1
|
Kitano E, Ueno H, Takahashi Y, Mori S, Murakami S, Wani K, Matsumoto Y, Ochi A, Hatano T, Okamoto M, Ishihara T. Postnatal sleep restriction in male mice impairs the development of parvalbumin-positive neurons in the prefrontal cortex and increases anxiety-like behaviour. Neuroscience 2025; 573:127-142. [PMID: 40113071 DOI: 10.1016/j.neuroscience.2025.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
Sleep is crucial for maintaining homeostasis and is conserved across the animal kingdom. Sleep restriction has emerged as a significant health concern, particularly in adolescents and adults. In infants and children, it is linked to disrupted brain development, impaired social-emotional growth, deficits in executive function, and increased anxiety and depression. However, the precise biological mechanisms remain unclear. This study aimed to investigate the effects of sleep restriction on parvalbumin (PV)-expressing inhibitory interneurons, which mature postnatally, and to clarify some of the developmental consequences of sleep restriction on brain function. Three hours of sleep restriction was induced daily from postnatal day (P) 10 until P14, P21, and P28. Behavioural abnormalities were assessed on P21, followed by brain histology and behavioural recovery analysis after sleep restoration. Our results showed that sleep restriction did not alter the development of PV-positive neurons in the somatosensory cortex or amygdala but significantly reduced PV-positive neurons in the prefrontal cortex. Moreover, sleep-restricted mice exhibited increased anxiety-like behaviour at P21. Upon sleep restoration, adult mice showed reduced activity in the open field test, indicating a persistent effect of early-life sleep restriction. These findings suggest that sleep restriction during postnatal development selectively affects certain brain regions, with potential long-lasting consequences. Early intervention to mitigate sleep restriction's impact on brain development may be crucial for reducing neurodevelopmental deficits.
Collapse
Affiliation(s)
- Eriko Kitano
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| | - Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, Okayama 701-0193, Japan.
| | - Yu Takahashi
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| | - Sachiko Mori
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Ayaka Ochi
- Department of Medical Technology, Kawasaki University of Medical Welfare, Okayama 701-0193, Japan.
| | - Tsukasa Hatano
- Department of Medical Technology, Kawasaki University of Medical Welfare, Okayama 701-0193, Japan.
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| |
Collapse
|
2
|
Wang T, Weng H, Li Y. Comparative study of the effects of prenatal sevoflurane exposure at different cortical stages on forebrain development and maturation in offspring. Front Neurosci 2025; 19:1556703. [PMID: 40248263 PMCID: PMC12003305 DOI: 10.3389/fnins.2025.1556703] [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: 01/07/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Brain development involves several critical stages, such as proliferation, neuronal migration, axonal pathfinding, and connection formation. Sevoflurane, a γ-aminobutyric acid (GABA) receptor agonist, is widely used as an inhaled general anesthetic. However, its impact on brain development has raised increasing concerns, particularly regarding prenatal exposure. This study aims to investigate the effects of prenatal sevoflurane exposure (PSE) at different cortical stages, focusing on its impact on the migration of glutamatergic and GABAergic neurons and neuronal behavior in offspring. Methods PSE was administered at two critical prenatal stages: embryonic day (E) 12.5 and E18.5. Double in situ hybridization was used to identify the coexpression of GABA receptors in Pax6- and Mash1-positive cells in the forebrain. The radial migration of glutamatergic neurons and the tangential migration of GABAergic neurons were analyzed. Behavioral tests, including the open-field test, elevated plus-maze test, forced swim test, tail suspension test, sucrose preference test, and Morris water maze, were performed on offspring to assess anxiety-like behaviors, depression, and learning and memory impairments. Results PSE inhibits the radial migration of glutamatergic neurons and promotes the tangential migration of GABAergic neurons. Specifically, early exposure (E12.5) inhibited the expression of the Pax6-Tbr2-Tbr1 cascade and the radial migration of Tbr1 in the ventral prefrontal cortex (PFC), whereas late exposure (E18.5) inhibited this process on the dorsal side. In addition, offspring mice with PSE exhibited increased anxiety-like behaviors, rather than depression, as demonstrated by reduced time spent in the center of the open-field test and in the open arms of the elevated plus-maze test. No significant differences were observed in the forced swim test, tail suspension test, or sucrose preference test. Furthermore, learning and memory impairments were observed in the Morris water maze. Conclusion Our results indicate that PSE at E12.5 and E18.5 leads to abnormalities in the migration of glutamatergic and GABAergic neurons, affecting long-term anxiety-like behaviors and causing learning and memory impairments in offspring mice.
Collapse
Affiliation(s)
- Tianyuan Wang
- School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Huandi Weng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yalan Li
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
3
|
Iida M, Tanaka M, Takagi T, Matsuki T, Kimura K, Shibata K, Kobayashi Y, Mizutani Y, Kuwamura H, Yamada K, Kitaura H, Kakita A, Sakakibara M, Asai N, Takahashi M, Asai M. Girdin deficiency causes developmental and epileptic encephalopathy with hippocampal sclerosis and interneuronopathy. Epilepsia 2025; 66:599-617. [PMID: 39675783 PMCID: PMC11827759 DOI: 10.1111/epi.18204] [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: 12/14/2023] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE Loss-of-function mutations in the GIRDIN/CCDC88A gene cause developmental epileptic encephalopathy (DEE) in humans. However, its pathogenesis is largely unknown. Global knockout mice of the corresponding orthologous gene (gKOs) have a preweaning lethal phenotype with growth failure, preventing longitudinal analysis. We aimed to overcome this lethality and elucidate DEE pathogenesis. METHODS We developed a novel lifelong feeding regimen (NLFR), which consists of providing mash food from postnatal day 14 (P14) until weaning (P28), followed by agar-bound food exclusively after weaning. Videography, electroencephalography (EEG), and histological analyses were performed. Conditional Girdin/Ccdc88a knockout mice (cKOs) of variable lineages (Nestin, Emx1, or Nkx2-1) were generated to identify the region responsible for epilepsy. RESULTS Under the NLFR, gKOs survived beyond 1 year and displayed fully penetrant, robust epileptic phenotypes, including early-onset (P22.3 in average) generalized tonic-clonic seizures (GTCSs) (averaging eight per day), which were completely synchronized with fast rhythms on EEG, frequent interictal electroencephalographic spikes (averaging 430 per hour), and progressive deformation of visceral organs. In addition, gKOs had absence seizures, which were not always time-locked to frequent spike waves on EEG. The frequent GTCSs and interictal spikes in gKOs were suppressed by known antiepileptic drugs. Histologically, bilateral hippocampi in gKOs exhibited congenital cornu-ammonis splitting, granule cell dispersion, and astrogliosis. Furthermore, analysis of conditional knockouts using multiple Cre-deleters identified a defect in the delivery of interneuron precursors from the medial ganglionic eminence into the hippocampal primordium during embryogenesis as a major cause of epileptogenesis. SIGNIFICANCE These findings give rise to a new approach of lifelong caregiving to overcome the problem of preweaning lethality in animal models. We propose a useful model for studying DEE with hippocampal sclerosis and interneuronopathy. gKOs with NLFR combine the contradictory properties of robust epileptic phenotypes and long-term survivability, which can be used to investigate spontaneous epileptic wave propagation and therapeutic intervention in hippocampal sclerosis.
Collapse
Affiliation(s)
- Machiko Iida
- Department of Disease Model, Institute for Developmental ResearchAichi Developmental Disability CenterKasugaiAichiJapan
| | - Motoki Tanaka
- Department of Disease Model, Institute for Developmental ResearchAichi Developmental Disability CenterKasugaiAichiJapan
| | - Tsuyoshi Takagi
- Department of Disease Model, Institute for Developmental ResearchAichi Developmental Disability CenterKasugaiAichiJapan
| | - Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental ResearchAichi Developmental Disability CenterKasugaiAichiJapan
| | - Kimihiro Kimura
- Pharmacology Research UnitSumitomo Pharma Co., Ltd.OsakaJapan
| | - Kazuki Shibata
- Pharmacology Research UnitSumitomo Pharma Co., Ltd.OsakaJapan
| | - Yohei Kobayashi
- Pharmacology Research UnitSumitomo Pharma Co., Ltd.OsakaJapan
| | - Yuka Mizutani
- Department of Disease Model, Institute for Developmental ResearchAichi Developmental Disability CenterKasugaiAichiJapan
| | - Haruki Kuwamura
- Department of Disease Model, Institute for Developmental ResearchAichi Developmental Disability CenterKasugaiAichiJapan
| | - Keitaro Yamada
- Department of Pediatric Neurology, Central HospitalAichi Developmental Disability CenterKasugaiAichiJapan
| | - Hiroki Kitaura
- Department of Pathology, Brain Research InstituteNiigata UniversityNiigataJapan
- Department of Clinical Engineering, Faculty of Health ScienceKomatsu UniversityKomatsuJapan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research InstituteNiigata UniversityNiigataJapan
| | - Mayu Sakakibara
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Naoya Asai
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Masahide Takahashi
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Masato Asai
- Department of Disease Model, Institute for Developmental ResearchAichi Developmental Disability CenterKasugaiAichiJapan
- Department of PathologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
4
|
Pavon N, Sun Y, Pak C. Cell type specification and diversity in subpallial organoids. Front Genet 2024; 15:1440583. [PMID: 39391063 PMCID: PMC11465425 DOI: 10.3389/fgene.2024.1440583] [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: 05/29/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Neural organoids have emerged as valuable tools for studying the developing brain, sparking enthusiasm and driving their adoption in disease modeling, drug screening, and investigating fetal neural development. The increasing popularity of neural organoids as models has led to a wide range of methodologies aimed at continuous improvement and refinement. Consequently, research groups often improve and reconfigure protocols to create region-specific organoids, resulting in diverse phenotypes, including variations in morphology, gene expression, and cell populations. While these improvements are exciting, routine adoptions of such modifications and protocols in the research laboratories are often challenging due to the reiterative empirical testing necessary to validate the cell types generated. To address this challenge, we systematically compare the similarities and differences that exist across published protocols that generates subpallial-specific organoids to date. In this review, we focus specifically on exploring the production of major GABAergic neuronal subtypes, especially Medium Spiny Neurons (MSNs) and Interneurons (INs), from multiple subpallial organoid protocols. Importantly, we look to evaluate the cell type diversity and the molecular pathways manipulated to generate them, thus broadening our understanding of the existing subpallial organoids as well as assessing the in vitro applicability of specific patterning factors. Lastly, we discuss the current challenges and outlook on the improved patterning of region-specific neural organoids. Given the critical roles MSN and IN dysfunction play in neurological disorders, comprehending the GABAergic neurons generated by neural organoids will undoubtedly facilitate clinical translation.
