconnected with the MNS and may be critically involved in its activities. We gathered evidence on the cerebellum's role in MNS functions, both theoretically and experimentally. Evidence shows that the cerebellum plays a major role during AO and MI and that its lesions impair MNS functions likely because, by modulating the activity of cortical inhibitory interneurons with mirror properties, the cerebellum may contribute to visuomotor matching, which is fundamental for shaping mirror properties. Indeed, the cerebellum may strengthen sensory-motor patterns that minimise the discrepancy between predicted and actual outcome, both during AE and AO. Furthermore, through its connections with the hippocampus, the cerebellum might be involved in internal simulations of motor programs during MI. Finally, as cerebellar neuromodulation might improve its impact on MNS activity, we explored its potential neurophysiological and neurorehabilitation implications.
Collapse
Affiliation(s)
- Annibale Antonioni
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy; Department of Neuroscience, Ferrara University Hospital, Ferrara 44124, Italy; Doctoral Program in Translational Neurosciences and Neurotechnologies, University of Ferrara, Ferrara 44121, Italy.
| | - Emanuela Maria Raho
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Sofia Straudi
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy; Department of Neuroscience, Ferrara University Hospital, Ferrara 44124, Italy
| | - Enrico Granieri
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy
| | - Giacomo Koch
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy; Center for Translational Neurophysiology of Speech and Communication (CTNSC), Italian Institute of Technology (IIT), Ferrara 44121 , Italy; Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, Rome 00179, Italy
| | - Luciano Fadiga
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara 44121, Italy; Center for Translational Neurophysiology of Speech and Communication (CTNSC), Italian Institute of Technology (IIT), Ferrara 44121 , Italy
| |
Collapse
|
15
|
Ma C, Li W, Ke S, Lv J, Zhou T, Zou L. Identification of autism spectrum disorder using multiple functional connectivity-based graph convolutional network. Med Biol Eng Comput 2024; 62:2133-2144. [PMID: 38457067 DOI: 10.1007/s11517-024-03060-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
Presently, the combination of graph convolutional networks (GCN) with resting-state functional magnetic resonance imaging (rs-fMRI) data is a promising approach for early diagnosis of autism spectrum disorder (ASD). However, the prevalent approach involves exclusively full-brain functional connectivity data for disease classification using GCN, while overlooking the prior information related to the functional connectivity of brain subnetworks associated with ASD. Therefore, in this study, the multiple functional connectivity-based graph convolutional network (MFC-GCN) framework is proposed, using not only full brain functional connectivity data but also the established functional connectivity data from networks of key brain subnetworks associated with ASD, and the GCN is adopted to acquire complementary feature information for the final classification task. Given the heterogeneity within the Autism Brain Imaging Data Exchange (ABIDE) dataset, a novel External Attention Network Readout (EANReadout) is introduced. This design enables the exploration of potential subject associations, effectively addressing the dataset's heterogeneity. Experiments were conducted on the ABIDE dataset using the proposed framework, involving 714 subjects, and the average accuracy of the framework was 70.31%. The experimental results show that the proposed EANReadout outperforms the best traditional readout layer and improves the average accuracy of the framework by 4.32%.
Collapse
Affiliation(s)
- Chaoran Ma
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, Jiangsu, China
| | - Wenjie Li
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, 213164, Jiangsu, China
| | - Sheng Ke
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, Jiangsu, China
| | - Jidong Lv
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, 213164, Jiangsu, China
| | - Tiantong Zhou
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, 213164, Jiangsu, China
| | - Ling Zou
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, Jiangsu, China.
- School of Microelectronics and Control Engineering, Changzhou University, Changzhou, 213164, Jiangsu, China.
| |
Collapse
|
16
|
Ren T, Zhang L, Liu Y, Zhang Q, Sun Y, Zhou W, Huang L, Wang M, Pu Y, Huang R, Chen J, He H, Zhu T, Wang S, Chen W, Zhang Q, Du W, Luo Q, Li F. Sex-specific associations of adolescent motherhood with cognitive function, behavioral problems, and autistic-like traits in offspring and the mediating roles of family conflict and altered brain structure. BMC Med 2024; 22:226. [PMID: 38840198 PMCID: PMC11155128 DOI: 10.1186/s12916-024-03442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Previous studies have linked adolescent motherhood to adverse neurodevelopmental outcomes in offspring, yet the sex-specific effect and underlying mechanisms remain unclear. METHODS This study included 6952 children aged 9-11 from the Adolescent Brain Cognitive Development study. The exposed group consisted of children of mothers < 20 years at the time of birth, while the unexposed group was composed of children of mothers aged 20-35 at birth. We employed a generalized linear mixed model to investigate the associations of adolescent motherhood with cognitive, behavioral, and autistic-like traits in offspring. We applied an inverse-probability-weighted marginal structural model to examine the potential mediating factors including adverse perinatal outcomes, family conflict, and brain structure alterations. RESULTS Our results revealed that children of adolescent mothers had significantly lower cognitive scores (β, - 2.11, 95% CI, - 2.90 to - 1.31), increased externalizing problems in male offspring (mean ratio, 1.28, 95% CI, 1.08 to 1.52), and elevated internalizing problems (mean ratio, 1.14, 95% CI, 0.99 to 1.33) and autistic-like traits (mean ratio, 1.22, 95% CI, 1.01 to 1.47) in female. A stressful family environment mediated ~ 70% of the association with internalizing problems in females, ~ 30% with autistic-like traits in females, and ~ 20% with externalizing problems in males. Despite observable brain morphometric changes related to adolescent motherhood, these did not act as mediating factors in our analysis, after adjusting for family environment. No elevated rate of adverse perinatal outcomes was observed in the offspring of adolescent mothers in this study. CONCLUSIONS Our results reveal distinct sex-specific neurodevelopmental outcomes impacts of being born to adolescent mothers, with a substantial mediating effect of family environment on behavioral outcomes. These findings highlight the importance of developing sex-tailored interventions and support the hypothesis that family environment significantly impacts the neurodevelopmental consequences of adolescent motherhood.
Collapse
Affiliation(s)
- Tai Ren
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lingli Zhang
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yongjie Liu
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Qingli Zhang
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yunjun Sun
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Wei Zhou
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Like Huang
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Ming Wang
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yiwei Pu
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Runqi Huang
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jingyu Chen
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hua He
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Tailin Zhu
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Susu Wang
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Weiran Chen
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Qianlong Zhang
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Wenchong Du
- NTU Psychology, School of Social Sciences, Nottingham Trent University, 50 Shakespeare Street, Nottingham, NG1 4FQ, UK.
| | - Qiang Luo
- Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, 220 Handan Road, Shanghai, 200433, China.
| | - Fei Li
- Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health & Department of Developmental and Behavioural Paediatric & Child Primary Care, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
17
|
Robinson K, Delhaye M, Craig AM. Mapping proteomic composition of excitatory postsynaptic sites in the cerebellar cortex. Front Mol Neurosci 2024; 17:1381534. [PMID: 38783902 PMCID: PMC11111907 DOI: 10.3389/fnmol.2024.1381534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Functions of the cerebellar cortex, from motor learning to emotion and cognition, depend on the appropriate molecular composition at diverse synapse types. Glutamate receptor distributions have been partially mapped using immunogold electron microscopy. However, information is lacking on the distribution of many other components, such as Shank2, a postsynaptic scaffolding protein whose cerebellar dysfunction is associated with autism spectrum disorders. Here, we used an adapted Magnified Analysis of the Proteome, an expansion microscopy approach, to map multiple glutamate receptors, scaffolding and signaling proteins at single synapse resolution in the cerebellar cortex. Multiple distinct synapse-selective distribution patterns were observed. For example, AMPA receptors were most concentrated at synapses on molecular layer interneurons and at climbing fiber synapses, Shank1 was most concentrated at parallel fiber synapses on Purkinje cells, and Shank2 at both climbing fiber and parallel fiber synapses on Purkinje cells but little on molecular layer interneurons. Our results are consistent with gene expression data but also reveal input-selective targeting within Purkinje cells. In specialized glomerular structures of the granule cell layer, AMPA receptors as well as most other synaptic components preferentially targeted to synapses. However, NMDA receptors and the synaptic GTPase activating protein SynGAP preferentially targeted to extrasynaptic sites. Thus, glomeruli may be considered integrative signaling units through which mossy fibers differentially activate synaptic AMPA and extrasynaptic NMDA receptor complexes. Furthermore, we observed NMDA receptors and SynGAP at adherens junctions, suggesting a role in structural plasticity of glomeruli. Altogether, these data contribute to mapping the cerebellar 'synaptome'.
