1
|
Parr AC, Perica MI, Calabro FJ, Foran W, Moon CH, Hetherington H, Luna B. Adolescent maturation of dorsolateral prefrontal cortex glutamate:GABA and cognitive function is supported by dopamine-related neurobiology. Mol Psychiatry 2025; 30:2558-2572. [PMID: 39653761 DOI: 10.1038/s41380-024-02860-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 05/22/2025]
Abstract
Developmental changes in prefrontal cortex (PFC) excitatory (glutamatergic, Glu) and inhibitory (gamma- aminobutryic acid, GABA) neurotransmitter balance (E:I) have been identified during human adolescence, potentially reflecting a critical period of plasticity that supports the maturation of PFC-dependent cognition. Animal models implicate increases in dopamine (DA) in regulating changes in PFC E:I during critical periods of development, however, mechanistic relationships between DA and E:I have not been studied in humans. Here, we used high field (7T) echo planar imaging (EPI) in combination with Magnetic Resonance Spectroscopic Imaging (MRSI) to assess the role of basal ganglia tissue iron-reflecting DA neurophysiology-in longitudinal trajectories of dorsolateral PFC Glu, GABA, and their relative levels (Glu:GABA) and working memory performance from adolescence to adulthood in 153 participants (ages 10-32 years old, 1-3 visits, 272 visits total). Using generalized additive mixed models (GAMMs) that capture linear and non-linear developmental processes, we show that basal ganglia tissue iron increases during adolescence, and Glu:GABA is biased towards heightened Glu relative to GABA early in adolescence, decreasing into adulthood. Critically, variation in basal ganglia tissue iron was linked to different age-related trajectories in Glu:GABA and working memory. Specifically, individuals with higher levels of tissue iron showed a greater degree of age-related declines in Glu and Glu:GABA, resulting in lower Glu relative to GABA (i.e., higher GABA relative to Glu) in young adulthood. Variation in tissue iron additionally moderated working memory trajectories, as higher levels of tissue iron were associated with steeper age-related improvements and better performance into adulthood. Our results provide novel evidence for a model of critical period plasticity whereby individual differences in DA may be involved in fine-tuning PFC E:I and PFC-dependent cognitive function at a critical transition from adolescence into adulthood.
Collapse
Affiliation(s)
- Ashley C Parr
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Maria I Perica
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Finnegan J Calabro
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Will Foran
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chan Hong Moon
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Orozco Valero A, Rodríguez-González V, Montobbio N, Casal MA, Tlaie A, Pelayo F, Morillas C, Poza J, Gómez C, Martínez-Cañada P. A Python toolbox for neural circuit parameter inference. NPJ Syst Biol Appl 2025; 11:45. [PMID: 40346107 PMCID: PMC12064716 DOI: 10.1038/s41540-025-00527-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
Computational research tools have reached a level of maturity that enables efficient simulation of neural activity across diverse scales. Concurrently, experimental neuroscience is experiencing an unprecedented scale of data generation. Despite these advancements, our understanding of the precise mechanistic relationship between neural recordings and key aspects of neural activity remains insufficient, including which specific features of electrophysiological population dynamics (i.e., putative biomarkers) best reflect properties of the underlying microcircuit configuration. We present ncpi, an open-source Python toolbox that serves as an all-in-one solution, effectively integrating well-established methods for both forward and inverse modeling of extracellular signals based on single-neuron network model simulations. Our tool serves as a benchmarking resource for model-driven interpretation of electrophysiological data and the evaluation of candidate biomarkers that plausibly index changes in neural circuit parameters. Using mouse LFP data and human EEG recordings, we demonstrate the potential of ncpi to uncover imbalances in neural circuit parameters during brain development and in Alzheimer's Disease.
Collapse
Affiliation(s)
- Alejandro Orozco Valero
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain
| | - Víctor Rodríguez-González
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valladolid, Spain
| | - Noemi Montobbio
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Miguel A Casal
- Research Center for Information and Communication Technologies (CITIC), University of A Coruña, A Coruña, Spain
| | - Alejandro Tlaie
- Ernst Strüngmann Institute for Neuroscience, Frankfurt am Main, Germany
| | - Francisco Pelayo
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain
| | - Christian Morillas
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain
| | - Jesús Poza
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valladolid, Spain
- IMUVA, Instituto de Investigación en Matemáticas, University of Valladolid, Valladolid, Spain
| | - Carlos Gómez
- Biomedical Engineering Group, University of Valladolid, Valladolid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valladolid, Spain
| | - Pablo Martínez-Cañada
- Research Center for Information and Communication Technologies (CITIC), University of Granada, Granada, Spain.
- Department of Computer Engineering, Automation and Robotics, University of Granada, Granada, Spain.
| |
Collapse
|
3
|
Kang J, Mao W, Wu J, Li X. Measurement of Excitation-Inhibition Imbalance in Autism spectrum Disorder Using EEG Proxy Markers: A Pilot Study. Clin EEG Neurosci 2025:15500594251333159. [PMID: 40223310 DOI: 10.1177/15500594251333159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Autism Spectrum Disorder (ASD) is a severe neurodevelopmental disorder characterized primarily by social impairments and repetitive behaviors. Imbalance in excitatory-inhibitory (E/I) activity within the central nervous system may be a key mechanism underlying ASD. Electroencephalography (EEG) is a useful tool for recording brain electrical signals, reflecting the activity of cortical neuron populations, and estimating both global and regional E/I balance. Various EEG methods can estimate E/I balance, including non-periodic exponent, corrected alpha power, sample entropy, average spatial phase synchronization (ASPS), and detrended fluctuation analysis (DFA) based on E/I indices. However, research on using EEG proxy markers to assess E/I imbalance in autism is limited, and there is no study indicating which method is most sensitive. Therefore, this study employed a high-density EEG acquisition system to collect data from a relatively large sample of autistic and typically developing (TD) children. We computed EEG proxy markers and used the Coefficient of Variation (CV) to compare the sensitivity of five EEG markers between the two groups. The results indicated that non-periodic exponent based on power spectra and corrected alpha power from non-periodic neural activity were more advantageous. The findings may provide theoretical support for the exploration of EEG biomarkers based on E/I balance theory.
Collapse
Affiliation(s)
- Jiannan Kang
- Child Rehabilitation Division, Ningbo Rehabilitation Hospital, Ningbo, China
| | - Wenqin Mao
- Child Rehabilitation Division, Ningbo Rehabilitation Hospital, Ningbo, China
| | - Juanmei Wu
- Child Rehabilitation Division, Ningbo Rehabilitation Hospital, Ningbo, China
| | - Xiaoli Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| |
Collapse
|
4
|
Murakami T. Spatial dynamics of spontaneous activity in the developing and adult cortices. Neurosci Res 2025; 212:1-10. [PMID: 39653148 DOI: 10.1016/j.neures.2024.12.002] [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: 03/29/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024]
Abstract
Even in the absence of external stimuli, the brain remains remarkably active, with neurons continuously firing and communicating with each other. It is not merely random firing of individual neurons but rather orchestrated patterns of activity that propagate throughout the intricate network. Over two decades, advancements in neuroscience observation tools for hemodynamics, membrane potential, and neural calcium signals, have allowed researchers to analyze the dynamics of spontaneous activity across different spatial scales, from individual neurons to macroscale brain networks. One of the remarkable findings from these studies is that the spatial patterns of spontaneous activity in the developing brain are vastly different from those in the mature adult brain. Spatial patterns of spontaneous activity during development are essential for connection refinement between brain regions, whereas the functional role in the adult brain is still controversial. In this paper, I review the differences in spatial dynamics of spontaneous activity between developing and adult cortices. Then, I delve into the cellular mechanisms underlying spontaneous activity, especially its generation and propagation manner, to contribute to a deeper understanding of brain function and its development.
Collapse
Affiliation(s)
- Tomonari Murakami
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
5
|
Lakhera S, Herbert E, Gjorgjieva J. Modeling the Emergence of Circuit Organization and Function during Development. Cold Spring Harb Perspect Biol 2025; 17:a041511. [PMID: 38858072 PMCID: PMC11864115 DOI: 10.1101/cshperspect.a041511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Developing neural circuits show unique patterns of spontaneous activity and structured network connectivity shaped by diverse activity-dependent plasticity mechanisms. Based on extensive experimental work characterizing patterns of spontaneous activity in different brain regions over development, theoretical and computational models have played an important role in delineating the generation and function of individual features of spontaneous activity and their role in the plasticity-driven formation of circuit connectivity. Here, we review recent modeling efforts that explore how the developing cortex and hippocampus generate spontaneous activity, focusing on specific connectivity profiles and the gradual strengthening of inhibition as the key drivers behind the observed developmental changes in spontaneous activity. We then discuss computational models that mechanistically explore how different plasticity mechanisms use this spontaneous activity to instruct the formation and refinement of circuit connectivity, from the formation of single neuron receptive fields to sensory feature maps and recurrent architectures. We end by highlighting several open challenges regarding the functional implications of the discussed circuit changes, wherein models could provide the missing step linking immature developmental and mature adult information processing capabilities.
Collapse
Affiliation(s)
- Shreya Lakhera
- School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Elizabeth Herbert
- School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Julijana Gjorgjieva
- School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
6
|
Simacek CA, Kirischuk S, Mittmann T. Postnatal development of vasoactive intestinal polypeptide-expressing GABAergic interneurons in mouse somatosensory cortex. Acta Physiol (Oxf) 2025; 241:e14265. [PMID: 39803724 PMCID: PMC11726421 DOI: 10.1111/apha.14265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/30/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
AIM Despite dysfunctional vasoactive intestinal polypeptide-positive interneurons (VIP-INs) being linked to the emergence of neurodevelopmental disorders, the temporal profile of VIP-IN functional maturation and cortical network integration remains unclear. METHODS Postnatal VIP-IN development was traced with patch clamp experiments in the somatosensory cortex of Vip-IRES-cre x tdTomato mice. Age groups were chosen during barrel field formation, before and after activation of main sensory inputs, and in adult animals (postnatal days (P) P3-4, P8-10, P14-16, and P30-36). RESULTS Changes in passive and active membrane properties show a maturation towards accelerated signal integrations. Excitatory and inhibitory postsynaptic currents (EPSCs and IPSCs) showed progressive VIP-IN integration into cortical networks, likely via synaptogenesis: mEPSC frequency increased before P8-10, while mIPSC frequency increased at P14-16. Only mIPSC kinetics became accelerated, and the E/I ratio of synaptic inputs, defined as a ratio of mEPSC to mIPSC charge transfer, remained constant throughout the investigated developmental stages. Evoked (e)EPSCs and (e)IPSCs showed increased amplitudes, while only eIPSCs demonstrated faster kinetics. eEPSCs and eIPSCs revealed a paired-pulse facilitation by P14-16, indicating probably a decrease in the presynaptic release probability (pr) and a paired-pulse depression in adulthood. eIPSCs also showed the latter, suggesting a decrease in pr for both signal transmission pathways at this time point. CONCLUSIONS VIP-INs mature towards faster signal integration and pursue different strategies to avoid overexcitation. Excitatory and inhibitory synaptic transmission become stronger and shorter via different pre- and postsynaptic alterations, likely promoting the execution of active whisking.
