1
|
Kheirkhah M, Hejazi NS, Nugent AC, Gilbert JR, Leistritz L, Walter M, Duncan WC, Goldman D, Zarate CA. Exploring the link between waking gamma and sleep delta power in healthy volunteers and individuals with treatment-resistant depression. J Affect Disord 2025; 385:119448. [PMID: 40398609 DOI: 10.1016/j.jad.2025.119448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/28/2025] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Sleep disruptions are a core feature of both major depressive disorder and treatment-resistant depression (TRD), which is defined by persistent symptoms despite multiple treatment efforts. In addition, disruptions in wakeful gamma power and sleep-related delta power have been observed in individuals with TRD. This study explored the association between gamma oscillations (30-100 Hz) occurring during wakefulness and delta power (0.5-4 Hz) in non-rapid eye movement (NREM) sleep-both of which have separately been implicated in plasticity-in healthy volunteers (HVs) and individuals with TRD. Specifically, the study explored whether a relationship exists between daytime wake gamma power and sleep NREM delta power in HVs and those with TRD. METHODS Brain activity was measured via electroencephalography (night-time) and magnetoencephalography (daytime resting state) in 23 HVs (9M/14F; 20-56 yrs) and 40 medication-free TRD participants (20 M/20F; 18-63 yrs). RESULTS In HVs, NREM episode (NREM1) sleep delta power correlated with daytime wake gamma power (r = 0.417; p = 0.04). This correlation was absent in TRD participants (r = 0.108; p = 0.50). LIMITATIONS The sample size of the HVs (n = 23) was smaller than the TRD participants (n = 40), and the measurement order for wake gamma power and sleep delta power varied. CONCLUSIONS These findings identify a possible link between daytime wake gamma power and NREM1 sleep delta power in HVs, supporting an association between gamma and delta power in sleep homeostasis. The lack of such a relationship in medication-free individuals with TRD suggests disrupted synaptic homeostasis that may contribute to impaired synaptic plasticity in TRD. Clinical Trials Identifiers: NCT00088699 and NCT01204918.
Collapse
Affiliation(s)
- Mina Kheirkhah
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA; Department of Psychiatry and Psychotherapy, Jena University Hospital, 07740 Jena, Germany
| | - Nadia S Hejazi
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Allison C Nugent
- Magnetoencephalography Core, National Institute of Mental Health, Bethesda, MD, USA.
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Lutz Leistritz
- Institute of Medical Statistics, Computer and Data Sciences, Jena University Hospital, 07740 Jena, Germany.
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, 07740 Jena, Germany.
| | - Wallace C Duncan
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - David Goldman
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
2
|
Madadi Asl M, Valizadeh A. Entrainment by transcranial alternating current stimulation: Insights from models of cortical oscillations and dynamical systems theory. Phys Life Rev 2025; 53:147-176. [PMID: 40106964 DOI: 10.1016/j.plrev.2025.03.008] [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: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Signature of neuronal oscillations can be found in nearly every brain function. However, abnormal oscillatory activity is linked with several brain disorders. Transcranial alternating current stimulation (tACS) is a non-invasive brain stimulation technique that can potentially modulate neuronal oscillations and influence behavior both in health and disease. Yet, a complete understanding of how interacting networks of neurons are affected by tACS remains elusive. Entrainment effects by which tACS synchronizes neuronal oscillations is one of the main hypothesized mechanisms, as evidenced in animals and humans. Computational models of cortical oscillations may shed light on the entrainment effects of tACS, but current modeling studies lack specific guidelines to inform experimental investigations. This study addresses the existing gap in understanding the mechanisms of tACS effects on rhythmogenesis within the brain by providing a comprehensive overview of both theoretical and experimental perspectives. We explore the intricate interactions between oscillators and periodic stimulation through the lens of dynamical systems theory. Subsequently, we present a synthesis of experimental findings that demonstrate the effects of tACS on both individual neurons and collective oscillatory patterns in animal models and humans. Our review extends to computational investigations that elucidate the interplay between tACS and neuronal dynamics across diverse cortical network models. To illustrate these concepts, we conclude with a simple oscillatory neuron model, showcasing how fundamental theories of oscillatory behavior derived from dynamical systems, such as phase response of neurons to external perturbation, can account for the entrainment effects observed with tACS. Studies reviewed here render the necessity of integrated experimental and computational approaches for effective neuromodulation by tACS in health and disease.
Collapse
Affiliation(s)
- Mojtaba Madadi Asl
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran; Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran, Iran.
| | - Alireza Valizadeh
- Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran, Iran; Department of Physics, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran; The Zapata-Briceño Institute of Neuroscience, Madrid, Spain
| |
Collapse
|
3
|
Saberi A, Wischnewski KJ, Jung K, Lotter LD, Schaare HL, Banaschewski T, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Papadopoulos Orfanos D, Lemaitre H, Poustka L, Hohmann S, Holz N, Baeuchl C, Smolka MN, Vaidya N, Walter H, Whelan R, Schumann G, IMAGEN Consortium, Paus T, Dukart J, Bernhardt BC, Popovych OV, Eickhoff SB, Valk SL. Adolescent maturation of cortical excitation-inhibition ratio based on individualized biophysical network modeling. SCIENCE ADVANCES 2025; 11:eadr8164. [PMID: 40465711 PMCID: PMC12136046 DOI: 10.1126/sciadv.adr8164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 04/25/2025] [Indexed: 06/11/2025]
Abstract
The excitation-inhibition ratio is a key functional property of cortical microcircuits which changes throughout an individual's lifespan. Adolescence is considered a critical period for maturation of excitation-inhibition ratio. This has primarily been observed in animal studies. However, there is limited human in vivo evidence for maturation of excitation-inhibition ratio at the individual level. Here, we developed an individualized in vivo marker of regional excitation-inhibition ratio in human adolescents, estimated using large-scale simulations of biophysical network models fitted to resting-state functional imaging data from both cross-sectional (n = 752) and longitudinal (n = 149) cohorts. In both datasets, we found a widespread decrease in excitation-inhibition ratio in association areas, paralleled by an increase or lack of change in sensorimotor areas. This developmental pattern was aligned with multiscale markers of sensorimotor-association differentiation. Although our main findings were robust across alternative modeling configurations, we observed local variations, highlighting the importance of methodological choices for future studies.
Collapse
Affiliation(s)
- Amin Saberi
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Kevin J. Wischnewski
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Mathematics, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Kyesam Jung
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Leon D. Lotter
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Max Planck School of Cognition, Stephanstrasse 1A, 04103 Leipzig, Germany
| | - H. Lina Schaare
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Gareth J. Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Arun L. W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, 68131 Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, Burlington, VT 05405, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, UK
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- German Center for Mental Health (DZPG), site Berlin-Potsdam, Berlin, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 “Trajectoires développementales en psychiatrie”, Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 “Trajectoires développementales en psychiatrie”, Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
- Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, AP-HP Sorbonne Université, Paris, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U1299 “Trajectoires développementales en psychiatrie”, Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli, Gif-sur-Yvette, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | | | - Herve Lemaitre
- NeuroSpin, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, 33076 Bordeaux, France
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry, Center for Psychosocial Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Nathalie Holz
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Christian Baeuchl
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Michael N. Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- German Center for Mental Health (DZPG), site Berlin-Potsdam, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- German Centre for Mental Health, Berlin, Germany
| | | | - Tomáš Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Juergen Dukart
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Boris C. Bernhardt
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Oleksandr V. Popovych
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Simon B. Eickhoff
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sofie L. Valk
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
4
|
van Bree S, Levenstein D, Krause MR, Voytek B, Gao R. Processes and measurements: a framework for understanding neural oscillations in field potentials. Trends Cogn Sci 2025; 29:448-466. [PMID: 39753446 DOI: 10.1016/j.tics.2024.12.003] [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: 07/17/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 05/09/2025]
Abstract
Various neuroscientific theories maintain that brain oscillations are important for neuronal computation, but opposing views claim that these macroscale dynamics are 'exhaust fumes' of more relevant processes. Here, we approach the question of whether oscillations are functional or epiphenomenal by distinguishing between measurements and processes, and by reviewing whether causal or inferentially useful links exist between field potentials, electric fields, and neurobiological events. We introduce a vocabulary for the role of brain signals and their underlying processes, demarcating oscillations as a distinct entity where both processes and measurements can exhibit periodicity. Leveraging this distinction, we suggest that electric fields, oscillating or not, are causally and computationally relevant, and that field potential signals can carry information even without causality.
Collapse
Affiliation(s)
- Sander van Bree
- Department of Medicine, Justus Liebig University, Giessen, Germany; Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; Centre for Cognitive Neuroimaging, School of Psychology and Neuroscience, University of Glasgow, Glasgow, UK
| | - Daniel Levenstein
- MILA - Quebec AI Institute, Montreal, QC, Canada; Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Matthew R Krause
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Bradley Voytek
- Department of Cognitive Science, Halıcıŏglu Data Science Institute, Kavli Institute for Brain & Mind, University of California, San Diego, La Jolla, CA, USA
| | - Richard Gao
- Machine Learning in Science, Excellence Cluster Machine Learning and Tübingen AI Center, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Lesch KP, Gorbunov N. Antisocial personality disorder:Failure to balance excitation/inhibition? Neuropharmacology 2025; 268:110321. [PMID: 39855295 DOI: 10.1016/j.neuropharm.2025.110321] [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: 08/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
While healthy brain function relies on a dynamic but tightly regulated interaction between excitation (E) and inhibition (I), a spectrum of social cognition disorders, including antisocial behavior and antisocial personality disorder (ASPD), frequently ensuing from irregular neurodevelopment, may be associated with E/I imbalance and concomitant alterations in neural connectivity. Technological advances in the evaluation of structural and functional E/I balance proxies in clinical settings and in human cell culture models provide a general basis for identification of biomarkers providing a powerful concept for prevention and intervention across different dimensions of mental health and disease. In this perspective we outline a framework for research to characterize neurodevelopmental pathways to antisocial behavior and ASPD driven by (epi)genetic factors across life, and to identify molecular targets for preventing the detrimental effects of cognitive dysfunction and maladaptive social behavior, considering psychosocial experience; to validate signatures of E/I imbalance and altered myelination proxies as biomarkers of pathogenic neural circuitry mechanisms to determine etiological processes in the transition from mental health to antisocial behavior and ASPD and in the switch from prevention to treatment; to develop a neurobiologically-grounded integrative model of antisocial behavior and ASPD resultant of disrupted E/I balance, allowing to establish objective diagnoses and monitoring tools, to personalize prevention and therapeutic decisions, to predict treatment response, and thus counteract relapse; and finally, to promote transformation of dimensional disorder taxonomy and to enhance societal awareness and reception of the neurobiological basis of antisocial behavior and ASPD.
Collapse
Affiliation(s)
- Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Child- and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
| | - Nikita Gorbunov
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany; Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
6
|
Bos H, Miehl C, Oswald AMM, Doiron B. Untangling stability and gain modulation in cortical circuits with multiple interneuron classes. eLife 2025; 13:RP99808. [PMID: 40304591 PMCID: PMC12043317 DOI: 10.7554/elife.99808] [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] [Indexed: 05/02/2025] Open
Abstract
Synaptic inhibition is the mechanistic backbone of a suite of cortical functions, not the least of which are maintaining network stability and modulating neuronal gain. In cortical models with a single inhibitory neuron class, network stabilization and gain control work in opposition to one another - meaning high gain coincides with low stability and vice versa. It is now clear that cortical inhibition is diverse, with molecularly distinguished cell classes having distinct positions within the cortical circuit. We analyze circuit models with pyramidal neurons (E) as well as parvalbumin (PV) and somatostatin (SOM) expressing interneurons. We show how, in E - PV - SOM recurrently connected networks, SOM-mediated modulation can lead to simultaneous increases in neuronal gain and network stability. Our work exposes how the impact of a modulation mediated by SOM neurons depends critically on circuit connectivity and the network state.