Collapse
Affiliation(s)
- Narciso Pavon
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Neuroscience and Behavior, University of Massachusetts Amherst, Amherst, MA, United States
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
5
|
Zavalin K, Hassan A, Zhang Y, Khera Z, Lagrange AH. Region and layer-specific expression of GABA A receptor isoforms and KCC2 in developing cortex. Front Cell Neurosci 2024; 18:1390742. [PMID: 38894703 PMCID: PMC11184147 DOI: 10.3389/fncel.2024.1390742] [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: 02/23/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction γ-Aminobutyric acid (GABA) type A receptors (GABAARs) are ligand-gated Cl-channels that mediate the bulk of inhibitory neurotransmission in the mature CNS and are targets of many drugs. During cortical development, GABAAR-mediated signals are significantly modulated by changing subunit composition and expression of Cl-transporters as part of developmental processes and early network activity. To date, this developmental evolution has remained understudied, particularly at the level of cortical layer-specific changes. In this study, we characterized the expression of nine major GABAAR subunits and K-Cl transporter 2 (KCC2) in mouse somatosensory cortex from embryonic development to postweaning maturity. Methods We evaluated expression of α1-5, β2-3, γ2, and δ GABAAR subunits using immunohistochemistry and Western blot techniques, and expression of KCC2 using immunohistochemistry in cortices from E13.5 to P25 mice. Results We found that embryonic cortex expresses mainly α3, α5, β3, and γ2, while expression of α1, α2, α4, β2, δ, and KCC2 begins at later points in development; however, many patterns of nuanced expression can be found in specific lamina, cortical regions, and cells and structures. Discussion While the general pattern of expression of each subunit and KCC2 is similar to previous studies, we found a number of unique temporal, regional, and laminar patterns that were previously unknown. These findings provide much needed knowledge of the intricate developmental evolution in GABAAR composition and KCC2 expression to accommodate developmental signals that transition to mature neurotransmission.
Collapse
Affiliation(s)
- Kirill Zavalin
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Anjana Hassan
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Yueli Zhang
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Zain Khera
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Andre H. Lagrange
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Neurology, TVH VA Medical Center, Nashville, TN, United States
| |
Collapse
|
6
|
Irala D, Wang S, Sakers K, Nagendren L, Ulloa Severino FP, Bindu DS, Savage JT, Eroglu C. Astrocyte-secreted neurocan controls inhibitory synapse formation and function. Neuron 2024; 112:1657-1675.e10. [PMID: 38574730 PMCID: PMC11098688 DOI: 10.1016/j.neuron.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 01/22/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
Astrocytes strongly promote the formation and maturation of synapses by secreted proteins. Several astrocyte-secreted synaptogenic proteins controlling excitatory synapse development were identified; however, those that induce inhibitory synaptogenesis remain elusive. Here, we identify neurocan as an astrocyte-secreted inhibitory synaptogenic protein. After secretion from astrocytes, neurocan is cleaved into N- and C-terminal fragments. We found that these fragments have distinct localizations in the extracellular matrix. The neurocan C-terminal fragment localizes to synapses and controls cortical inhibitory synapse formation and function. Neurocan knockout mice lacking the whole protein or only its C-terminal synaptogenic domain have reduced inhibitory synapse numbers and function. Through super-resolution microscopy, in vivo proximity labeling by secreted TurboID, and astrocyte-specific rescue approaches, we discovered that the synaptogenic domain of neurocan localizes to somatostatin-positive inhibitory synapses and strongly regulates their formation. Together, our results unveil a mechanism through which astrocytes control circuit-specific inhibitory synapse development in the mammalian brain.
Collapse
Affiliation(s)
- Dolores Irala
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Shiyi Wang
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristina Sakers
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Leykashree Nagendren
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Francesco Paolo Ulloa Severino
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Psychology and Neuroscience, Duke University, Durham, NC 27710, USA; Instituto Cajal, CSIC 28002 Madrid, Spain
| | | | - Justin T Savage
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Psychology and Neuroscience, Duke University, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Duke Institute for Brain Sciences (DIBS), Durham, NC 27710, USA; Howard Hughes Medical Institute, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
7
|
Marchetta P, Dapper K, Hess M, Calis D, Singer W, Wertz J, Fink S, Hage SR, Alam M, Schwabe K, Lukowski R, Bourien J, Puel JL, Jacob MH, Munk MHJ, Land R, Rüttiger L, Knipper M. Dysfunction of specific auditory fibers impacts cortical oscillations, driving an autism phenotype despite near-normal hearing. FASEB J 2024; 38:e23411. [PMID: 38243766 DOI: 10.1096/fj.202301995r] [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: 09/29/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/21/2024]
Abstract
Autism spectrum disorder is discussed in the context of altered neural oscillations and imbalanced cortical excitation-inhibition of cortical origin. We studied here whether developmental changes in peripheral auditory processing, while preserving basic hearing function, lead to altered cortical oscillations. Local field potentials (LFPs) were recorded from auditory, visual, and prefrontal cortices and the hippocampus of BdnfPax2 KO mice. These mice develop an autism-like behavioral phenotype through deletion of BDNF in Pax2+ interneuron precursors, affecting lower brainstem functions, but not frontal brain regions directly. Evoked LFP responses to behaviorally relevant auditory stimuli were weaker in the auditory cortex of BdnfPax2 KOs, connected to maturation deficits of high-spontaneous rate auditory nerve fibers. This was correlated with enhanced spontaneous and induced LFP power, excitation-inhibition imbalance, and dendritic spine immaturity, mirroring autistic phenotypes. Thus, impairments in peripheral high-spontaneous rate fibers alter spike synchrony and subsequently cortical processing relevant for normal communication and behavior.
Collapse
Affiliation(s)
- Philine Marchetta
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Konrad Dapper
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Morgan Hess
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Dila Calis
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Wibke Singer
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Jakob Wertz
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Stefan Fink
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Steffen R Hage
- Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, Tübingen, Germany
| | - Mesbah Alam
- Experimental Neurosurgery, Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Kerstin Schwabe
- Experimental Neurosurgery, Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Robert Lukowski
- Institute of Pharmacy, Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Jerome Bourien
- Institute for Neurosciences Montpellier, Institut National de la Santé et de la Recherche Médical, University of Montpellier, Montpellier, France
| | - Jean-Luc Puel
- Institute for Neurosciences Montpellier, Institut National de la Santé et de la Recherche Médical, University of Montpellier, Montpellier, France
| | - Michele H Jacob
- Department of Neuroscience, Tufts University School of Medicine, Sackler School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Matthias H J Munk
- Department of Psychiatry & Psychotherapy, University of Tübingen, Tübingen, Germany
- Department of Biology, Technical University Darmstadt, Darmstadt, Germany
| | - Rüdiger Land
- Department of Experimental Otology, Institute of Audioneurotechnology, Hannover Medical School, Hannover, Germany
| | - Lukas Rüttiger
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| | - Marlies Knipper
- Molecular Physiology of Hearing, Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, University of Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Pavon N, Diep K, Yang F, Sebastian R, Martinez-Martin B, Ranjan R, Sun Y, Pak C. Patterning ganglionic eminences in developing human brain organoids using a morphogen-gradient-inducing device. CELL REPORTS METHODS 2024; 4:100689. [PMID: 38228151 PMCID: PMC10831957 DOI: 10.1016/j.crmeth.2023.100689] [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: 06/19/2023] [Revised: 10/21/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
In early neurodevelopment, the central nervous system is established through the coordination of various neural organizers directing tissue patterning and cell differentiation. Better recapitulation of morphogen gradient production and signaling will be crucial for establishing improved developmental models of the brain in vitro. Here, we developed a method by assembling polydimethylsiloxane devices capable of generating a sustained chemical gradient to produce patterned brain organoids, which we termed morphogen-gradient-induced brain organoids (MIBOs). At 3.5 weeks, MIBOs replicated dorsal-ventral patterning observed in the ganglionic eminences (GE). Analysis of mature MIBOs through single-cell RNA sequencing revealed distinct dorsal GE-derived CALB2+ interneurons, medium spiny neurons, and medial GE-derived cell types. Finally, we demonstrate long-term culturing capabilities with MIBOs maintaining stable neural activity in cultures grown up to 5.5 months. MIBOs demonstrate a versatile approach for generating spatially patterned brain organoids for embryonic development and disease modeling.
Collapse
Affiliation(s)
- Narciso Pavon
- Graduate Program in Neuroscience and Behavior, UMass Amherst, Amherst, MA 01003, USA; Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Karmen Diep
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Feiyu Yang
- Department of Mechanical and Industrial Engineering, UMass Amherst, Amherst, MA 01003, USA
| | - Rebecca Sebastian
- Graduate Program in Neuroscience and Behavior, UMass Amherst, Amherst, MA 01003, USA; Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Beatriz Martinez-Martin
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA; Graduate Program in Molecular and Cellular Biology, UMass Amherst, Amherst, MA 01003, USA
| | - Ravi Ranjan
- Genomics Core, Institute of Applied Life Sciences, UMass Amherst, Amherst, MA 01003, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, UMass Amherst, Amherst, MA 01003, USA.
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
9
|
Irala D, Wang S, Sakers K, Nagendren L, Ulloa-Severino FP, Bindu DS, Eroglu C. Astrocyte-Secreted Neurocan Controls Inhibitory Synapse Formation and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535448. [PMID: 37066164 PMCID: PMC10104008 DOI: 10.1101/2023.04.03.535448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Astrocytes strongly promote the formation and maturation of synapses by secreted proteins. To date, several astrocyte-secreted synaptogenic proteins controlling different stages of excitatory synapse development have been identified. However, the identities of astrocytic signals that induce inhibitory synapse formation remain elusive. Here, through a combination of in vitro and in vivo experiments, we identified Neurocan as an astrocyte-secreted inhibitory synaptogenic protein. Neurocan is a chondroitin sulfate proteoglycan that is best known as a protein localized to the perineuronal nets. However, Neurocan is cleaved into two after secretion from astrocytes. We found that the resulting N- and C-terminal fragments have distinct localizations in the extracellular matrix. While the N-terminal fragment remains associated with perineuronal nets, the Neurocan C-terminal fragment localizes to synapses and specifically controls cortical inhibitory synapse formation and function. Neurocan knockout mice lacking the whole protein or only its C-terminal synaptogenic region have reduced inhibitory synapse numbers and function. Through super-resolution microscopy and in vivo proximity labeling by secreted TurboID, we discovered that the synaptogenic domain of Neurocan localizes to somatostatin-positive inhibitory synapses and strongly regulates their formation. Together, our results unveil a mechanism through which astrocytes control circuit-specific inhibitory synapse development in the mammalian brain.