Collapse
Affiliation(s)
- Clara A. Simacek
- Institute for PhysiologyUniversity Medical Centre of the Johannes Gutenberg University MainzMainzGermany
| | - Sergei Kirischuk
- Institute for PhysiologyUniversity Medical Centre of the Johannes Gutenberg University MainzMainzGermany
| | - Thomas Mittmann
- Institute for PhysiologyUniversity Medical Centre of the Johannes Gutenberg University MainzMainzGermany
| |
Collapse
|
7
|
Mediane DH, Basu S, Cahill EN, Anastasiades PG. Medial prefrontal cortex circuitry and social behaviour in autism. Neuropharmacology 2024; 260:110101. [PMID: 39128583 DOI: 10.1016/j.neuropharm.2024.110101] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Autism spectrum disorder (ASD) has proven to be highly enigmatic due to the diversity of its underlying genetic causes and the huge variability in symptom presentation. Uncovering common phenotypes across people with ASD and pre-clinical models allows us to better understand the influence on brain function of the many different genetic and cellular processes thought to contribute to ASD aetiology. One such feature of ASD is the convergent evidence implicating abnormal functioning of the medial prefrontal cortex (mPFC) across studies. The mPFC is a key part of the 'social brain' and may contribute to many of the changes in social behaviour observed in people with ASD. Here we review recent evidence for mPFC involvement in both ASD and social behaviours. We also highlight how pre-clinical mouse models can be used to uncover important cellular and circuit-level mechanisms that may underly atypical social behaviours in ASD. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- Diego H Mediane
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Shinjini Basu
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Emma N Cahill
- Department of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - Paul G Anastasiades
- Department of Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom.
| |
Collapse
|
8
|
Asim M, Qianqian G, Waris A, Wang H, Lai Y, Chen X. Unraveling the role of cholecystokinin in epilepsy: Mechanistic insight into neuroplasticity. Neurochem Int 2024; 180:105870. [PMID: 39343303 DOI: 10.1016/j.neuint.2024.105870] [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: 08/15/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Epilepsy is a disorder characterized by an imbalance between excitability and inhibition, leading to uncontrolled hyperexcitability of neurons in the central nervous system. Despite the prevalence of epileptic seizures, the underlying mechanisms driving this hyperexcitability remain poorly understood. This review article aims to enhance our understanding of the mechanisms of epilepsy, with a specific focus on the role of cholecystokinin (CCK) in this debilitating disease. We will begin with an introduction to the topic, followed by an examination of the role of GABAergic neurons and the synaptic plasticity mechanisms associated with seizures. As we delve deeper, we will elucidate how CCK and its receptors contribute to seizure behavior. Finally, we will discuss the CCK-dependent synaptic plasticity mechanisms and highlight their potential implications in seizure activity. Through a comprehensive examination of these aspects, this review provides valuable insights into the involvement of CCK and its receptors in epilepsy. By improving our understanding of the mechanisms underlying this condition, particularly the role of CCK, we aim to contribute to the development of more effective treatment strategies.
Collapse
Affiliation(s)
- Muhammad Asim
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong.
| | - Gao Qianqian
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Abdul Waris
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Huajie Wang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Yuanying Lai
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong; Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong
| |
Collapse
|
9
|
Bernardo D, Xie X, Verma P, Kim J, Liu V, Numis AL, Wu Y, Glass HC, Yap PT, Nagarajan SS, Raj A. Simulation-based Inference of Developmental EEG Maturation with the Spectral Graph Model. ARXIV 2024:arXiv:2405.02524v3. [PMID: 39040639 PMCID: PMC11261974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
The spectral content of macroscopic neural activity evolves throughout development, yet how this maturation relates to underlying brain network formation and dynamics remains unknown. Here, we assess the developmental maturation of electroencephalogram spectra via Bayesian model inversion of the spectral graph model, a parsimonious whole-brain model of spatiospectral neural activity derived from linearized neural field models coupled by the structural connectome. Simulation-based inference was used to estimate age-varying spectral graph model parameter posterior distributions from electroencephalogram spectra spanning the developmental period. This model-fitting approach accurately captures observed developmental electroencephalogram spectral maturation via a neurobiologically consistent progression of key neural parameters: long-range coupling, axonal conduction speed, and excitatory:inhibitory balance. These results suggest that the spectral maturation of macroscopic neural activity observed during typical development is supported by age-dependent functional adaptations in localized neural dynamics and their long-range coupling across the macroscopic structural network.
Collapse
Affiliation(s)
- Danilo Bernardo
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Xihe Xie
- Department of Neuroscience, Weill Cornell Medicine, New York, NY, USA
| | - Parul Verma
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan Kim
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Virginia Liu
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Adam L. Numis
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Ye Wu
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Hannah C. Glass
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Pew-Thian Yap
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Srikantan S. Nagarajan
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
10
|
Chini M, Hnida M, Kostka JK, Chen YN, Hanganu-Opatz IL. Preconfigured architecture of the developing mouse brain. Cell Rep 2024; 43:114267. [PMID: 38795344 DOI: 10.1016/j.celrep.2024.114267] [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: 12/20/2023] [Revised: 03/13/2024] [Accepted: 05/08/2024] [Indexed: 05/27/2024] Open
Abstract
In the adult brain, structural and functional parameters, such as synaptic sizes and neuronal firing rates, follow right-skewed and heavy-tailed distributions. While this organization is thought to have significant implications, its development is still largely unknown. Here, we address this knowledge gap by investigating a large-scale dataset recorded from the prefrontal cortex and the olfactory bulb of mice aged 4-60 postnatal days. We show that firing rates and spike train interactions have a largely stable distribution shape throughout the first 60 postnatal days and that the prefrontal cortex displays a functional small-world architecture. Moreover, early brain activity exhibits an oligarchical organization, where high-firing neurons have hub-like properties. In a neural network model, we show that analogously right-skewed and heavy-tailed synaptic parameters are instrumental to consistently recapitulate the experimental data. Thus, functional and structural parameters in the developing brain are already extremely distributed, suggesting that this organization is preconfigured and not experience dependent.
Collapse
Affiliation(s)
- Mattia Chini
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Marilena Hnida
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johanna K Kostka
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yu-Nan Chen
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Mòdol L, Moissidis M, Selten M, Oozeer F, Marín O. Somatostatin interneurons control the timing of developmental desynchronization in cortical networks. Neuron 2024; 112:2015-2030.e5. [PMID: 38599213 DOI: 10.1016/j.neuron.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/21/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Synchronous neuronal activity is a hallmark of the developing brain. In the mouse cerebral cortex, activity decorrelates during the second week of postnatal development, progressively acquiring the characteristic sparse pattern underlying the integration of sensory information. The maturation of inhibition seems critical for this process, but the interneurons involved in this crucial transition of network activity in the developing cortex remain unknown. Using in vivo longitudinal two-photon calcium imaging during the period that precedes the change from highly synchronous to decorrelated activity, we identify somatostatin-expressing (SST+) interneurons as critical modulators of this switch in mice. Modulation of the activity of SST+ cells accelerates or delays the decorrelation of cortical network activity, a process that involves regulating the maturation of parvalbumin-expressing (PV+) interneurons. SST+ cells critically link sensory inputs with local circuits, controlling the neural dynamics in the developing cortex while modulating the integration of other interneurons into nascent cortical circuits.
Collapse
Affiliation(s)
- Laura Mòdol
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Monika Moissidis
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
12
|
McKeon SD, Perica MI, Parr AC, Calabro FJ, Foran W, Hetherington H, Moon CH, Luna B. Aperiodic EEG and 7T MRSI evidence for maturation of E/I balance supporting the development of working memory through adolescence. Dev Cogn Neurosci 2024; 66:101373. [PMID: 38574406 PMCID: PMC11000172 DOI: 10.1016/j.dcn.2024.101373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/06/2024] Open
Abstract
Adolescence has been hypothesized to be a critical period for the development of human association cortex and higher-order cognition. A defining feature of critical period development is a shift in the excitation: inhibition (E/I) balance of neural circuitry, however how changes in E/I may enhance cortical circuit function to support maturational improvements in cognitive capacities is not known. Harnessing ultra-high field 7 T MR spectroscopy and EEG in a large, longitudinal cohort of youth (N = 164, ages 10-32 years old, 347 neuroimaging sessions), we delineate biologically specific associations between age-related changes in excitatory glutamate and inhibitory GABA neurotransmitters and EEG-derived measures of aperiodic neural activity reflective of E/I balance in prefrontal association cortex. Specifically, we find that developmental increases in E/I balance reflected in glutamate:GABA balance are linked to changes in E/I balance assessed by the suppression of prefrontal aperiodic activity, which in turn facilitates robust improvements in working memory. These findings indicate a role for E/I-engendered changes in prefrontal signaling mechanisms in the maturation of cognitive maintenance. More broadly, this multi-modal imaging study provides evidence that human association cortex undergoes physiological changes consistent with critical period plasticity during adolescence.
Collapse
Affiliation(s)
- Shane D McKeon
- Department of Bioengineering, University of Pittsburgh, PA, USA; The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA.
| | - Maria I Perica
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA; Department of Psychology, University of Pittsburgh, PA, USA
| | - Ashley C Parr
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Finnegan J Calabro
- Department of Bioengineering, University of Pittsburgh, PA, USA; The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Will Foran
- Department of Psychiatry, University of Pittsburgh, PA, USA
| | - Hoby Hetherington
- Resonance Research Incorporated, Billerica, MA, USA; Department of Radiology, University of Missouri, Columbia, MO, USA
| | - Chan-Hong Moon
- Department of Radiology, University of Pittsburgh, PA, USA
| | - Beatriz Luna
- The Center for the Neural Basis of Cognition, University of Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Thomson AR, Hwa H, Pasanta D, Hopwood B, Powell HJ, Lawrence R, Tabuenca ZG, Arichi T, Edden RAE, Chai X, Puts NA. The developmental trajectory of 1H-MRS brain metabolites from childhood to adulthood. Cereb Cortex 2024; 34:bhae046. [PMID: 38430105 PMCID: PMC10908220 DOI: 10.1093/cercor/bhae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 03/03/2024] Open
Abstract
Human brain development is ongoing throughout childhood, with for example, myelination of nerve fibers and refinement of synaptic connections continuing until early adulthood. 1H-Magnetic Resonance Spectroscopy (1H-MRS) can be used to quantify the concentrations of endogenous metabolites (e.g. glutamate and γ -aminobutyric acid (GABA)) in the human brain in vivo and so can provide valuable, tractable insight into the biochemical processes that support postnatal neurodevelopment. This can feasibly provide new insight into and aid the management of neurodevelopmental disorders by providing chemical markers of atypical development. This study aims to characterize the normative developmental trajectory of various brain metabolites, as measured by 1H-MRS from a midline posterior parietal voxel. We find significant non-linear trajectories for GABA+ (GABA plus macromolecules), Glx (glutamate + glutamine), total choline (tCho) and total creatine (tCr) concentrations. Glx and GABA+ concentrations steeply decrease across childhood, with more stable trajectories across early adulthood. tCr and tCho concentrations increase from childhood to early adulthood. Total N-acetyl aspartate (tNAA) and Myo-Inositol (mI) concentrations are relatively stable across development. Trajectories likely reflect fundamental neurodevelopmental processes (including local circuit refinement) which occur from childhood to early adulthood and can be associated with cognitive development; we find GABA+ concentrations significantly positively correlate with recognition memory scores.