Collapse
Affiliation(s)
- Hannah Bos
- Department of Mathematics, University of PittsburghPittsburghUnited States
| | - Christoph Miehl
- Department of Neurobiology, University of ChicagoChicagoUnited States
- Grossman Center for Quantitative Biology and Human Behavior, University of ChicagoChicagoUnited States
| | - Anne-Marie Michelle Oswald
- Department of Neurobiology, University of ChicagoChicagoUnited States
- Grossman Center for Quantitative Biology and Human Behavior, University of ChicagoChicagoUnited States
| | - Brent Doiron
- Department of Mathematics, University of PittsburghPittsburghUnited States
- Department of Neurobiology, University of ChicagoChicagoUnited States
- Grossman Center for Quantitative Biology and Human Behavior, University of ChicagoChicagoUnited States
- Department of Neuroscience, University of PittsburghPittsburghUnited States
- Department of Statistics, University of ChicagoChicagoUnited States
| |
Collapse
|
7
|
Fournier Z, Alonso LM, Marder E. Rhythmic circuit function is more robust to changes in synaptic than intrinsic conductances. eLife 2025; 13:RP102938. [PMID: 40293432 PMCID: PMC12037179 DOI: 10.7554/elife.102938] [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] [Indexed: 04/30/2025] Open
Abstract
Circuit function results from both intrinsic conductances of network neurons and the synaptic conductances that connect them. In models of neural circuits, different combinations of maximal conductances can give rise to similar activity. We compared the robustness of a neural circuit to changes in their intrinsic versus synaptic conductances. To address this, we performed a sensitivity analysis on a population of conductance-based models of the pyloric network from the crustacean stomatogastric ganglion (STG). The model network consists of three neurons with nine currents: a sodium current (Na), three potassium currents (Kd, KCa, KA), two calcium currents (CaS and CaT), a hyperpolarization-activated current (H), a non-voltage-gated leak current (leak), and a neuromodulatory current (MI). The model cells are connected by seven synapses of two types, glutamatergic and cholinergic. We produced one hundred models of the pyloric network that displayed similar activities with values of maximal conductances distributed over wide ranges. We evaluated the robustness of each model to changes in their maximal conductances. We found that individual models have different sensitivities to changes in their maximal conductances, both in their intrinsic and synaptic conductances. As expected, the models become less robust as the extent of the changes increases. Despite quantitative differences in their robustness, we found that in all cases, the model networks are more sensitive to the perturbation of their intrinsic conductances than their synaptic conductances.
Collapse
Affiliation(s)
- Zachary Fournier
- Volen Center and Biology Department, Brandeis UniversityWalthamUnited States
| | - Leandro M Alonso
- Volen Center and Biology Department, Brandeis UniversityWalthamUnited States
| | - Eve Marder
- Volen Center and Biology Department, Brandeis UniversityWalthamUnited States
| |
Collapse
|
8
|
Liang X, Ma Y, Wang R, Wu H, Liu C, Cao F, An N, Xiang M, Zhai Y, Ning X. An Exploration on Aperiodic Activities and Transient Oscillations During Semantic Processing: A Study With Wearable MEG. IEEE Trans Neural Syst Rehabil Eng 2025; 33:1473-1485. [PMID: 40238608 DOI: 10.1109/tnsre.2025.3561356] [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: 04/18/2025]
Abstract
The processing of semantic information is pivotal in language cognition. However, there is a scarcity of research exploring the semantic-related patterns associated with aperiodic and transient periodic brain activities. In this study, recently developed algorithms were employed to parameterize the time-frequency characteristics of neural activities captured with optically pumped magnetometers-based wearable Magnetoencephalography from participants engaged in a Chinese semantic-based task. This study elucidated the neural mechanisms during semantic processing, in relation to transient oscillations and aperiodic activity. Additionally, the results demonstrated that these parameterized features could serve as indicators for decoding semantics. These findings may offer novel contribution to analyzing the mechanism of semantic perception, which will be potential to rehabilitation of language disorders with OPM-MEG.
Collapse
|
9
|
Papatheodoropoulos C. Compensatory Regulation of Excitation/Inhibition Balance in the Ventral Hippocampus: Insights from Fragile X Syndrome. BIOLOGY 2025; 14:363. [PMID: 40282228 PMCID: PMC12025323 DOI: 10.3390/biology14040363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
The excitation/inhibition (E/I) balance is a critical feature of neural circuits, which is crucial for maintaining optimal brain function by ensuring network stability and preventing neural hyperexcitability. The hippocampus exhibits the particularly interesting characteristics of having different functions and E/I profiles between its dorsal and ventral segments. Furthermore, the hippocampus is particularly vulnerable to epilepsy and implicated in Fragile X Syndrome (FXS), disorders associated with heightened E/I balance and possible deficits in GABA-mediated inhibition. In epilepsy, the ventral hippocampus shows heightened susceptibility to seizures, while in FXS, recent evidence suggests differential alterations in excitability and inhibition between dorsal and ventral regions. This article explores the mechanisms underlying E/I balance regulation, focusing on the hippocampus in epilepsy and FXS, and emphasizing the possible mechanisms that may confer homeostatic flexibility to the ventral hippocampus in maintaining E/I balance. Notably, the ventral hippocampus in adult FXS models shows enhanced GABAergic inhibition, resistance to epileptiform activity, and physiological network pattern (sharp wave-ripples, SWRs), potentially representing a homeostatic adaptation. In contrast, the dorsal hippocampus in these FXS models is more vulnerable to aberrant discharges and displays altered SWRs. These findings highlight the complex, region-specific nature of E/I balance disruptions in neurological disorders and suggest that the ventral hippocampus may possess unique compensatory mechanisms. Specifically, it is proposed that the ventral hippocampus, the brain region most prone to hyperexcitability, may have unique adaptive capabilities at the cellular and network levels that maintain the E/I balance within a normal range to prevent the transition to hyperexcitability and preserve normal function. Investigating the mechanisms underlying these compensatory responses in the ventral hippocampus and their developmental trajectories may offer novel insights into strategies for mitigating E/I imbalances in epilepsy, FXS, and potentially other neuropsychiatric and neurodevelopmental disorders.
Collapse
|
10
|
Onorato I, Tzanou A, Schneider M, Uran C, Broggini AC, Vinck M. Distinct roles of PV and Sst interneurons in visually induced gamma oscillations. Cell Rep 2025; 44:115385. [PMID: 40048428 DOI: 10.1016/j.celrep.2025.115385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/26/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
Gamma-frequency oscillations are a hallmark of active information processing and are generated by interactions between excitatory and inhibitory neurons. To examine the contribution of distinct inhibitory interneurons to visually induced gamma oscillations, we recorded from optogenetically identified PV+ (parvalbumin) and Sst+ (somatostatin) interneurons in mouse primary visual cortex (V1). PV and Sst inhibitory interneurons exhibited distinct correlations to gamma oscillations. PV cells were strongly phase locked, while Sst cells were weakly phase locked, except for narrow-waveform Sst cells. PV cells fired at a substantially earlier phase in the gamma cycle (≈6 ms) than Sst cells. PV cells fired shortly after the onset of tightly synchronized burst events in excitatory cells, while Sst interneurons fired after subsequent burst spikes or single spikes. These findings indicate a main role of PV interneurons in synchronizing network activity and suggest that PV and Sst interneurons control the excitability of somatic and dendritic neural compartments with precise time delays coordinated by gamma oscillations.
Collapse
Affiliation(s)
- Irene Onorato
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany; Max Planck Institute for Brain Research, 60438 Frankfurt, Germany; Neuroscience Research Center, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.
| | - Athanasia Tzanou
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany
| | - Marius Schneider
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany; Max Planck Institute for Brain Research, 60438 Frankfurt, Germany
| | - Cem Uran
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany; Donders Centre for Neuroscience, Department of Neurophysics, Radboud University Nijmegen, 6525 Nijmegen, the Netherlands; Max Planck Institute for Brain Research, 60438 Frankfurt, Germany
| | - Ana Clara Broggini
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany
| | - Martin Vinck
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt am Main, Germany; Donders Centre for Neuroscience, Department of Neurophysics, Radboud University Nijmegen, 6525 Nijmegen, the Netherlands.
| |
Collapse
|
11
|
Silverstein BH, Kolbman N, Nelson A, Liu T, Guzzo P, Gilligan J, Lee U, Mashour GA, Vanini G, Pal D. Intravenous psilocybin induces dose-dependent changes in functional network organization in rat cortex. Transl Psychiatry 2025; 15:93. [PMID: 40128190 PMCID: PMC11933319 DOI: 10.1038/s41398-025-03308-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 01/23/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
Psilocybin produces an altered state of consciousness in humans and is associated with complex spatiotemporal changes in cortical networks. Given the emphasis on rodent models for mechanistic studies, there is a need for characterization of the effect of psilocybin on cortex-wide network dynamics. Previous electroencephalographic studies of psychedelics in rodents have primarily used sparse electrode arrays with limited spatial resolution, precluding network level analysis, and have been restricted to lower gamma frequencies. Therefore, in this study, we used electroencephalographic recordings from 27 sites/electrodes across rat cortex (n = 6 male, 6 female) to characterize the effect of psilocybin (0.1, 1, and 10 mg/kg delivered over an hour) on brain network organization as inferred through changes in node degree (an index of network density) and connection strength (via weighted phase-lag index). The removal of aperiodic component from the electroencephalogram localized the primary oscillatory changes to theta (4-10 Hz), medium gamma (70-110 Hz), and high gamma (110-150 Hz) bands, which were used for the network analysis. Additionally, we determined the concurrent changes in theta-gamma phase-amplitude coupling. We report that psilocybin, in a dose-dependent manner, 1) disrupted theta-gamma coupling [p < 0.05], 2) increased frontal high gamma connectivity [p < 0.05] and posterior theta connectivity [p ≤ 0.049], and 3) increased frontal high gamma [p < 0.05] and posterior theta [p ≤ 0.046] network density. The behavioral activity and the medium gamma frontoparietal connectivity showed an inverted-U relationship with psilocybin dose. Our results suggest that high-frequency network organization, decoupled from local theta-phase, may be an important signature of psilocybin-induced non-ordinary state of consciousness.
Collapse
Affiliation(s)
- Brian H Silverstein
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas Kolbman
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Amanda Nelson
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Tiecheng Liu
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - UnCheol Lee
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI, USA
| | - George A Mashour
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Giancarlo Vanini
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Dinesh Pal
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA.
- Center for Consciousness Science, University of Michigan, Ann Arbor, MI, USA.
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI, USA.