Collapse
|
10
|
D’Arcy BR, Lennox AL, Manso Musso C, Bracher A, Escobar-Tomlienovich C, Perez-Sanchez S, Silver DL. Non-muscle myosins control radial glial basal endfeet to mediate interneuron organization. PLoS Biol 2023; 21:e3001926. [PMID: 36854011 PMCID: PMC9974137 DOI: 10.1371/journal.pbio.3001926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/17/2023] [Indexed: 03/02/2023] Open
Abstract
Radial glial cells (RGCs) are essential for the generation and organization of neurons in the cerebral cortex. RGCs have an elongated bipolar morphology with basal and apical endfeet that reside in distinct niches. Yet, how this subcellular compartmentalization of RGCs controls cortical development is largely unknown. Here, we employ in vivo proximity labeling, in the mouse, using unfused BirA to generate the first subcellular proteome of RGCs and uncover new principles governing local control of cortical development. We discover a cohort of proteins that are significantly enriched in RGC basal endfeet, with MYH9 and MYH10 among the most abundant. Myh9 and Myh10 transcripts also localize to endfeet with distinct temporal dynamics. Although they each encode isoforms of non-muscle myosin II heavy chain, Myh9 and Myh10 have drastically different requirements for RGC integrity. Myh9 loss from RGCs decreases branching complexity and causes endfoot protrusion through the basement membrane. In contrast, Myh10 controls endfoot adhesion, as mutants have unattached apical and basal endfeet. Finally, we show that Myh9- and Myh10-mediated regulation of RGC complexity and endfoot position non-cell autonomously controls interneuron number and organization in the marginal zone. Our study demonstrates the utility of in vivo proximity labeling for dissecting local control of complex systems and reveals new mechanisms for dictating RGC integrity and cortical architecture.
Collapse
Affiliation(s)
- Brooke R. D’Arcy
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Ashley L. Lennox
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Camila Manso Musso
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Annalise Bracher
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Carla Escobar-Tomlienovich
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Stephany Perez-Sanchez
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Debra L. Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Institute for Brain Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Regeneration Center, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
11
|
Banerjee T, Pati S, Tiwari P, Vaidya VA. Chronic hM3Dq-DREADD-mediated chemogenetic activation of parvalbumin-positive inhibitory interneurons in postnatal life alters anxiety and despair-like behavior in adulthood in a task- and sex-dependent manner. J Biosci 2022. [DOI: 10.1007/s12038-022-00308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
12
|
Khozhai LI, Otellin VA. Distribution of GABAergic Neurons and Expression Levels of GABA Transporter 1 in the Rat Neocortex during the Neonatal Period after Perinatal Hypoxic Exposure. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
13
|
Hui KK, Chater TE, Goda Y, Tanaka M. How Staying Negative Is Good for the (Adult) Brain: Maintaining Chloride Homeostasis and the GABA-Shift in Neurological Disorders. Front Mol Neurosci 2022; 15:893111. [PMID: 35875665 PMCID: PMC9305173 DOI: 10.3389/fnmol.2022.893111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/10/2022] [Indexed: 01/27/2023] Open
Abstract
Excitatory-inhibitory (E-I) imbalance has been shown to contribute to the pathogenesis of a wide range of neurodevelopmental disorders including autism spectrum disorders, epilepsy, and schizophrenia. GABA neurotransmission, the principal inhibitory signal in the mature brain, is critically coupled to proper regulation of chloride homeostasis. During brain maturation, changes in the transport of chloride ions across neuronal cell membranes act to gradually change the majority of GABA signaling from excitatory to inhibitory for neuronal activation, and dysregulation of this GABA-shift likely contributes to multiple neurodevelopmental abnormalities that are associated with circuit dysfunction. Whilst traditionally viewed as a phenomenon which occurs during brain development, recent evidence suggests that this GABA-shift may also be involved in neuropsychiatric disorders due to the "dematuration" of affected neurons. In this review, we will discuss the cell signaling and regulatory mechanisms underlying the GABA-shift phenomenon in the context of the latest findings in the field, in particular the role of chloride cotransporters NKCC1 and KCC2, and furthermore how these regulatory processes are altered in neurodevelopmental and neuropsychiatric disorders. We will also explore the interactions between GABAergic interneurons and other cell types in the developing brain that may influence the GABA-shift. Finally, with a greater understanding of how the GABA-shift is altered in pathological conditions, we will briefly outline recent progress on targeting NKCC1 and KCC2 as a therapeutic strategy against neurodevelopmental and neuropsychiatric disorders associated with improper chloride homeostasis and GABA-shift abnormalities.
Collapse
Affiliation(s)
- Kelvin K. Hui
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Thomas E. Chater
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
| | - Yukiko Goda
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako, Japan
- Synapse Biology Unit, Okinawa Institute for Science and Technology Graduate University, Onna, Japan
| | - Motomasa Tanaka
- Laboratory for Protein Conformation Diseases, RIKEN Center for Brain Science, Wako, Japan
| |
Collapse
|
14
|
Llorca A, Deogracias R. Origin, Development, and Synaptogenesis of Cortical Interneurons. Front Neurosci 2022; 16:929469. [PMID: 35833090 PMCID: PMC9272671 DOI: 10.3389/fnins.2022.929469] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex represents one of the most recent and astonishing inventions of nature, responsible of a large diversity of functions that range from sensory processing to high-order cognitive abilities, such as logical reasoning or language. Decades of dedicated study have contributed to our current understanding of this structure, both at structural and functional levels. A key feature of the neocortex is its outstanding richness in cell diversity, composed by multiple types of long-range projecting neurons and locally connecting interneurons. In this review, we will describe the great diversity of interneurons that constitute local neocortical circuits and summarize the mechanisms underlying their development and their assembly into functional networks.
Collapse
Affiliation(s)
- Alfredo Llorca
- Visual Neuroscience Laboratory, Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of Edinburgh, Edinburg, United Kingdom
- *Correspondence: Alfredo Llorca
| | - Ruben Deogracias
- Neuronal Circuits Formation and Brain Disorders Laboratory, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Department of Cell Biology and Pathology, School of Medicine, University of Salamanca, Salamanca, Spain
- Ruben Deogracias
| |
Collapse
|
15
|
Cossart R, Garel S. Step by step: cells with multiple functions in cortical circuit assembly. Nat Rev Neurosci 2022; 23:395-410. [DOI: 10.1038/s41583-022-00585-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2022] [Indexed: 12/23/2022]
|
16
|
Genescu I, Aníbal-Martínez M, Kouskoff V, Chenouard N, Mailhes-Hamon C, Cartonnet H, Lokmane L, Rijli FM, López-Bendito G, Gambino F, Garel S. Dynamic interplay between thalamic activity and Cajal-Retzius cells regulates the wiring of cortical layer 1. Cell Rep 2022; 39:110667. [PMID: 35417707 PMCID: PMC9035679 DOI: 10.1016/j.celrep.2022.110667] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/17/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Abstract
Cortical wiring relies on guidepost cells and activity-dependent processes that are thought to act sequentially. Here, we show that the construction of layer 1 (L1), a main site of top-down integration, is regulated by crosstalk between transient Cajal-Retzius cells (CRc) and spontaneous activity of the thalamus, a main driver of bottom-up information. While activity was known to regulate CRc migration and elimination, we found that prenatal spontaneous thalamic activity and NMDA receptors selectively control CRc early density, without affecting their demise. CRc density, in turn, regulates the distribution of upper layer interneurons and excitatory synapses, thereby drastically impairing the apical dendrite activity of output pyramidal neurons. In contrast, postnatal sensory-evoked activity had a limited impact on L1 and selectively perturbed basal dendrites synaptogenesis. Collectively, our study highlights a remarkable interplay between thalamic activity and CRc in L1 functional wiring, with major implications for our understanding of cortical development. Prenatal thalamic waves of activity regulate CRc density in L1 Prenatal and postnatal CRc manipulations alter specific interneuron populations Postnatal CRc shape L5 apical dendrite structural and functional properties Early sensory activity selectively regulates L5 basal dendrite spine formation
Collapse
Affiliation(s)
- Ioana Genescu
- Institut de Biologie de l'École Normale Supérieure (IBENS), Département de Biologie, École Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Mar Aníbal-Martínez
- Instituto de Neurosciencias de Alicante, Universidad Miguel Hernandez, Sant Joan d'Alacant, Spain
| | - Vladimir Kouskoff
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Nicolas Chenouard
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Caroline Mailhes-Hamon
- Acute Transgenesis Facility, Institut de Biologie de l'École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, PSL Université Paris, 75005 Paris, France
| | - Hugues Cartonnet
- Institut de Biologie de l'École Normale Supérieure (IBENS), Département de Biologie, École Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Ludmilla Lokmane
- Institut de Biologie de l'École Normale Supérieure (IBENS), Département de Biologie, École Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; University of Basel, 4056 Basel, Switzerland
| | | | - Frédéric Gambino
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS UMR 5297, 33000 Bordeaux, France
| | - Sonia Garel
- Institut de Biologie de l'École Normale Supérieure (IBENS), Département de Biologie, École Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France; Collège de France, 75005 Paris, France.
| |
Collapse
|
17
|
Wang Q, Li Y, Tan H, Wang Y. Sevoflurane-Induced Apoptosis in the Mouse Cerebral Cortex Follows Similar Characteristics of Physiological Apoptosis. Front Mol Neurosci 2022; 15:873658. [PMID: 35465098 PMCID: PMC9024292 DOI: 10.3389/fnmol.2022.873658] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
General anesthetics are capable of inducing neuronal apoptosis during the rapid synaptogenesis of immature mammalian brains. In this vulnerable time window, physiological apoptosis also occurs to eliminate excess and inappropriately integrated neurons. We previously showed that physiological and ketamine-induced apoptosis in mouse primary somatosensory cortex (S1) followed similar developmental patterns. However, since sevoflurane is more widely used in pediatric anesthesia, and targets mainly on different receptors, as compared with ketamine, it is important to determine whether sevoflurane-induced apoptosis also follows similar developmental patterns as physiological apoptosis or not. Mice at postnatal days 5 (P5) and P9 were anesthetized with 1.5% sevoflurane for 4 h, and the apoptotic neurons in S1 were quantitated by immunohistochemistry. The results showed that sevoflurane raised the levels of apoptosis in S1 without interfering with the developmental patterns of physiological apoptosis. The cells more vulnerable to both physiological and sevoflurane-induced apoptosis shifted from layer V pyramidal neurons at P5 to layers II–IV GABAergic neurons by P9. The magnitude of both sevoflurane-induced and physiological apoptosis was more attenuated at P9 than P5. To determine whether the Akt-FoxO1-PUMA pathway contributes to the developmental decrease in magnitude of both physiological and sevoflurane-induced apoptosis, Western blot was used to measure the levels of related proteins in S1 of P5 and P9 mice. We observed higher levels of antiapoptotic phosphorylated Akt (p-Akt) and phosphorylated FoxO1 (p-FoxO1), and lower levels of the downstream proapoptotic factor PUMA in control and anesthetized mice at P9 than P5. In addition, the Akt-FoxO1-PUMA pathway may also be responsible for sevoflurane-induced apoptosis. Together, these results suggest that magnitude, lamination pattern and cell-type specificity to sevoflurane-induced apoptosis are age-dependent and follow physiological apoptosis pattern. Moreover, The Akt-FoxO1-PUMA pathway may mediate the developmental decreases in magnitude of both physiological and sevoflurane-induced apoptosis in neonatal mouse S1.