Collapse
Affiliation(s)
- Alice R Thomson
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Department of Neurodevelopmental Disorders, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, United Kingdom
| | - Hannah Hwa
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Duanghathai Pasanta
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Benjamin Hopwood
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Helen J Powell
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Ross Lawrence
- Division of Cognitive Neurology, Department of Neurology, Johns Hopkins University, 1629 Thames Street Suite 350, Baltimore, MD 21231, United States
| | - Zeus G Tabuenca
- Department of Statistical Methods, University of Zaragoza, Pedro Cerbuna 12, Zaragoza, 50009, Spain
| | - Tomoki Arichi
- MRC Centre for Neurodevelopmental Disorders, Department of Neurodevelopmental Disorders, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, United Kingdom
- Centre for the Developing Brain, Department of Perinatal Imaging & Health, 1st Floor, South Wing, St Thomas’ Hospital, London, SE1 7EH, United Kingdom
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 601 North Caroline Street, Baltimore, MD 21287, United States
- F.M. Kirby Research Centre for Functional Brain Imaging, Kennedy Krieger Institute, 707 North Broadway, Baltimore, MD 21205, United States
| | - Xiaoqian Chai
- Department of Neurology and Neurosurgery, McGill University, QC H3A2B4, Canada
| | - Nicolaas A Puts
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Department of Neurodevelopmental Disorders, New Hunt's House, Guy's Campus, King's College London, London, SE1 1UL, United Kingdom
| |
Collapse
|
14
|
Pochinok I, Stöber TM, Triesch J, Chini M, Hanganu-Opatz IL. A developmental increase of inhibition promotes the emergence of hippocampal ripples. Nat Commun 2024; 15:738. [PMID: 38272901 PMCID: PMC10810866 DOI: 10.1038/s41467-024-44983-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024] Open
Abstract
Sharp wave-ripples (SPW-Rs) are a hippocampal network phenomenon critical for memory consolidation and planning. SPW-Rs have been extensively studied in the adult brain, yet their developmental trajectory is poorly understood. While SPWs have been recorded in rodents shortly after birth, the time point and mechanisms of ripple emergence are still unclear. Here, we combine in vivo electrophysiology with optogenetics and chemogenetics in 4 to 12-day-old mice to address this knowledge gap. We show that ripples are robustly detected and induced by light stimulation of channelrhodopsin-2-transfected CA1 pyramidal neurons only from postnatal day 10 onwards. Leveraging a spiking neural network model, we mechanistically link the maturation of inhibition and ripple emergence. We corroborate these findings by reducing ripple rate upon chemogenetic silencing of CA1 interneurons. Finally, we show that early SPW-Rs elicit a more robust prefrontal cortex response than SPWs lacking ripples. Thus, development of inhibition promotes ripples emergence.
Collapse
Affiliation(s)
- Irina Pochinok
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology (ZMNH), Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Tristan M Stöber
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
| | - Jochen Triesch
- Frankfurt Institute for Advanced Studies, 60438, Frankfurt am Main, Germany
| | - Mattia Chini
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology (ZMNH), Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology (ZMNH), Hamburg Center of Neuroscience (HCNS), University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
15
|
Kalinowski D, Bogus-Nowakowska K, Kozłowska A, Równiak M. The Co-Expression Pattern of Calcium-Binding Proteins with γ-Aminobutyric Acid and Glutamate Transporters in the Amygdala of the Guinea Pig: Evidence for Glutamatergic Subpopulations. Int J Mol Sci 2023; 24:15025. [PMID: 37834473 PMCID: PMC10573686 DOI: 10.3390/ijms241915025] [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: 08/04/2023] [Revised: 09/27/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
The amygdala has large populations of neurons utilizing specific calcium-binding proteins such as parvalbumin (PV), calbindin (CB), or calretinin (CR). They are considered specialized subsets of γ-aminobutyric acid (GABA) interneurons; however, many of these cells are devoid of GABA or glutamate decarboxylase. The neurotransmitters used by GABA-immunonegative cells are still unknown, but it is suggested that a part may use glutamate. Thus, this study investigates in the amygdala of the guinea pig relationships between PV, CB, or CR-containing cells and GABA transporter (VGAT) or glutamate transporter type 2 (VGLUT2), markers of GABAergic and glutamatergic neurons, respectively. The results show that although most neurons using PV, CB, and CR co-expressed VGAT, each of these populations also had a fraction of VGLUT2 co-expressing cells. For almost all neurons using PV (~90%) co-expressed VGAT, while ~1.5% of them had VGLUT2. The proportion of neurons using CB and VGAT was smaller than that for PV (~80%), while the percentage of cells with VGLUT2 was larger (~4.5%). Finally, only half of the neurons using CR (~53%) co-expressed VGAT, while ~3.5% of them had VGLUT2. In conclusion, the populations of neurons co-expressing PV, CB, and CR are in the amygdala, primarily GABAergic. However, at least a fraction of neurons in each of them co-express VGLUT2, suggesting that these cells may use glutamate. Moreover, the number of PV-, CB-, and CR-containing neurons that may use glutamate is probably larger as they can utilize VGLUT1 or VGLUT3, which are also present in the amygdala.
Collapse
Affiliation(s)
- Daniel Kalinowski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, pl. Łódzki 3, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| | - Krystyna Bogus-Nowakowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, pl. Łódzki 3, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Maciej Równiak
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, pl. Łódzki 3, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| |
Collapse
|
16
|
Choi JS, Ayupe AC, Beckedorff F, Catanuto P, McCartan R, Levay K, Park KK. Single-nucleus RNA sequencing of developing superior colliculus identifies neuronal diversity and candidate mediators of circuit assembly. Cell Rep 2023; 42:113037. [PMID: 37624694 PMCID: PMC10592058 DOI: 10.1016/j.celrep.2023.113037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/26/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The superior colliculus (SC) is a sensorimotor structure in the midbrain that integrates input from multiple sensory modalities to initiate motor commands. It undergoes well-characterized steps of circuit assembly during development, rendering the mouse SC a popular model to study establishment of neural connectivity. Here we perform single-nucleus RNA-sequencing analysis of the mouse SC isolated at various developmental time points. Our study provides a transcriptomic landscape of the cell types that comprise the SC across murine development with particular emphasis on neuronal heterogeneity. We report a repertoire of genes differentially expressed across the different postnatal ages, many of which are known to regulate axon guidance and synapse formation. Using these data, we find that Pax7 expression is restricted to a subset of GABAergic neurons. Our data provide a valuable resource for interrogating the mechanisms of circuit development and identifying markers for manipulating specific SC neuronal populations and circuits.
Collapse
Affiliation(s)
- James S Choi
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Ter., Miami, FL 33136, USA
| | - Ana C Ayupe
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Ter., Miami, FL 33136, USA
| | - Felipe Beckedorff
- Department of Human Genetics, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, 1501 NW 10th Avenue, Miami, FL 33136, USA
| | - Paola Catanuto
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Ter., Miami, FL 33136, USA
| | - Robyn McCartan
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Ter., Miami, FL 33136, USA
| | - Konstantin Levay
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Ter., Miami, FL 33136, USA
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Ter., Miami, FL 33136, USA.
| |
Collapse
|
17
|
Lombardi A, Wang Q, Stüttgen MC, Mittmann T, Luhmann HJ, Kilb W. Recovery kinetics of short-term depression of GABAergic and glutamatergic synapses at layer 2/3 pyramidal cells in the mouse barrel cortex. Front Cell Neurosci 2023; 17:1254776. [PMID: 37817883 PMCID: PMC10560857 DOI: 10.3389/fncel.2023.1254776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction Short-term synaptic plasticity (STP) is a widespread mechanism underlying activity-dependent modifications of cortical networks. Methods To investigate how STP influences excitatory and inhibitory synapses in layer 2/3 of mouse barrel cortex, we combined whole-cell patch-clamp recordings from visually identified pyramidal neurons (PyrN) and parvalbumin-positive interneurons (PV-IN) of cortical layer 2/3 in acute slices with electrical stimulation of afferent fibers in layer 4 and optogenetic activation of PV-IN. Results These experiments revealed that electrical burst stimulation (10 pulses at 10 Hz) of layer 4 afferents to layer 2/3 neurons induced comparable short-term depression (STD) of glutamatergic postsynaptic currents (PSCs) in PyrN and in PV-IN, while disynaptic GABAergic PSCs in PyrN showed a stronger depression. Burst-induced depression of glutamatergic PSCs decayed within <4 s, while the decay of GABAergic PSCs required >11 s. Optogenetically-induced GABAergic PSCs in PyrN also demonstrated STD after burst stimulation, with a decay of >11 s. Excitatory postsynaptic potentials (EPSPs) in PyrN were unaffected after electrical burst stimulation, while a selective optogenetic STD of GABAergic synapses caused a transient increase of electrically evoked EPSPs in PyrN. Discussion In summary, these results demonstrate substantial short-term plasticity at all synapses investigated and suggest that the prominent STD observed in GABAergic synapses can moderate the functional efficacy of glutamatergic STD after repetitive synaptic stimulations. This mechanism may contribute to a reliable information flow toward the integrative layer 2/3 for complex time-varying sensory stimuli.
Collapse
Affiliation(s)
- Aniello Lombardi
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Qiang Wang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Maik C. Stüttgen
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Mittmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Heiko J. Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
18
|
Yakovlev AV, Kurmashova E, Gataulina E, Gerasimova E, Khalilov I, Sitdikova GF. Maternal hyperhomocysteinemia increases seizures susceptibility of neonatal rats. Life Sci 2023; 329:121953. [PMID: 37467884 DOI: 10.1016/j.lfs.2023.121953] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023]
Abstract
AIMS Neonatal seizures are severe pathologies which may result in long-term neurological consequences. High plasma concentrations of homocysteine - hyperhomocysteinemia (hHCy) - are associated with epilepsy. In the present study, we evaluated susceptibility to seizure of neonatal rats with prenatal hHCy. MAIN METHODS Prenatal hHCy was induced by feeding females with a high-methionine diet. Experiments were performed on pups during the first three postnatal weeks. Flurothyl-induced epileptic behavior was assessed according to Racine's scale. Epileptiform activity in the hippocampus was recorded using electrophysiological methods. The balance of excitation/inhibition, functional GABAergic inhibition and GABA reversal potential in hippocampal neurons were analyzed. KEY FINDINGS Rats with hHCy developed more severe stages of behavioral patterns during flurothyl-induced epilepsy with shorter latency. Electrophysiological recordings demonstrated higher background neuronal activity in rats with hHCy. Seizure-like events triggered by flurothyl (in vivo) or 4-aminopyridine (in vitro) showed shorter latency, higher power and amplitude. An increased glutamate/GABA synaptic ratio was shown in the pyramidal neurons of rats with hHCy and more slices demonstrated excitation by isoguvacine, a selective GABA(A) receptor agonist, during the first and second postnatal weeks. The GABA driving force and the reversal potential of GABA(A) currents were more positive during the second postnatal week for hHCy rats. SIGNIFICANCE The higher susceptibility to seizures in rats with prenatal hHCy due to a shift in the balance of excitation/inhibition toward excitation may underlie the clinical evidence about the association of hHCy with an increased risk of epilepsy.