Collapse
Affiliation(s)
| | | | - Ann Marie Craig
- Djavad Mowafaghian Centre for Brain Health and Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
King C, Mali I, Strating H, Fangman E, Neyhard J, Payne M, Bossmann SH, Plakke B. Region-Specific Brain Volume Changes Emerge in Adolescence in the Valproic Acid Model of Autism and Parallel Human Findings. Dev Neurosci 2024; 47:68-80. [PMID: 38679020 PMCID: PMC11511791 DOI: 10.1159/000538932] [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: 01/11/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and communication deficits, cognitive dysfunction, and stereotyped repetitive behaviors. Regional volume changes are commonly observed in individuals with ASD. To examine volumetric dysregulation across adolescence, the valproic acid (VPA) model was used to induce ASD-like phenotypes in rats. METHOD Regional volumes were obtained via magnetic resonance imaging at either postnatal day 28 or postnatal day 40 (P40), which correspond to early and late adolescence, respectively. RESULTS Consistent with prior research, VPA animals had reduced total brain volume compared to control animals. A novel outcome was that VPA animals had overgrown right hippocampi at P40. Differences in the pattern of development of the anterior cingulate cortex were also observed in VPA animals. Differences for the posterior cingulate were only observed in males, but not females. CONCLUSION These results demonstrate differences in region-specific developmental trajectories between control and VPA animals and suggest that the VPA model may capture regional volume changes consistent with human ASD. INTRODUCTION Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and communication deficits, cognitive dysfunction, and stereotyped repetitive behaviors. Regional volume changes are commonly observed in individuals with ASD. To examine volumetric dysregulation across adolescence, the valproic acid (VPA) model was used to induce ASD-like phenotypes in rats. METHOD Regional volumes were obtained via magnetic resonance imaging at either postnatal day 28 or postnatal day 40 (P40), which correspond to early and late adolescence, respectively. RESULTS Consistent with prior research, VPA animals had reduced total brain volume compared to control animals. A novel outcome was that VPA animals had overgrown right hippocampi at P40. Differences in the pattern of development of the anterior cingulate cortex were also observed in VPA animals. Differences for the posterior cingulate were only observed in males, but not females. CONCLUSION These results demonstrate differences in region-specific developmental trajectories between control and VPA animals and suggest that the VPA model may capture regional volume changes consistent with human ASD.
Collapse
Affiliation(s)
- Cole King
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| | - Ivina Mali
- Department of Chemistry, Kansas State University, Manhattan, KS, USA
| | - Hunter Strating
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| | | | - Jenna Neyhard
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| | - Macy Payne
- Department of Chemistry, Kansas State University, Manhattan, KS, USA
| | | | - Bethany Plakke
- Psychological Sciences, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
19
|
Gao J, Xu Y, Li Y, Lu F, Wang Z. Comprehensive exploration of multi-modal and multi-branch imaging markers for autism diagnosis and interpretation: insights from an advanced deep learning model. Cereb Cortex 2024; 34:bhad521. [PMID: 38220572 DOI: 10.1093/cercor/bhad521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/16/2024] Open
Abstract
Autism spectrum disorder is a complex neurodevelopmental condition with diverse genetic and brain involvement. Despite magnetic resonance imaging advances, autism spectrum disorder diagnosis and understanding its neurogenetic factors remain challenging. We propose a dual-branch graph neural network that effectively extracts and fuses features from bimodalities, achieving 73.9% diagnostic accuracy. To explain the mechanism distinguishing autism spectrum disorder from healthy controls, we establish a perturbation model for brain imaging markers and perform a neuro-transcriptomic joint analysis using partial least squares regression and enrichment to identify potential genetic biomarkers. The perturbation model identifies brain imaging markers related to structural magnetic resonance imaging in the frontal, temporal, parietal, and occipital lobes, while functional magnetic resonance imaging markers primarily reside in the frontal, temporal, occipital lobes, and cerebellum. The neuro-transcriptomic joint analysis highlights genes associated with biological processes, such as "presynapse," "behavior," and "modulation of chemical synaptic transmission" in autism spectrum disorder's brain development. Different magnetic resonance imaging modalities offer complementary information for autism spectrum disorder diagnosis. Our dual-branch graph neural network achieves high accuracy and identifies abnormal brain regions and the neuro-transcriptomic analysis uncovers important genetic biomarkers. Overall, our study presents an effective approach for assisting in autism spectrum disorder diagnosis and identifying genetic biomarkers, showing potential for enhancing the diagnosis and treatment of this condition.
Collapse
Affiliation(s)
- Jingjing Gao
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yuhang Xu
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yanling Li
- School of Electrical Engineering and Electronic Information, Xihua University, Chengdu 610039, China
| | - Fengmei Lu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Zhengning Wang
- School of Information and Communication Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
20
|
Jimenez-Gomez A, Nguyen MX, Gill JS. Understanding the role of AMPA receptors in autism: insights from circuit and synapse dysfunction. Front Psychiatry 2024; 15:1304300. [PMID: 38352654 PMCID: PMC10861716 DOI: 10.3389/fpsyt.2024.1304300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Autism spectrum disorders represent a diverse etiological spectrum that converge on a syndrome characterized by discrepant deficits in developmental domains often highlighted by concerns in socialization, sensory integration, and autonomic functioning. Importantly, the incidence and prevalence of autism spectrum disorders have seen sharp increases since the syndrome was first described in the 1940s. The wide etiological spectrum and rising number of individuals being diagnosed with the condition lend urgency to capturing a more nuanced understanding of the pathogenic mechanisms underlying the autism spectrum disorders. The current review seeks to understand how the disruption of AMPA receptor (AMPAr)-mediated neurotransmission in the cerebro-cerebellar circuit, particularly in genetic autism related to SHANK3 or SYNGAP1 protein dysfunction function and autism associated with in utero exposure to the anti-seizure medications valproic acid and topiramate, may contribute to the disease presentation. Initially, a discussion contextualizing AMPAr signaling in the cerebro-cerebellar circuitry and microstructural circuit considerations is offered. Subsequently, a detailed review of the literature implicating mutations or deletions of SHANK3 and SYNGAP1 in disrupted AMPAr signaling reveals how bidirectional pathogenic modulation of this key circuit may contribute to autism. Finally, how pharmacological exposure may interact with this pathway, via increased risk of autism diagnosis with valproic acid and topiramate exposure and potential treatment of autism using AMPAr modulator perampanel, is discussed. Through the lens of the review, we will offer speculation on how neuromodulation may be used as a rational adjunct to therapy. Together, the present review seeks to synthesize the disparate considerations of circuit understanding, genetic etiology, and pharmacological modulation to understand the mechanistic interaction of this important and complex disorder.
Collapse
Affiliation(s)
- Andres Jimenez-Gomez
- Neurodevelopmental Disabilities Program, Department of Neurology, Joe DiMaggio Children’s Hospital, Hollywood, FL, United States
| | - Megan X. Nguyen
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Jason S. Gill
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
21
|
Hirata R, Yoshimura S, Kobayashi K, Aki M, Shibata M, Ueno T, Miyagi T, Oishi N, Murai T, Fujiwara H. Differences between subclinical attention-deficit/hyperactivity and autistic traits in default mode, salience, and frontoparietal network connectivities in young adult Japanese. Sci Rep 2023; 13:19724. [PMID: 37957246 PMCID: PMC10643712 DOI: 10.1038/s41598-023-47034-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) are associated with attentional impairments, with both commonalities and differences in the nature of their attention deficits. This study aimed to investigate the neural correlates of ADHD and ASD traits in healthy individuals, focusing on the functional connectivity (FC) of attention-related large-scale brain networks (LSBNs). The participants were 61 healthy individuals (30 men; age, 21.9 ± 1.9 years). The Adult ADHD Self-Report Scale (ASRS) and Autism Spectrum Quotient (AQ) were administered as indicators of ADHD and ASD traits, respectively. Performance in the continuous performance test (CPT) was used as a behavioural measure of sustained attentional function. Functional magnetic resonance imaging scans were performed during the resting state (Rest) and auditory oddball task (Odd). Considering the critical role in attention processing, we focused our analyses on the default mode (DMN), frontoparietal (FPN), and salience (SN) networks. Region of interest (ROI)-to-ROI analyses (false discovery rate < 0.05) were performed to determine relationships between psychological measures with within-network FC (DMN, FPN, and SN) as well as with between-network FC (DMN-FPN, DMN-SN, and FPN-SN). ASRS scores, but not AQ scores, were correlated with less frequent commission errors and shorter reaction times in the CPT. During Odd, significant positive correlations with ASRS were demonstrated in multiple FCs within DMN, while significant positive correlations with AQ were demonstrated in multiple FCs within FPN. AQs were negatively correlated with FPN-SN FCs. During Rest, AQs were negatively and positively correlated with one FC within the SN and multiple FCs between the DMN and SN, respectively. These findings of the ROI-to-ROI analysis were only partially replicated in a split-half replication analysis, a replication analysis with open-access data sets, and a replication analysis with a structure-based atlas. The better CPT performance by individuals with subclinical ADHD traits suggests positive effects of these traits on sustained attention. Differential associations between LSBN FCs and ASD/ADHD traits corroborate the notion of differences in sustained and selective attention between clinical ADHD and ASD.
Collapse
Affiliation(s)
- Risa Hirata
- Department of Neuropsychiatry, Kyoto University Hospital, 54 Shogoinkawaracho, Sakyo-ku, Kyoto, 6068397, Japan
| | - Sayaka Yoshimura
- Faculty of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Organization for Promotion of Neurodevelopmental Disorder Research, Kyoto, Japan
| | - Key Kobayashi
- Department of Neuropsychiatry, Graduate School of Medicine, University of Kyoto, Kyoto, Japan
| | - Morio Aki
- Department of Neuropsychiatry, Graduate School of Medicine, University of Kyoto, Kyoto, Japan
| | - Mami Shibata
- Department of Neuropsychiatry, Graduate School of Medicine, University of Kyoto, Kyoto, Japan
| | - Tsukasa Ueno
- Department of Neuropsychiatry, Graduate School of Medicine, University of Kyoto, Kyoto, Japan
- Integrated Clinical Education Center, Kyoto University Hospital, Kyoto, Japan
| | - Takashi Miyagi
- Department of Neuropsychiatry, Graduate School of Medicine, University of Kyoto, Kyoto, Japan
| | - Naoya Oishi
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshiya Murai
- Department of Neuropsychiatry, Kyoto University Hospital, 54 Shogoinkawaracho, Sakyo-ku, Kyoto, 6068397, Japan
- Department of Neuropsychiatry, Graduate School of Medicine, University of Kyoto, Kyoto, Japan
| | - Hironobu Fujiwara
- Department of Neuropsychiatry, Kyoto University Hospital, 54 Shogoinkawaracho, Sakyo-ku, Kyoto, 6068397, Japan.