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Ryu J, Kao JC, Bari A. Spontaneous pain dynamics characterized by stochasticity in neural recordings of awake humans with chronic pain. Pain 2025:00006396-990000000-00862. [PMID: 40112191 DOI: 10.1097/j.pain.0000000000003592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025]
Abstract
ABSTRACT Chronic pain is characterized by spontaneous fluctuations in pain intensity, a phenomenon that remains poorly understood. The aim of this study is to elucidate the neural mechanisms underlying pain fluctuations in patients with chronic pain undergoing deep brain stimulation surgery. We recorded local field potentials (LFPs) from pain-processing hub structures, including the ventral posteromedial nucleus of the thalamus, subgenual cingulate cortex, and periventricular and periaqueductal gray, while patients continuously reported their pain levels. Using novel auto-mutual information metrics to analyze LFP stochastic patterns, we found that pain intensity correlated with both increased regularity of spike-like events and greater past-dependency of neural oscillations in the 4- to 15-Hz frequency band. In addition, during periods of higher pain states, we observed enhanced functional connectivity between the examined hub structures and the prefrontal cortex, suggesting a more focused flow of pain-related information within the pain circuit. By characterizing the dynamic nature of pain fluctuations, this study bridges the gap in understanding moment-to-moment pain variations and their underlying neural mechanisms, paving the way for improved chronic pain management strategies.
Collapse
Affiliation(s)
- Jihye Ryu
- Department of Neurosurgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| | - Jonathan C Kao
- Department of Electrical and Computer Engineering, University of California Los Angeles, Los Angeles, CA, United States
| | - Ausaf Bari
- Department of Neurosurgery, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
13
|
Kim SH, Choi H. Inhibitory cell type heterogeneity in a spatially structured mean-field model of V1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643046. [PMID: 40161661 PMCID: PMC11952513 DOI: 10.1101/2025.03.13.643046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Inhibitory interneurons in the cortex are classified into cell types differing in their morphology, electrophysiology, and connectivity. Although it is known that parvalbumin (PV), somatostatin (SST), and vasoactive intestinal polypeptide-expressing neurons (VIP), the major inhibitory neuron subtypes in the cortex, have distinct modulatory effects on excitatory neurons, how heterogeneous spatial connectivity properties relate to network computations is not well understood. Here, we study the implications of heterogeneous inhibitory neurons on the dynamics and computations of spatially-structured neural networks. We develop a mean-field model of the system in order to systematically examine excitation-inhibition balance, dynamical stability, and cell-type specific gain modulations. The model incorporates three inhibitory cell types and excitatory neurons with distinct connectivity probabilities and recent evidence of long-range spatial projections of SST neurons. Position-dependent firing rate predictions are validated against simulations, and balanced solutions under Gaussian assumptions are derived from scaling arguments. Stability analysis shows that while long-range inhibitory projections in E-I circuits with a homogeneous inhibitory population result in instability, the heterogeneous network maintains stability with long-range SST projections. This suggests that a mixture of short and long-range inhibitions may be key to providing diverse computations while maintaining stability. We further find that conductance-based synaptic transmissions are necessary to reproduce experimentally observed cell-type-specific gain modulations of inhibition by PV and SST neurons. The mechanisms underlying cell-type-specific gain changes are elucidated using linear response theory. Our theoretical approach offers insight into the computational function of cell-type-specific and distance-dependent network structure.
Collapse
Affiliation(s)
- Soon Ho Kim
- School of Mathematics, Georgia Institute of Technology, Atlanta, GA 30332-0160
| | - Hannah Choi
- School of Mathematics, Georgia Institute of Technology, Atlanta, GA 30332-0160
| |
Collapse
|
14
|
Qu T. The effects of amyloidosis and aging on glutamatergic and GABAergic synapses, and interneurons in the barrel cortex and non-neocortical brain regions. Front Neuroanat 2025; 19:1526962. [PMID: 40012738 PMCID: PMC11863279 DOI: 10.3389/fnana.2025.1526962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Previous studies on changes in the distribution of GABAergic interneurons and excitation/inhibition (E/I) balance in Alzheimer's disease (AD) and aging were mainly conducted in the neocortex and hippocampus. However, the limbic system is the primary and crucial location for AD progression. Therefore, in this study, we utilized AD and aging mouse models to investigate the E/I balance and the distribution of parvalbumin (PV)- and somatostatin (SST)-expressing cells in S1BF (barrel field of primary somatosensory cortex, barrel cortex), CA1 hippocampal area and brain regions beyond the neocortex and hippocampus, including retrosplenial cortex (RSC, which is composed of RSG and RSA), piriform cortex (Pir), amygdala (BMA), and hypothalamus (DM). We discovered that amyloidosis may disrupt the alignment of excitatory pre- and postsynaptic quantities. Amyloidosis reduces the quantity of synapses and SST cells, but does not impact the counts of PV cells. By contrast, aging is linked to a decline in synapses, I/E ratios, SST and PV cells. Amyloidosis affects the S1BF and BMA, while aging may harm all studied regions, including the S1BF, RSC, hippocampus, Pir, BMA, and DM. Aging mostly affects synapses and I/E ratios in Pir, BMA, and DM, and PV and SST interneurons in the hippocampus.
Collapse
Affiliation(s)
- Tao Qu
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
15
|
Fournier Z, Alonso LM, Marder E. Rhythmic circuit function is more robust to changes in synaptic than intrinsic conductances. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.03.611139. [PMID: 39282429 PMCID: PMC11398330 DOI: 10.1101/2024.09.03.611139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Circuit function results from both intrinsic conductances of network neurons and the synaptic conductances that connect them. In models of neural circuits, different combinations of maximal conductances can give rise to similar activity. We compared the robustness of a neural circuit to changes in their intrinsic versus synaptic conductances. To address this, we performed a sensitivity analysis on a population of conductance-based models of the pyloric network from the crustacean stomatogastric ganglion (STG). The model network consists of three neurons with nine currents: a sodium current (Na), three potassium currents (Kd, KCa, A-type), two calcium currents (CaS and CaT), a hyperpolarization-activated current (H), a non-voltage-gated leak current (leak), and a neuromodulatory current (MI). The model cells are connected by seven synapses of two types, glutamatergic and cholinergic. We produced one hundred models of the pyloric network that displayed similar activities with values of maximal conductances distributed over wide ranges. We evaluated the robustness of each model to changes in their maximal conductances. We found that individual models have different sensitivities to changes in their maximal conductances, both in their intrinsic and synaptic conductances. As expected the models become less robust as the extent of the changes increase. Despite quantitative differences in their robustness, we found that in all cases, the model networks are more sensitive to the perturbation of their intrinsic conductances than their synaptic conductances.
Collapse
Affiliation(s)
| | - Leandro M. Alonso
- Volen Center and Biology Department, Brandeis University, Waltham, MA, 02454, USA
| | - Eve Marder
- Volen Center and Biology Department, Brandeis University, Waltham, MA, 02454, USA
| |
Collapse
|
16
|
Singh MF, Braver TS, Cole M, Ching S. Precision data-driven modeling of cortical dynamics reveals person-specific mechanisms underpinning brain electrophysiology. Proc Natl Acad Sci U S A 2025; 122:e2409577121. [PMID: 39823302 PMCID: PMC11761305 DOI: 10.1073/pnas.2409577121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 11/02/2024] [Indexed: 01/19/2025] Open
Abstract
Task-free brain activity affords unique insight into the functional structure of brain network dynamics and has been used to identify neural markers of individual differences. In this work, we present an algorithmic optimization framework that directly inverts and parameterizes brain-wide dynamical-systems models involving hundreds of interacting neural populations, from single-subject M/EEG time-series recordings. This technique provides a powerful neurocomputational tool for interrogating mechanisms underlying individual brain dynamics ("precision brain models") and making quantitative predictions. We extensively validate the models' performance in forecasting future brain activity and predicting individual variability in key M/EEG metrics. Last, we demonstrate the power of our technique in resolving individual differences in the generation of alpha and beta-frequency oscillations. We characterize subjects based upon model attractor topology and a dynamical-systems mechanism by which these topologies generate individual variation in the expression of alpha vs. beta rhythms. We trace these phenomena back to global variation in excitatory-inhibitory balance, highlighting the explanatory power of our framework to generate mechanistic insights.
Collapse
Affiliation(s)
- Matthew F. Singh
- Department of Statistics, University of Illinois, Urbana-Champaign, Champaign, IL61820
- Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Champaign, IL61801
- Department of Psychology, University of Illinois, Urbana-Champaign, Champaign, IL61820
- Department of Electrical & Systems Engineering, Washington University in St. Louis, St. Louis, MO63130
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO63130
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ07102
| | - Todd S. Braver
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO63130
| | - Michael Cole
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ07102
| | - ShiNung Ching
- Department of Electrical & Systems Engineering, Washington University in St. Louis, St. Louis, MO63130
| |
Collapse
|
17
|
Schuessler F, Mastrogiuseppe F, Ostojic S, Barak O. Aligned and oblique dynamics in recurrent neural networks. eLife 2024; 13:RP93060. [PMID: 39601404 DOI: 10.7554/elife.93060] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
The relation between neural activity and behaviorally relevant variables is at the heart of neuroscience research. When strong, this relation is termed a neural representation. There is increasing evidence, however, for partial dissociations between activity in an area and relevant external variables. While many explanations have been proposed, a theoretical framework for the relationship between external and internal variables is lacking. Here, we utilize recurrent neural networks (RNNs) to explore the question of when and how neural dynamics and the network's output are related from a geometrical point of view. We find that training RNNs can lead to two dynamical regimes: dynamics can either be aligned with the directions that generate output variables, or oblique to them. We show that the choice of readout weight magnitude before training can serve as a control knob between the regimes, similar to recent findings in feedforward networks. These regimes are functionally distinct. Oblique networks are more heterogeneous and suppress noise in their output directions. They are furthermore more robust to perturbations along the output directions. Crucially, the oblique regime is specific to recurrent (but not feedforward) networks, arising from dynamical stability considerations. Finally, we show that tendencies toward the aligned or the oblique regime can be dissociated in neural recordings. Altogether, our results open a new perspective for interpreting neural activity by relating network dynamics and their output.
Collapse
Affiliation(s)
- Friedrich Schuessler
- Faculty of Electrical Engineering and Computer Science, Technical University of Berlin, Berlin, Germany
- Science of Intelligence, Research Cluster of Excellence, Berlin, Germany
| | | | - Srdjan Ostojic
- Laboratoire de Neurosciences Cognitives et Computationnelles, INSERM U960, Ecole Normale Superieure-PSL Research University, Paris, France
| | - Omri Barak
- Rappaport Faculty of Medicine and Network Biology Research Laboratories, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
18
|
Masharipov R, Knyazeva I, Korotkov A, Cherednichenko D, Kireev M. Comparison of whole-brain task-modulated functional connectivity methods for fMRI task connectomics. Commun Biol 2024; 7:1402. [PMID: 39462101 PMCID: PMC11513045 DOI: 10.1038/s42003-024-07088-3] [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: 02/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Higher brain functions require flexible integration of information across widely distributed brain regions depending on the task context. Resting-state functional magnetic resonance imaging (fMRI) has provided substantial insight into large-scale intrinsic brain network organisation, yet the principles of rapid context-dependent reconfiguration of that intrinsic network organisation are much less understood. A major challenge for task connectome mapping is the absence of a gold standard for deriving whole-brain task-modulated functional connectivity matrices. Here, we perform biophysically realistic simulations to control the ground-truth task-modulated functional connectivity over a wide range of experimental settings. We reveal the best-performing methods for different types of task designs and their fundamental limitations. Importantly, we demonstrate that rapid (100 ms) modulations of oscillatory neuronal synchronisation can be recovered from sluggish haemodynamic fluctuations even at typically low fMRI temporal resolution (2 s). Finally, we provide practical recommendations on task design and statistical analysis to foster task connectome mapping.