Collapse
Affiliation(s)
- A V Yakovlev
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan 420008, Russia.
| | - E Kurmashova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan 420008, Russia
| | - E Gataulina
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan 420008, Russia
| | - E Gerasimova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan 420008, Russia
| | - I Khalilov
- Institut de Neurobiologie de la Méditerranée, INMED UMR901 Parc scientifique de Luminy, 163 avenue de Luminy BP13 - 13273, Marseille cedex 09, France; Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan 420008, Russia
| | - G F Sitdikova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kremlevskaya str 18, Kazan 420008, Russia
| |
Collapse
|
19
|
Vasilopoulos N, Kaplanian A, Vinos M, Katsaiti Y, Christodoulou O, Denaxa M, Skaliora I. The role of selective SATB1 deletion in somatostatin expressing interneurons on endogenous network activity and the transition to epilepsy. J Neurosci Res 2023; 101:424-447. [PMID: 36541427 DOI: 10.1002/jnr.25156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/24/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
Somatostatin (SST) expressing interneurons are the second most abundant group of inhibitory neurons in the neocortex. They mainly target the apical dendrites of excitatory pyramidal cells and are implicated in feedforward and feedback inhibition. In the present study, we employ a conditional knockout mouse, in which the transcription factor Satb1 is selectively deleted in SST-expressing interneurons resulting to the reduction of their number across the somatosensory barrel field. Our goal was to investigate the effect of the reduced number of Satb1 mutant SST-interneurons on (i) the endogenous cortical network activity (spontaneously recurring Up/Down states), and (ii) the transition to epileptiform activity. By conducting LFP recordings in acute brain slices from young male and female mice, we demonstrate that mutant animals exhibit significant changes in network excitability, reflected in increased Up state occurrence, decreased Up state duration and higher levels of extracellular spiking activity. Epileptiform activity was induced through two distinct and widely used in vitro protocols: the low magnesium and the 4-Aminopyridine (4-AP) model. In the former, slices from mutant animals manifested shorter latency for the expression of stable seizure-like events. In contrast, when epilepsy was induced by 4-AP, no significant differences were reported. We conclude that normal SST-interneuron function has a significant role both in the regulation of the endogenous network activity, and in the transition to seizure-like discharges in a context-dependent manner.
Collapse
Affiliation(s)
- Nikos Vasilopoulos
- Centre for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Ani Kaplanian
- Centre for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Michael Vinos
- Centre for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.,Department of History and Philosophy of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Yolanda Katsaiti
- Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Department of Biology, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Ourania Christodoulou
- Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Department of Biology, University of Crete, Heraklion, Greece
| | - Myrto Denaxa
- Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Irini Skaliora
- Centre for Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.,Department of History and Philosophy of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| |
Collapse
|
20
|
McSweeney M, Morales S, Valadez EA, Buzzell GA, Yoder L, Fifer WP, Pini N, Shuffrey LC, Elliott AJ, Isler JR, Fox NA. Age-related trends in aperiodic EEG activity and alpha oscillations during early- to middle-childhood. Neuroimage 2023; 269:119925. [PMID: 36739102 DOI: 10.1016/j.neuroimage.2023.119925] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Age-related structural and functional changes that occur during brain development are critical for cortical development and functioning. Previous electroencephalography (EEG) and magnetoencephalography (MEG) studies have highlighted the utility of power spectra analyses and have uncovered age-related trends that reflect perceptual, cognitive, and behavioural states as well as their underlying neurophysiology. The aim of the current study was to investigate age-related change in aperiodic and periodic alpha activity across a large sample of pre- and school-aged children (N = 502, age range 4 -11-years-of-age). Power spectra were extracted from baseline EEG recordings (eyes closed, eyes open) for each participant and parameterized into aperiodic activity to derive the offset and exponent parameters and periodic alpha oscillatory activity to derive the alpha peak frequency and the associated power estimates. Multilevel models were run to investigate age-related trends and condition-dependent changes for each of these measures. We found quadratic age-related effects for both the aperiodic offset and exponent. In addition, we observed increases in periodic alpha peak frequency as a function of age. Aperiodic measures and periodic alpha power were larger in magnitude during eyes closed compared to the eyes open baseline condition. Taken together, these results advance our understanding of the maturational patterns/trajectories of brain development during early- to middle-childhood.
Collapse
Affiliation(s)
- Marco McSweeney
- Department of Human Development and Quantitative Methodology, University of Maryland, 3304 Benjamin Building, College Park, MD 20742, USA.
| | - Santiago Morales
- Department of Psychology, University of Southern California, USA
| | - Emilio A Valadez
- Department of Human Development and Quantitative Methodology, University of Maryland, 3304 Benjamin Building, College Park, MD 20742, USA
| | - George A Buzzell
- Department of Psychology and the Center for Children and Families, Florida International University, USA
| | - Lydia Yoder
- Department of Human Development and Quantitative Methodology, University of Maryland, 3304 Benjamin Building, College Park, MD 20742, USA
| | - William P Fifer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, USA; Department of Paediatrics, Columbia University Irving Medical Center, New York, USA; Division of Developmental Neuroscience, New York State Psychiatric Institute, USA
| | - Nicolò Pini
- Department of Psychiatry, Columbia University Irving Medical Center, New York, USA; Division of Developmental Neuroscience, New York State Psychiatric Institute, USA
| | - Lauren C Shuffrey
- Department of Psychiatry, Columbia University Irving Medical Center, New York, USA; Division of Developmental Neuroscience, New York State Psychiatric Institute, USA
| | - Amy J Elliott
- Avera Research Institute, USA; Department of Paediatrics, University of South Dakota School of Medicine, USA
| | - Joseph R Isler
- Department of Paediatrics, Columbia University Irving Medical Center, New York, USA
| | - Nathan A Fox
- Department of Human Development and Quantitative Methodology, University of Maryland, 3304 Benjamin Building, College Park, MD 20742, USA
| |
Collapse
|
21
|
Arutiunian V, Arcara G, Buyanova I, Davydova E, Pereverzeva D, Sorokin A, Tyushkevich S, Mamokhina U, Danilina K, Dragoy O. Neuromagnetic 40 Hz Auditory Steady-State Response in the left auditory cortex is related to language comprehension in children with Autism Spectrum Disorder. Prog Neuropsychopharmacol Biol Psychiatry 2023; 122:110690. [PMID: 36470421 DOI: 10.1016/j.pnpbp.2022.110690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 11/06/2022] [Accepted: 11/29/2022] [Indexed: 12/08/2022]
Abstract
Language impairment is comorbid in most children with Autism Spectrum Disorder (ASD), but its neural mechanisms are still poorly understood. Some studies hypothesize that the atypical low-level sensory perception in the auditory cortex accounts for the abnormal language development in these children. One of the potential non-invasive measures of such low-level perception can be the cortical gamma-band oscillations registered with magnetoencephalography (MEG), and 40 Hz Auditory Steady-State Response (40 Hz ASSR) is a reliable paradigm for eliciting auditory gamma response. Although there is research in children with and without ASD using 40 Hz ASSR, nothing is known about the relationship between this auditory response in children with ASD and their language abilities measured directly in formal assessment. In the present study, we used MEG and individual brain models to investigate 40 Hz ASSR in primary-school-aged children with and without ASD. It was also used to assess how the strength of the auditory response is related to language abilities of children with ASD, their non-verbal IQ, and social functioning. A total of 40 children were included in the study. The results demonstrated that 40 Hz ASSR was reduced in the right auditory cortex in children with ASD when comparing them to typically developing controls. Importantly, our study provides the first evidence of the association between 40 Hz ASSR in the language-dominant left auditory cortex and language comprehension in children with ASD. This link was domain-specific because the other brain-behavior correlations were non-significant.
Collapse
Affiliation(s)
| | | | - Irina Buyanova
- Center for Language and Brain, HSE University, Moscow, Russia
| | - Elizaveta Davydova
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia; Chair of Differential Psychology and Psychophysiology, Moscow State University of Psychology and Education, Moscow, Russia
| | - Darya Pereverzeva
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Alexander Sorokin
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia; Haskins Laboratories, New Haven, CT, United States of America
| | - Svetlana Tyushkevich
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Uliana Mamokhina
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Kamilla Danilina
- Federal Resource Center for ASD, Moscow State University of Psychology and Education, Moscow, Russia
| | - Olga Dragoy
- Center for Language and Brain, HSE University, Moscow, Russia; Institute of Linguistics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
22
|
Zhou L, Sun X, Duan J. NMDARs regulate the excitatory-inhibitory balance within neural circuits. BRAIN SCIENCE ADVANCES 2023. [DOI: 10.26599/bsa.2022.9050020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Excitatory-inhibitory (E/I) balance is essential for normal neural development, behavior and cognition. E/I imbalance leads to a variety of neurological disorders, such as autism and schizophrenia. NMDA receptors (NMDARs) regulate AMPAR-mediated excitatory and GABAAR-mediated inhibitory synaptic transmission, suggesting that NMDARs play an important role in the establishment and maintenance of the E/I balance. In this review, we briefly introduced NMDARs, AMPARs and GABAARs, summarized the current studies on E/I balance mediated by NMDARs, and discussed the current advances in NMDAR-mediated AMPAR and GABAAR development. Specifically, we analyzed the role of NMDAR subunits in the establishment and maintenance of E/I balance, which may provide new therapeutic strategies for the recovery of E/I imbalance in neurological disorders.
Collapse
Affiliation(s)
- Liang Zhou
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Xiaohui Sun
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jingjing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
23
|
Bhattacharya D, Bartley AF, Li Q, Dobrunz LE. Bicuculline restores frequency-dependent hippocampal I/E ratio and circuit function in PGC-1ɑ null mice. Neurosci Res 2022; 184:9-18. [PMID: 35842011 PMCID: PMC10865982 DOI: 10.1016/j.neures.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/22/2022] [Accepted: 07/12/2022] [Indexed: 10/31/2022]
Abstract
Altered inhibition/excitation (I/E) balance contributes to various brain disorders. Dysfunctional GABAergic interneurons enhance or reduce inhibition, resulting in I/E imbalances. Differences in short-term plasticity between excitation and inhibition cause frequency-dependence of the I/E ratio, which can be altered by GABAergic dysfunction. However, it is unknown whether I/E imbalances can be rescued pharmacologically using a single dose when the imbalance magnitude is frequency-dependent. Loss of PGC-1α (peroxisome proliferator activated receptor γ coactivator 1α) causes transcriptional dysregulation in hippocampal GABAergic interneurons. PGC-1α-/- slices have enhanced baseline inhibition onto CA1 pyramidal cells, causing increased I/E ratio and impaired circuit function. High frequency stimulation reduces the I/E ratio and recovers circuit function in PGC-1α-/- slices. Here we tested if using a low dose of bicuculline that can restore baseline I/E ratio can also rescue the frequency-dependent I/E imbalances in these mice. Remarkably, bicuculline did not reduce the I/E ratio below that of wild type during high frequency stimulation. Interestingly, bicuculline enhanced the paired-pulse ratio (PPR) of disynaptic inhibition without changing the monosynaptic inhibition PPR, suggesting that bicuculline modifies interneuron recruitment and not GABA release. Bicuculline improved CA1 output in PGC-1α-/- slices, enhancing EPSP-spike coupling to wild type levels at high and low frequencies. Our results show that it is possible to rescue frequency-dependent I/E imbalances in an animal model of transcriptional dysregulation with a single treatment.