- Department of Neuropsychiatry, Graduate School of Medicine, University of Kyoto, Kyoto, Japan.
- Artificial Intelligence Ethics and Society Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan.
- The General Research Division, Osaka University Research Center on Ethical, Legal and Social Issues, Kyoto, Japan.
| |
Collapse
|
22
|
Razzak R, Li J, He S, Sokhadze E. Investigating Sex-Based Neural Differences in Autism and Their Extended Reality Intervention Implications. Brain Sci 2023; 13:1571. [PMID: 38002531 PMCID: PMC10670246 DOI: 10.3390/brainsci13111571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Autism Spectrum Disorder (ASD) affects millions of individuals worldwide, and there is growing interest in the use of extended reality (XR) technologies for intervention. Despite the promising potential of XR interventions, there remain gaps in our understanding of the neurobiological mechanisms underlying ASD, particularly in relation to sex-based differences. This scoping review synthesizes the current research on brain activity patterns in ASD, emphasizing the implications for XR interventions and neurofeedback therapy. We examine the brain regions commonly affected by ASD, the potential benefits and drawbacks of XR technologies, and the implications of sex-specific differences for designing effective interventions. Our findings underscore the need for ongoing research into the neurobiological underpinnings of ASD and sex-based differences, as well as the importance of developing tailored interventions that consider the unique needs and experiences of autistic individuals.
Collapse
Affiliation(s)
- Rehma Razzak
- Department of Computer Science, Kennesaw State University, Marietta, GA 30060, USA; (R.R.); (S.H.)
| | - Joy Li
- Department of Software Engineering and Game Development, Kennesaw State University, Marietta, GA 30060, USA;
| | - Selena He
- Department of Computer Science, Kennesaw State University, Marietta, GA 30060, USA; (R.R.); (S.H.)
| | - Estate Sokhadze
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
23
|
Rudolph S, Badura A, Lutzu S, Pathak SS, Thieme A, Verpeut JL, Wagner MJ, Yang YM, Fioravante D. Cognitive-Affective Functions of the Cerebellum. J Neurosci 2023; 43:7554-7564. [PMID: 37940582 PMCID: PMC10634583 DOI: 10.1523/jneurosci.1451-23.2023] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
The cerebellum, traditionally associated with motor coordination and balance, also plays a crucial role in various aspects of higher-order function and dysfunction. Emerging research has shed light on the cerebellum's broader contributions to cognitive, emotional, and reward processes. The cerebellum's influence on autonomic function further highlights its significance in regulating motivational and emotional states. Perturbations in cerebellar development and function have been implicated in various neurodevelopmental disorders, including autism spectrum disorder and attention deficit hyperactivity disorder. An increasing appreciation for neuropsychiatric symptoms that arise from cerebellar dysfunction underscores the importance of elucidating the circuit mechanisms that underlie complex interactions between the cerebellum and other brain regions for a comprehensive understanding of complex behavior. By briefly discussing new advances in mapping cerebellar function in affective, cognitive, autonomic, and social processing and reviewing the role of the cerebellum in neuropathology beyond the motor domain, this Mini-Symposium review aims to provide a broad perspective of cerebellar intersections with the limbic brain in health and disease.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Stefano Lutzu
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
| | - Andreas Thieme
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, D-45147, Germany
| | - Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, Arizona 85287
| | - Mark J Wagner
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, Maryland 20814
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Diasynou Fioravante
- Center for Neuroscience, University of California-Davis, Davis, California 95618
- Department of Neurobiology, Physiology and Behavior, University of California-Davis, Davis, California 95618
| |
Collapse
|
24
|
Reichlmeir M, Canet-Pons J, Koepf G, Nurieva W, Duecker RP, Doering C, Abell K, Key J, Stokes MP, Zielen S, Schubert R, Ivics Z, Auburger G. In Cerebellar Atrophy of 12-Month-Old ATM-Null Mice, Transcriptome Upregulations Concern Most Neurotransmission and Neuropeptide Pathways, While Downregulations Affect Prominently Itpr1, Usp2 and Non-Coding RNA. Cells 2023; 12:2399. [PMID: 37830614 PMCID: PMC10572167 DOI: 10.3390/cells12192399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2023] Open
Abstract
The autosomal recessive disorder Ataxia-Telangiectasia is caused by a dysfunction of the stress response protein, ATM. In the nucleus of proliferating cells, ATM senses DNA double-strand breaks and coordinates their repair. This role explains T-cell dysfunction and tumour risk. However, it remains unclear whether this function is relevant for postmitotic neurons and underlies cerebellar atrophy, since ATM is cytoplasmic in postmitotic neurons. Here, we used ATM-null mice that survived early immune deficits via bone-marrow transplantation, and that reached initial neurodegeneration stages at 12 months of age. Global cerebellar transcriptomics demonstrated that ATM depletion triggered upregulations in most neurotransmission and neuropeptide systems. Downregulated transcripts were found for the ATM interactome component Usp2, many non-coding RNAs, ataxia genes Itpr1, Grid2, immediate early genes and immunity factors. Allelic splice changes affected prominently the neuropeptide machinery, e.g., Oprm1. Validation experiments with stressors were performed in human neuroblastoma cells, where ATM was localised only to cytoplasm, similar to the brain. Effect confirmation in SH-SY5Y cells occurred after ATM depletion and osmotic stress better than nutrient/oxidative stress, but not after ATM kinase inhibition or DNA stressor bleomycin. Overall, we provide pioneer observations from a faithful A-T mouse model, which suggest general changes in synaptic and dense-core vesicle stress adaptation.
Collapse
Affiliation(s)
- Marina Reichlmeir
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Júlia Canet-Pons
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Gabriele Koepf
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Wasifa Nurieva
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Ruth Pia Duecker
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Claudia Doering
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany;
| | - Kathryn Abell
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Matthew P. Stokes
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Stefan Zielen
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
- Respiratory Research Institute, Medaimun GmbH, 60596 Frankfurt am Main, Germany
| | - Ralf Schubert
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Zoltán Ivics
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| |
Collapse
|
25
|
Jun S, Kim M, Park H, Hwang E, Yamamoto Y, Tanaka-Yamamoto K. Organization of Purkinje cell development by neuronal MEGF11 in cerebellar granule cells. Cell Rep 2023; 42:113137. [PMID: 37708022 DOI: 10.1016/j.celrep.2023.113137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/24/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023] Open
Abstract
As cerebellar granule cells (GCs) coordinate the formation of regular cerebellar networks during postnatal development, molecules in GCs are expected to be involved. Here, we test the effects of the knockdown (KD) of multiple epidermal growth factor-like domains protein 11 (MEGF11), which is a homolog of proteins mediating astrocytic phagocytosis but is substantially increased at the later developmental stages of GCs on cerebellar development. MEGF11-KD in GCs of developing mice results in abnormal cerebellar structures, including extensively ectopic Purkinje cell (PC) somas, and in impaired motor functions. MEGF11-KD also causes abnormally asynchronous synaptic release from GC axons, parallel fibers, before the appearance of abnormal cerebellar structures. Interestingly, blockade of this abnormal synaptic release restores most of the cerebellar structures. Thus, apart from phagocytic functions of its related homologs in astrocytes, MEGF11 in GCs promotes proper PC development and cerebellar network formation by regulating immature synaptic transmission.
Collapse
Affiliation(s)
- Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Muwoong Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Eunmi Hwang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
26
|
Nardi L, Chhabra S, Leukel P, Krueger-Burg D, Sommer CJ, Schmeisser MJ. Neuroanatomical changes of ionotropic glutamatergic and GABAergic receptor densities in male mice modeling idiopathic and syndromic autism spectrum disorder. Front Psychiatry 2023; 14:1199097. [PMID: 37547211 PMCID: PMC10401048 DOI: 10.3389/fpsyt.2023.1199097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023] Open
Abstract
Autism spectrum disorder (ASD) comprises a wide range of neurodevelopment conditions primarily characterized by impaired social interaction and repetitive behavior, accompanied by a variable degree of neuropsychiatric characteristics. Synaptic dysfunction is undertaken as one of the key underlying mechanisms in understanding the pathophysiology of ASD. The excitatory/inhibitory (E/I) hypothesis is one of the most widely held theories for its pathogenesis. Shifts in E/I balance have been proven in several ASD models. In this study, we investigated three mouse lines recapitulating both idiopathic (the BTBR strain) and genetic (Fmr1 and Shank3 mutants) forms of ASD at late infancy and early adulthood. Using receptor autoradiography for ionotropic excitatory (AMPA and NMDA) and inhibitory (GABAA) receptors, we mapped the receptor binding densities in brain regions known to be associated with ASD such as prefrontal cortex, dorsal and ventral striatum, dorsal hippocampus, and cerebellum. The individual mouse lines investigated show specific alterations in excitatory ionotropic receptor density, which might be accounted as specific hallmark of each individual line. Across all the models investigated, we found an increased binding density to GABAA receptors at adulthood in the dorsal hippocampus. Interestingly, reduction in the GABAA receptor binding density was observed in the cerebellum. Altogether, our findings suggest that E/I disbalance individually affects several brain regions in ASD mouse models and that alterations in GABAergic transmission might be accounted as unifying factor.