Collapse
Affiliation(s)
- Ruslan Masharipov
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia.
| | - Irina Knyazeva
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| | - Alexander Korotkov
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| | - Denis Cherednichenko
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| | - Maxim Kireev
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
19
|
Masharipov R, Knyazeva I, Korotkov A, Cherednichenko D, Kireev M. Comparison of whole-brain task-modulated functional connectivity methods for fMRI task connectomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576622. [PMID: 39464064 PMCID: PMC11507666 DOI: 10.1101/2024.01.22.576622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Higher brain functions require flexible integration of information across widely distributed brain regions depending on the task context. Resting-state functional magnetic resonance imaging (fMRI) has provided substantial insight into large-scale intrinsic brain network organisation, yet the principles of rapid context-dependent reconfiguration of that intrinsic network organisation are much less understood. A major challenge for task connectome mapping is the absence of a gold standard for deriving whole-brain task-modulated functional connectivity matrices. Here, we perform biophysically realistic simulations to control the ground-truth task-modulated functional connectivity over a wide range of experimental settings. We reveal the best-performing methods for different types of task designs and their fundamental limitations. Importantly, we demonstrate that rapid (100 ms) modulations of oscillatory neuronal synchronisation can be recovered from sluggish haemodynamic fluctuations even at typically low fMRI temporal resolution (2 s). Finally, we provide practical recommendations on task design and statistical analysis to foster task connectome mapping.
Collapse
Affiliation(s)
- Ruslan Masharipov
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| | - Irina Knyazeva
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| | - Alexander Korotkov
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| | - Denis Cherednichenko
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| | - Maxim Kireev
- N.P. Bechtereva Institute of the Human Brain, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
20
|
Mockevičius A, Voicikas A, Jurkuvėnas V, Tarailis P, Griškova-Bulanova I. Individualized EEG-Based Neurofeedback Targeting Auditory Steady-State Responses: A Proof-of-Concept Study. Appl Psychophysiol Biofeedback 2024:10.1007/s10484-024-09662-1. [PMID: 39183248 DOI: 10.1007/s10484-024-09662-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
Gamma-band (> 30 Hz) brain oscillatory activity is linked with sensory and cognitive processes and exhibits abnormalities in neuropsychiatric disorders. Therefore, neuromodulation techniques targeting gamma activity are being developed. One promising approach is neurofeedback (NFB) which is based on the alteration of brain responses via online feedback. However, the existing gamma-based NFB systems lack individualized approach. In the present work, we developed and tested an individualized EEG-NFB system. 46 healthy volunteers participated in three sessions on separate days. Before NFB training, individual gamma frequency (IGF) was estimated using chirp-modulated auditory stimulation (30-60 Hz). Participants were subjected to IGF-increase (if IGF was ≤ 45 Hz) or IGF-decrease conditions (if IGF was > 45 Hz). Gamma-band responses were targeted during NFB training, in which participants received auditory steady-state stimulation at frequency slightly above or below IGF and were instructed to try to increase their response while receiving real-time visual feedback. Each time a pre-defined response goal was reached, stimulation frequency was either increased or decreased. After training, IGF was reassessed. Experimental group participants were divided into equal groups based on the median success rate during NFB training. The results showed that high-responders had a significantly higher IGF modulation compared to control group, while low-responders did not differ from controls. No differences in IGF modulation were found between sessions and between NFB repetitions in all participant groups. The initial evaluation of the proposed EEG-NFB system showed potential to modulate IGF. Future studies could investigate longer-lasting electrophysiological and behavioural effects of the application of ASSR/IGF-based NFB system in clinical populations.
Collapse
|
21
|
Süzen E, Şavklıyıldız A, Özkan Ö, Çolak ÖH, Apaydın Doğan E, Özkan Ö, Şimşek B, Uluşar ÜD, Carlak HF, Polat Ö, Uysal H. Delta waves as a sign of cortical plasticity after full-face transplantation. Sci Rep 2024; 14:16454. [PMID: 39014053 PMCID: PMC11252439 DOI: 10.1038/s41598-024-67469-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/11/2024] [Indexed: 07/18/2024] Open
Abstract
This study focused on detecting the reflections of healing and change in cortex activation in full-face transplantation and lesions patients on EEG activity. Face transplant patients have facial lesions before transplantation and, to identify pre-face transplant patients' brain activity in the absence of pre-transplant recordings, we used data obtained from pre-transplant facial lesion patients. Ten healthy, four facial lesion and three full-face transplant patients participated in this study. EEG data recorded for four different sensory stimuli (brush from the right face, right hand, left face, and left-hand regions) were analyzed using wavelet packet transform method. EEG waves were analyzed for standard bands. Our findings indicate significant change in the 2-4 Hz frequency range which may be a result of ongoing or previous cortical reorganization for face lesion and transplant patients. Alterations of the delta wave seen in patients with facial lesion and face transplant can also be explained by the intense central plasticity. Our findings show that the delta band differences might be used as a marker in the evaluation of post-transplant cortical plasticity in the future.
Collapse
Affiliation(s)
- Esra Süzen
- Faculty of Engineering, Department of Electrical and Electronics Engineering, Akdeniz University, Pınarbasi Blvd., Antalya, Turkey
| | - Ayhan Şavklıyıldız
- Faculty of Engineering, Department of Electrical and Electronics Engineering, Akdeniz University, Pınarbasi Blvd., Antalya, Turkey
| | - Ömer Özkan
- Faculty of Medicine, Department of Plastic and Reconstructive Surgery, Akdeniz University, Antalya, Turkey
| | - Ömer Halil Çolak
- Faculty of Engineering, Department of Electrical and Electronics Engineering, Akdeniz University, Pınarbasi Blvd., Antalya, Turkey.
| | - Ebru Apaydın Doğan
- Faculty of Medicine, Department of Neurology, Akdeniz University, Antalya, Turkey
| | - Özlenen Özkan
- Faculty of Medicine, Department of Plastic and Reconstructive Surgery, Akdeniz University, Antalya, Turkey
| | - Buket Şimşek
- Faculty of Engineering, Department of Electrical and Electronics Engineering, Akdeniz University, Pınarbasi Blvd., Antalya, Turkey
| | - Ümit Deniz Uluşar
- Faculty of Engineering, Department of Computer Engineering, Akdeniz University, Antalya, Turkey
| | - Hamza Feza Carlak
- Faculty of Engineering, Department of Electrical and Electronics Engineering, Akdeniz University, Pınarbasi Blvd., Antalya, Turkey
| | - Övünç Polat
- Faculty of Engineering, Department of Electrical and Electronics Engineering, Akdeniz University, Pınarbasi Blvd., Antalya, Turkey
| | - Hilmi Uysal
- Faculty of Medicine, Department of Neurology, Akdeniz University, Antalya, Turkey
| |
Collapse
|
22
|
Saberi A, Wischnewski KJ, Jung K, Lotter LD, Schaare HL, Banaschewski T, Barker GJ, Bokde ALW, Desrivières S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Brühl R, Martinot JL, Martinot MLP, Artiges E, Nees F, Orfanos DP, Lemaitre H, Poustka L, Hohmann S, Holz N, Baeuchl C, Smolka MN, Vaidya N, Walter H, Whelan R, Schumann G, IMAGEN Consortium, Paus T, Dukart J, Bernhardt BC, Popovych OV, Eickhoff SB, Valk SL. Adolescent maturation of cortical excitation-inhibition balance based on individualized biophysical network modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599509. [PMID: 38948771 PMCID: PMC11213014 DOI: 10.1101/2024.06.18.599509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The balance of excitation and inhibition is a key functional property of cortical microcircuits which changes through the lifespan. Adolescence is considered a crucial period for the maturation of excitation-inhibition balance. This has been primarily observed in animal studies, yet human in vivo evidence on adolescent maturation of the excitation-inhibition balance at the individual level is limited. Here, we developed an individualized in vivo marker of regional excitation-inhibition balance in human adolescents, estimated using large-scale simulations of biophysical network models fitted to resting-state functional magnetic resonance imaging data from two independent cross-sectional (N = 752) and longitudinal (N = 149) cohorts. We found a widespread relative increase of inhibition in association cortices paralleled by a relative age-related increase of excitation, or lack of change, in sensorimotor areas across both datasets. This developmental pattern co-aligned with multiscale markers of sensorimotor-association differentiation. The spatial pattern of excitation-inhibition development in adolescence was robust to inter-individual variability of structural connectomes and modeling configurations. Notably, we found that alternative simulation-based markers of excitation-inhibition balance show a variable sensitivity to maturational change. Taken together, our study highlights an increase of inhibition during adolescence in association areas using cross sectional and longitudinal data, and provides a robust computational framework to estimate microcircuit maturation in vivo at the individual level.