Collapse
Affiliation(s)
- Dwipayan Bhattacharya
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States
| | - Aundrea F Bartley
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States
| | - Qin Li
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States
| | - Lynn E Dobrunz
- Department of Neurobiology, Civitan International Research Center, and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, 1825 University Blvd., Birmingham, AL, United States.
| |
Collapse
|
24
|
Chini M, Pfeffer T, Hanganu-Opatz I. An increase of inhibition drives the developmental decorrelation of neural activity. eLife 2022; 11:78811. [PMID: 35975980 PMCID: PMC9448324 DOI: 10.7554/elife.78811] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Throughout development, the brain transits from early highly synchronous activity patterns to a mature state with sparse and decorrelated neural activity, yet the mechanisms underlying this process are poorly understood. The developmental transition has important functional consequences, as the latter state is thought to allow for more efficient storage, retrieval, and processing of information. Here, we show that, in the mouse medial prefrontal cortex (mPFC), neural activity during the first two postnatal weeks decorrelates following specific spatial patterns. This process is accompanied by a concomitant tilting of excitation-inhibition (E-I) ratio toward inhibition. Using optogenetic manipulations and neural network modeling, we show that the two phenomena are mechanistically linked, and that a relative increase of inhibition drives the decorrelation of neural activity. Accordingly, in mice mimicking the etiology of neurodevelopmental disorders, subtle alterations in E-I ratio are associated with specific impairments in the correlational structure of spike trains. Finally, capitalizing on EEG data from newborn babies, we show that an analogous developmental transition takes place also in the human brain. Thus, changes in E-I ratio control the (de)correlation of neural activity and, by these means, its developmental imbalance might contribute to the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Mattia Chini
- Institute of Developmental Neurophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Pfeffer
- Center for Brain and Cognition, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ileana Hanganu-Opatz
- Institute of Developmental Neurophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
25
|
GABAergic and Glutamatergic Phenotypes of Neurons Expressing Calcium-Binding Proteins in the Preoptic Area of the Guinea Pig. Int J Mol Sci 2022; 23:ijms23147963. [PMID: 35887305 PMCID: PMC9320123 DOI: 10.3390/ijms23147963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The mammalian preoptic area (POA) has large populations of calbindin (CB), calretinin (CR) and parvalbumin (PV) neurons, but phenotypes of these cells are unknown. Therefore, the question is whether neurons expressing CB, CR, and/or PV are GABAergic or glutamatergic. Double-immunofluorescence staining followed by epifluorescence and confocal microscopy was used to determine the coexpression patterns of CB, CR and PV expressing neurons with vesicular GABA transporters (VGAT) as specific markers of GABAergic neurons and vesicular glutamate transporters (VGLUT 2) as specific markers of glutamatergic neurons. The guinea pig was adopted as, like humans, it has a reproductive cycle with a true luteal phase and a long gestation period. The results demonstrated that in the guinea pig POA of both sexes, ~80% of CB+ and ~90% of CR+ neurons coexpress VGAT; however, one-fifth of CB+ neurons and one-third of CR+ cells coexpress VGLUT. About two-thirds of PV+ neurons express VGAT, and similar proportion of them coexpress VGLUT. Thus, many CB+, CR+ and PV+ neurons may be exclusively GABAergic (VGAT-expressing cells) or glutamatergic (VGLUT-expressing cells); however, at least a small fraction of CR+ cells and at least one-third of PV+ cells are likely neurons with a dual GABA/glutamate phenotype that may coexpress both transporters.
Collapse
|
26
|
Arutiunian V, Arcara G, Buyanova I, Gomozova M, Dragoy O. The age-related changes in 40 Hz Auditory Steady-State Response and sustained Event-Related Fields to the same amplitude-modulated tones in typically developing children: A magnetoencephalography study. Hum Brain Mapp 2022; 43:5370-5383. [PMID: 35833318 PMCID: PMC9812253 DOI: 10.1002/hbm.26013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 01/15/2023] Open
Abstract
Recent studies have revealed that gamma-band oscillatory and transient evoked potentials may change with age during childhood. It is hypothesized that these changes can be associated with a maturation of GABAergic neurotransmission and, subsequently, the age-related changes of excitation-inhibition balance in the neural circuits. One of the reliable paradigms for investigating these effects in the auditory cortex is 40 Hz Auditory Steady-State Response (ASSR), where participants are presented with the periodic auditory stimuli. It is known that such stimuli evoke two types of responses in magnetoencephalography (MEG)-40 Hz steady-state gamma response (or 40 Hz ASSR) and auditory evoked response called sustained Event-Related Field (ERF). Although several studies have been conducted in children, focusing on the changes of 40 Hz ASSR with age, almost nothing is known about the age-related changes of the sustained ERF to the same periodic stimuli and their relationships with changes in the gamma strength. Using MEG, we investigated the association between 40 Hz steady-state gamma response and sustained ERF response to the same stimuli and also their age-related changes in the group of 30 typically developing 7-to-12-year-old children. The results revealed a tight relationship between 40 Hz ASSR and ERF, indicating that the age-related increase in strength of 40 Hz ASSR was associated with the age-related decrease of the amplitude of ERF. These effects were discussed in the light of the maturation of the GABAergic system and excitation-inhibition balance development, which may contribute to the changes in ASSR and ERF.
Collapse
Affiliation(s)
| | | | | | | | - Olga Dragoy
- Center for Language and BrainHSE UniversityMoscowRussia,Institute of LinguisticsRussian Academy of SciencesMoscowRussia
| |
Collapse
|
27
|
Transcranial Direct Current Stimulation as an Approach to Mitigate Neurodevelopmental Disorders Affecting Excitation/Inhibition Balance: Focus on Autism Spectrum Disorder, Schizophrenia, and Attention Deficit/Hyperactivity Disorder. J Clin Med 2022; 11:jcm11102839. [PMID: 35628965 PMCID: PMC9143428 DOI: 10.3390/jcm11102839] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
Transcranial direct current stimulation (tDCS) has been proposed as a promising therapy for rehabilitation of neurodevelopmental disorders. In this review, we discuss studies on the impact of tDCS as a therapy for autism, schizophrenia, and attention deficit/hyperactivity disorder, as well as the tDCS' mechanism of action, and propose future paths of research to optimize tDCS treatment protocols. The mechanism underlying tDCS effects is the modulation of excitatory and/or inhibitory activity, making it a valuable tool for restoring the excitation/inhibition (E/I) balance which is disrupted in many neurodevelopmental disorders. Clinical studies have shown that tDCS therapy is well-tolerated by patients and seems to ameliorate behavior and cognitive functions. Alterations in early development of neuronal circuits lead to disruptions in brain activity in neurodevelopmental disorders. An increasing amount of research into the effects of tDCS on neuronal activity has provided a foundation for its use as a therapy for behavior and cognitive characteristics of neurodevelopmental disorders. Clinical studies show that tDCS appears to ameliorate behavioral and cognitive outcomes of patients with autism, schizophrenia, and attention deficit/hyperactivity disorder. More research is needed to understand the mechanisms of action of tDCS and to optimize treatment protocols.
Collapse
|
28
|
Homeostatic plasticity and excitation-inhibition balance: The good, the bad, and the ugly. Curr Opin Neurobiol 2022; 75:102553. [PMID: 35594578 DOI: 10.1016/j.conb.2022.102553] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/15/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022]
Abstract
In this review, we discuss the significance of the synaptic excitation/inhibition (E/I) balance in the context of homeostatic plasticity, whose primary goal is thought to maintain neuronal firing rates at a set point. We first provide an overview of the processes through which patterned input activity drives synaptic E/I tuning and maturation of circuits during development. Next, we emphasize the importance of the E/I balance at the synaptic level (homeostatic control of message reception) as a means to achieve the goal (homeostatic control of information transmission) at the network level and consider how compromised homeostatic plasticity associated with neurological diseases leads to hyperactivity, network instability, and ultimately improper information processing. Lastly, we highlight several pathological conditions related to sensory deafferentation and describe how, in some cases, homeostatic compensation without appropriate sensory inputs can result in phantom perceptions.
Collapse
|
29
|
Karalunas SL, Ostlund BD, Alperin BR, Figuracion M, Gustafsson HC, Deming EM, Foti D, Antovich D, Dude J, Nigg J, Sullivan E. Electroencephalogram aperiodic power spectral slope can be reliably measured and predicts ADHD risk in early development. Dev Psychobiol 2022; 64:e22228. [PMID: 35312046 PMCID: PMC9707315 DOI: 10.1002/dev.22228] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/20/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The aperiodic exponent of the electroencephalogram (EEG) power spectrum has received growing attention as a physiological marker of neurodevelopmental psychopathology, including attention-deficit/hyperactivity disorder (ADHD). However, its use as a marker of ADHD risk across development, and particularly in very young children, is limited by unknown reliability, difficulty in aligning canonical band-based measures across development periods, and unclear effects of treatment in later development. Here, we investigate the internal consistency of the aperiodic EEG power spectrum slope and its association with ADHD risk in both infants (n = 69, 1-month-old) and adolescents (n = 262, ages 11-17 years). Results confirm good to excellent internal consistency in infancy and adolescence. In infancy, a larger aperiodic exponent was associated with greater family history of ADHD. In contrast, in adolescence, ADHD diagnosis was associated with a smaller aperiodic exponent, but only in children with ADHD who had not received stimulant medication treatment. Results suggest that disruptions in cortical development associated with ADHD risk may be detectable shortly after birth via this approach. Together, findings imply a dynamic developmental shift in which the developmentally normative flattening of the EEG power spectrum is exaggerated in ADHD, potentially reflecting imbalances in cortical excitation and inhibition that could contribute to long-lasting differences in brain connectivity.
Collapse
Affiliation(s)
- Sarah L Karalunas
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Brendan D Ostlund
- Department of Psychology, Pennsylvania State University, State College, Pennsylvania, USA
| | - Brittany R Alperin
- Department of Psychology, University of Richmond, Richmond, Virginia, USA
| | - McKenzie Figuracion
- Department of Psychiatry, Oregon Health and Science University, Portland, Oregon, USA
| | - Hanna C Gustafsson
- Department of Psychiatry, Oregon Health and Science University, Portland, Oregon, USA
| | - Erika Michiko Deming
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Dan Foti
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Dylan Antovich
- Department of Psychiatry, Oregon Health and Science University, Portland, Oregon, USA
| | - Jason Dude
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Joel Nigg
- Department of Psychiatry, Oregon Health and Science University, Portland, Oregon, USA
| | - Elinor Sullivan
- Department of Psychiatry, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
30
|
Kalinowski D, Bogus-Nowakowska K, Kozłowska A, Równiak M. Expression of Calbindin, a Marker of Gamma-Aminobutyric Acid Neurons, Is Reduced in the Amygdala of Oestrogen Receptor β-Deficient Female Mice. J Clin Med 2022; 11:jcm11071760. [PMID: 35407369 PMCID: PMC8999607 DOI: 10.3390/jcm11071760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/12/2022] [Accepted: 03/19/2022] [Indexed: 12/15/2022] Open
Abstract
Oestrogen receptor β (ERβ) knock-out female mice display increased anxiety and decreased threshold for synaptic plasticity induction in the basolateral amygdala. This may suggest that the γ-aminobutyric acid (GABA) inhibitory system is altered. Therefore, the immunoreactivity of main GABAergic markers-i.e., calbindin, parvalbumin, calretinin, somatostatin, α1 subunit-containing GABAA receptor and vesicular GABA transporter-were compared in the six subregions (LA, BL, BM, ME, CE and CO) of the amygdala of adult female wild-type and ERβ knock-out mice using immunohistochemistry and quantitative methods. The influence of ERβ knock-out on neuronal loss and glia was also elucidated using pan-neuronal and astrocyte markers. The results show severe neuronal deficits in all main amygdala regions in ERβ knock-out mice accompanied by astroglia overexpression only in the medial, basomedial and cortical nuclei and a decrease in calbindin-expressing neurons (CB+) in the amygdala in ERβ knock-out mice compared with controls, while other markers of the GABAergic system remain unchanged. Concluding, the lack of ERβ led to failure in the structural integrity of the CB+ subpopulation, reducing interneuron firing and resulting in a disinhibitory effect over pyramidal function. This fear-promoting excitatory/inhibitory alteration may lead to the increased anxiety observed in these mice. The impact of neuronal deficits and astroglia overexpression on the amygdala functions remains unknown.