Collapse
|
18
|
Tufo C, Poopalasundaram S, Dorrego-Rivas A, Ford MC, Graham A, Grubb MS. Development of the mammalian main olfactory bulb. Development 2022; 149:274348. [PMID: 35147186 PMCID: PMC8918810 DOI: 10.1242/dev.200210] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The mammalian main olfactory bulb is a crucial processing centre for the sense of smell. The olfactory bulb forms early during development and is functional from birth. However, the olfactory system continues to mature and change throughout life as a target of constitutive adult neurogenesis. Our Review synthesises current knowledge of prenatal, postnatal and adult olfactory bulb development, focusing on the maturation, morphology, functions and interactions of its diverse constituent glutamatergic and GABAergic cell types. We highlight not only the great advances in the understanding of olfactory bulb development made in recent years, but also the gaps in our present knowledge that most urgently require addressing. Summary: This Review describes the morphological and functional maturation of cells in the mammalian main olfactory bulb, from embryonic development to adult neurogenesis.
Collapse
Affiliation(s)
- Candida Tufo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Subathra Poopalasundaram
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Ana Dorrego-Rivas
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Marc C Ford
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Anthony Graham
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Matthew S Grubb
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| |
Collapse
|
19
|
Kalemaki K, Velli A, Christodoulou O, Denaxa M, Karagogeos D, Sidiropoulou K. The Developmental Changes in Intrinsic and Synaptic Properties of Prefrontal Neurons Enhance Local Network Activity from the Second to the Third Postnatal Weeks in Mice. Cereb Cortex 2021; 32:3633-3650. [PMID: 34905772 DOI: 10.1093/cercor/bhab438] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
The prefrontal cortex (PFC) is characterized by protracted maturation. The cellular mechanisms controlling the early development of prefrontal circuits are still largely unknown. Our study delineates the developmental cellular processes in the mouse medial PFC (mPFC) during the second and the third postnatal weeks and characterizes their contribution to the changes in network activity. We show that spontaneous inhibitory postsynaptic currents (sIPSC) are increased, whereas spontaneous excitatory postsynaptic currents (sEPSC) are reduced from the second to the third postnatal week. Drug application suggested that the increased sEPSC frequency in mPFC at postnatal day 10 (P10) is due to depolarizing γ-aminobutyric acid (GABA) type A receptor function. To further validate this, perforated patch-clamp recordings were obtained and the expression levels of K-Cl cotransporter 2 (KCC2) protein were examined. The reversal potential of IPSCs in response to current stimulation was significantly more depolarized at P10 than P20 while KCC2 expression is decreased. Moreover, the number of parvalbumin-expressing GABAergic interneurons increases and their intrinsic electrophysiological properties significantly mature in the mPFC from P10 to P20. Using computational modeling, we show that the developmental changes in synaptic and intrinsic properties of mPFC neurons contribute to the enhanced network activity in the juvenile compared with neonatal mPFC.
Collapse
Affiliation(s)
- Katerina Kalemaki
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion GR70013, Greece.,Institute of Molecular Biology and Biotechnology (IMBB), FORTH, Heraklion GR70013, Greece
| | - Angeliki Velli
- Institute of Molecular Biology and Biotechnology (IMBB), FORTH, Heraklion GR70013, Greece.,Department of Biology, University of Crete, Heraklion GR70013, Greece
| | - Ourania Christodoulou
- Department of Biology, University of Crete, Heraklion GR70013, Greece.,Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center 'Alexander Fleming', Heraklion GR70013, Greece
| | - Myrto Denaxa
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center 'Alexander Fleming', Heraklion GR70013, Greece
| | - Domna Karagogeos
- Department of Basic Science, Faculty of Medicine, University of Crete, Heraklion GR70013, Greece.,Institute of Molecular Biology and Biotechnology (IMBB), FORTH, Heraklion GR70013, Greece
| | - Kyriaki Sidiropoulou
- Institute of Molecular Biology and Biotechnology (IMBB), FORTH, Heraklion GR70013, Greece.,Department of Biology, University of Crete, Heraklion GR70013, Greece
| |
Collapse
|
20
|
Postnatal Sox6 Regulates Synaptic Function of Cortical Parvalbumin-Expressing Neurons. J Neurosci 2021; 41:8876-8886. [PMID: 34503995 PMCID: PMC8549537 DOI: 10.1523/jneurosci.0021-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 11/29/2022] Open
Abstract
Cortical parvalbumin-expressing (Pvalb+) neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. This class of inhibitory neurons undergoes extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. While several transcription factors, such as Nkx2-1, Lhx6, and Sox6, are known to be necessary for the differentiation of progenitors into Pvalb+ neurons, which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons' innervation and synaptic function remains largely unknown. Because Sox6 is continuously expressed in Pvalb+ neurons until adulthood, we used conditional knock-out strategies to investigate its putative role in the postnatal maturation and synaptic function of cortical Pvalb+ neurons in mice of both sexes. We found that early postnatal loss of Sox6 in Pvalb+ neurons leads to failure of synaptic bouton growth, whereas later removal in mature Pvalb+ neurons in the adult causes shrinkage of already established synaptic boutons. Paired recordings between Pvalb+ neurons and pyramidal neurons revealed reduced release probability and increased failure rate of Pvalb+ neurons' synaptic output. Furthermore, Pvalb+ neurons lacking Sox6 display reduced expression of full-length tropomyosin-receptor kinase B (TrkB), a key modulator of GABAergic transmission. Once re-expressed in neurons lacking Sox6, TrkB was sufficient to rescue the morphologic synaptic phenotype. Finally, we showed that Sox6 mRNA levels were increased by motor training. Our data thus suggest a constitutive role for Sox6 in the maintenance of synaptic output from Pvalb+ neurons into adulthood. SIGNIFICANCE STATEMENT Cortical parvalbumin-expressing (Pvalb+) inhibitory neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. These inhibitory neurons undergo extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. However, it remains largely unknown which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons. Here, we show that the transcription factor Sox6 cell-autonomously regulates the synaptic maintenance and output of Pvalb+ neurons until adulthood, leaving unaffected other maturational features of this neuronal population.
Collapse
|
21
|
White AK, Baumgartner M, Lee MF, Drake KD, Aquino GS, Kanadia RN. Trp53 ablation fails to prevent microcephaly in mouse pallium with impaired minor intron splicing. Development 2021; 148:272517. [PMID: 34557915 DOI: 10.1242/dev.199591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/17/2021] [Indexed: 12/21/2022]
Abstract
Minor spliceosome inhibition due to mutations in RNU4ATAC are linked to primary microcephaly. Ablation of Rnu11, which encodes a minor spliceosome snRNA, inhibits the minor spliceosome in the developing mouse pallium, causing microcephaly. There, cell cycle defects and p53-mediated apoptosis in response to DNA damage resulted in loss of radial glial cells (RGCs), underpinning microcephaly. Here, we ablated Trp53 to block cell death in Rnu11 cKO mice. We report that Trp53 ablation failed to prevent microcephaly in these double knockout (dKO) mice. We show that the transcriptome of the dKO pallium was more similar to the control compared with the Rnu11 cKO. We find aberrant minor intron splicing in minor intron-containing genes involved in cell cycle regulation, resulting in more severely impaired mitotic progression and cell cycle lengthening of RGCs in the dKO that was detected earlier than in the Rnu11 cKO. Furthermore, we discover a potential role of p53 in causing DNA damage in the developing pallium, as detection of γH2aX+ was delayed in the dKO. Thus, we postulate that microcephaly in minor spliceosome-related diseases is primarily caused by cell cycle defects.
Collapse
Affiliation(s)
- Alisa K White
- Physiology and Neurobiology Department, University of Connecticut, Storrs, CT 06269, USA
| | | | - Madisen F Lee
- Physiology and Neurobiology Department, University of Connecticut, Storrs, CT 06269, USA
| | - Kyle D Drake
- Physiology and Neurobiology Department, University of Connecticut, Storrs, CT 06269, USA
| | - Gabriela S Aquino
- Physiology and Neurobiology Department, University of Connecticut, Storrs, CT 06269, USA
| | - Rahul N Kanadia
- Physiology and Neurobiology Department, University of Connecticut, Storrs, CT 06269, USA.,Institute of Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
22
|
Capsoni S, Fogli Iseppe A, Casciano F, Pignatelli A. Unraveling the Role of Dopaminergic and Calretinin Interneurons in the Olfactory Bulb. Front Neural Circuits 2021; 15:718221. [PMID: 34690707 PMCID: PMC8531203 DOI: 10.3389/fncir.2021.718221] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/31/2021] [Indexed: 11/27/2022] Open
Abstract
The perception and discriminating of odors are sensory activities that are an integral part of our daily life. The first brain region where odors are processed is the olfactory bulb (OB). Among the different cell populations that make up this brain area, interneurons play an essential role in this sensory activity. Moreover, probably because of their activity, they represent an exception compared to other parts of the brain, since OB interneurons are continuously generated in the postnatal and adult period. In this review, we will focus on periglomerular (PG) cells which are a class of interneurons found in the glomerular layer of the OB. These interneurons can be classified into distinct subtypes based on their neurochemical nature, based on the neurotransmitter and calcium-binding proteins expressed by these cells. Dopaminergic (DA) periglomerular cells and calretinin (CR) cells are among the newly generated interneurons and play an important role in the physiology of OB. In the OB, DA cells are involved in the processing of odors and the adaptation of the bulbar network to external conditions. The main role of DA cells in OB appears to be the inhibition of glutamate release from olfactory sensory fibers. Calretinin cells are probably the best morphologically characterized interneurons among PG cells in OB, but little is known about their function except for their inhibitory effect on noisy random excitatory signals arriving at the main neurons. In this review, we will mainly describe the electrophysiological properties related to the excitability profiles of DA and CR cells, with a particular view on the differences that characterize DA mature interneurons from cells in different stages of adult neurogenesis.
Collapse
Affiliation(s)
- Simona Capsoni
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Alex Fogli Iseppe
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Fabio Casciano
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
- Interdepartmental Research Centre for the Study of Multiple Sclerosis and Inflammatory and Degenerative Diseases of the Nervous System, University of Ferrara, Ferrara, Italy
| | - Angela Pignatelli
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| |
Collapse
|
23
|
Esumi S, Nasu M, Kawauchi T, Miike K, Morooka K, Yanagawa Y, Seki T, Sakimura K, Fukuda T, Tamamaki N. Characterization and Stage-Dependent Lineage Analysis of Intermediate Progenitors of Cortical GABAergic Interneurons. Front Neurosci 2021; 15:607908. [PMID: 34305510 PMCID: PMC8297055 DOI: 10.3389/fnins.2021.607908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Abstract
Intermediate progenitors of both excitatory and inhibitory neurons, which can replenish neurons in the adult brain, were recently identified. However, the generation of intermediate progenitors of GABAergic inhibitory neurons (IPGNs) has not been studied in detail. Here, we characterized the spatiotemporal distribution of IPGNs in mouse cerebral cortex. IPGNs generated neurons during both embryonic and postnatal stages, but the embryonic IPGNs were more proliferative. Our lineage tracing analyses showed that the embryonically proliferating IPGNs tended to localize to the superficial layers rather than the deep cortical layers at 3 weeks after birth. We also found that embryonic IPGNs derived from the medial and caudal ganglionic eminence (CGE) but more than half of the embryonic IPGNs were derived from the CGE and broadly distributed in the cerebral cortex. Taken together, our data indicate that the broadly located IPGNs during embryonic and postnatal stages exhibit a different proliferative property and layer distribution.