Collapse
Affiliation(s)
- Amin Saberi
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Kevin J Wischnewski
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Mathematics, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Dusseldorf, Germany
| | - Kyesam Jung
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Leon D Lotter
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Max Planck School of Cognition, Stephanstrasse 1A, 04103 Leipzig, Germany
| | - H Lina Schaare
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Tobias Banaschewski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom
| | - Arun L W Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sylvane Desrivières
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom
| | - Herta Flor
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Department of Psychology, School of Social Sciences, University of Mannheim, 68131 Mannheim, Germany
| | - Antoine Grigis
- NeuroSpin, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Hugh Garavan
- Departments of Psychiatry and Psychology, University of Vermont, 05405 Burlington, Vermont, USA
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre School of Physics and Astronomy, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Center for Mental Health (DZPG), site Berlin-Potsdam, Germany
| | - Rüdiger Brühl
- Physikalisch-Technische Bundesanstalt (PTB), Braunschweig and Berlin, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 "Trajectoires développementales en psychiatrie"; Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli; Gif-sur-Yvette, France
| | - Marie-Laure Paillère Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 "Trajectoires développementales en psychiatrie"; Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli; Gif-sur-Yvette, France
- AP-HP. Sorbonne Université, Department of Child and Adolescent Psychiatry, Pitié-Salpêtrière Hospital, Paris, France
| | - Eric Artiges
- Institut National de la Santé et de la Recherche Médicale, INSERM U A10 "Trajectoires développementales en psychiatrie"; Université Paris-Saclay, Ecole Normale supérieure Paris-Saclay, CNRS, Centre Borelli; Gif-sur-Yvette, France
- Psychiatry Department, EPS Barthélémy Durand, Etampes, France
| | - Frauke Nees
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
- Institute of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, Mannheim, Germany
- Institute of Medical Psychology and Medical Sociology, University Medical Center Schleswig-Holstein, Kiel University, Kiel, Germany
| | | | - Herve Lemaitre
- NeuroSpin, CEA, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, CEA, Université de Bordeaux, 33076 Bordeaux, France
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry, Center for Psychosocial Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sarah Hohmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Nathalie Holz
- Department of Child and Adolescent Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Square J5, 68159 Mannheim, Germany
| | - Christian Baeuchl
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Michael N Smolka
- Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Nilakshi Vaidya
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Robert Whelan
- School of Psychology and Global Brain Health Institute, Trinity College Dublin, Ireland
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin Berlin, Germany
- Centre for Population Neuroscience and Precision Medicine (PONS), Institute for Science and Technology of Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China
| | | | - Tomáš Paus
- Departments of Psychiatry and Neuroscience, Faculty of Medicine and Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| | - Juergen Dukart
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Boris C Bernhardt
- Multimodal Imaging and Connectome Analysis Laboratory, McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Canada
| | - Oleksandr V Popovych
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sofie L Valk
- Institute of Neuroscience and Medicine - Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
23
|
Zhang H, Wang Z, Qiao X, Peng N, Wu J, Chen Y, Cheng C. Unveiling the therapeutic potential of IHMT-337 in glioma treatment: targeting the EZH2-SLC12A5 axis. Mol Med 2024; 30:91. [PMID: 38886655 PMCID: PMC11184773 DOI: 10.1186/s10020-024-00857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
Glioma is the most common malignant tumor of the central nervous system, with EZH2 playing a crucial regulatory role. This study further explores the abnormal expression of EZH2 and its mechanisms in regulating glioma progression. Additionally, it was found that IHMT-337 can potentially be a therapeutic agent for glioma. The prognosis, expression, and localization of EZH2 were determined using bioinformatics, IHC staining, Western blot (WB) analysis, and immunofluorescence (IF) localization. The effects of EZH2 on cell function were assessed using CCK-8 assays, Transwell assays, and wound healing assays. Public databases and RT-qPCR were utilized to identify downstream targets. The mechanisms regulating these downstream targets were elucidated using MS-PCR and WB analysis. The efficacy of IHMT-337 was demonstrated through IC50 measurements, WB analysis, and RT-qPCR. The effects of IHMT-337 on glioma cells in vitro were evaluated using Transwell assays, EdU incorporation assays, and flow cytometry. The potential of IHMT-337 as a treatment for glioma was assessed using a blood-brain barrier (BBB) model and an orthotopic glioma model. Our research confirms significantly elevated EZH2 expression in gliomas, correlating with patient prognosis. EZH2 facilitates glioma proliferation, migration, and invasion alongside promoting SLC12A5 DNA methylation. By regulating SLC12A5 expression, EZH2 activates the WNK1-OSR1-NKCC1 pathway, enhancing its interaction with ERM to promote glioma migration. IHMT-337 targets EZH2 in vitro to inhibit WNK1 activation, thereby suppressing glioma cell migration. Additionally, it inhibits cell proliferation and arrests the cell cycle. IHMT-337 has the potential to cross the BBB and has successfully inhibited glioma progression in vivo. This study expands our understanding of the EZH2-SLC12A5 axis in gliomas, laying a new foundation for the clinical translation of IHMT-337 and offering new insights for precision glioma therapy.
Collapse
Affiliation(s)
- Hongwei Zhang
- Anhui University of Science and Technology, Huainan, 232001, Anhui, China
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zixuan Wang
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Dalian Medical University, Dalian, 116044, Liaoning, China
| | - Xiaolong Qiao
- Anhui University of Science and Technology, Huainan, 232001, Anhui, China
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Nan Peng
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Jiaxing Wu
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
- Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Yinan Chen
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Chuandong Cheng
- Anhui University of Science and Technology, Huainan, 232001, Anhui, China.
- Division of Life Sciences and Medicine, Department of Neurosurgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
24
|
Zhang S, Larsen B, Sydnor VJ, Zeng T, An L, Yan X, Kong R, Kong X, Gur RC, Gur RE, Moore TM, Wolf DH, Holmes AJ, Xie Y, Zhou JH, Fortier MV, Tan AP, Gluckman P, Chong YS, Meaney MJ, Deco G, Satterthwaite TD, Yeo BTT. In vivo whole-cortex marker of excitation-inhibition ratio indexes cortical maturation and cognitive ability in youth. Proc Natl Acad Sci U S A 2024; 121:e2318641121. [PMID: 38814872 PMCID: PMC11161789 DOI: 10.1073/pnas.2318641121] [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: 10/26/2023] [Accepted: 04/04/2024] [Indexed: 06/01/2024] Open
Abstract
A balanced excitation-inhibition ratio (E/I ratio) is critical for healthy brain function. Normative development of cortex-wide E/I ratio remains unknown. Here, we noninvasively estimate a putative marker of whole-cortex E/I ratio by fitting a large-scale biophysically plausible circuit model to resting-state functional MRI (fMRI) data. We first confirm that our model generates realistic brain dynamics in the Human Connectome Project. Next, we show that the estimated E/I ratio marker is sensitive to the gamma-aminobutyric acid (GABA) agonist benzodiazepine alprazolam during fMRI. Alprazolam-induced E/I changes are spatially consistent with positron emission tomography measurement of benzodiazepine receptor density. We then investigate the relationship between the E/I ratio marker and neurodevelopment. We find that the E/I ratio marker declines heterogeneously across the cerebral cortex during youth, with the greatest reduction occurring in sensorimotor systems relative to association systems. Importantly, among children with the same chronological age, a lower E/I ratio marker (especially in the association cortex) is linked to better cognitive performance. This result is replicated across North American (8.2 to 23.0 y old) and Asian (7.2 to 7.9 y old) cohorts, suggesting that a more mature E/I ratio indexes improved cognition during normative development. Overall, our findings open the door to studying how disrupted E/I trajectories may lead to cognitive dysfunction in psychopathology that emerges during youth.
Collapse
Affiliation(s)
- Shaoshi Zhang
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
- Department of Pediatrics, University of Minnesota, Minneapolis, MN55455
| | - Valerie J. Sydnor
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
| | - Tianchu Zeng
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Lijun An
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Xiaoxuan Yan
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Ru Kong
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Xiaolu Kong
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
- ByteDance, Singapore048583, Singapore
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA19104
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiology, University of Pennsylvania, Philadelphia, PA19104
| | - Tyler M. Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
| | - Avram J. Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ07103
- Wu Tsai Institute, Yale University, New Haven, CT06520
| | - Yapei Xie
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
| | - Marielle V. Fortier
- Department of Diagnostic and Interventional Imaging, Kandang Kerbau Women’s and Children’s Hospital, Singapore229899, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
| | - Ai Peng Tan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore119074, Singapore
| | - Peter Gluckman
- Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland1142, New Zealand
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore119228, Singapore
| | - Michael J. Meaney
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore138632, Singapore
- Department of Neurology and Neurosurgery, McGill University, Montreal, QCH3A1A1, Canada
| | - Gustavo Deco
- Center for Brain and Cognition, Department of Technology and Information, Universitat Pompeu Fabra, Barcelona08002, Spain
- Institució Catalana de la Recerca i Estudis Avançats, Universitat Barcelona, Barcelona08010, Spain
| | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA19104
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Lifespan Brain Institute of Penn Medicine and Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA19104
| | - B. T. Thomas Yeo
- Centre for Sleep and Cognition and Centre for Translational Magnetic Resonance Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore117594, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore117583, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore119077, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Signapore117456, Signapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hopstial, Charlestown, MA02129
| |
Collapse
|
25
|
Tan E, Troller-Renfree SV, Morales S, Buzzell GA, McSweeney M, Antúnez M, Fox NA. Theta activity and cognitive functioning: Integrating evidence from resting-state and task-related developmental electroencephalography (EEG) research. Dev Cogn Neurosci 2024; 67:101404. [PMID: 38852382 PMCID: PMC11214181 DOI: 10.1016/j.dcn.2024.101404] [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: 11/02/2023] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024] Open
Abstract
The theta band is one of the most prominent frequency bands in the electroencephalography (EEG) power spectrum and presents an interesting paradox: while elevated theta power during resting state is linked to lower cognitive abilities in children and adolescents, increased theta power during cognitive tasks is associated with higher cognitive performance. Why does theta power, measured during resting state versus cognitive tasks, show differential correlations with cognitive functioning? This review provides an integrated account of the functional correlates of theta across different contexts. We first present evidence that higher theta power during resting state is correlated with lower executive functioning, attentional abilities, language skills, and IQ. Next, we review research showing that theta power increases during memory, attention, and cognitive control, and that higher theta power during these processes is correlated with better performance. Finally, we discuss potential explanations for the differential correlations between resting/task-related theta and cognitive functioning, and offer suggestions for future research in this area.
Collapse
Affiliation(s)
- Enda Tan
- Department of Human Development and Quantitative Methodology, University of Maryland, College Park, MD 20740, USA; Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD 20740, USA.
| | | | - Santiago Morales
- Department of Psychology, University of Southern California, CA 90007, USA
| | - George A Buzzell
- Department of Psychology, Florida International University, FL 33199, USA
| | - Marco McSweeney
- Department of Human Development and Quantitative Methodology, University of Maryland, College Park, MD 20740, USA
| | - Martín Antúnez
- Department of Human Development and Quantitative Methodology, University of Maryland, College Park, MD 20740, USA
| | - Nathan A Fox
- Department of Human Development and Quantitative Methodology, University of Maryland, College Park, MD 20740, USA; Neuroscience and Cognitive Science Program, University of Maryland, College Park, MD 20740, USA
| |
Collapse
|
26
|
Wise DL, Greene SB, Escobedo-Lozoya Y, Van Hooser SD, Nelson SB. Progressive Circuit Hyperexcitability in Mouse Neocortical Slice Cultures with Increasing Duration of Activity Silencing. eNeuro 2024; 11:ENEURO.0362-23.2024. [PMID: 38653560 PMCID: PMC11079856 DOI: 10.1523/eneuro.0362-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Forebrain neurons deprived of activity become hyperactive when activity is restored. Rebound activity has been linked to spontaneous seizures in vivo following prolonged activity blockade. Here, we measured the time course of rebound activity and the contributing circuit mechanisms using calcium imaging, synaptic staining, and whole-cell patch clamp in organotypic slice cultures of mouse neocortex. Calcium imaging revealed hypersynchronous activity increasing in intensity with longer periods of deprivation. While activity partially recovered 3 d after slices were released from 5 d of deprivation, they were less able to recover after 10 d of deprivation. However, even after the longer period of deprivation, activity patterns eventually returned to baseline levels. The degree of deprivation-induced rebound was age-dependent, with the greatest effects occurring when silencing began in the second week. Pharmacological blockade of NMDA receptors indicated that hypersynchronous rebound activity did not require activation of Hebbian plasticity. In single-neuron recordings, input resistance roughly doubled with a concomitant increase in intrinsic excitability. Synaptic imaging of pre- and postsynaptic proteins revealed dramatic reductions in the number of presumptive synapses with a larger effect on inhibitory than excitatory synapses. Putative excitatory synapses colocalizing PSD-95 and Bassoon declined by 39 and 56% following 5 and 10 d of deprivation, but presumptive inhibitory synapses colocalizing gephyrin and VGAT declined by 55 and 73%, respectively. The results suggest that with prolonged deprivation, a progressive reduction in synapse number is accompanied by a shift in the balance between excitation and inhibition and increased cellular excitability.