Collapse
Affiliation(s)
- Daniel Kalinowski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
- Correspondence: ; Tel./Fax: +48-89-523-4301
| | - Krystyna Bogus-Nowakowska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland;
| | - Maciej Równiak
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-727 Olsztyn, Poland; (K.B.-N.); (M.R.)
| |
Collapse
|
31
|
Smith LM, Kim JZ, Lu Z, Bassett DS. Learning continuous chaotic attractors with a reservoir computer. CHAOS (WOODBURY, N.Y.) 2022; 32:011101. [PMID: 35105129 DOI: 10.1063/5.0075572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/19/2021] [Indexed: 06/14/2023]
Abstract
Neural systems are well known for their ability to learn and store information as memories. Even more impressive is their ability to abstract these memories to create complex internal representations, enabling advanced functions such as the spatial manipulation of mental representations. While recurrent neural networks (RNNs) are capable of representing complex information, the exact mechanisms of how dynamical neural systems perform abstraction are still not well-understood, thereby hindering the development of more advanced functions. Here, we train a 1000-neuron RNN-a reservoir computer (RC)-to abstract a continuous dynamical attractor memory from isolated examples of dynamical attractor memories. Furthermore, we explain the abstraction mechanism with a new theory. By training the RC on isolated and shifted examples of either stable limit cycles or chaotic Lorenz attractors, the RC learns a continuum of attractors as quantified by an extra Lyapunov exponent equal to zero. We propose a theoretical mechanism of this abstraction by combining ideas from differentiable generalized synchronization and feedback dynamics. Our results quantify abstraction in simple neural systems, enabling us to design artificial RNNs for abstraction and leading us toward a neural basis of abstraction.
Collapse
Affiliation(s)
- Lindsay M Smith
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jason Z Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Zhixin Lu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Dani S Bassett
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
32
|
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
|
33
|
McSweeney M, Morales S, Valadez EA, Buzzell GA, Fox NA. Longitudinal age- and sex-related change in background aperiodic activity during early adolescence. Dev Cogn Neurosci 2021; 52:101035. [PMID: 34781249 PMCID: PMC8605214 DOI: 10.1016/j.dcn.2021.101035] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 02/08/2023] Open
Abstract
Aperiodic activity contains important and meaningful physiological information that has been shown to dynamically change with age. However, no longitudinal studies have examined its development during early-to-mid adolescence. The current study closes this gap by investigating age- and sex-related longitudinal change in aperiodic activity across early-to-mid adolescence (N = 186; 54.3% female). Participants completed a resting state task and a Flanker task while EEG was record at age 13 years and again at age 15 years. Across different tasks and two time points, we observed significant age-related reductions in aperiodic offset and exponent. In addition, we observed significant sex-related differences in the aperiodic offset and exponent over time. We did not find any significant correlation between aperiodic activity and behavioral measures, nor did we find any significant condition-dependent change in aperiodic activity during the Flanker task. However, we did observe significant correlations between aperiodic activity across tasks and over time, suggesting that aperiodic activity may demonstrate stable trait-like characteristics. Collectively, these results may suggest a developmental parallelism between decreases in aperiodic components alongside adolescent brain development during this period; changes to cortical and subcortical brain structure and organization during early adolescence may have been responsible for the observed sex-related effects. Early adolescence is associated with changes in the aperiodic signal. We observed significant sex-related differences in the aperiodic signal over time. Aperiodic activity is significantly correlated within/between tasks and over time.
Collapse
|
34
|
Sydnor VJ, Larsen B, Bassett DS, Alexander-Bloch A, Fair DA, Liston C, Mackey AP, Milham MP, Pines A, Roalf DR, Seidlitz J, Xu T, Raznahan A, Satterthwaite TD. Neurodevelopment of the association cortices: Patterns, mechanisms, and implications for psychopathology. Neuron 2021; 109:2820-2846. [PMID: 34270921 PMCID: PMC8448958 DOI: 10.1016/j.neuron.2021.06.016] [Citation(s) in RCA: 324] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/24/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022]
Abstract
The human brain undergoes a prolonged period of cortical development that spans multiple decades. During childhood and adolescence, cortical development progresses from lower-order, primary and unimodal cortices with sensory and motor functions to higher-order, transmodal association cortices subserving executive, socioemotional, and mentalizing functions. The spatiotemporal patterning of cortical maturation thus proceeds in a hierarchical manner, conforming to an evolutionarily rooted, sensorimotor-to-association axis of cortical organization. This developmental program has been characterized by data derived from multimodal human neuroimaging and is linked to the hierarchical unfolding of plasticity-related neurobiological events. Critically, this developmental program serves to enhance feature variation between lower-order and higher-order regions, thus endowing the brain's association cortices with unique functional properties. However, accumulating evidence suggests that protracted plasticity within late-maturing association cortices, which represents a defining feature of the human developmental program, also confers risk for diverse developmental psychopathologies.
Collapse
Affiliation(s)
- Valerie J Sydnor
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Danielle S Bassett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Electrical & Systems Engineering, School of Engineering & Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Physics & Astronomy, College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Aaron Alexander-Bloch
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Damien A Fair
- Masonic Institute for the Developing Brain, Institute of Child Development, College of Education and Human Development, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN 55414, USA
| | - Conor Liston
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Allyson P Mackey
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P Milham
- Center for the Developing Brain, Child Mind Institute, New York, NY 10022, USA; Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY 10962, USA
| | - Adam Pines
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David R Roalf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jakob Seidlitz
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Child and Adolescent Psychiatry and Behavioral Science, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ting Xu
- Center for the Developing Brain, Child Mind Institute, New York, NY 10022, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, NIMH Intramural Research Program, NIH, Bethesda, MD 20892, USA
| | - Theodore D Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Kim JZ, Lu Z, Nozari E, Pappas GJ, Bassett DS. Teaching recurrent neural networks to infer global temporal structure from local examples. NAT MACH INTELL 2021. [DOI: 10.1038/s42256-021-00321-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
36
|
Jamann N, Dannehl D, Lehmann N, Wagener R, Thielemann C, Schultz C, Staiger J, Kole MHP, Engelhardt M. Sensory input drives rapid homeostatic scaling of the axon initial segment in mouse barrel cortex. Nat Commun 2021; 12:23. [PMID: 33397944 PMCID: PMC7782484 DOI: 10.1038/s41467-020-20232-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
The axon initial segment (AIS) is a critical microdomain for action potential initiation and implicated in the regulation of neuronal excitability during activity-dependent plasticity. While structural AIS plasticity has been suggested to fine-tune neuronal activity when network states change, whether it acts in vivo as a homeostatic regulatory mechanism in behaviorally relevant contexts remains poorly understood. Using the mouse whisker-to-barrel pathway as a model system in combination with immunofluorescence, confocal analysis and electrophysiological recordings, we observed bidirectional AIS plasticity in cortical pyramidal neurons. Furthermore, we find that structural and functional AIS remodeling occurs in distinct temporal domains: Long-term sensory deprivation elicits an AIS length increase, accompanied with an increase in neuronal excitability, while sensory enrichment results in a rapid AIS shortening, accompanied by a decrease in action potential generation. Our findings highlight a central role of the AIS in the homeostatic regulation of neuronal input-output relations.
Collapse
Affiliation(s)
- Nora Jamann
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, The Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dominik Dannehl
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nadja Lehmann
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Robin Wagener
- Clinic of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Corinna Thielemann
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian Schultz
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jochen Staiger
- Institute of Neuroanatomy, University Medical Center, Georg August University of Göttingen, Göttingen, Germany
| | - Maarten H P Kole
- Axonal Signaling Group, Netherlands Institute for Neurosciences (NIN), Royal Netherlands Academy for Arts and Sciences (KNAW), Amsterdam, The Netherlands.
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| | - Maren Engelhardt
- Institute of Neuroanatomy, Mannheim Center for Translational Neuroscience (MCTN), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
37
|
Dutta A, Karanth SS, Bhattacharya M, Liput M, Augustyniak J, Cheung M, Stachowiak EK, Stachowiak MK. A proof of concept 'phase zero' study of neurodevelopment using brain organoid models with Vis/near-infrared spectroscopy and electrophysiology. Sci Rep 2020; 10:20987. [PMID: 33268815 PMCID: PMC7710726 DOI: 10.1038/s41598-020-77929-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/18/2020] [Indexed: 12/14/2022] Open
Abstract
Homeostatic control of neuronal excitability by modulation of synaptic inhibition (I) and excitation (E) of the principal neurons is important during brain maturation. The fundamental features of in-utero brain development, including local synaptic E-I ratio and bioenergetics, can be modeled by cerebral organoids (CO) that have exhibited highly regular nested oscillatory network events. Therefore, we evaluated a 'Phase Zero' clinical study platform combining broadband Vis/near-infrared(NIR) spectroscopy and electrophysiology with studying E-I ratio based on the spectral exponent of local field potentials and bioenergetics based on the activity of mitochondrial Cytochrome-C Oxidase (CCO). We found a significant effect of the age of the healthy controls iPSC CO from 23 days to 3 months on the CCO activity (chi-square (2, N = 10) = 20, p = 4.5400e-05), and spectral exponent between 30-50 Hz (chi-square (2, N = 16) = 13.88, p = 0.001). Also, a significant effect of drugs, choline (CHO), idebenone (IDB), R-alpha-lipoic acid plus acetyl-L-carnitine (LCLA), was found on the CCO activity (chi-square (3, N = 10) = 25.44, p = 1.2492e-05), spectral exponent between 1 and 20 Hz (chi-square (3, N = 16) = 43.5, p = 1.9273e-09) and 30-50 Hz (chi-square (3, N = 16) = 23.47, p = 3.2148e-05) in 34 days old CO from schizophrenia (SCZ) patients iPSC. We present the feasibility of a multimodal approach, combining electrophysiology and broadband Vis-NIR spectroscopy, to monitor neurodevelopment in brain organoid models that can complement traditional drug design approaches to test clinically meaningful hypotheses.
Collapse
Affiliation(s)
- Anirban Dutta
- Department of Biomedical Engineering, University at Buffalo, Buffalo, 14260, USA.
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, 14260, USA.
| | | | | | - Michal Liput
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, 14260, USA
- Department of Stem Cells Bioengineering, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Justyna Augustyniak
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, 14260, USA
- Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Mancheung Cheung
- Department of Biomedical Engineering, University at Buffalo, Buffalo, 14260, USA
| | - Ewa K Stachowiak
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, 14260, USA
| | - Michal K Stachowiak
- Department of Biomedical Engineering, University at Buffalo, Buffalo, 14260, USA.