Collapse
Affiliation(s)
- Shigeyuki Esumi
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Makoto Nasu
- Department of Health Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Kawauchi
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| | - Koichiro Miike
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | | | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takaichi Fukuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Nobuaki Tamamaki
- Department of Morphological Neural Science, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
24
|
De Gregorio R, Chen X, Petit EI, Dobrenis K, Sze JY. Disruption of Transient SERT Expression in Thalamic Glutamatergic Neurons Alters Trajectory of Postnatal Interneuron Development in the Mouse Cortex. Cereb Cortex 2021; 30:1623-1636. [PMID: 31504267 DOI: 10.1093/cercor/bhz191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/29/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
In mice, terminal differentiation of subpopulations of interneurons occurs in late postnatal stages, paralleling the emergence of the adult cortical architecture. Here, we investigated the effects of altered initial cortical architecture on later interneuron development. We identified that a class of somatostatin (SOM)-expressing GABAergic interneurons undergoes terminal differentiation between 2nd and 3rd postnatal week in the mouse somatosensory barrel cortex and upregulates Reelin expression during neurite outgrowth. Our previous work demonstrated that transient expression (E15-P10) of serotonin uptake transporter (SERT) in thalamocortical projection neurons regulates barrel elaboration during cortical map establishment. We show here that in thalamic neuron SERT knockout mice, these SOM-expressing interneurons develop at the right time, reach correct positions and express correct neurochemical markers, but only 70% of the neurons remain in the adult barrel cortex. Moreover, those neurons that remain display altered dendritic patterning. Our data indicate that a precise architecture at the cortical destination is not essential for specifying late-developing interneuron identities, their cortical deposition, and spatial organization, but dictates their number and dendritic structure ultimately integrated into the cortex. Our study illuminates how disruption of temporal-specific SERT function and related key regulators during cortical map establishment can alter interneuron development trajectory that persists to adult central nervous system.
Collapse
Affiliation(s)
- Roberto De Gregorio
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Xiaoning Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Emilie I Petit
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| |
Collapse
|
25
|
The role of GABAergic signalling in neurodevelopmental disorders. Nat Rev Neurosci 2021; 22:290-307. [PMID: 33772226 PMCID: PMC9001156 DOI: 10.1038/s41583-021-00443-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
GABAergic inhibition shapes the connectivity, activity and plasticity of the brain. A series of exciting new discoveries provides compelling evidence that disruptions in a number of key facets of GABAergic inhibition have critical roles in the aetiology of neurodevelopmental disorders (NDDs). These facets include the generation, migration and survival of GABAergic neurons, the formation of GABAergic synapses and circuit connectivity, and the dynamic regulation of the efficacy of GABAergic signalling through neuronal chloride transporters. In this Review, we discuss recent work that elucidates the functions and dysfunctions of GABAergic signalling in health and disease, that uncovers the contribution of GABAergic neural circuit dysfunction to NDD aetiology and that leverages such mechanistic insights to advance precision medicine for the treatment of NDDs.
Collapse
|
26
|
Sevoflurane impairs m6A-mediated mRNA translation and leads to fine motor and cognitive deficits. Cell Biol Toxicol 2021; 38:347-369. [PMID: 33928466 DOI: 10.1007/s10565-021-09601-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 03/10/2021] [Indexed: 12/14/2022]
Abstract
Clinical surgical practices have found that children who undergo multiple anesthesia may have an increased risk of deficiencies in cognition and fine motor control. Here, we report that YT521-B homology domain family 1 (YTHDF1), a critical reader protein for N6-methyladenosine-modified mRNA, was significantly downregulated in the prefrontal cortex of young mice after multiple sevoflurane anesthesia exposures. Importantly, sevoflurane led to a decrease in protein synthesis in mouse cortical neurons that was fully rescued by YTHDF1, suggesting that anesthesia may affect early brain development by affecting m6A-dependent mRNA translation. Transcriptome-wide experiments showed that numerous mRNA targets related to synaptic functions in the prefrontal mouse cortex were associated with m6A methylation and YTHDF1. In particular, we found that synaptophysin, a critical presynaptic protein, was specifically modified by m6A methylation and associated with YTHDF1, and m6A methylation of synaptophysin decreased with multiple sevoflurane exposures. Importantly, we showed that fine motor control skills and cognitive functions were impaired in mice with multiple anesthesia exposures, and these effects were fully reversed by reintroducing YTHDF1 through a blood-brain barrier (BBB)-crossing viral delivery system. Finally, we found that the fine motor skills in children who underwent prolonged anesthesia were compromised 6 months after surgery. Our findings indicated that impairment in the translational regulation of mRNA via N6-methyladenosine methylation is a potential mechanism underlying the effects of anesthesia on neural development in the young brain. 1. N6-methyladenosine (m6A) modifications were involved in anesthesia-induced neurotoxicity. 2. Sevoflurane impairs m6A-mediated mRNA translation and leads to fine motor deficits in young mice. 3. YTHDF1, a m6A reader protein, rescued sevoflurane-induced protein synthesis inhibition and fine motor deficits in young mice.
Collapse
|
27
|
Vasistha NA, Khodosevich K. The impact of (ab)normal maternal environment on cortical development. Prog Neurobiol 2021; 202:102054. [PMID: 33905709 DOI: 10.1016/j.pneurobio.2021.102054] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/01/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
The cortex in the mammalian brain is the most complex brain region that integrates sensory information and coordinates motor and cognitive processes. To perform such functions, the cortex contains multiple subtypes of neurons that are generated during embryogenesis. Newly born neurons migrate to their proper location in the cortex, grow axons and dendrites, and form neuronal circuits. These developmental processes in the fetal brain are regulated to a large extent by a great variety of factors derived from the mother - starting from simple nutrients as building blocks and ending with hormones. Thus, when the normal maternal environment is disturbed due to maternal infection, stress, malnutrition, or toxic substances, it might have a profound impact on cortical development and the offspring can develop a variety of neurodevelopmental disorders. Here we first describe the major developmental processes which generate neuronal diversity in the cortex. We then review our knowledge of how most common maternal insults affect cortical development, perturb neuronal circuits, and lead to neurodevelopmental disorders. We further present a concept of selective vulnerability of cortical neuronal subtypes to maternal-derived insults, where the vulnerability of cortical neurons and their progenitors to an insult depends on the time (developmental period), place (location in the developing brain), and type (unique features of a cell type and an insult). Finally, we provide evidence for the existence of selective vulnerability during cortical development and identify the most vulnerable neuronal types, stages of differentiation, and developmental time for major maternal-derived insults.
Collapse
Affiliation(s)
- Navneet A Vasistha
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
28
|
Timely Inhibitory Circuit Formation Controlled by Abl1 Regulates Innate Olfactory Behaviors in Mouse. Cell Rep 2021; 30:187-201.e4. [PMID: 31914386 DOI: 10.1016/j.celrep.2019.12.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 10/16/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022] Open
Abstract
More than one-half of the interneurons in a mouse olfactory bulb (OB) develop during the first week after birth and predominantly connect to excitatory tufted cells near the superficial granule cell layer (sGCL), unlike late-born interneurons. However, the molecular mechanisms underlying the temporal specification are yet to be identified. In this study, we determined the role of Abelson tyrosine-protein kinase 1 (Abl1) in the temporal development of early-born OB interneurons. Lentiviral knockdown of Abl1 disrupts the sGCL circuit of early-born interneurons through defects in function and circuit integration, resulting in olfactory hyper-sensitivity. We show that doublecortin (Dcx) is phosphorylated by Abl1, which contributes to the stabilization of Dcx, thereby regulating microtubule dynamics. Finally, Dcx overexpression rescues Abl1 knockdown-induced anatomic or functional defects. In summary, specific signaling by Abl1-Dcx in early-born interneurons facilitates the temporal development of the sGCL circuit to regulate innate olfactory functions, such as detection and sensitivity.
Collapse
|
29
|
Matsui TK, Tsuru Y, Kuwako KI. Challenges in Modeling Human Neural Circuit Formation via Brain Organoid Technology. Front Cell Neurosci 2020; 14:607399. [PMID: 33362473 PMCID: PMC7756199 DOI: 10.3389/fncel.2020.607399] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/12/2020] [Indexed: 01/12/2023] Open
Abstract
Human brain organoids are three-dimensional self-organizing tissues induced from pluripotent cells that recapitulate some aspects of early development and some of the early structure of the human brain in vitro. Brain organoids consist of neural lineage cells, such as neural stem/precursor cells, neurons, astrocytes and oligodendrocytes. Additionally, brain organoids contain fluid-filled ventricle-like structures surrounded by a ventricular/subventricular (VZ/SVZ) zone-like layer of neural stem cells (NSCs). These NSCs give rise to neurons, which form multiple outer layers. Since these structures resemble some aspects of structural arrangements in the developing human brain, organoid technology has attracted great interest in the research fields of human brain development and disease modeling. Developmental brain disorders have been intensely studied through the use of human brain organoids. Relatively early steps in human brain development, such as differentiation and migration, have also been studied. However, research on neural circuit formation with brain organoids has just recently began. In this review, we summarize the current challenges in studying neural circuit formation with organoids and discuss future perspectives.
Collapse
Affiliation(s)
- Takeshi K Matsui
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, Izumo, Japan
| | - Yuichiro Tsuru
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, Izumo, Japan
| | - Ken-Ichiro Kuwako
- Department of Neural and Muscular Physiology, Shimane University School of Medicine, Izumo, Japan
| |
Collapse
|
30
|
Genescu I, Garel S. Being superficial: a developmental viewpoint on cortical layer 1 wiring. Curr Opin Neurobiol 2020; 66:125-134. [PMID: 33186879 DOI: 10.1016/j.conb.2020.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/03/2020] [Accepted: 10/04/2020] [Indexed: 01/01/2023]
Abstract
Functioning of the neocortex relies on a complex architecture of circuits, as illustrated by the causal link between neocortical excitation/inhibition imbalance and the etiology of several neurodevelopmental disorders. An important entry point to cortical circuits is located in the superficial layer 1 (L1), which contains mostly local and long-range inputs and sparse inhibitory interneurons that collectively regulate cerebral functions. While increasing evidence indicates that L1 has important physiological roles, our understanding of how it wires up during development remains limited. Here, we provide an integrated overview of L1 anatomy, function and development, with a focus on transient early born Cajal-Retzius neurons, and highlight open questions key for progressing our understanding of this essential yet understudied layer of the cerebral cortex.