Collapse
Affiliation(s)
- Derek L Wise
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| | - Samuel B Greene
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| | | | | | - Sacha B Nelson
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| |
Collapse
|
27
|
Khanjanianpak M, Azimi-Tafreshi N, Valizadeh A. Emergence of complex oscillatory dynamics in the neuronal networks with long activity time of inhibitory synapses. iScience 2024; 27:109401. [PMID: 38532887 PMCID: PMC10963234 DOI: 10.1016/j.isci.2024.109401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/30/2023] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
The brain displays complex dynamics, including collective oscillations, and extensive research has been conducted to understand their generation. However, our understanding of how biological constraints influence these oscillations is incomplete. This study investigates the essential properties of neuronal networks needed to generate oscillations resembling those in the brain. A simple discrete-time model of interconnected excitable elements is developed, capable of closely resembling the complex oscillations observed in biological neural networks. In the model, synaptic connections remain active for a duration exceeding individual neuron activity. We show that the inhibitory synapses must exhibit longer activity than excitatory synapses to produce a diverse range of the dynamical states, including biologically plausible oscillations. Upon meeting this condition, the transition between different dynamical states can be controlled by external stochastic input to the neurons. The study provides a comprehensive explanation for the emergence of distinct dynamical states in neural networks based on specific parameters.
Collapse
Affiliation(s)
- Mozhgan Khanjanianpak
- Physics Department, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
- Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran 1991633357, Iran
| | - Nahid Azimi-Tafreshi
- Physics Department, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
| | - Alireza Valizadeh
- Physics Department, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 45137-66731, Iran
- Pasargad Institute for Advanced Innovative Solutions (PIAIS), Tehran 1991633357, Iran
| |
Collapse
|
28
|
Zhao Z, Shirinpour S, Tran H, Wischnewski M, Opitz A. intensity- and frequency-specific effects of transcranial alternating current stimulation are explained by network dynamics. J Neural Eng 2024; 21:026024. [PMID: 38530297 DOI: 10.1088/1741-2552/ad37d9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/26/2024] [Indexed: 03/27/2024]
Abstract
Objective. Transcranial alternating current stimulation (tACS) can be used to non-invasively entrain neural activity and thereby cause changes in local neural oscillatory power. Despite its increased use in cognitive and clinical neuroscience, the fundamental mechanisms of tACS are still not fully understood.Approach. We developed a computational neuronal network model of two-compartment pyramidal neurons (PY) and inhibitory interneurons, which mimic the local cortical circuits. We modeled tACS with electric field strengths that are achievable in human applications. We then simulated intrinsic network activity and measured neural entrainment to investigate how tACS modulates ongoing endogenous oscillations.Main results. The intensity-specific effects of tACS are non-linear. At low intensities (<0.3 mV mm-1), tACS desynchronizes neural firing relative to the endogenous oscillations. At higher intensities (>0.3 mV mm-1), neurons are entrained to the exogenous electric field. We then further explore the stimulation parameter space and find that the entrainment of ongoing cortical oscillations also depends on stimulation frequency by following an Arnold tongue. Moreover, neuronal networks can amplify the tACS-induced entrainment via synaptic coupling and network effects. Our model shows that PY are directly entrained by the exogenous electric field and drive the inhibitory neurons.Significance. The results presented in this study provide a mechanistic framework for understanding the intensity- and frequency-specific effects of oscillating electric fields on neuronal networks. This is crucial for rational parameter selection for tACS in cognitive studies and clinical applications.
Collapse
Affiliation(s)
- Zhihe Zhao
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Sina Shirinpour
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Harry Tran
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Miles Wischnewski
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| | - Alexander Opitz
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States of America
| |
Collapse
|
29
|
Zhang S, Larsen B, Sydnor VJ, Zeng T, An L, Yan X, Kong R, Kong X, Gur RC, Gur RE, Moore TM, Wolf DH, Holmes AJ, Xie Y, Zhou JH, Fortier MV, Tan AP, Gluckman P, Chong YS, Meaney MJ, Deco G, Satterthwaite TD, Yeo BT. In-vivo whole-cortex marker of excitation-inhibition ratio indexes cortical maturation and cognitive ability in youth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.22.546023. [PMID: 38586012 PMCID: PMC10996460 DOI: 10.1101/2023.06.22.546023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
A balanced excitation-inhibition ratio (E/I ratio) is critical for healthy brain function. Normative development of cortex-wide E/I ratio remains unknown. Here we non-invasively estimate a putative marker of whole-cortex E/I ratio by fitting a large-scale biophysically-plausible circuit model to resting-state functional MRI (fMRI) data. We first confirm that our model generates realistic brain dynamics in the Human Connectome Project. Next, we show that the estimated E/I ratio marker is sensitive to the GABA-agonist benzodiazepine alprazolam during fMRI. Alprazolam-induced E/I changes are spatially consistent with positron emission tomography measurement of benzodiazepine receptor density. We then investigate the relationship between the E/I ratio marker and neurodevelopment. We find that the E/I ratio marker declines heterogeneously across the cerebral cortex during youth, with the greatest reduction occurring in sensorimotor systems relative to association systems. Importantly, among children with the same chronological age, a lower E/I ratio marker (especially in association cortex) is linked to better cognitive performance. This result is replicated across North American (8.2 to 23.0 years old) and Asian (7.2 to 7.9 years old) cohorts, suggesting that a more mature E/I ratio indexes improved cognition during normative development. Overall, our findings open the door to studying how disrupted E/I trajectories may lead to cognitive dysfunction in psychopathology that emerges during youth.
Collapse
Affiliation(s)
- Shaoshi Zhang
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Valerie J. Sydnor
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tianchu Zeng
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Lijun An
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Xiaoxuan Yan
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Ru Kong
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Xiaolu Kong
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
- ByteDance, Singapore
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler M. Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, United States
- Wu Tsai Institute, Yale University, New Haven, CT, United States
| | - Yapei Xie
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
| | - Marielle V Fortier
- Department of Diagnostic and Interventional Imaging, KK Women’s and Children’s Hospital, Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Ai Peng Tan
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Peter Gluckman
- UK Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Michael J Meaney
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Gustavo Deco
- Center for Brain and Cognition, Department of Technology and Information, Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avançats, Universitat Barcelona, Barcelona, Spain
| | - Theodore D. Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - B.T. Thomas Yeo
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National Univeristy of Singapore, Signapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hopstial, Charlestown, MA, USA
| |
Collapse
|
30
|
Ryu J, Choi JW, Niketeghad S, Torres EB, Pouratian N. Irregularity of instantaneous gamma frequency in the motor control network characterize visuomotor and proprioceptive information processing. J Neural Eng 2024; 21:10.1088/1741-2552/ad2e1d. [PMID: 38417152 PMCID: PMC11025688 DOI: 10.1088/1741-2552/ad2e1d] [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: 10/20/2023] [Accepted: 02/28/2024] [Indexed: 03/01/2024]
Abstract
Objective.The study aims to characterize movements with different sensory goals, by contrasting the neural activity involved in processing proprioceptive and visuo-motor information. To accomplish this, we have developed a new methodology that utilizes the irregularity of the instantaneous gamma frequency parameter for characterization.Approach.In this study, eight essential tremor patients undergoing an awake deep brain stimulation implantation surgery repetitively touched the clinician's finger (forward visually-guided/FV movement) and then one's own chin (backward proprioceptively-guided/BP movement). Neural electrocorticographic recordings from the motor (M1), somatosensory (S1), and posterior parietal cortex (PPC) were obtained and band-pass filtered in the gamma range (30-80 Hz). The irregularity of the inter-event intervals (IEI; inverse of instantaneous gamma frequency) were examined as: (1) auto-information of the IEI time series and (2) correlation between the amplitude and its proceeding IEI. We further explored the network connectivity after segmenting the FV and BP movements by periods of accelerating and decelerating forces, and applying the IEI parameter to transfer entropy methods.Main results.Conceptualizing that the irregularity in IEI reflects active new information processing, we found the highest irregularity in M1 during BP movement, highest in PPC during FV movement, and the lowest during rest at all sites. Also, connectivity was the strongest from S1 to M1 and from S1 to PPC during FV movement with accelerating force and weakest during rest.Significance. We introduce a novel methodology that utilize the instantaneous gamma frequency (i.e. IEI) parameter in characterizing goal-oriented movements with different sensory goals, and demonstrate its use to inform the directional connectivity within the motor cortical network. This method successfully characterizes different movement types, while providing interpretations to the sensory-motor integration processes.
Collapse
Affiliation(s)
- Jihye Ryu
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Jeong Woo Choi
- Department of Neurological Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Soroush Niketeghad
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Elizabeth B. Torres
- Psychology Department, Rutgers University Center for Cognitive Science, Computational Biomedicine Imaging and Modeling Center at Computer Science Department, Rutgers University, Piscataway, NJ 08854
| | - Nader Pouratian
- Department of Neurological Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
31
|
Adaikkan C, Joseph J, Foustoukos G, Wang J, Polygalov D, Boehringer R, Middleton SJ, Huang AJY, Tsai LH, McHugh TJ. Silencing CA1 pyramidal cells output reveals the role of feedback inhibition in hippocampal oscillations. Nat Commun 2024; 15:2190. [PMID: 38467602 PMCID: PMC10928166 DOI: 10.1038/s41467-024-46478-3] [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: 08/20/2023] [Accepted: 02/20/2024] [Indexed: 03/13/2024] Open
Abstract
The precise temporal coordination of neural activity is crucial for brain function. In the hippocampus, this precision is reflected in the oscillatory rhythms observed in CA1. While it is known that a balance between excitatory and inhibitory activity is necessary to generate and maintain these oscillations, the differential contribution of feedforward and feedback inhibition remains ambiguous. Here we use conditional genetics to chronically silence CA1 pyramidal cell transmission, ablating the ability of these neurons to recruit feedback inhibition in the local circuit, while recording physiological activity in mice. We find that this intervention leads to local pathophysiological events, with ripple amplitude and intrinsic frequency becoming significantly larger and spatially triggered local population spikes locked to the trough of the theta oscillation appearing during movement. These phenotypes demonstrate that feedback inhibition is crucial in maintaining local sparsity of activation and reveal the key role of lateral inhibition in CA1 in shaping circuit function.
Collapse
Affiliation(s)
| | - Justin Joseph
- Centre for Brain Research, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Georgios Foustoukos
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wakoshi, Saitama, Japan
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Jun Wang
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Denis Polygalov
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wakoshi, Saitama, Japan
| | - Roman Boehringer
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wakoshi, Saitama, Japan
| | - Steven J Middleton
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wakoshi, Saitama, Japan
| | - Arthur J Y Huang
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wakoshi, Saitama, Japan
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas J McHugh
- Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, Wakoshi, Saitama, Japan.
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
32
|
Silverstein BH, Kolbman N, Nelson A, Liu T, Guzzo P, Gilligan J, Lee U, Mashour GA, Vanini G, Pal D. Psilocybin induces dose-dependent changes in functional network organization in rat cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579718. [PMID: 38405722 PMCID: PMC10888735 DOI: 10.1101/2024.02.09.579718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Psilocybin produces an altered state of consciousness in humans and is associated with complex spatiotemporal changes in brain networks. Given the emphasis on rodent models for mechanistic studies, there is a need for characterization of the effect of psilocybin on brain-wide network dynamics. Previous rodent studies of psychedelics, using electroencephalogram, have primarily been done with sparse electrode arrays that offered limited spatial resolution precluding network level analysis, and have been restricted to lower gamma frequencies. Therefore, in the study, we used electroencephalographic recordings from 27 sites (electrodes) across rat cortex (n=6 male, 6 female) to characterize the effect of psilocybin (0.1 mg/kg, 1 mg/kg, and 10 mg/kg delivered over an hour) on network organization as inferred through changes in node degree (index of network density) and connection strength (weighted phase-lag index). The removal of aperiodic component from the electroencephalogram localized the primary oscillatory changes to theta (4-10 Hz), medium gamma (70-110 Hz), and high gamma (110-150 Hz) bands, which were used for the network analysis. Additionally, we determined the concurrent changes in theta-gamma phase-amplitude coupling. We report that psilocybin, in a dose-dependent manner, 1) disrupted theta-gamma coupling [p<0.05], 2) increased frontal high gamma connectivity [p<0.05] and posterior theta connectivity [p≤0.049], and 3) increased frontal high gamma [p<0.05] and posterior theta [p≤0.046] network density. The medium gamma frontoparietal connectivity showed a nonlinear relationship with psilocybin dose. Our results suggest that high-frequency network organization, decoupled from local theta-phase, may be an important signature of psilocybin-induced non-ordinary state of consciousness.