Collapse
Affiliation(s)
- Leonardo Nardi
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Stuti Chhabra
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Petra Leukel
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dilja Krueger-Burg
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Clemens J. Sommer
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Michael J. Schmeisser
- Institute of Anatomy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
27
|
Brunert D, Quintela RM, Rothermel M. The anterior olfactory nucleus revisited - an emerging role for neuropathological conditions? Prog Neurobiol 2023:102486. [PMID: 37343762 DOI: 10.1016/j.pneurobio.2023.102486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023]
Abstract
Olfaction is an important sensory modality for many species and greatly influences animal and human behavior. Still, much about olfactory perception remains unknown. The anterior olfactory nucleus is one of the brain's central early olfactory processing areas. Located directly posterior to the olfactory bulb in the olfactory peduncle with extensive in- and output connections and unique cellular composition, it connects olfactory processing centers of the left and right hemispheres. Almost 20 years have passed since the last comprehensive review on the anterior olfactory nucleus has been published and significant advances regarding its anatomy, function, and pathophysiology have been made in the meantime. Here we briefly summarize previous knowledge on the anterior olfactory nucleus, give detailed insights into the progress that has been made in recent years, and map out its emerging importance in translational research of neurological diseases.
Collapse
Affiliation(s)
- Daniela Brunert
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | | | - Markus Rothermel
- Institute of Physiology, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany.
| |
Collapse
|
28
|
Xi K, Cai SQ, Yan HF, Tian Y, Cai J, Yang XM, Wang JM, Xing GG. CSMD3 Deficiency Leads to Motor Impairments and Autism-Like Behaviors via Dysfunction of Cerebellar Purkinje Cells in Mice. J Neurosci 2023; 43:3949-3969. [PMID: 37037606 PMCID: PMC10219040 DOI: 10.1523/jneurosci.1835-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 03/18/2023] [Accepted: 04/05/2023] [Indexed: 04/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with highly heritable heterogeneity. Mutations of CUB and sushi multiple domains 3 (CSMD3) gene have been reported in individuals with ASD. However, the underlying mechanisms of CSMD3 for the onset of ASD remain unexplored. Here, using male CSMD3 knock-out (CSMD3 -/-) mice, we found that genetic deletion of CSMD3 produced core autistic-like symptoms (social interaction deficits, restricted interests, and repetitive and stereotyped behaviors) and motor dysfunction in mice, indicating that the CSMD3 gene can be considered as a candidate for ASD. Moreover, we discovered that the ablation of CSMD3 in mice led to abnormal cerebellar Purkinje cell (PC) morphology in Crus I/II lobules, including aberrant developmental dendritogenesis and spinogenesis of PCs. Furthermore, combining in vivo fiber photometry calcium imaging and ex vivo electrophysiological recordings, we showed that the CSMD3 -/- mice exhibited an increased neuronal activity (calcium fluorescence signals) in PCs of Crus I/II lobules in response to movement activity, as well as an enhanced intrinsic excitability of PCs and an increase of excitatory rather than inhibitory synaptic input to the PCs, and an impaired long-term depression at the parallel fiber-PC synapse. These results suggest that CSMD3 plays an important role in the development of cerebellar PCs. Loss of CSMD3 causes abnormal PC morphology and dysfunction in the cerebellum, which may underlie the pathogenesis of motor deficits and core autistic-like symptoms in CSMD3 -/- mice. Our findings provide novel insight into the pathophysiological mechanisms by which CSMD3 mutations cause impairments in cerebellar function that may contribute to ASD.SIGNIFICANCE STATEMENT Autism spectrum disorder (ASD) is a neurodevelopmental disorder with highly heritable heterogeneity. Advances in genomic analysis have contributed to numerous candidate genes for the risk of ASD. Recently, a novel giant gene CSMD3 encoding a protein with CUB and sushi multiple domains (CSMDs) has been identified as a candidate gene for ASD. However, the underlying mechanisms of CSMD3 for the onset of ASD remain largely unknown. Here, we unravel that loss of CSMD3 results in abnormal morphology, increased intrinsic excitabilities, and impaired synaptic plasticity in cerebellar PCs, subsequently leading to motor deficits and ASD-like behaviors in mice. These results provide novel insight into the pathophysiological mechanisms by which CSMD3 mutations cause impairments in cerebellar function that may contribute to ASD.
Collapse
Affiliation(s)
- Ke Xi
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Si-Qing Cai
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Hui-Fang Yan
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Yue Tian
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Jie Cai
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
| | - Xiao-Mei Yang
- Department of Human Anatomy and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
| | - Jing-Min Wang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, People's Republic of China
| | - Guo-Gang Xing
- Neuroscience Research Institute, Peking University, Beijing 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, People's Republic of China
- Health Science Center, Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of China, Beijing 100191, People's Republic of China
- Second Affiliated Hospital of Xinxiang Medical University, Henan 453002, People's Republic of China
| |
Collapse
|
29
|
Streng ML, Froula JM, Krook-Magnuson E. The cerebellum's understated role and influences in the epilepsies. Neurobiol Dis 2023; 183:106160. [PMID: 37209926 DOI: 10.1016/j.nbd.2023.106160] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023] Open
Abstract
Approximately 1 in 26 people will develop epilepsy in their lifetime, but current treatment options leave as many as half of all epilepsy patients with uncontrolled seizures. In addition to the burden of the seizures themselves, chronic epilepsy can be associated with cognitive deficits, structural changes, and devastating negative outcomes such as sudden unexpected death in epilepsy (SUDEP). Thus, major challenges in epilepsy research surround the need to both develop new therapeutic targets for intervention as well as shed light on the mechanisms by which chronic epilepsy can lead to comorbidities and negative outcomes. Despite not being traditionally associated with epilepsy or seizures, the cerebellum has emerged as not only a brain region that can serve as an important target for seizure control, but one that may also be profoundly impacted by chronic epilepsy. Here, we discuss targeting the cerebellum for potential therapeutic intervention and discuss pathway insights gained from recent optogenetic studies. We then review observations of cerebellar alterations during seizures and in chronic epilepsy, as well as the potential for the cerebellum to be a seizure focus. Cerebellar alterations in epilepsy may be critical to patient outcomes, highlighting the need for a more comprehensive understanding and appreciation of the cerebellum in the epilepsies.
Collapse
Affiliation(s)
- Martha L Streng
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
| | - Jessica M Froula
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
30
|
van der Heijden ME, Rey Hipolito AG, Kim LH, Kizek DJ, Perez RM, Lin T, Sillitoe RV. Glutamatergic cerebellar neurons differentially contribute to the acquisition of motor and social behaviors. Nat Commun 2023; 14:2771. [PMID: 37188723 PMCID: PMC10185563 DOI: 10.1038/s41467-023-38475-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
Insults to the developing cerebellum can cause motor, language, and social deficits. Here, we investigate whether developmental insults to different cerebellar neurons constrain the ability to acquire cerebellar-dependent behaviors. We perturb cerebellar cortical or nuclei neuron function by eliminating glutamatergic neurotransmission during development, and then we measure motor and social behaviors in early postnatal and adult mice. Altering cortical and nuclei neurons impacts postnatal motor control and social vocalizations. Normalizing neurotransmission in cortical neurons but not nuclei neurons restores social behaviors while the motor deficits remain impaired in adults. In contrast, manipulating only a subset of nuclei neurons leaves social behaviors intact but leads to early motor deficits that are restored by adulthood. Our data uncover that glutamatergic neurotransmission from cerebellar cortical and nuclei neurons differentially control the acquisition of motor and social behaviors, and that the brain can compensate for some but not all perturbations to the developing cerebellum.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Alejandro G Rey Hipolito
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Linda H Kim
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Dominic J Kizek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Ross M Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Tao Lin
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Roy V Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
31
|
Butler DF, Skibo J, Traudt CM, Millen KJ. Neonatal subarachnoid hemorrhage disrupts multiple aspects of cerebellar development. Front Mol Neurosci 2023; 16:1161086. [PMID: 37187957 PMCID: PMC10175619 DOI: 10.3389/fnmol.2023.1161086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
Over the past decade, survival rates for extremely low gestational age neonates (ELGANs; <28 weeks gestation) has markedly improved. Unfortunately, a significant proportion of ELGANs will suffer from neurodevelopmental dysfunction. Cerebellar hemorrhagic injury (CHI) has been increasingly recognized in the ELGANs population and may contribute to neurologic dysfunction; however, the underlying mechanisms are poorly understood. To address this gap in knowledge, we developed a novel model of early isolated posterior fossa subarachnoid hemorrhage (SAH) in neonatal mice and investigated both acute and long-term effects. Following SAH on postnatal day 6 (P6), we found significant decreased levels of proliferation with the external granular layer (EGL), thinning of the EGL, decreased Purkinje cell (PC) density, and increased Bergmann glial (BG) fiber crossings at P8. At P42, CHI resulted in decreased PC density, decreased molecular layer interneuron (MLI) density, and increased BG fiber crossings. Results from both Rotarod and inverted screen assays did not demonstrate significant effects on motor strength or learning at P35-38. Treatment with the anti-inflammatory drug Ketoprofen did not significantly alter our findings after CHI, suggesting that treatment of neuro-inflammation does not provide significant neuroprotection post CHI. Further studies are required to fully elucidate the mechanisms through which CHI disrupts cerebellar developmental programming in order to develop therapeutic strategies for neuroprotection in ELGANs.