Collapse
Affiliation(s)
- Ioana Genescu
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Sonia Garel
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France; Collège de France, Paris, France.
| |
Collapse
|
31
|
Ekins TG, Mahadevan V, Zhang Y, D'Amour JA, Akgül G, Petros TJ, McBain CJ. Emergence of non-canonical parvalbumin-containing interneurons in hippocampus of a murine model of type I lissencephaly. eLife 2020; 9:e62373. [PMID: 33150866 PMCID: PMC7673787 DOI: 10.7554/elife.62373] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022] Open
Abstract
Type I lissencephaly is a neuronal migration disorder caused by haploinsuffiency of the PAFAH1B1 (mouse: Pafah1b1) gene and is characterized by brain malformation, developmental delays, and epilepsy. Here, we investigate the impact of Pafah1b1 mutation on the cellular migration, morphophysiology, microcircuitry, and transcriptomics of mouse hippocampal CA1 parvalbumin-containing inhibitory interneurons (PV+INTs). We find that WT PV+INTs consist of two physiological subtypes (80% fast-spiking (FS), 20% non-fast-spiking (NFS)) and four morphological subtypes. We find that cell-autonomous mutations within interneurons disrupts morphophysiological development of PV+INTs and results in the emergence of a non-canonical 'intermediate spiking (IS)' subset of PV+INTs. We also find that now dominant IS/NFS cells are prone to entering depolarization block, causing them to temporarily lose the ability to initiate action potentials and control network excitation, potentially promoting seizures. Finally, single-cell nuclear RNAsequencing of PV+INTs revealed several misregulated genes related to morphogenesis, cellular excitability, and synapse formation.
Collapse
Affiliation(s)
- Tyler G Ekins
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
- NIH-Brown University Graduate Partnership ProgramProvidenceUnited States
| | - Vivek Mahadevan
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Yajun Zhang
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - James A D'Amour
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
- Postdoctoral Research Associate Training Program, National Institute of General Medical SciencesBethesdaUnited States
| | - Gülcan Akgül
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Timothy J Petros
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Chris J McBain
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
32
|
GABAergic Interneuron and Neurotransmission Are mTOR-Dependently Disturbed in Experimental Focal Cortical Dysplasia. Mol Neurobiol 2020; 58:156-169. [PMID: 32909150 DOI: 10.1007/s12035-020-02086-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 08/21/2020] [Indexed: 01/13/2023]
Abstract
Focal cortical dysplasia (FCD) is a major cause for drug-resistant epilepsies. The molecular and cellular mechanisms of epileptogenesis in FCD are still poorly understood. Some studies have suggested that deficiencies of γ-aminobutyric acid (GABA) system may play an important role in type II FCD, but it remains controversial. In order to examine whether and how GABAergic interneurons and synaptic function are affected, we generated a somatic mTOR hyperactivation-based mouse model of type II FCD by in utero electroporation, quantified densities of interneurons in the malformed cortices, and recorded miniature inhibitory postsynaptic currents in dysmorphic neurons. We detected 20-25% reduction of GABAergic interneurons within malformed cortices, independent of cortical regions and cell subtypes but proportionate to the decrease of global neuron counts. GABAergic synaptic transmission from interneurons to mTOR hyperactivated dysmorphic neurons was dramatically disrupted, outweighing the decrease of interneuron counts. Postnatal mTOR inhibition partially rescued these alterations of GABAergic system. We also quantified the expression of GABAA receptor, GABA transporter, and chloridion transporter encoding genes and found that their expression was relatively intact within the malformed cortices. Taken together, these results confirmed that GABAergic interneuron and synapse transmission are disturbed profoundly in an mTOR-dependent manner in type II FCD. Our study suggests that postsynaptic mechanisms independent of interneuron reduction or altered expression of GABA synapse genes might be accountable for the impaired GABAergic neurotransmission in type II FCD as well as other mTOR-related epilepsies.
Collapse
|
33
|
Nguyen VT, Bhalla R, Cowin G, Stimson DHR, Song X, Chong S, Jackson A, Trigg WJ, Tieng QM, Mardon K, Galloway GJ, Kurniawan ND. GABA a receptor density alterations revealed in a mouse model of early moderate prenatal ethanol exposure using [ 18F]AH114726. Nucl Med Biol 2020; 88-89:44-51. [PMID: 32777548 DOI: 10.1016/j.nucmedbio.2020.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Prenatal ethanol exposure (PEE) has been shown to alter the level and function of receptors in the brain, one of which is GABAa receptors (GABAaR), the major inhibitory ligand gated ion channels that mediate neuronal inhibition. High dose PEE in animals resulted in the upregulation of GABAaR, but the effects of low and moderate dose PEE at early gestation have not been investigated. This study aimed at examining GABAaR density in the adult mouse brain following PEE during a period equivalent to the first 3 to 4 weeks in human gestation. It was hypothesized that early moderate PEE would cause alterations in brain GABAaR levels in the adult offspring. METHODS C57BL/6J mice were given 10% v/v ethanol during the first 8 gestational days. Male offspring were studied using in-vivo Positron Emission Tomography (PET)/Magnetic Resonance Imaging (MRI), biodistribution, in-vitro autoradiography using [18F]AH114726, a novel flumazenil analogue with a high affinity for the benzodiazepine-binding site, and validated using immunohistochemistry. RESULTS In vivo PET and biodistribution did not detect alteration in brain tracer uptake. In vitro radiotracer studies detected significantly reduced GABAaR in the olfactory bulbs. Immunohistochemistry detected reduced GABAaR in the cerebral cortex, cerebellum and hippocampus, while Nissl staining showed that cell density was significantly higher in the striatum following PEE. CONCLUSION Early moderate PEE may induce long-term alterations in the GABAaR system that persisted into adulthood.
Collapse
Affiliation(s)
- Van T Nguyen
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia; Hanoi University of Science and Technology, Hanoi, Viet Nam
| | - Rajiv Bhalla
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Gary Cowin
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Damion H R Stimson
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Xin Song
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Suyinn Chong
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia
| | - Alexander Jackson
- Core Imaging R&D, Life Sciences, GE Healthcare, Chalfont St Giles, Buckinghamshire, UK
| | - William J Trigg
- Core Imaging R&D, Life Sciences, GE Healthcare, Chalfont St Giles, Buckinghamshire, UK
| | - Quang M Tieng
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Karine Mardon
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia; National Imaging Facility, University of Queensland, Brisbane, Queensland, Australia
| | - Graham J Galloway
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia; National Imaging Facility, University of Queensland, Brisbane, Queensland, Australia
| | - Nyoman D Kurniawan
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
34
|
Horigane S, Ozawa Y, Zhang J, Todoroki H, Miao P, Haijima A, Yanagawa Y, Ueda S, Nakamura S, Kakeyama M, Takemoto‐Kimura S. A mouse model of Timothy syndrome exhibits altered social competitive dominance and inhibitory neuron development. FEBS Open Bio 2020; 10:1436-1446. [PMID: 32598571 PMCID: PMC7396430 DOI: 10.1002/2211-5463.12924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/13/2020] [Accepted: 06/25/2020] [Indexed: 01/17/2023] Open
Abstract
Multiple genetic factors related to autism spectrum disorder (ASD) have been identified, but the biological mechanisms remain obscure. Timothy syndrome (TS), associated with syndromic ASD, is caused by a gain-of-function mutation, G406R, in the pore-forming subunit of L-type Ca2+ channels, Cav 1.2. In this study, a mouse model of TS, TS2-neo, was used to enhance behavioral phenotyping and to identify developmental anomalies in inhibitory neurons. Using the IntelliCage, which enables sequential behavioral tasks without human handling and mouse isolation stress, high social competitive dominance was observed in TS2-neo mice. Furthermore, histological analysis demonstrated inhibitory neuronal abnormalities in the neocortex, including an excess of smaller-sized inhibitory presynaptic terminals in the somatosensory cortex of young adolescent mice and higher numbers of migrating inhibitory neurons from the medial ganglionic eminence during embryonic development. In contrast, no obvious changes in excitatory synaptic terminals were found. These novel neural abnormalities in inhibitory neurons of TS2-neo mice may result in a disturbed excitatory/inhibitory (E/I) balance, a key feature underlying ASD.
Collapse
Affiliation(s)
- Shin‐ichiro Horigane
- Department of Neuroscience IResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Molecular/Cellular NeuroscienceNagoya University Graduate School of MedicineNagoyaJapan
| | - Yukihiro Ozawa
- Department of Neuroscience IResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Molecular/Cellular NeuroscienceNagoya University Graduate School of MedicineNagoyaJapan
- Department of Pathology and Laboratory MedicineNagoya University HospitalNagoyaJapan
| | - Jun Zhang
- Department of Neuroscience IResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Molecular/Cellular NeuroscienceNagoya University Graduate School of MedicineNagoyaJapan
| | - Hiroe Todoroki
- Laboratory for Systems Neurosciences and Preventive MedicineFaculty of Human SciencesWaseda UniversityTokorozawaJapan
| | - Pan Miao
- Department of Neuroscience IResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Molecular/Cellular NeuroscienceNagoya University Graduate School of MedicineNagoyaJapan
| | - Asahi Haijima
- Laboratory for Systems Neurosciences and Preventive MedicineFaculty of Human SciencesWaseda UniversityTokorozawaJapan
- Research Institute for Environmental Medical SciencesWaseda UniversityTokorozawaJapan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral NeuroscienceGunma University Graduate School of MedicineMaebashiJapan
| | - Shuhei Ueda
- Department of Neuroscience IResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Molecular/Cellular NeuroscienceNagoya University Graduate School of MedicineNagoyaJapan
| | - Shigeo Nakamura
- Department of Pathology and Laboratory MedicineNagoya University HospitalNagoyaJapan
| | - Masaki Kakeyama
- Laboratory for Systems Neurosciences and Preventive MedicineFaculty of Human SciencesWaseda UniversityTokorozawaJapan
- Research Institute for Environmental Medical SciencesWaseda UniversityTokorozawaJapan
| | - Sayaka Takemoto‐Kimura
- Department of Neuroscience IResearch Institute of Environmental MedicineNagoya UniversityNagoyaJapan
- Molecular/Cellular NeuroscienceNagoya University Graduate School of MedicineNagoyaJapan
- Precursory Research for Embryonic Science and Technology (PRESTO)Japan Science and Technology AgencySaitamaJapan
| |
Collapse
|
35
|
Gallo NB, Paul A, Van Aelst L. Shedding Light on Chandelier Cell Development, Connectivity, and Contribution to Neural Disorders. Trends Neurosci 2020; 43:565-580. [PMID: 32564887 PMCID: PMC7392791 DOI: 10.1016/j.tins.2020.05.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/06/2020] [Accepted: 05/07/2020] [Indexed: 02/04/2023]
Abstract
Chandelier cells (ChCs) are a unique type of GABAergic interneuron that selectively innervate the axon initial segment (AIS) of excitatory pyramidal neurons; the subcellular domain where action potentials are initiated. The proper genesis and maturation of ChCs is critical for regulating neural ensemble firing in the neocortex throughout development and adulthood. Recently, genetic and molecular studies have shed new light on the complex innerworkings of ChCs in health and disease. This review presents an overview of recent studies on the developmental origins, migratory properties, and morphology of ChCs. In addition, attention is given to newly identified molecules regulating ChC morphogenesis and connectivity as well as recent work linking ChC dysfunction to neural disorders, including schizophrenia, epilepsy, and autism spectrum disorder (ASD).