Collapse
Affiliation(s)
- Brian H. Silverstein
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Nicholas Kolbman
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Amanda Nelson
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Tiecheng Liu
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Peter Guzzo
- Tryp Therapeutics, Kelowna, British Columbia, V1Y 7T2, Canada
| | - Jim Gilligan
- Tryp Therapeutics, Kelowna, British Columbia, V1Y 7T2, Canada
| | - UnCheol Lee
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, Michigan 48109, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - George A. Mashour
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, Michigan 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Giancarlo Vanini
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, Michigan 48109, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, Michigan 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Dinesh Pal
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan 48109, USA
- Center for Consciousness Science, University of Michigan, Ann Arbor, Michigan 48109, USA
- Michigan Psychedelic Center, University of Michigan, Ann Arbor, Michigan 48109, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
33
|
Hadler MD, Tzilivaki A, Schmitz D, Alle H, Geiger JRP. Gamma oscillation plasticity is mediated via parvalbumin interneurons. SCIENCE ADVANCES 2024; 10:eadj7427. [PMID: 38295164 PMCID: PMC10830109 DOI: 10.1126/sciadv.adj7427] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024]
Abstract
Understanding the plasticity of neuronal networks is an emerging field of (patho-) physiological research, yet the underlying cellular mechanisms remain poorly understood. Gamma oscillations (30 to 80 hertz), a biomarker of cognitive performance, require and potentiate glutamatergic transmission onto parvalbumin-positive interneurons (PVIs), suggesting an interface for cell-to-network plasticity. In ex vivo local field potential recordings, we demonstrate long-term potentiation of hippocampal gamma power. Gamma potentiation obeys established rules of PVI plasticity, requiring calcium-permeable AMPA receptors (CP-AMPARs) and metabotropic glutamate receptors (mGluRs). A microcircuit computational model of CA3 gamma oscillations predicts CP-AMPAR plasticity onto PVIs critically outperforms pyramidal cell plasticity in increasing gamma power and completely accounts for gamma potentiation. We reaffirm this ex vivo in three PVI-targeting animal models, demonstrating that gamma potentiation requires PVI-specific signaling via a Gq/PKC pathway comprising mGluR5 and a Gi-sensitive, PKA-dependent pathway. Gamma activity-dependent, metabotropically mediated CP-AMPAR plasticity on PVIs may serve as a guiding principle in understanding network plasticity in health and disease.
Collapse
Affiliation(s)
- Michael D. Hadler
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Neurocure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Neurocure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle-Straße 10, 13125 Berlin, Germany
| | - Henrik Alle
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg R. P. Geiger
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
34
|
Shirani F, Choi H. On the physiological and structural contributors to the overall balance of excitation and inhibition in local cortical networks. J Comput Neurosci 2024; 52:73-107. [PMID: 37837534 PMCID: PMC11582336 DOI: 10.1007/s10827-023-00863-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/25/2023] [Accepted: 09/08/2023] [Indexed: 10/16/2023]
Abstract
Overall balance of excitation and inhibition in cortical networks is central to their functionality and normal operation. Such orchestrated co-evolution of excitation and inhibition is established through convoluted local interactions between neurons, which are organized by specific network connectivity structures and are dynamically controlled by modulating synaptic activities. Therefore, identifying how such structural and physiological factors contribute to establishment of overall balance of excitation and inhibition is crucial in understanding the homeostatic plasticity mechanisms that regulate the balance. We use biologically plausible mathematical models to extensively study the effects of multiple key factors on overall balance of a network. We characterize a network's baseline balanced state by certain functional properties, and demonstrate how variations in physiological and structural parameters of the network deviate this balance and, in particular, result in transitions in spontaneous activity of the network to high-amplitude slow oscillatory regimes. We show that deviations from the reference balanced state can be continuously quantified by measuring the ratio of mean excitatory to mean inhibitory synaptic conductances in the network. Our results suggest that the commonly observed ratio of the number of inhibitory to the number of excitatory neurons in local cortical networks is almost optimal for their stability and excitability. Moreover, the values of inhibitory synaptic decay time constants and density of inhibitory-to-inhibitory network connectivity are critical to overall balance and stability of cortical networks. However, network stability in our results is sufficiently robust against modulations of synaptic quantal conductances, as required by their role in learning and memory. Our study based on extensive bifurcation analyses thus reveal the functional optimality and criticality of structural and physiological parameters in establishing the baseline operating state of local cortical networks.
Collapse
Affiliation(s)
- Farshad Shirani
- School of Mathematics, Georgia Institute of Technology, Atlanta, 30332, Georgia, USA.
| | - Hannah Choi
- School of Mathematics, Georgia Institute of Technology, Atlanta, 30332, Georgia, USA
| |
Collapse
|
35
|
Zheng Y, Kang S, O'Neill J, Bojak I. Spontaneous slow wave oscillations in extracellular field potential recordings reflect the alternating dominance of excitation and inhibition. J Physiol 2024; 602:713-736. [PMID: 38294945 DOI: 10.1113/jp284587] [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: 02/23/2023] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
In the resting state, cortical neurons can fire action potentials spontaneously but synchronously (Up state), followed by a quiescent period (Down state) before the cycle repeats. Extracellular recordings in the infragranular layer of cortex with a micro-electrode display a negative deflection (depth-negative) during Up states and a positive deflection (depth-positive) during Down states. The resulting slow wave oscillation (SWO) has been studied extensively during sleep and under anaesthesia. However, recent research on the balanced nature of synaptic excitation and inhibition has highlighted our limited understanding of its genesis. Specifically, are excitation and inhibition balanced during SWOs? We analyse spontaneous local field potentials (LFPs) during SWOs recorded from anaesthetised rats via a multi-channel laminar micro-electrode and show that the Down state consists of two distinct synaptic states: a Dynamic Down state associated with depth-positive LFPs and a prominent dipole in the extracellular field, and a Static Down state with negligible (≈ 0 mV $ \approx 0{\mathrm{\;mV}}$ ) LFPs and a lack of dipoles extracellularly. We demonstrate that depth-negative and -positive LFPs are generated by a shift in the balance of synaptic excitation and inhibition from excitation dominance (depth-negative) to inhibition dominance (depth-positive) in the infragranular layer neurons. Thus, although excitation and inhibition co-tune overall, differences in their timing lead to an alternation of dominance, manifesting as SWOs. We further show that Up state initiation is significantly faster if the preceding Down state is dynamic rather than static. Our findings provide a coherent picture of the dependence of SWOs on synaptic activity. KEY POINTS: Cortical neurons can exhibit repeated cycles of spontaneous activity interleaved with periods of relative silence, a phenomenon known as 'slow wave oscillation' (SWO). During SWOs, recordings of local field potentials (LFPs) in the neocortex show depth-negative deflection during the active period (Up state) and depth-positive deflection during the silent period (Down state). Here we further classified the Down state into a dynamic phase and a static phase based on a novel method of classification and revealed non-random, stereotypical sequences of the three states occurring with significantly different transitional kinetics. Our results suggest that the positive and negative deflections in the LFP reflect the shift of the instantaneous balance between excitatory and inhibitory synaptic activity of the local cortical neurons. The differences in transitional kinetics may imply distinct synaptic mechanisms for Up state initiation. The study may provide a new approach for investigating spontaneous brain rhythms.
Collapse
Affiliation(s)
- Ying Zheng
- School of Biological Sciences, Whiteknights, University of Reading, Reading, UK
- Centre for Integrative Neuroscience and Neurodynamics (CINN), University of Reading, Reading, UK
| | - Sungmin Kang
- School of Psychology, Cardiff University, Cardiff, UK
| | | | - Ingo Bojak
- Centre for Integrative Neuroscience and Neurodynamics (CINN), University of Reading, Reading, UK
- School of Psychology and Clinical Language Science, Whiteknights, University of Reading, Reading, UK
| |
Collapse
|
36
|
Kim HR, Martina M. Bidirectional Regulation of GABA A Reversal Potential in the Adult Brain: Physiological and Pathological Implications. Life (Basel) 2024; 14:143. [PMID: 38276272 PMCID: PMC10817304 DOI: 10.3390/life14010143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
In physiological conditions, the intracellular chloride concentration is much lower than the extracellular. As GABAA channels are permeable to anions, the reversal potential of GABAA is very close to that of Cl-, which is the most abundant free anion in the intra- and extracellular spaces. Intracellular chloride is regulated by the activity ratio of NKCC1 and KCC2, two chloride-cation cotransporters that import and export Cl-, respectively. Due to the closeness between GABAA reversal potential and the value of the resting membrane potential in most neurons, small changes in intracellular chloride have a major functional impact, which makes GABAA a uniquely flexible signaling system. In most neurons of the adult brain, the GABAA reversal potential is slightly more negative than the resting membrane potential, which makes GABAA hyperpolarizing. Alterations in GABAA reversal potential are a common feature in numerous conditions as they are the consequence of an imbalance in the NKCC1-KCC2 activity ratio. In most conditions (including Alzheimer's disease, schizophrenia, and Down's syndrome), GABAA becomes depolarizing, which causes network desynchronization and behavioral impairment. In other conditions (neonatal inflammation and neuropathic pain), however, GABAA reversal potential becomes hypernegative, which affects behavior through a potent circuit deactivation.
Collapse
Affiliation(s)
- Haram R. Kim
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, 300 E. Superior, Chicago, IL 60611, USA;
| | - Marco Martina
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, 300 E. Superior, Chicago, IL 60611, USA;
- Department of Psychiatry, Feinberg School of Medicine, Northwestern University, 300 E. Superior, Chicago, IL 60611, USA
| |
Collapse
|
37
|
Carlos-Lima E, Higa GSV, Viana FJC, Tamais AM, Cruvinel E, Borges FDS, Francis-Oliveira J, Ulrich H, De Pasquale R. Serotonergic Modulation of the Excitation/Inhibition Balance in the Visual Cortex. Int J Mol Sci 2023; 25:519. [PMID: 38203689 PMCID: PMC10778629 DOI: 10.3390/ijms25010519] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Serotonergic neurons constitute one of the main systems of neuromodulators, whose diffuse projections regulate the functions of the cerebral cortex. Serotonin (5-HT) is known to play a crucial role in the differential modulation of cortical activity related to behavioral contexts. Some features of the 5-HT signaling organization suggest its possible participation as a modulator of activity-dependent synaptic changes during the critical period of the primary visual cortex (V1). Cells of the serotonergic system are among the first neurons to differentiate and operate. During postnatal development, ramifications from raphe nuclei become massively distributed in the visual cortical area, remarkably increasing the availability of 5-HT for the regulation of excitatory and inhibitory synaptic activity. A substantial amount of evidence has demonstrated that synaptic plasticity at pyramidal neurons of the superficial layers of V1 critically depends on a fine regulation of the balance between excitation and inhibition (E/I). 5-HT could therefore play an important role in controlling this balance, providing the appropriate excitability conditions that favor synaptic modifications. In order to explore this possibility, the present work used in vitro intracellular electrophysiological recording techniques to study the effects of 5-HT on the E/I balance of V1 layer 2/3 neurons, during the critical period. Serotonergic action on the E/I balance has been analyzed on spontaneous activity, evoked synaptic responses, and long-term depression (LTD). Our results pointed out that the predominant action of 5-HT implies a reduction in the E/I balance. 5-HT promoted LTD at excitatory synapses while blocking it at inhibitory synaptic sites, thus shifting the Hebbian alterations of synaptic strength towards lower levels of E/I balance.