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, 14260, USA.
| |
Collapse
|
38
|
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
|
39
|
Chow CY, Absalom N, Biggs K, King GF, Ma L. Venom-derived modulators of epilepsy-related ion channels. Biochem Pharmacol 2020; 181:114043. [PMID: 32445870 DOI: 10.1016/j.bcp.2020.114043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/18/2020] [Indexed: 12/18/2022]
Abstract
Epilepsy is characterised by spontaneous recurrent seizures that are caused by an imbalance between neuronal excitability and inhibition. Since ion channels play fundamental roles in the generation and propagation of action potentials as well as neurotransmitter release at a subset of excitatory and inhibitory synapses, their dysfunction has been linked to a wide variety of epilepsies. Indeed, these unique proteins are the major biological targets for antiepileptic drugs. Selective targeting of a specific ion channel subtype remains challenging for small molecules, due to the high level of homology among members of the same channel family. As a consequence, there is a growing trend to target ion channels with biologics. Venoms are the best known natural source of ion channel modulators, and venom peptides are increasingly recognised as potential therapeutics due to their high selectivity and potency gained through millions of years of evolutionary selection pressure. Here we describe the major ion channel families involved in the pathogenesis of various types of epilepsy, including voltage-gated Na+, K+, Ca2+ channels, Cys-loop receptors, ionotropic glutamate receptors and P2X receptors, and currently available venom-derived peptides that target these channel proteins. Although only a small number of venom peptides have successfully progressed to the clinic, there is reason to be optimistic about their development as antiepileptic drugs, notwithstanding the challenges associated with development of any class of peptide drug.
Collapse
Affiliation(s)
- Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nathan Absalom
- Brain and Mind Centre, School of Pharmacy, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW 2050, Australia
| | - Kimberley Biggs
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Linlin Ma
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia.
| |
Collapse
|
40
|
Rodríguez G, Chakraborty D, Schrode KM, Saha R, Uribe I, Lauer AM, Lee HK. Cross-Modal Reinstatement of Thalamocortical Plasticity Accelerates Ocular Dominance Plasticity in Adult Mice. Cell Rep 2019; 24:3433-3440.e4. [PMID: 30257205 PMCID: PMC6233297 DOI: 10.1016/j.celrep.2018.08.072] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/19/2018] [Accepted: 08/24/2018] [Indexed: 01/09/2023] Open
Abstract
Plasticity of thalamocortical (TC) synapses is robust during early
development and becomes limited in the adult brain. We previously reported that
a short duration of deafening strengthens TC synapses in the primary visual
cortex (V1) of adult mice. Here, we demonstrate that deafening restores NMDA
receptor (NMDAR)-dependent long-term potentiation (LTP) of TC synapses onto
principal neurons in V1 layer 4 (L4), which is accompanied by an increase in
NMDAR function. In contrast, deafening did not recover long-term depression
(LTD) at TC synapses. Potentiation of TC synapses by deafening is absent in
parvalbumin-positive (PV+) interneurons, resulting in an increase in feedforward
excitation to inhibition (E/I) ratio. Furthermore, we found that a brief
duration of deafening adult mice recovers rapid ocular dominance plasticity
(ODP) mainly by accelerating potentiation of the open-eye responses. Our results
suggest that cross-modal sensory deprivation promotes adult cortical plasticity
by specifically recovering TC-LTP and increasing the E/I ratio. Plasticity of thalamocortical (TC) synapses is limited in adults.
Rodríguez et al. demonstrate that a brief period of deafening adults
recovers LTP at TC synapses in visual cortex and accelerates ocular dominance
plasticity. These results suggest that cross-modal sensory deprivation may be an
effective way to promote adult cortical plasticity.
Collapse
Affiliation(s)
- Gabriela Rodríguez
- Mind/Brain Institute, Department of Neuroscience, Johns Hopkins University, 3400 N. Charles Street, Dunning Hall, Baltimore, MD 21218, USA; Cellular Molecular Developmental Biology and Biophysics Program, Johns Hopkins University, Mudd Hall, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Darpan Chakraborty
- Mind/Brain Institute, Department of Neuroscience, Johns Hopkins University, 3400 N. Charles Street, Dunning Hall, Baltimore, MD 21218, USA
| | - Katrina M Schrode
- Department of Otolaryngology-Head and Neck Surgery and Center for Hearing and Balance, Johns Hopkins School of Medicine, 720 Rutland Ave., Traylor Building, Baltimore, MD 21205, USA
| | - Rinki Saha
- Mind/Brain Institute, Department of Neuroscience, Johns Hopkins University, 3400 N. Charles Street, Dunning Hall, Baltimore, MD 21218, USA
| | - Isabel Uribe
- Mind/Brain Institute, Department of Neuroscience, Johns Hopkins University, 3400 N. Charles Street, Dunning Hall, Baltimore, MD 21218, USA
| | - Amanda M Lauer
- Department of Otolaryngology-Head and Neck Surgery and Center for Hearing and Balance, Johns Hopkins School of Medicine, 720 Rutland Ave., Traylor Building, Baltimore, MD 21205, USA
| | - Hey-Kyoung Lee
- Mind/Brain Institute, Department of Neuroscience, Johns Hopkins University, 3400 N. Charles Street, Dunning Hall, Baltimore, MD 21218, USA; Cellular Molecular Developmental Biology and Biophysics Program, Johns Hopkins University, Mudd Hall, 3400 N. Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
41
|
Excitation/inhibition imbalance and impaired neurogenesis in neurodevelopmental and neurodegenerative disorders. Rev Neurosci 2019; 30:807-820. [DOI: 10.1515/revneuro-2019-0014] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022]
Abstract
AbstractThe excitation/inhibition (E/I) balance controls the synaptic inputs to prevent the inappropriate responses of neurons to input strength, and is required to restore the initial pattern of network activity. Various neurotransmitters affect synaptic plasticity within neural networks via the modulation of neuronal E/I balance in the developing and adult brain. Less is known about the role of E/I balance in the control of the development of the neural stem and progenitor cells in the course of neurogenesis and gliogenesis. Recent findings suggest that neural stem and progenitor cells appear to be the target for the action of GABA within the neurogenic or oligovascular niches. The same might be true for the role of neuropeptides (i.e. oxytocin) in neurogenic niches. This review covers current understanding of the role of E/I balance in the regulation of neuroplasticity associated with social behavior in normal brain, and in neurodevelopmental and neurodegenerative diseases. Further studies are required to decipher the GABA-mediated regulation of postnatal neurogenesis and synaptic integration of newly-born neurons as a potential target for the treatment of brain diseases.
Collapse
|
42
|
Kroon T, van Hugte E, van Linge L, Mansvelder HD, Meredith RM. Early postnatal development of pyramidal neurons across layers of the mouse medial prefrontal cortex. Sci Rep 2019; 9:5037. [PMID: 30911152 PMCID: PMC6433913 DOI: 10.1038/s41598-019-41661-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/12/2019] [Indexed: 12/24/2022] Open
Abstract
Mammalian neocortex is a highly layered structure. Each layer is populated by distinct subtypes of principal cells that are born at different times during development. While the differences between principal cells across layers have been extensively studied, it is not known how the developmental profiles of neurons in different layers compare. Here, we provide a detailed morphological and functional characterisation of pyramidal neurons in mouse mPFC during the first postnatal month, corresponding to known critical periods for synapse and neuron formation in mouse sensory neocortex. Our data demonstrate similar maturation profiles of dendritic morphology and intrinsic properties of pyramidal neurons in both deep and superficial layers. In contrast, the balance of synaptic excitation and inhibition differs in a layer-specific pattern from one to four postnatal weeks of age. Our characterisation of the early development and maturation of pyramidal neurons in mouse mPFC not only demonstrates a comparable time course of postnatal maturation to that in other neocortical circuits, but also implies that consideration of layer- and time-specific changes in pyramidal neurons may be relevant for studies in mouse models of neuropsychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tim Kroon
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands.
- MRC Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| | - Eline van Hugte
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Department Cognitive Neurosciences, Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Geert Grooteplein 10 Noord, 6500 HB, Nijmegen, The Netherlands
| | - Lola van Linge
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Department of Functional Genomics, Center for Neurogenomics & Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Rhiannon M Meredith
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
43
|
da Silva Lantyer A, Calcini N, Bijlsma A, Kole K, Emmelkamp M, Peeters M, Scheenen WJJ, Zeldenrust F, Celikel T. A databank for intracellular electrophysiological mapping of the adult somatosensory cortex. Gigascience 2018; 7:5232232. [PMID: 30521020 PMCID: PMC6302958 DOI: 10.1093/gigascience/giy147] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/18/2018] [Indexed: 02/04/2023] Open
Abstract
Background Neurons in the supragranular layers of the somatosensory cortex integrate sensory (bottom-up) and cognitive/perceptual (top-down) information as they orchestrate communication across cortical columns. It has been inferred, based on intracellular recordings from juvenile animals, that supragranular neurons are electrically mature by the fourth postnatal week. However, the dynamics of the neuronal integration in adulthood is largely unknown. Electrophysiological characterization of the active properties of these neurons throughout adulthood will help to address the biophysical and computational principles of the neuronal integration. Findings Here, we provide a database of whole-cell intracellular recordings from 315 neurons located in the supragranular layers (L2/3) of the primary somatosensory cortex in adult mice (9–45 weeks old) from both sexes (females, N = 195; males, N = 120). Data include 361 somatic current-clamp (CC) and 476 voltage-clamp (VC) experiments, recorded using a step-and-hold protocol (CC, N = 257; VC, N = 46), frozen noise injections (CC, N = 104) and triangular voltage sweeps (VC, 10 (N = 132), 50 (N = 146) and 100 ms (N = 152)), from regular spiking (N = 169) and fast-spiking neurons (N = 66). Conclusions The data can be used to systematically study the properties of somatic integration and the principles of action potential generation across sexes and across electrically characterized neuronal classes in adulthood. Understanding the principles of the somatic transformation of postsynaptic potentials into action potentials will shed light onto the computational principles of intracellular information transfer in single neurons and information processing in neuronal networks, helping to recreate neuronal functions in artificial systems.
Collapse
Affiliation(s)
- Angelica da Silva Lantyer
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Niccolò Calcini
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Ate Bijlsma
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Koen Kole
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Melanie Emmelkamp
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Manon Peeters
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Wim J J Scheenen
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Fleur Zeldenrust
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| | - Tansu Celikel
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyedaalseweg 135, 6525 HJ, Nijmegen - the Netherlands
| |
Collapse
|
44
|
Larsen B, Luna B. Adolescence as a neurobiological critical period for the development of higher-order cognition. Neurosci Biobehav Rev 2018; 94:179-195. [PMID: 30201220 PMCID: PMC6526538 DOI: 10.1016/j.neubiorev.2018.09.005] [Citation(s) in RCA: 416] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/29/2018] [Accepted: 09/06/2018] [Indexed: 01/08/2023]
Abstract
The transition from adolescence to adulthood is characterized by improvements in higher-order cognitive abilities and corresponding refinements of the structure and function of the brain regions that support them. Whereas the neurobiological mechanisms that govern early development of sensory systems are well-understood, the mechanisms that drive developmental plasticity of association cortices, such as prefrontal cortex (PFC), during adolescence remain to be explained. In this review, we synthesize neurodevelopmental findings at the cellular, circuit, and systems levels in PFC and evaluate them through the lens of established critical period (CP) mechanisms that guide early sensory development. We find remarkable correspondence between these neurodevelopmental processes and the mechanisms driving CP plasticity, supporting the hypothesis that adolescent development is driven by CP mechanisms that guide the rapid development of neurobiology and cognitive ability during adolescence and their subsequent stability in adulthood. Critically, understanding adolescence as a CP not only provides a mechanism for normative adolescent development, it provides a framework for understanding the role of experience and neurobiology in the emergence of psychopathology that occurs during this developmental period.