Collapse
Affiliation(s)
- Nicholas B Gallo
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 11724, USA; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Anirban Paul
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Linda Van Aelst
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 11724, USA.
| |
Collapse
|
36
|
Fazzari P, Mortimer N, Yabut O, Vogt D, Pla R. Cortical distribution of GABAergic interneurons is determined by migration time and brain size. Development 2020; 147:dev.185033. [PMID: 32586977 DOI: 10.1242/dev.185033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 06/15/2020] [Indexed: 11/20/2022]
Abstract
Cortical interneurons (CINs) originate in the ganglionic eminences (GEs) and migrate tangentially to the cortex guided by different attractive and repulsive cues. Once inside the cortex, the cellular and molecular mechanisms determining the migration of CINs along the rostrocaudal axis are less well understood. Here, we investigated the cortical distribution of CINs originating in the medial and caudal GEs at different time points. Using molecular and genetic labeling, we showed that, in the mouse, early- and late-born CINs (E12 versus E15) are differentially distributed along the rostrocaudal axis. Specifically, late-born CINs are preferentially enriched in cortical areas closer to their respective sites of origin in the medial or caudal GE. Surprisingly, our in vitro experiments failed to show a preferential migration pattern along the rostrocaudal axis for medial- or caudal-born CINs. Moreover, in utero transplantation experiments suggested that the rostrocaudal dispersion of CINs depends on the developmental stage of the host brain and is limited by the migration time and the increasing size of the developing brain. These data suggest that the embryonic expansion of the cortex contributes to the rostrocaudal distribution of CINs.
Collapse
Affiliation(s)
- Pietro Fazzari
- Laboratory of Cortical Circuits in Health and Disease, CIPF Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Niall Mortimer
- Department of Psychiatry, Neuroscience Program and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94158, USA.,Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97070 Würzburg, Germany.,Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain.,Department of Psychiatry, Hospital Universitari Vall d'Hebron, 08035 Barcelona, Spain
| | - Odessa Yabut
- Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Daniel Vogt
- Department of Psychiatry, Neuroscience Program and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94158, USA.,Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
| | - Ramon Pla
- Department of Psychiatry, Neuroscience Program and the Nina Ireland Laboratory of Developmental Neurobiology, University of California San Francisco, San Francisco, CA 94158, USA .,Instituto de investigación en discapacidades neurológicas (IDINE), University of Castile-la-Mancha, 02006 Albacete, Spain
| |
Collapse
|
37
|
Manzano Nieves G, Bravo M, Baskoylu S, Bath KG. Early life adversity decreases pre-adolescent fear expression by accelerating amygdala PV cell development. eLife 2020; 9:55263. [PMID: 32692310 PMCID: PMC7413666 DOI: 10.7554/elife.55263] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
Early life adversity (ELA) is associated with increased risk for stress-related disorders later in life. The link between ELA and risk for psychopathology is well established but the developmental mechanisms remain unclear. Using a mouse model of resource insecurity, limited bedding (LB), we tested the effects of LB on the development of fear learning and neuronal structures involved in emotional regulation, the medial prefrontal cortex (mPFC) and basolateral amygdala (BLA). LB delayed the ability of peri-weanling (21 days old) mice to express, but not form, an auditory conditioned fear memory. LB accelerated the developmental emergence of parvalbumin (PV)-positive cells in the BLA and increased anatomical connections between PL and BLA. Fear expression in LB mice was rescued through optogenetic inactivation of PV-positive cells in the BLA. The current results provide a model of transiently blunted emotional reactivity in early development, with latent fear-associated memories emerging later in adolescence.
Collapse
Affiliation(s)
| | - Marilyn Bravo
- Department of Neuroscience, Brown University, Providence, United States
| | - Saba Baskoylu
- Department of Neuroscience, Brown University, Providence, United States
| | - Kevin G Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, United States
| |
Collapse
|
38
|
Bartholome O, de la Brassinne Bonardeaux O, Neirinckx V, Rogister B. A Composite Sketch of Fast-Spiking Parvalbumin-Positive Neurons. Cereb Cortex Commun 2020; 1:tgaa026. [PMID: 34296100 PMCID: PMC8153048 DOI: 10.1093/texcom/tgaa026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 01/28/2023] Open
Abstract
Parvalbumin-positive neurons are inhibitory neurons that release GABA and are mostly represented by fast-spiking basket or chandelier cells. They constitute a minor neuronal population, yet their peculiar profiles allow them to react quickly to any event in the brain under normal or pathological conditions. In this review, we will summarize the current knowledge about the fundamentals of fast-spiking parvalbumin-positive neurons, focusing on their morphology and specific channel/protein content. Next, we will explore their development, maturation, and migration in the brain. Finally, we will unravel their potential contribution to the physiopathology of epilepsy.
Collapse
Affiliation(s)
| | | | | | - Bernard Rogister
- GIGA-Neurosciences, University of Liege, 4000 Liège, Belgium
- Neurology Department, CHU, Academic Hospital, University of Liege, 4000 Liège, Belgium
| |
Collapse
|
39
|
Refinement of Cerebellar Network Organization by Extracellular Signaling During Development. Neuroscience 2020; 462:44-55. [PMID: 32502568 DOI: 10.1016/j.neuroscience.2020.05.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022]
Abstract
The cerebellum forms regular neural network structures consisting of a few major types of neurons, such as Purkinje cells, granule cells, and molecular layer interneurons, and receives two major inputs from climbing fibers and mossy fibers. Its regular structures consist of three well-defined layers, with each type of neuron designated to a specific location and forming specific synaptic connections. During the first few weeks of postnatal development in rodents, the cerebellum goes through dynamic changes via proliferation, migration, differentiation, synaptogenesis, and maturation, to create such a network structure. The development of this organized network structure presumably relies on the communication between developing elements in the network, including not only individual neurons, but also their dendrites, axons, and synapses. Therefore, it is reasonable that extracellular signaling via synaptic transmission, secreted molecules, and cell adhesion molecules, plays important roles in cerebellar network development. Although it is not yet clear as to how overall cerebellar development is orchestrated, there is indeed accumulating lines of evidence that extracellular signaling acts toward the development of individual elements in the cerebellar networks. In this article, we introduce what we have learned from many studies regarding the extracellular signaling required for cerebellar network development, including our recent study suggesting the importance of unbiased synaptic inputs from parallel fibers.
Collapse
|
40
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
41
|
Shen W, Ba R, Su Y, Ni Y, Chen D, Xie W, Pleasure SJ, Zhao C. Foxg1 Regulates the Postnatal Development of Cortical Interneurons. Cereb Cortex 2020; 29:1547-1560. [PMID: 29912324 DOI: 10.1093/cercor/bhy051] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/23/2018] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
Abnormalities in cortical interneurons are closely associated with neurological diseases. Most patients with Foxg1 syndrome experience seizures, suggesting a possible role of Foxg1 in the cortical interneuron development. Here, by conditional deletion of Foxg1, which was achieved by crossing Foxg1fl/fl with the Gad2-CreER line, we found the postnatal distributions of somatostatin-, calretinin-, and neuropeptide Y-positive interneurons in the cortex were impaired. Further investigations revealed an enhanced dendritic complexity and decreased migration capacity of Foxg1-deficient interneurons, accompanied by remarkable downregulation of Dlx1 and CXCR4. Overexpression of Dlx1 or knock down its downstream Pak3 rescued the differentiation detects, demonstrated that Foxg1 functioned upstream of Dlx1-Pak3 signal pathway to regulate the postnatal development of cortical interneurons. Due to the imbalanced neural circuit, Foxg1 mutants showed increased seizure susceptibility. These findings will improve our understanding of the postnatal development of interneurons and help to elucidate the mechanisms underlying seizure in patients carrying Foxg1 mutations.
Collapse
Affiliation(s)
- Wei Shen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Ru Ba
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Yan Su
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Yang Ni
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Dongsheng Chen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China
| | - Wei Xie
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Institute of Life Science, Southeast University, Nanjing, P. R. China
| | - Samuel J Pleasure
- Department of Neurology, Weill Institute for Neuroscience, Programs in Neuroscience and Developmental Stem Cell Biology, UCSF, San Francisco, CA, USA
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, School of Medicine, Southeast University, Nanjing, P. R. China.,Center of Depression, Beijing Institute for Brain Disorders, Beijing 100069, People's Republic of China
| |
Collapse
|
42
|
Chen A, Guo Z, Fang L, Bian S. Application of Fused Organoid Models to Study Human Brain Development and Neural Disorders. Front Cell Neurosci 2020; 14:133. [PMID: 32670022 PMCID: PMC7326106 DOI: 10.3389/fncel.2020.00133] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Human brain organoids cultured from human pluripotent stem cells provide a promising platform to recapitulate histological features of the human brain and model neural disorders. However, unlike animal models, brain organoids lack a reproducible topographic organization, which limits their application in modeling intricate biology, such as the interaction between different brain regions. To overcome these drawbacks, brain organoids have been pre-patterned into specific brain regions and fused to form an assembloid that represents reproducible models recapitulating more complex biological processes of human brain development and neurological diseases. This approach has been applied to model interneuron migration, neuronal projections, tumor invasion, oligodendrogenesis, forebrain axis establishment, and brain vascularization. In this review article, we will summarize the usage of this technology to understand the fundamental biology underpinning human brain development and disorders.
Collapse
Affiliation(s)
- Augustin Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China.,Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Zhenming Guo
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China.,Bio-X Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lipao Fang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China.,Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Shan Bian
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| |
Collapse
|
43
|
Kim JY, Choe J, Moon C. Distinct Developmental Features of Olfactory Bulb Interneurons. Mol Cells 2020; 43:215-221. [PMID: 32208366 PMCID: PMC7103883 DOI: 10.14348/molcells.2020.0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 01/20/2023] Open
Abstract
The olfactory bulb (OB) has an extremely higher proportionof interneurons innervating excitatory neurons than otherbrain regions, which is evolutionally conserved across species.Despite the abundance of OB interneurons, little is knownabout the diversification and physiological functions ofOB interneurons compared to cortical interneurons. In thisreview, an overview of the general developmental processof interneurons from the angles of the spatial and temporalspecifications was presented. Then, the distinct featuresshown exclusively in OB interneurons development andmolecular machinery recently identified were discussed.Finally, we proposed an evolutionary meaning for thediversity of OB interneurons.