Collapse
Affiliation(s)
- David F. Butler
- Division of Pediatric Critical Care, Seattle Children's Hospital, University of Washington, Seattle, WA, United States
| | - Jonathan Skibo
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
| | | | - Kathleen J. Millen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington Medical School, Seattle, WA, United States
| |
Collapse
|
32
|
Bylemans T, Heleven E, Baetens K, Deroost N, Baeken C, Van Overwalle F. Mentalizing and narrative coherence in autistic adults: Cerebellar sequencing and prediction. Neurosci Biobehav Rev 2023; 146:105045. [PMID: 36646260 DOI: 10.1016/j.neubiorev.2023.105045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/11/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
BYLEMANS, T., et al. Mentalizing and narrative coherence in autistic adults: Cerebellar sequencing and prediction. NEUROSCI BIOBEHAV REV, 2022. - This review focuses on autistic adults and serves 4 purposes: (1) providing an overview of their difficulties regarding mentalizing (understanding others' mental states) and narrative coherence (structured storytelling), (2) highlighting the relations between both skills by examining behavioral observations and shared neural substrates, (3) providing an integrated perspective regarding novel diagnostic tools and support services, and (4) raising awareness of adult autism. We suggest that mentalizing and narrative coherence are related at the behavioral level and neural level. In addition to the traditional mentalizing network, the cerebellum probably serves as an important hub in shared cerebral networks implicated in mentalizing and narrative coherence. Future autism research and support services should tackle new questions within a framework of social cerebellar (dys)functioning.
Collapse
Affiliation(s)
- Tom Bylemans
- Brain, Body and Cognition, Department of Psychology, and Center for Neuroscience, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Elien Heleven
- Brain, Body and Cognition, Department of Psychology, and Center for Neuroscience, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Kris Baetens
- Brain, Body and Cognition, Department of Psychology, and Center for Neuroscience, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Natacha Deroost
- Brain, Body and Cognition, Department of Psychology, and Center for Neuroscience, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Chris Baeken
- Ghent University: Department of Head and Skin (UZGent), Ghent Experimental Psychiatry (GHEP) Lab, Belgium; Vrije Universiteit Brussel (VUB), Department of Psychiatry, University Hospital (UZ Brussel), Brussels, Belgium; Eindhoven University of Technology, Department of Electrical Engineering, Eindhoven, the Netherlands.
| | - Frank Van Overwalle
- Brain, Body and Cognition, Department of Psychology, and Center for Neuroscience, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
33
|
Butler DF, Skibo J, Traudt CM, Millen KJ. Neonatal Subarachnoid Hemorrhage Disrupts Multiple Aspects of Cerebellar Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528048. [PMID: 36798230 PMCID: PMC9934646 DOI: 10.1101/2023.02.10.528048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Over the past decade, survival rates for extremely low gestational age neonates (ELGANs; <28 weeks gestation) has markedly improved. Unfortunately, a significant proportion of ELGANs will suffer from neurodevelopmental dysfunction. Cerebellar hemorrhagic injury (CHI) has been increasingly recognized in the ELGANs population and may contribute to neurologic dysfunction; however, the underlying mechanisms are poorly understood. To address this gap in knowledge, we developed a novel model of early isolated posterior fossa subarachnoid hemorrhage (SAH) in neonatal mice and investigated both acute and long-term effects. Following SAH on postnatal day 6 (P6), we found significant decreased levels of proliferation with the external granular layer (EGL), thinning of the EGL, decreased Purkinje cell (PC) density, and increased Bergmann glial (BG) fiber crossings at P8. At P42, CHI resulted in decreased PC density, decreased molecular layer interneuron (MLI) density, and increased BG fiber crossings. Results from both Rotarod and inverted screen assays did not demonstrate significant effects on motor strength or learning at P35-38. Treatment with the anti-inflammatory drug Ketoprofen did not significantly alter our findings after CHI, suggesting that treatment of neuro-inflammation does not provide significant neuroprotection post CHI. Further studies are required to fully elucidate the mechanisms through which CHI disrupts cerebellar developmental programming in order to develop therapeutic strategies for neuroprotection in ELGANs.
Collapse
|
34
|
Bagnall-Moreau C, Spielman B, Brimberg L. Maternal brain reactive antibodies profile in autism spectrum disorder: an update. Transl Psychiatry 2023; 13:37. [PMID: 36737600 PMCID: PMC9898547 DOI: 10.1038/s41398-023-02335-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder with multifactorial etiologies involving both genetic and environmental factors. In the past two decades it has become clear that in utero exposure to toxins, inflammation, microbiome, and antibodies (Abs), may play a role in the etiology of ASD. Maternal brain-reactive Abs, present in 10-20% of mothers of a child with ASD, pose a potential risk to the developing brain because they can gain access to the brain during gestation, altering brain development during a critical period. Different maternal anti-brain Abs have been associated with ASD and have been suggested to bind extracellular or intracellular neuronal antigens. Clinical data from various cohorts support the increase in prevalence of such maternal brain-reactive Abs in mothers of a child with ASD compared to mothers of a typically developing child. Animal models of both non-human primates and rodents have provided compelling evidence supporting a pathogenic role of these Abs. In this review we summarize the data from clinical and animal models addressing the role of pathogenic maternal Abs in ASD. We propose that maternal brain-reactive Abs are an overlooked and promising field of research, representing a modifiable risk factor that may account for up to 20% of cases of ASD. More studies are needed to better characterize the Abs that contribute to the risk of having a child with ASD, to understand whether we can we predict such cases of ASD, and to better pinpoint the antigenic specificity of these Abs and their mechanisms of pathogenicity.
Collapse
Affiliation(s)
- Ciara Bagnall-Moreau
- grid.250903.d0000 0000 9566 0634Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY USA
| | - Benjamin Spielman
- grid.250903.d0000 0000 9566 0634Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY USA ,grid.512756.20000 0004 0370 4759Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Lior Brimberg
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, New York, NY, USA. .,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
35
|
Obara K, Abe E, Mamiya S, Toyoshima I. Cerebellar Hypoperfusion in Two Patients with Cornelia de Lange Syndrome with Novel NIPBL Variants. Mol Syndromol 2023; 14:51-58. [PMID: 36777704 PMCID: PMC9911990 DOI: 10.1159/000525681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/21/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction Cornelia de Lange syndrome (CdLS) is a rare congenital malformation characterized by distinctive facial features, short stature, and limb defects. In addition, half of the patients with CdLS exhibit repetitive self-injurious behaviors (SIBs) related to intellectual disability with autistic traits. CdLS is caused by pathogenic variants of genes encoding the cohesin complex pathway, with 70% of these variants identified in the nipped-B-like (NIPBL) gene. Case Presentation We report 2 patients with CdLS who exhibited repetitive SIBs. Patient 1, a 40-year-old male, carried a novel heterozygous duplication variant, c.1458dup, p.(Glu487*), in exon 9 of the NIPBL gene. Patient 2, a 49-year-old female, carried a novel heterozygous insertion variant, c.1751_1752ins[A;1652_1751], p.(Asp584Glufs*8), in exon 10 of the NIPBL gene. These variants were predicted to confer loss of function to the protein because of a premature stop codon. In both patients, single-photon emission computed tomography using N-isopropyl-p-[123I] iodoamphetamine (IMP-SPECT) revealed diffuse hypoperfusion in the cerebellum. Discussion This report identified 2 novel pathogenic variants in the NIPBL gene and the relationship between SIBs and cerebellar hypoperfusion in patients with CdLS. The cerebellar hypoperfusion might have been caused by the dysfunction of the cohesin complex via the downregulation of the NIPBL gene products. Further studies should be conducted to elucidate the contribution of the NIPBL gene to the development of the cerebello-cerebral cortical circuits associated with behavioral disorders.