Collapse
Affiliation(s)
- Bart Larsen
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Center for the Neural Basis of Cognition, Pittsburgh, PA, 15213, United States.
| | - Beatriz Luna
- Center for the Neural Basis of Cognition, Pittsburgh, PA, 15213, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, United States
| |
Collapse
|
45
|
Foxp2 loss of function increases striatal direct pathway inhibition via increased GABA release. Brain Struct Funct 2018; 223:4211-4226. [PMID: 30187194 PMCID: PMC6267273 DOI: 10.1007/s00429-018-1746-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/31/2018] [Indexed: 12/19/2022]
Abstract
Heterozygous mutations of the Forkhead-box protein 2 (FOXP2) gene in humans cause childhood apraxia of speech. Loss of Foxp2 in mice is known to affect striatal development and impair motor skills. However, it is unknown if striatal excitatory/inhibitory balance is affected during development and if the imbalance persists into adulthood. We investigated the effect of reduced Foxp2 expression, via a loss-of-function mutation, on striatal medium spiny neurons (MSNs). Our data show that heterozygous loss of Foxp2 decreases excitatory (AMPA receptor-mediated) and increases inhibitory (GABA receptor-mediated) currents in D1 dopamine receptor positive MSNs of juvenile and adult mice. Furthermore, reduced Foxp2 expression increases GAD67 expression, leading to both increased presynaptic content and release of GABA. Finally, pharmacological blockade of inhibitory activity in vivo partially rescues motor skill learning deficits in heterozygous Foxp2 mice. Our results suggest a novel role for Foxp2 in the regulation of striatal direct pathway activity through managing inhibitory drive.
Collapse
|
46
|
van der Bourg A, Yang JW, Reyes-Puerta V, Laurenczy B, Wieckhorst M, Stüttgen MC, Luhmann HJ, Helmchen F. Layer-Specific Refinement of Sensory Coding in Developing Mouse Barrel Cortex. Cereb Cortex 2018; 27:4835-4850. [PMID: 27620976 DOI: 10.1093/cercor/bhw280] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/17/2016] [Indexed: 12/20/2022] Open
Abstract
Rodent rhythmic whisking behavior matures during a critical period around 2 weeks after birth. The functional adaptations of neocortical circuitry during this developmental period remain poorly understood. Here, we characterized stimulus-evoked neuronal activity across all layers of mouse barrel cortex before, during, and after the onset of whisking behavior. Employing multi-electrode recordings and 2-photon calcium imaging in anesthetized mice, we tested responses to rostro-caudal whisker deflections, axial "tapping" stimuli, and their combination from postnatal day 10 (P10) to P28. Within this period, whisker-evoked activity of neurons displayed a general decrease in layer 2/3 (L2/3) and L4, but increased in L5 and L6. Distinct alterations in neuronal response adaptation during the 2-s period of stimulation at ~5 Hz accompanied these changes. Moreover, single-unit analysis revealed that response selectivity in favor of either lateral deflection or axial tapping emerges in deeper layers within the critical period around P14. For superficial layers we confirmed this finding using calcium imaging of L2/3 neurons, which also exhibited emergence of response selectivity as well as progressive sparsification and decorrelation of evoked responses around P14. Our results demonstrate layer-specific development of sensory responsiveness and response selectivity in mouse somatosensory cortex coinciding with the onset of exploratory behavior.
Collapse
Affiliation(s)
- Alexander van der Bourg
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Jenq-Wei Yang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, D-55128 Mainz, Germany
| | - Vicente Reyes-Puerta
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, D-55128 Mainz, Germany
| | - Balazs Laurenczy
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| | - Martin Wieckhorst
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, CH-8057 Zurich, Switzerland
| | - Maik C Stüttgen
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University, D-55128 Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, D-55128 Mainz, Germany
| | - Fritjof Helmchen
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zurich, CH-8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, CH-8057 Zurich, Switzerland
| |
Collapse
|
47
|
Choi SJ, Mukai J, Kvajo M, Xu B, Diamantopoulou A, Pitychoutis PM, Gou B, Gogos JA, Zhang H. A Schizophrenia-Related Deletion Leads to KCNQ2-Dependent Abnormal Dopaminergic Modulation of Prefrontal Cortical Interneuron Activity. Cereb Cortex 2018; 28:2175-2191. [PMID: 28525574 PMCID: PMC6018968 DOI: 10.1093/cercor/bhx123] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 03/25/2017] [Indexed: 02/06/2023] Open
Abstract
Altered prefrontal cortex function is implicated in schizophrenia (SCZ) pathophysiology and could arise from imbalance between excitation and inhibition (E/I) in local circuits. It remains unclear whether and how such imbalances relate to genetic etiologies. We used a mouse model of the SCZ-predisposing 22q11.2 deletion (Df(16)A+/- mice) to evaluate how this genetic lesion affects the excitability of layer V prefrontal pyramidal neurons and its modulation by dopamine (DA). Df(16)A+/- mice have normal balance between E/I at baseline but are unable to maintain it upon dopaminergic challenge. Specifically, in wild-type mice, D1 receptor (D1R) activation enhances excitability of layer V prefrontal pyramidal neurons and D2 receptor (D2R) activation reduces it. Whereas the excitatory effect upon D1R activation is enhanced in Df(16)A+/- mice, the inhibitory effect upon D2R activation is reduced. The latter is partly due to the inability of mutant mice to activate GABAergic parvalbumin (PV)+ interneurons through D2Rs. We further demonstrate that reduced KCNQ2 channel function in PV+ interneurons in Df(16)A+/- mice renders them less capable of inhibiting pyramidal neurons upon D2 modulation. Thus, DA modulation of PV+ interneurons and control of E/I are altered in Df(16)A+/- mice with a higher excitation and lower inhibition during dopaminergic modulation.
Collapse
Affiliation(s)
- Se Joon Choi
- Department of Neurology, Columbia University, New York, NY10032, USA
| | - Jun Mukai
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Mirna Kvajo
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Bin Xu
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Anastasia Diamantopoulou
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Pothitos M Pitychoutis
- Department of Biology, Center for Tissue Regeneration and Engineering (TREND), University of Dayton, 300 College Park, Dayton, OH 45469, USA
| | - Bin Gou
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Joseph A Gogos
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Hui Zhang
- Department of Neurology, Columbia University, New York, NY10032, USA
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
48
|
Yang JW, Kilb W, Kirischuk S, Unichenko P, Stüttgen MC, Luhmann HJ. Development of the whisker-to-barrel cortex system. Curr Opin Neurobiol 2018; 53:29-34. [PMID: 29738998 DOI: 10.1016/j.conb.2018.04.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 01/30/2023]
Abstract
This review provides an overview on the development of the rodent whisker-to-barrel cortex system from late embryonic stage to the end of the first postnatal month. During this period the system shows a remarkable transition from a mostly genetic-molecular driven generation of crude connectivity, providing the template for activity-dependent structural and functional maturation and plasticity, to the manifestation of a complex behavioral repertoire including social interactions. Spontaneous and sensory-evoked activity is present in neonatal barrel cortex and control the generation of the cortical architecture. Half a century after its first description by Woolsey and van der Loos the whisker-to-barrel cortex system with its unique and clear topographic organization still offers the exceptional opportunity to study sensory processing and complex behavior.
Collapse
Affiliation(s)
- Jenq-Wei Yang
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany
| | - Werner Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany
| | - Sergei Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany
| | - Petr Unichenko
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany
| | - Maik C Stüttgen
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany.
| |
Collapse
|
49
|
Farhy-Tselnicker I, Allen NJ. Astrocytes, neurons, synapses: a tripartite view on cortical circuit development. Neural Dev 2018; 13:7. [PMID: 29712572 PMCID: PMC5928581 DOI: 10.1186/s13064-018-0104-y] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/17/2018] [Indexed: 01/09/2023] Open
Abstract
In the mammalian cerebral cortex neurons are arranged in specific layers and form connections both within the cortex and with other brain regions, thus forming a complex mesh of specialized synaptic connections comprising distinct circuits. The correct establishment of these connections during development is crucial for the proper function of the brain. Astrocytes, a major type of glial cell, are important regulators of synapse formation and function during development. While neurogenesis precedes astrogenesis in the cortex, neuronal synapses only begin to form after astrocytes have been generated, concurrent with neuronal branching and process elaboration. Here we provide a combined overview of the developmental processes of synapse and circuit formation in the rodent cortex, emphasizing the timeline of both neuronal and astrocytic development and maturation. We further discuss the role of astrocytes at the synapse, focusing on astrocyte-synapse contact and the role of synapse-related proteins in promoting formation of distinct cortical circuits.
Collapse
Affiliation(s)
- Isabella Farhy-Tselnicker
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA.
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
50
|
Lopez AY, Wang X, Xu M, Maheshwari A, Curry D, Lam S, Adesina AM, Noebels JL, Sun QQ, Cooper EC. Ankyrin-G isoform imbalance and interneuronopathy link epilepsy and bipolar disorder. Mol Psychiatry 2017; 22:1464-1472. [PMID: 27956739 PMCID: PMC5798616 DOI: 10.1038/mp.2016.233] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/04/2016] [Accepted: 10/31/2016] [Indexed: 12/12/2022]
Abstract
ANK3, encoding the adaptor protein Ankyrin-G (AnkG), has been implicated in bipolar disorder by genome-wide association studies. ANK3 has multiple alternative first exons, and a bipolar disorder-associated ANK3 variant has been shown to reduce the expression of exon 1b. Here we identify mechanisms through which reduced ANK3 exon 1b isoform expression disrupts neuronal excitation-inhibition balance. We find that parvalbumin (PV) interneurons and principal cells differentially express ANK3 first exon subtypes. PV interneurons express only isoforms containing exon 1b, whereas excitatory principal cells express exon 1e alone or both 1e and 1b. In transgenic mice deficient for exon 1b, PV interneurons lack voltage-gated sodium channels at their axonal initial segments and have increased firing thresholds and diminished action potential dynamic range. These mice exhibit an Ank3 gene dosage-dependent phenotype including behavior changes modeling bipolar disorder, epilepsy and sudden death. Thus ANK3's important association with human bipolar susceptibility may arise from imbalance between AnkG function in interneurons and principal cells and resultant excessive circuit sensitivity and output. AnkG isoform imbalance is a novel molecular endophenotype and potential therapeutic target.
Collapse
Affiliation(s)
- Angel Y. Lopez
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Xinjun Wang
- Department of Neuroscience, University of Wyoming, Laramie, WY, USA
| | - Mingxuan Xu
- Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Atul Maheshwari
- Neurology, Baylor College of Medicine, Houston, TX, USA,Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Daniel Curry
- Department of Neurosurgery, Texas Children’s Hospital, Houston, TX, USA
| | - Sandi Lam
- Department of Neurosurgery, Texas Children’s Hospital, Houston, TX, USA
| | | | - Jeffrey L. Noebels
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA,Neurology, Baylor College of Medicine, Houston, TX, USA,Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Qian-Quan Sun
- Department of Neuroscience, University of Wyoming, Laramie, WY, USA
| | - Edward C. Cooper
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA,Neurology, Baylor College of Medicine, Houston, TX, USA,Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|