Collapse
Affiliation(s)
- Jae Yeon Kim
- Department of Brain and Cognitive Sciences, Graduate School, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Korea
| | - Jiyun Choe
- Department of Brain and Cognitive Sciences, Graduate School, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Korea
| | - Cheil Moon
- Department of Brain and Cognitive Sciences, Graduate School, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Korea
- Convergence Research Advanced Centre for Olfaction, Daegu Gyeongbuk Institute of Science and Technology, Daegu 4988, Korea
- Korea Brain Research Institute, Daegu 41062, Korea
| |
Collapse
|
44
|
Guo T, Liu G, Du H, Wen Y, Wei S, Li Z, Tao G, Shang Z, Song X, Zhang Z, Xu Z, You Y, Chen B, Rubenstein JL, Yang Z. Dlx1/2 are Central and Essential Components in the Transcriptional Code for Generating Olfactory Bulb Interneurons. Cereb Cortex 2019; 29:4831-4849. [PMID: 30796806 PMCID: PMC6917526 DOI: 10.1093/cercor/bhz018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/03/2019] [Accepted: 01/26/2019] [Indexed: 12/22/2022] Open
Abstract
Generation of olfactory bulb (OB) interneurons requires neural stem/progenitor cell specification, proliferation, differentiation, and young interneuron migration and maturation. Here, we show that the homeobox transcription factors Dlx1/2 are central and essential components in the transcriptional code for generating OB interneurons. In Dlx1/2 constitutive null mutants, the differentiation of GSX2+ and ASCL1+ neural stem/progenitor cells in the dorsal lateral ganglionic eminence is blocked, resulting in a failure of OB interneuron generation. In Dlx1/2 conditional mutants (hGFAP-Cre; Dlx1/2F/- mice), GSX2+ and ASCL1+ neural stem/progenitor cells in the postnatal subventricular zone also fail to differentiate into OB interneurons. In contrast, overexpression of Dlx1&2 in embryonic mouse cortex led to ectopic production of OB-like interneurons that expressed Gad1, Sp8, Sp9, Arx, Pbx3, Etv1, Tshz1, and Prokr2. Pax6 mutants generate cortical ectopia with OB-like interneurons, but do not do so in compound Pax6; Dlx1/2 mutants. We propose that DLX1/2 promote OB interneuron development mainly through activating the expression of Sp8/9, which further promote Tshz1 and Prokr2 expression. Based on this study, in combination with earlier ones, we propose a transcriptional network for the process of OB interneuron development.
Collapse
Affiliation(s)
- Teng Guo
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Heng Du
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yan Wen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Song Wei
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Guangxu Tao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zicong Shang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Xiaolei Song
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Zhejun Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yan You
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - John L Rubenstein
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontier Research Center for Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| |
Collapse
|
45
|
Hsing HW, Zhuang ZH, Niou ZX, Chou SJ. Temporal Differences in Interneuron Invasion of Neocortex and Piriform Cortex during Mouse Cortical Development. Cereb Cortex 2019; 30:3015-3029. [PMID: 31838488 DOI: 10.1093/cercor/bhz291] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/28/2022] Open
Abstract
Establishing a balance between excitation and inhibition is critical for brain functions. However, how inhibitory interneurons (INs) generated in the ventral telencephalon integrate with the excitatory neurons generated in the dorsal telencephalon remains elusive. Previous studies showed that INs migrating tangentially to enter the neocortex (NCx), remain in the migratory stream for days before invading the cortical plate during late corticogenesis. Here we show that in developing mouse cortices, INs in the piriform cortex (PCx; the major olfactory cortex) distribute differently from those in the NCx. We provide evidence that during development INs invade and mature earlier in PCx than in NCx, likely owing to the lack of CXCR4 expression in INs from PCx compared to those in NCx. We analyzed IN distribution patterns in Lhx2 cKO mice, where projection neurons in the lateral NCx are re-fated to generate an ectopic PCx (ePCx). The PCx-specific IN distribution patterns found in ePCx suggest that properties of PCx projection neurons regulate IN distribution. Collectively, our results show that the timing of IN invasion in the developing PCx fundamentally differs from what is known in the NCx. Further, our results suggest that projection neurons instruct the PCx-specific pattern of IN distribution.
Collapse
Affiliation(s)
- Hsiang-Wei Hsing
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529 Taiwan
| | - Zi-Hui Zhuang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529 Taiwan
| | - Zhen-Xian Niou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529 Taiwan
| | - Shen-Ju Chou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529 Taiwan
| |
Collapse
|
46
|
Costamagna G, Andreoli L, Corti S, Faravelli I. iPSCs-Based Neural 3D Systems: A Multidimensional Approach for Disease Modeling and Drug Discovery. Cells 2019; 8:E1438. [PMID: 31739555 PMCID: PMC6912470 DOI: 10.3390/cells8111438] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/26/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs)-based two-dimensional (2D) protocols have offered invaluable insights into the pathophysiology of neurological diseases. However, these systems are unable to reproduce complex cytoarchitectural features, cell-cell and tissue-tissue interactions like their in vivo counterpart. Three-dimensional (3D)-based culture protocols, though in their infancy, have offered new insights into modeling human diseases. Human neural organoids try to recapitulate the cellular diversity of complex tissues and can be generated from iPSCs to model the pathophysiology of a wide spectrum of pathologies. The engraftment of iPSCs into mice models and the improvement of differentiation protocols towards 3D cultures has enabled the generation of more complex multicellular systems. Consequently, models of neuropsychiatric disorders, infectious diseases, brain cancer and cerebral hypoxic injury can now be investigated from new perspectives. In this review, we consider the advancements made in modeling neuropsychiatric and neurological diseases with iPSC-derived organoids and their potential use to develop new drugs.
Collapse
Affiliation(s)
| | | | | | - Irene Faravelli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.C.); (L.A.); (S.C.)
| |
Collapse
|
47
|
Li J, Wang C, Zhang Z, Wen Y, An L, Liang Q, Xu Z, Wei S, Li W, Guo T, Liu G, Tao G, You Y, Du H, Fu Z, He M, Chen B, Campbell K, Alvarez-Buylla A, Rubenstein JL, Yang Z. Transcription Factors Sp8 and Sp9 Coordinately Regulate Olfactory Bulb Interneuron Development. Cereb Cortex 2019; 28:3278-3294. [PMID: 28981617 DOI: 10.1093/cercor/bhx199] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/13/2017] [Indexed: 01/01/2023] Open
Abstract
Neural stem cells in the postnatal telencephalic ventricular-subventricular zone (V-SVZ) generate new interneurons, which migrate tangentially through the rostral migratory stream (RMS) into the olfactory bulb (OB). The Sp8 and Sp9 transcription factors are expressed in neuroblasts, as well as in the immature and mature interneurons in the V-SVZ-RMS-OB system. Here we show that Sp8 and Sp9 coordinately regulate OB interneuron development: although Sp9 null mutants show no major OB interneuron defect, conditional deletion of both Sp8 and Sp9 resulted in a much more severe reduction of OB interneuron number than that observed in the Sp8 conditional mutant mice, due to defects in neuronal differentiation, tangential and radial migration, and increased cell death in the V-SVZ-RMS-OB system. RNA-Seq and RNA in situ hybridization reveal that, in Sp8/Sp9 double mutant mice, but not in Sp8 or Sp9 single mutant mice, newly born neuroblasts in the V-SVZ-RMS-OB system fail to express Prokr2 and Tshz1 expression, genes with known roles in promoting OB interneuron differentiation and migration, and that are involved in human Kallmann syndrome.
Collapse
Affiliation(s)
- Jiwen Li
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Chunyang Wang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhuangzhi Zhang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yan Wen
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Lei An
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Qifei Liang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhejun Xu
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Song Wei
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Weiwei Li
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Teng Guo
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guoping Liu
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guangxu Tao
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yan You
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Heng Du
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhuoning Fu
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Miao He
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Bin Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
| | - Kenneth Campbell
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - John L Rubenstein
- Department of Psychiatry, Nina Ireland Laboratory of Developmental Neurobiology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Zhengang Yang
- Department of Translational Neuroscience, Shanghai Pudong Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
48
|
Park H, Kim T, Kim J, Yamamoto Y, Tanaka-Yamamoto K. Inputs from Sequentially Developed Parallel Fibers Are Required for Cerebellar Organization. Cell Rep 2019; 28:2939-2954.e5. [DOI: 10.1016/j.celrep.2019.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/10/2019] [Accepted: 07/30/2019] [Indexed: 11/15/2022] Open
|
49
|
Edens BM, Vissers C, Su J, Arumugam S, Xu Z, Shi H, Miller N, Rojas Ringeling F, Ming GL, He C, Song H, Ma YC. FMRP Modulates Neural Differentiation through m 6A-Dependent mRNA Nuclear Export. Cell Rep 2019; 28:845-854.e5. [PMID: 31340148 PMCID: PMC6687293 DOI: 10.1016/j.celrep.2019.06.072] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/09/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
N6-methyladenosine (m6A) modification of mRNA is emerging as a vital mechanism regulating RNA function. Here, we show that fragile X mental retardation protein (FMRP) reads m6A to promote nuclear export of methylated mRNA targets during neural differentiation. Fmr1 knockout (KO) mice show delayed neural progenitor cell cycle progression and extended maintenance of proliferating neural progenitors into postnatal stages, phenocopying methyltransferase Mettl14 conditional KO (cKO) mice that have no m6A modification. RNA-seq and m6A-seq reveal that both Mettl14cKO and Fmr1KO lead to the nuclear retention of m6A-modified FMRP target mRNAs regulating neural differentiation, indicating that both m6A and FMRP are required for the nuclear export of methylated target mRNAs. FMRP preferentially binds m6A-modified RNAs to facilitate their nuclear export through CRM1. The nuclear retention defect can be mitigated by wild-type but not nuclear export-deficient FMRP, establishing a critical role for FMRP in mediating m6A-dependent mRNA nuclear export during neural differentiation.
Collapse
Affiliation(s)
- Brittany M Edens
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Caroline Vissers
- Biochemistry, Molecular and Cellular Biology Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jing Su
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Saravanan Arumugam
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Zhaofa Xu
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Han Shi
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Nimrod Miller
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | | | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neuroscience, Department of Cell and Developmental Biology, Institute for Regeneration, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chuan He
- Departments of Chemistry and Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neuroscience, Department of Cell and Developmental Biology, Institute for Regeneration, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Yongchao C Ma
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
| |
Collapse
|
50
|
Johnstone M, Hillary RF, St Clair D. Stem Cells to Inform the Neurobiology of Mental Illness. Curr Top Behav Neurosci 2019; 40:13-43. [PMID: 30030769 DOI: 10.1007/7854_2018_57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The inception of human-induced pluripotent stem cell (hiPSCs) technology has provided an exciting platform upon which the modelling and treatment of human neurodevelopmental and neuropsychiatric disorders may be expedited. Although the genetic architecture of these disorders is far more complex than previously imagined, many key loci have at last been identified. This has allowed in vivo and in vitro technologies to be refined to model specific high-penetrant genetic loci involved in both disorders. Animal models of neurodevelopmental disorders, such as schizophrenia and autism spectrum disorders, show limitations in recapitulating the full complexity and heterogeneity of human neurodevelopmental disease states. Indeed, patient-derived hiPSCs offer distinct advantages over classical animal models in the study of human neuropathologies. Here we have discussed the current, relative translational merit of hiPSCs in investigating human neurodevelopmental and neuropsychiatric disorders with a specific emphasis on the utility of such systems to aid in the identification of biomarkers. We have highlighted the promises and pitfalls of reprogramming cell fate for the study of these disorders and provide recommendations for future directions in this field in order to overcome current limitations. Ultimately, this will aid in the development of effective clinical strategies for diverse patient populations affected by these disorders with the aim of also leading to biomarker identification.
Collapse
Affiliation(s)
- Mandy Johnstone
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK.
| | - Robert F Hillary
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David St Clair
- Division of Psychiatry, Royal Edinburgh Hospital, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|