Collapse
Affiliation(s)
- Koji Obara
- Department of Neurology, National Hospital Organization Akita National Hospital, Yurihonjo, Japan,*Koji Obara,
| | - Erika Abe
- Department of Neurology, National Hospital Organization Akita National Hospital, Yurihonjo, Japan
| | - Shigeo Mamiya
- Department of Internal Medicine, National Hospital Organization Akita National Hospital, Yurihonjo, Japan
| | - Itaru Toyoshima
- Department of Neurology, National Hospital Organization Akita National Hospital, Yurihonjo, Japan
| |
Collapse
|
36
|
Kainer D, Templeton AR, Prates ET, Jacboson D, Allan ER, Climer S, Garvin MR. Structural variants identified using non-Mendelian inheritance patterns advance the mechanistic understanding of autism spectrum disorder. HGG ADVANCES 2023; 4:100150. [PMID: 36340933 PMCID: PMC9634371 DOI: 10.1016/j.xhgg.2022.100150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
The heritability of autism spectrum disorder (ASD), based on 680,000 families and five countries, is estimated to be nearly 80%, yet heritability reported from SNP-based studies are consistently lower, and few significant loci have been identified with genome-wide association studies. This gap in genomic information may reside in rare variants, interaction among variants (epistasis), or cryptic structural variation (SV) and may provide mechanisms that underlie ASD. Here we use a method to identify potential SVs based on non-Mendelian inheritance patterns in pedigrees using parent-child genotypes from ASD families and demonstrate that they are enriched in ASD-risk genes. Most are in non-coding genic space and are over-represented in expression quantitative trait loci, suggesting that they affect gene regulation, which we confirm with their overlap of differentially expressed genes in postmortem brain tissue of ASD individuals. We then identify an SV in the GRIK2 gene that alters RNA splicing and a regulatory region of the ACMSD gene in the kynurenine pathway as significantly associated with a non-verbal ASD phenotype, supporting our hypothesis that these currently excluded loci can provide a clearer mechanistic understanding of ASD. Finally, we use an explainable artificial intelligence approach to define subgroups demonstrating their use in the context of precision medicine.
Collapse
Affiliation(s)
- David Kainer
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Alan R. Templeton
- Department of Biology, Washington University – St Louis, St. Louis, MO, USA
| | - Erica T. Prates
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Daniel Jacboson
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | - Sharlee Climer
- Department of Computer Science, University of Missouri, St. Louis, MO, USA
| | - Michael R. Garvin
- Computational Systems Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
- Williwaw Biosciences, LLC, Clarkston, MI, USA
| |
Collapse
|
37
|
Anatomical Development of the Cerebellothalamic Tract in Embryonic Mice. Cells 2022; 11:cells11233800. [PMID: 36497060 PMCID: PMC9738252 DOI: 10.3390/cells11233800] [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: 10/02/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 11/29/2022] Open
Abstract
The main connection from cerebellum to cerebrum is formed by cerebellar nuclei axons that synapse in the thalamus. Apart from its role in coordinating sensorimotor integration in the adult brain, the cerebello-thalamic tract (CbT) has also been implicated in developmental disorders, such as autism spectrum disorders. Although the development of the cerebellum, thalamus and cerebral cortex have been studied, there is no detailed description of the ontogeny of the mammalian CbT. Here we investigated the development of the CbT at embryonic stages using transgenic Ntsr1-Cre/Ai14 mice and in utero electroporation of wild type mice. Wide-field, confocal and 3D light-sheet microscopy of immunohistochemical stainings showed that CbT fibers arrive in the prethalamus between E14.5 and E15.5, but only invade the thalamus after E16.5. We quantified the spread of CbT fibers throughout the various thalamic nuclei and found that at E17.5 and E18.5 the ventrolateral, ventromedial and parafascicular nuclei, but also the mediodorsal and posterior complex, become increasingly innervated. Several CbT fiber varicosities express vesicular glutamate transporter type 2 at E18.5, indicating cerebello-thalamic synapses. Our results provide the first quantitative data on the developing murine CbT, which provides guidance for future investigations of the impact that cerebellum has on thalamo-cortical networks during development.
Collapse
|
38
|
Alterations in Cerebellar Microtubule Cytoskeletal Network in a ValproicAcid-Induced Rat Model of Autism Spectrum Disorders. Biomedicines 2022; 10:biomedicines10123031. [PMID: 36551785 PMCID: PMC9776106 DOI: 10.3390/biomedicines10123031] [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: 10/13/2022] [Revised: 11/15/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental diseases characterised by deficits in social communication, restricted interests, and repetitive behaviours. The growing body of evidence points to a role for cerebellar changes in ASD pathology. Some of the findings suggest that not only motor problems but also social deficits, repetitive behaviours, and mental inflexibility associated with ASD are connected with damage to the cerebellum. However, the understanding of this brain structure's functions in ASD pathology needs future investigations. Therefore, in this study, we generated a rodent model of ASD through a single prenatal administration of valproic acid (VPA) into pregnant rats, followed by cerebellar morphological studies of the offspring, focusing on the alterations of key cytoskeletal elements. The expression (Western blot) of α/β-tubulin and the major neuronal MT-associated proteins (MAP) such as MAP-Tau and MAP1B, MAP2, MAP6 (STOP) along with actin-crosslinking αII-spectrin and neurofilament light polypeptide (NF-L) was investigated. We found that maternal exposure to VPA induces a significant decrease in the protein levels of α/β-tubulin, MAP-Tau, MAP1B, MAP2, and αII-spectrin. Moreover, excessive MAP-Tau phosphorylation at (Ser396) along with key Tau-kinases activation was indicated. Immunohistochemical staining showed chromatolysis in the cerebellum of autistic-like rats and loss of Purkinje cells shedding light on one of the possible molecular mechanisms underpinning neuroplasticity alterations in the ASD brain.
Collapse
|
39
|
Deficits in Cerebellum-Dependent Learning and Cerebellar Morphology in Male and Female BTBR Autism Model Mice. NEUROSCI 2022. [DOI: 10.3390/neurosci3040045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Recently, there has been increased interest in the role of the cerebellum in autism spectrum disorder (ASD). To better understand the pathophysiological role of the cerebellum in ASD, it is necessary to have a variety of mouse models that have face validity for cerebellar disruption in humans. Here, we add to the literature on the cerebellum in mouse models of autism with the characterization of the cerebellum in the idiopathic BTBR T + Itpr3tf/J (BTBR) inbred mouse strain, which has behavioral phenotypes that are reminiscent of ASD in patients. When we examined both male and female BTBR mice in comparison to C57BL/6J (C57) controls, we noted that both sexes of BTBR mice showed motor coordination deficits characteristic of cerebellar dysfunction, but only the male mice showed differences in delay eyeblink conditioning, a cerebellum-dependent learning task that is known to be disrupted in ASD patients. Both male and female BTBR mice showed considerable expansion of, and abnormal foliation in, the cerebellum vermis—including a significant expansion of specific lobules in the anterior cerebellum. In addition, we found a slight but significant decrease in Purkinje cell density in both male and female BTBR mice, irrespective of the lobule. Finally, there was a marked reduction of Purkinje cell dendritic spine density in both male and female BTBR mice. These findings suggest that, for the most part, the BTBR mouse model phenocopies many of the characteristics of the subpopulation of ASD patients that have a hypertrophic cerebellum. We discuss the significance of strain differences in the cerebellum as well as the importance of this first effort to identify both similarities and differences between male and female BTBR mice with regard to the cerebellum.
Collapse
|
40
|
Tartaglione AM, Villani A, Ajmone-Cat MA, Minghetti L, Ricceri L, Pazienza V, De Simone R, Calamandrei G. Maternal immune activation induces autism-like changes in behavior, neuroinflammatory profile and gut microbiota in mouse offspring of both sexes. Transl Psychiatry 2022; 12:384. [PMID: 36104346 PMCID: PMC9474453 DOI: 10.1038/s41398-022-02149-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/09/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a sex-biased neurodevelopmental disorder with a male to female prevalence of 4:1, characterized by persistent deficits in social communication and interaction and restricted-repetitive patterns of behavior, interests or activities. Microbiota alterations as well as signs of neuroinflammation have been also reported in ASD. The involvement of immune dysregulation in ASD is further supported by evidence suggesting that maternal immune activation (MIA), especially during early pregnancy, may be a risk factor for ASD. The present study was aimed at characterizing the effects of MIA on behavior, gut microbiota and neuroinflammation in the mouse offspring also considering the impact of MIA in the two sexes. MIA offspring exhibited significant ASD-like behavioral alterations (i.e., deficits in sociability and sensorimotor gating, perseverative behaviors). The analysis of microbiota revealed changes in specific microbial taxa that recapitulated those seen in ASD children. In addition, molecular analyses indicated sex-related differences in the neuroinflammatory responses triggered by MIA, with a more prominent effect in the cerebellum. Our data suggest that both sexes should be included in the experimental designs of preclinical studies in order to identify those mechanisms that confer different vulnerability to ASD to males and females.
Collapse
Affiliation(s)
- Anna Maria Tartaglione
- Centre for Behavioral Sciences and Mental Health, Italian National Institute of Health (ISS), Rome, Italy.
| | - Annacandida Villani
- grid.413503.00000 0004 1757 9135Gastroenterology Unit IRCCS “Casa Sollievo della Sofferenza”, Hospital San Giovanni Rotondo, Foggia, Italy
| | - Maria Antonietta Ajmone-Cat
- grid.416651.10000 0000 9120 6856National Centre for Drug Research and Evaluation, Italian National Institute of Health (ISS), Rome, Italy
| | - Luisa Minghetti
- grid.416651.10000 0000 9120 6856Research Coordination and Support Service, Italian National Institute of Health (ISS), Rome, Italy
| | - Laura Ricceri
- grid.416651.10000 0000 9120 6856Centre for Behavioral Sciences and Mental Health, Italian National Institute of Health (ISS), Rome, Italy
| | - Valerio Pazienza
- grid.413503.00000 0004 1757 9135Gastroenterology Unit IRCCS “Casa Sollievo della Sofferenza”, Hospital San Giovanni Rotondo, Foggia, Italy
| | - Roberta De Simone
- grid.416651.10000 0000 9120 6856National Centre for Drug Research and Evaluation, Italian National Institute of Health (ISS), Rome, Italy
| | - Gemma Calamandrei
- grid.416651.10000 0000 9120 6856Centre for Behavioral Sciences and Mental Health, Italian National Institute of Health (ISS), Rome, Italy
| |
Collapse
|
41
|
Deficiency of the ywhaz gene, involved in neurodevelopmental disorders, alters brain activity and behaviour in zebrafish. Mol Psychiatry 2022; 27:3739-3748. [PMID: 35501409 DOI: 10.1038/s41380-022-01577-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 02/08/2023]
Abstract
Genetic variants in YWHAZ contribute to psychiatric disorders such as autism spectrum disorder and schizophrenia, and have been related to an impaired neurodevelopment in humans and mice. Here, we have used zebrafish to investigate the mechanisms by which YWHAZ contributes to neurodevelopmental disorders. We observed that ywhaz expression was pan-neuronal during developmental stages and restricted to Purkinje cells in the adult cerebellum, cells that are described to be reduced in number and size in autistic patients. We then performed whole-brain imaging in wild-type and ywhaz CRISPR/Cas9 knockout (KO) larvae and found altered neuronal activity and connectivity in the hindbrain. Adult ywhaz KO fish display decreased levels of monoamines in the hindbrain and freeze when exposed to novel stimuli, a phenotype that can be reversed with drugs that target monoamine neurotransmission. These findings suggest an important role for ywhaz in establishing neuronal connectivity during development and modulating both neurotransmission and behaviour in adults.
Collapse
|
42
|
Sreelakshmy R, Titus A, Sasirekha N, Logashanmugam E, Begam RB, Ramkumar G, Raju R. An Automated Deep Learning Model for the Cerebellum Segmentation from Fetal Brain Images. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8342767. [PMID: 35757468 PMCID: PMC9225853 DOI: 10.1155/2022/8342767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/25/2022] [Accepted: 06/04/2022] [Indexed: 01/03/2023]
Abstract
Cerebellum measures taken from routinely obtained ultrasound (US) images have been frequently employed to determine gestational age and identify developing central nervous system's anatomical abnormalities. Standardized cerebellar assessments from large-scale clinical datasets are required to investigate correlations between the growing cerebellum and postnatal neurodevelopmental results. These studies could uncover structural abnormalities that could be employed as indicators to forecast neurodevelopmental and growth consequences. To achieve this, higher-throughput, precise, and impartial measures must be used to replace the existing human, semiautomatic, and advanced algorithms, which seem to be time-consuming and inaccurate. In this article, we presented an innovative deep learning (DL) technique for automatic fetal cerebellum segmentation from 2-dimensional (2D) US brain images. We present ReU-Net, a semantic segmentation network tailored to the anatomy of the fetal cerebellum. Moreover, we use U-Net as a foundation models with the incorporation of residual blocks and Wiener filter over the last 2 layers to segregate the cerebellum (c) from the noisy US data. 590 images for training and 150 images for testing were taken; also, we employed a 5-fold cross-assessment method. Our ReU-Net scored 91%, 92%, 25.42, 98%, 92%, and 94% for Dice Score Coefficient (DSC), F1-score, Hausdorff Distance (HD), accuracy, recall, and precision, correspondingly. The suggested method outperforms the other U-Net predicated techniques by a quantitatively significant margin (p 0.001). Our presented approach can be used to allow high bandwidth imaging techniques in medical study fetal US images as well as biometric evaluation on a broader scale in fetal US images.
Collapse
Affiliation(s)
- R. Sreelakshmy
- Department of Electronics and Communication Engineering, Veltech Rangarajan Dr Sagunthala R&D Institute of Science and Technology, Avadi, Chennai, 600062 Tamil Nadu, India
| | - Anita Titus
- Department of Electronics and Communication Engineering, Jeppiaar Engineering College, Semmenchery, Raghiv Gandhi Salai, OMR, Jeppiaar Nagar, Chennai 600119, India
| | - N. Sasirekha
- Department of Electronics and Communication Engineering, Sona College of Technology, Salem, 636005 Tamil Nadu, India
| | - E. Logashanmugam
- Department of Electronics and Communication Engineering, Sathyabama Institute of Science and Technology, Chennai, 600119 Tamil Nadu, India
| | - R. Benazir Begam
- Department of Electronics and Communication Engineering, Rajalakshmi Engineering College, Chennai, 602105 Tamil Nadu, India
| | - G. Ramkumar
- Department of Electronics and Communication Engineering, Saveetha School of Engineering, SIMATS, Chennai, 602 105 Tamil Nadu, India
| | - Raja Raju
- Department of Mechanical Engineering, St. Joseph College of Engineering and Technology, St. Joseph University, Tanzania
| |
Collapse
|
43
|
Yeh CH, Tseng RY, Ni HC, Cocchi L, Chang JC, Hsu MY, Tu EN, Wu YY, Chou TL, Gau SSF, Lin HY. White matter microstructural and morphometric alterations in autism: implications for intellectual capabilities. Mol Autism 2022; 13:21. [PMID: 35585645 PMCID: PMC9118608 DOI: 10.1186/s13229-022-00499-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/30/2022] [Indexed: 12/13/2022] Open
Abstract
Background Neuroimage literature of autism spectrum disorder (ASD) has a moderate-to-high risk of bias, partially because those combined with intellectual impairment (II) and/or minimally verbal (MV) status are generally ignored. We aimed to provide more comprehensive insights into white matter alterations of ASD, inclusive of individuals with II (ASD-II-Only) or MV expression (ASD-MV). Methods Sixty-five participants with ASD (ASD-Whole; 16.6 ± 5.9 years; comprising 34 intellectually able youth, ASD-IA, and 31 intellectually impaired youth, ASD-II, including 24 ASD-II-Only plus 7 ASD-MV) and 38 demographic-matched typically developing controls (TDC; 17.3 ± 5.6 years) were scanned in accelerated diffusion-weighted MRI. Fixel-based analysis was undertaken to investigate the categorical differences in fiber density (FD), fiber cross section (FC), and a combined index (FDC), and brain symptom/cognition associations. Results ASD-Whole had reduced FD in the anterior and posterior corpus callosum and left cerebellum Crus I, and smaller FDC in right cerebellum Crus II, compared to TDC. ASD-IA, relative to TDC, had no significant discrepancies, while ASD-II showed almost identical alterations to those from ASD-Whole vs. TDC. ASD-II-Only had greater FD/FDC in the isthmus splenium of callosum than ASD-MV. Autistic severity negatively correlated with FC in right Crus I. Nonverbal full-scale IQ positively correlated with FC/FDC in cerebellum VI. FD/FDC of the right dorsolateral prefrontal cortex showed a diagnosis-by-executive function interaction. Limitations We could not preclude the potential effects of age and sex from the ASD cohort, although statistical tests suggested that these factors were not influential. Our results could be confounded by variable psychiatric comorbidities and psychotropic medication uses in our ASD participants recruited from outpatient clinics, which is nevertheless closer to a real-world presentation of ASD. The outcomes related to ASD-MV were considered preliminaries due to the small sample size within this subgroup. Finally, our study design did not include intellectual impairment-only participants without ASD to disentangle the mixture of autistic and intellectual symptoms. Conclusions ASD-associated white matter alterations appear driven by individuals with II and potentially further by MV. Results suggest that changes in the corpus callosum and cerebellum are key for psychopathology and cognition associated with ASD. Our work highlights an essential to include understudied subpopulations on the spectrum in research. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-022-00499-1.
Collapse
Affiliation(s)
- Chun-Hung Yeh
- Institute for Radiological Research, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, 333, Taoyuan City, Taiwan. .,Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Rung-Yu Tseng
- Institute for Radiological Research, Chang Gung University, No. 259, Wenhua 1st Road, Guishan District, 333, Taoyuan City, Taiwan.,Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hsing-Chang Ni
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Luca Cocchi
- Clinical Brain Networks Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jung-Chi Chang
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | | | - En-Nien Tu
- Department of Psychiatry, University of Oxford, Oxford, UK.,Department of Psychiatry, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | | | - Tai-Li Chou
- Department of Psychology, National Taiwan University, Taipei, Taiwan
| | - Susan Shur-Fen Gau
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Hsiang-Yuan Lin
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan. .,Azrieli Adult Neurodevelopmental Centre, Campbell Family Mental Health Research Institute, and Adult Neurodevelopmental and Geriatric Psychiatry Division, Centre for Addiction and Mental Health, 1025 Queen St W - 3314, Toronto, ON, M6J 1H4, Canada. .,Department of Psychiatry and Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
44
|
Serra I, Stravs A, Osório C, Oyaga MR, Schonewille M, Tudorache C, Badura A. Tsc1 Haploinsufficiency Leads to Pax2 Dysregulation in the Developing Murine Cerebellum. Front Mol Neurosci 2022; 15:831687. [PMID: 35645731 PMCID: PMC9137405 DOI: 10.3389/fnmol.2022.831687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/04/2022] [Indexed: 12/03/2022] Open
Abstract
Tuberous sclerosis complex 1 (TSC1) is a tumor suppressor that promotes the inhibition of mechanistic target of rapamycin (mTOR) pathway, and mutations in TSC1 lead to a rare complex disorder of the same name. Despite phenotype heterogeneity, up to 50% of TSC patients present with autism spectrum disorder (ASD). Consequently, TSC models are often used to probe molecular and behavioral mechanisms of ASD development. Amongst the different brain areas proposed to play a role in the development of ASD, the cerebellum is commonly reported to be altered, and cerebellar-specific deletion of Tsc1 in mice is sufficient to induce ASD-like phenotypes. However, despite these functional changes, whether Tsc1 haploinsufficiency affects cerebellar development is still largely unknown. Given that the mTOR pathway is a master regulator of cell replication and migration, we hypothesized that dysregulation of this pathway would also disrupt the development of cell populations during critical periods of cerebellar development. Here, we used a mouse model of TSC to investigate gene and protein expression during embryonic and early postnatal periods of cerebellar development. We found that, at E18 and P7, mRNA levels of the cerebellar inhibitory interneuron marker paired box gene 2 (Pax2) were dysregulated. This dysregulation was accompanied by changes in the expression of mTOR pathway-related genes and downstream phosphorylation of S6. Differential gene correlation analysis revealed dynamic changes in correlated gene pairs across development, with an overall loss of correlation between mTOR- and cerebellar-related genes in Tsc1 mutants compared to controls. We corroborated the genetic findings by characterizing the mTOR pathway and cerebellar development on protein and cellular levels with Western blot and immunohistochemistry. We found that Pax2-expressing cells were largely unchanged at E18 and P1, while at P7, their number was increased and maturation into parvalbumin-expressing cells delayed. Our findings indicate that, in mice, Tsc1 haploinsufficiency leads to altered cerebellar development and that cerebellar interneuron precursors are particularly susceptible to mTOR pathway dysregulation.
Collapse
Affiliation(s)
- Ines Serra
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Ana Stravs
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Catarina Osório
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Maria Roa Oyaga
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
- *Correspondence: Aleksandra Badura,
| |
Collapse
|
45
|
Structure, Function, and Genetics of the Cerebellum in Autism. JOURNAL OF PSYCHIATRY AND BRAIN SCIENCE 2022; 7:e220008. [PMID: 36425354 PMCID: PMC9683352 DOI: 10.20900/jpbs.20220008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Autism spectrum disorders are common neurodevelopmental disorders that are defined by core behavioral symptoms but have diverse genetic and environmental risk factors. Despite its etiological heterogeneity, several unifying theories of autism have been proposed, including a central role for cerebellar dysfunction. The cerebellum follows a protracted course of development that culminates in an exquisitely crafted brain structure containing over half of the neurons in the entire brain densely packed into a highly organized structure. Through its complex network of connections with cortical and subcortical brain regions, the cerebellum acts as a sensorimotor regulator and affects changes in executive and limbic processing. In this review, we summarize the structural, functional, and genetic contributions of the cerebellum to autism.
Collapse
|
46
|
Cerebellar Structure and Function in Autism Spectrum Disorder. JOURNAL OF PSYCHIATRY AND BRAIN SCIENCE 2022; 7. [PMID: 35978711 PMCID: PMC9380863 DOI: 10.20900/jpbs.20220003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental condition characterized by early-onset repetitive behaviors, restricted interests, sensory and motor difficulties, and impaired social interactions. Converging evidence from neuroimaging, lesion and postmortem studies, and rodent models suggests cerebellar involvement in ASD and points to promising targets for therapeutic interventions for the disorder. This review elucidates understanding of cerebellar mechanisms in ASD by integrating and contextualizing recent structural and functional cerebellar research.
Collapse
|
47
|
Van Der Heijden ME, Gill JS, Rey Hipolito AG, Salazar Leon LE, Sillitoe RV. Quantification of Behavioral Deficits in Developing Mice With Dystonic Behaviors. DYSTONIA 2022; 1:10494. [PMID: 36960404 PMCID: PMC10032351 DOI: 10.3389/dyst.2022.10494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Converging evidence from structural imaging studies in patients, the function of dystonia-causing genes, and the comorbidity of neuronal and behavioral defects all suggest that pediatric-onset dystonia is a neurodevelopmental disorder. However, to fully appreciate the contribution of altered development to dystonia, a mechanistic understanding of how networks become dysfunctional is required for early-onset dystonia. One current hurdle is that many dystonia animal models are ideally suited for studying adult phenotypes, as the neurodevelopmental features can be subtle or are complicated by broad developmental deficits. Furthermore, most assays that are used to measure dystonia are not suited for developing postnatal mice. Here, we characterize the early-onset dystonia in Ptf1a Cre ;Vglut2 fl/fl mice, which is caused by the absence of neurotransmission from inferior olive neurons onto cerebellar Purkinje cells. We investigate motor control with two paradigms that examine how altered neural function impacts key neurodevelopmental milestones seen in postnatal pups (postnatal day 7-11). We find that Ptf1a Cre ;Vglut2 fl/fl mice have poor performance on the negative geotaxis assay and the surface righting reflex. Interestingly, we also find that Ptf1a Cre ;Vglut2 fl/fl mice make fewer ultrasonic calls when socially isolated from their nests. Ultrasonic calls are often impaired in rodent models of autism spectrum disorders, a condition that can be comorbid with dystonia. Together, we show that these assays can serve as useful quantitative tools for investigating how neural dysfunction during development influences neonatal behaviors in a dystonia mouse model. Our data implicate a shared cerebellar circuit mechanism underlying dystonia-related motor signs and social impairments in mice.
Collapse
Affiliation(s)
- Meike E. Van Der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
| | - Jason S. Gill
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Alejandro G. Rey Hipolito
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Luis E. Salazar Leon
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Roy V. Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, United States
- Correspondence: Roy V. Sillitoe,
| |
Collapse
|
48
|
Pereida-Jaramillo E, Gómez-González GB, Espino-Saldaña AE, Martínez-Torres A. Calcium Signaling in the Cerebellar Radial Glia and Its Association with Morphological Changes during Zebrafish Development. Int J Mol Sci 2021; 22:ijms222413509. [PMID: 34948305 PMCID: PMC8706707 DOI: 10.3390/ijms222413509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/08/2021] [Accepted: 12/12/2021] [Indexed: 01/02/2023] Open
Abstract
Radial glial cells are a distinct non-neuronal cell type that, during development, span the entire width of the brain walls of the ventricular system. They play a central role in the origin and placement of neurons, since their processes form structural scaffolds that guide and facilitate neuronal migration. Furthermore, glutamatergic signaling in the radial glia of the adult cerebellum (i.e., Bergmann glia), is crucial for precise motor coordination. Radial glial cells exhibit spontaneous calcium activity and functional coupling spread calcium waves. However, the origin of calcium activity in relation to the ontogeny of cerebellar radial glia has not been widely explored, and many questions remain unanswered regarding the role of radial glia in brain development in health and disease. In this study we used a combination of whole mount immunofluorescence and calcium imaging in transgenic (gfap-GCaMP6s) zebrafish to determine how development of calcium activity is related to morphological changes of the cerebellum. We found that the morphological changes in cerebellar radial glia are quite dynamic; the cells are remarkably larger and more elaborate in their soma size, process length and numbers after 7 days post fertilization. Spontaneous calcium events were scarce during the first 3 days of development and calcium waves appeared on day 5, which is associated with the onset of more complex morphologies of radial glia. Blockage of gap junction coupling inhibited the propagation of calcium waves, but not basal local calcium activity. This work establishes crucial clues in radial glia organization, morphology and calcium signaling during development and provides insight into its role in complex behavioral paradigms.
Collapse
|
49
|
The GM2 gangliosidoses: Unlocking the mysteries of pathogenesis and treatment. Neurosci Lett 2021; 764:136195. [PMID: 34450229 PMCID: PMC8572160 DOI: 10.1016/j.neulet.2021.136195] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/28/2022]
|
50
|
van der Heijden ME, Lackey EP, Perez R, Ișleyen FS, Brown AM, Donofrio SG, Lin T, Zoghbi HY, Sillitoe RV. Maturation of Purkinje cell firing properties relies on neurogenesis of excitatory neurons. eLife 2021; 10:e68045. [PMID: 34542409 PMCID: PMC8452305 DOI: 10.7554/elife.68045] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/31/2021] [Indexed: 01/18/2023] Open
Abstract
Preterm infants that suffer cerebellar insults often develop motor disorders and cognitive difficulty. Excitatory granule cells, the most numerous neuron type in the brain, are especially vulnerable and likely instigate disease by impairing the function of their targets, the Purkinje cells. Here, we use regional genetic manipulations and in vivo electrophysiology to test whether excitatory neurons establish the firing properties of Purkinje cells during postnatal mouse development. We generated mutant mice that lack the majority of excitatory cerebellar neurons and tracked the structural and functional consequences on Purkinje cells. We reveal that Purkinje cells fail to acquire their typical morphology and connectivity, and that the concomitant transformation of Purkinje cell firing activity does not occur either. We also show that our mutant pups have impaired motor behaviors and vocal skills. These data argue that excitatory cerebellar neurons define the maturation time-window for postnatal Purkinje cell functions and refine cerebellar-dependent behaviors.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Elizabeth P Lackey
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Ross Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Fatma S Ișleyen
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
| | - Amanda M Brown
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Sarah G Donofrio
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Tao Lin
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Howard Hughes Medical Institute, Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of MedicineHoustonUnited States
| |
Collapse
|