Collapse
Affiliation(s)
- Estevão Carlos-Lima
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
- Departamento de Bioquímica, Instituto de Química (USP), São Paulo 05508-900, SP, Brazil;
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo 09210-580, SP, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Alicia Moraes Tamais
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| | - Fernando da Silva Borges
- Department of Physiology & Pharmacology, SUNY Downstate Health Sciences University, New York, NY 11203, USA;
| | - José Francis-Oliveira
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), São Paulo 05508-900, SP, Brazil;
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil; (E.C.-L.); (G.S.V.H.); (E.C.); (J.F.-O.)
| |
Collapse
|
38
|
Singh MF, Braver TS, Cole MW, Ching S. Precision data-driven modeling of cortical dynamics reveals idiosyncratic mechanisms underlying canonical oscillations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567088. [PMID: 38077097 PMCID: PMC10705281 DOI: 10.1101/2023.11.14.567088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Task-free brain activity affords unique insight into the functional structure of brain network dynamics and is a strong marker of individual differences. In this work, we present an algorithmic optimization framework that makes it possible to directly invert and parameterize brain-wide dynamical-systems models involving hundreds of interacting brain areas, from single-subject time-series recordings. This technique provides a powerful neurocomputational tool for interrogating mechanisms underlying individual brain dynamics ("precision brain models") and making quantitative predictions. We extensively validate the models' performance in forecasting future brain activity and predicting individual variability in key M/EEG markers. Lastly, we demonstrate the power of our technique in resolving individual differences in the generation of alpha and beta-frequency oscillations. We characterize subjects based upon model attractor topology and a dynamical-systems mechanism by which these topologies generate individual variation in the expression of alpha vs. beta rhythms. We trace these phenomena back to global variation in excitation-inhibition balance, highlighting the explanatory power of our framework in generating mechanistic insights.
Collapse
Affiliation(s)
- Matthew F Singh
- Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, 63130, MO, USA
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, 07102, NJ, USA
- Psychological and Brain Science, Washington University in St. Louis, St. Louis, 63130, MO, USA
| | - Todd S Braver
- Psychological and Brain Science, Washington University in St. Louis, St. Louis, 63130, MO, USA
| | - Michael W Cole
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, 07102, NJ, USA
| | - ShiNung Ching
- Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, 63130, MO, USA
| |
Collapse
|
39
|
Yang Y, Chen D, Wang J, Wang J, Yan Z, Deng Q, Zhang L, Luan G, Wang M, Li T. Dynamic evolution of the anterior cingulate-insula network during seizures. CNS Neurosci Ther 2023; 29:3901-3912. [PMID: 37309272 PMCID: PMC10651990 DOI: 10.1111/cns.14310] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/14/2023] Open
Abstract
OBJECTIVES In physiological situations, the anterior cingulate cortex (ACC) and anterior insular cortex (AIC) are prone to coactivation. The functional connectivity and interaction between ACC and AIC in the context of epilepsy remain unclear. This study aimed to investigate the dynamic coupling between these two brain regions during seizures. METHODS Patients who underwent stereoelectroencephalography (SEEG) recording were included in this study. The SEEG data were visually inspected and quantitatively analyzed. The narrowband oscillations and aperiodic components at seizure onset were parameterized. The frequency-specific non-linear correlation analysis was applied to the functional connectivity. The excitation/inhibition ratio (E:I ratio) reflected by the aperiodic slope was performed to evaluate the excitability. RESULTS Twenty patients were included in the study, with 10 diagnosed with anterior cingulate epilepsy and 10 with anterior insular epilepsy. In both types of epilepsy, the correlation coefficient (h2 ) between the ACC and AIC at seizure onset exhibited a significantly higher value than that during interictal and preictal periods (p < 0.05). The direction index (D) showed a significant increase at seizure onset, serving as an indicator for the direction of information flow between these two brain regions with up to 90% accuracy. The E:I ratio increased significantly at seizure onset, with the seizure-onset zone (SOZ) demonstrating a more pronounced increase compared to non-SOZ (p < 0.05). For seizures originating from AIC, the E:I ratio was significantly higher in the AIC than in the ACC (p = 0.0364). CONCLUSIONS In the context of epilepsy, the ACC and AIC are dynamically coupled during seizures. The functional connectivity and excitability exhibit a significant increase at seizure onset. By analyzing connectivity and excitability, the SOZ in ACC and AIC can be identified. The direction index (D) serves as an indicator for the direction of information flow from SOZ to non-SOZ. Notably, the excitability of SOZ changes more significantly than that of non-SOZ.
Collapse
Affiliation(s)
- Yujiao Yang
- Department of Neurology, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| | - Dong Chen
- Key Laboratory of Mental HealthInstitute of Psychology, Chinese Academy of SciencesBeijingChina
| | - Jing Wang
- Department of Neurology, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| | - Jie Wang
- Department of ElectrophysiologyCapital Institute of PediatricsBeijingChina
| | - Zhaofen Yan
- Department of Neurology, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| | - Qinqin Deng
- Department of Neurology, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| | - Liping Zhang
- Department of Neurology, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| | - Guoming Luan
- Department of Functional Neurosurgery, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Epilepsy, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
- Beijing Institute for Brain Disorders, Capital Medical UniversityBeijingChina
| | - Mengyang Wang
- Department of Neurology, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
| | - Tianfu Li
- Department of Neurology, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Epilepsy, Sanbo Brain HospitalCapital Medical UniversityBeijingChina
- Beijing Institute for Brain Disorders, Capital Medical UniversityBeijingChina
| |
Collapse
|
40
|
Hijazi S, Smit AB, van Kesteren RE. Fast-spiking parvalbumin-positive interneurons in brain physiology and Alzheimer's disease. Mol Psychiatry 2023; 28:4954-4967. [PMID: 37419975 PMCID: PMC11041664 DOI: 10.1038/s41380-023-02168-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/09/2023]
Abstract
Fast-spiking parvalbumin (PV) interneurons are inhibitory interneurons with unique morphological and functional properties that allow them to precisely control local circuitry, brain networks and memory processing. Since the discovery in 1987 that PV is expressed in a subset of fast-spiking GABAergic inhibitory neurons, our knowledge of the complex molecular and physiological properties of these cells has been expanding. In this review, we highlight the specific properties of PV neurons that allow them to fire at high frequency and with high reliability, enabling them to control network oscillations and shape the encoding, consolidation and retrieval of memories. We next discuss multiple studies reporting PV neuron impairment as a critical step in neuronal network dysfunction and cognitive decline in mouse models of Alzheimer's disease (AD). Finally, we propose potential mechanisms underlying PV neuron dysfunction in AD and we argue that early changes in PV neuron activity could be a causal step in AD-associated network and memory impairment and a significant contributor to disease pathogenesis.
Collapse
Affiliation(s)
- Sara Hijazi
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Burman RJ, Brodersen PJN, Raimondo JV, Sen A, Akerman CJ. Active cortical networks promote shunting fast synaptic inhibition in vivo. Neuron 2023; 111:3531-3540.e6. [PMID: 37659408 PMCID: PMC11913778 DOI: 10.1016/j.neuron.2023.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/03/2023] [Accepted: 08/04/2023] [Indexed: 09/04/2023]
Abstract
Fast synaptic inhibition determines neuronal response properties in the mammalian brain and is mediated by chloride-permeable ionotropic GABA-A receptors (GABAARs). Despite their fundamental role, it is still not known how GABAARs signal in the intact brain. Here, we use in vivo gramicidin recordings to investigate synaptic GABAAR signaling in mouse cortical pyramidal neurons under conditions that preserve native transmembrane chloride gradients. In anesthetized cortex, synaptic GABAARs exert classic hyperpolarizing effects. In contrast, GABAAR-mediated synaptic signaling in awake cortex is found to be predominantly shunting. This is due to more depolarized GABAAR equilibrium potentials (EGABAAR), which are shown to result from the high levels of synaptic activity that characterize awake cortical networks. Synaptic EGABAAR observed in awake cortex facilitates the desynchronizing effects of inhibitory inputs upon local networks, which increases the flexibility of spiking responses to external inputs. Our findings therefore suggest that GABAAR signaling adapts to optimize cortical functions.
Collapse
Affiliation(s)
- Richard J Burman
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK; Oxford Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | | | - Joseph V Raimondo
- Division of Cell Biology, Department of Human Biology, Neuroscience Institute and Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, 7935, South Africa
| | - Arjune Sen
- Oxford Epilepsy Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Colin J Akerman
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK.
| |
Collapse
|
42
|
Burns TF. Does inhibitory (dys)function account for involuntary autobiographical memory and déjà vu experience? Behav Brain Sci 2023; 46:e360. [PMID: 37961769 DOI: 10.1017/s0140525x23000146] [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: 11/15/2023]
Abstract
External cues and internal configuration states are the likely instigators of involuntary autobiographical memories (IAMs) and déjà vu experience. Indeed, Barzykowski and Moulin discuss relevant neuroscientific evidence in this direction. A complementary line of enquiry and evidence is the study of inhibition and its role in memory retrieval, and particularly how its (dys)function may contribute to IAMs and déjà vu.
Collapse
Affiliation(s)
- Thomas F Burns
- Okinawa Institute of Science and Technology Graduate University, Onna-son, Kunigami-gun, Okinawa, Japan ://tfburns.com/
| |
Collapse
|
43
|
Chan MMY, Choi CXT, Tsoi TCW, Shea CKS, Yiu KWK, Han YMY. Effects of multisession cathodal transcranial direct current stimulation with cognitive training on sociocognitive functioning and brain dynamics in autism: A double-blind, sham-controlled, randomized EEG study. Brain Stimul 2023; 16:1604-1616. [PMID: 37918630 DOI: 10.1016/j.brs.2023.10.012] [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: 07/04/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Few treatment options are available for targeting core symptoms of autism spectrum disorder (ASD). The development of treatments that target common neural circuit dysfunctions caused by known genetic defects, namely, disruption of the excitation/inhibition (E/I) balance, is promising. Transcranial direct current stimulation (tDCS) is capable of modulating the E/I balance in healthy individuals, yet its clinical and neurobiological effects in ASD remain elusive. OBJECTIVE This double-blind, randomized, sham-controlled trial investigated the effects of multisession cathodal prefrontal tDCS coupled with online cognitive remediation on social functioning, information processing efficiency and the E/I balance in ASD patients aged 14-21 years. METHODS Sixty individuals were randomly assigned to receive either active or sham tDCS (10 sessions in total, 20 min/session, stimulation intensity: 1.5 mA, cathode: F3, anode: Fp2, size of electrodes: 25 cm2) combined with 20 min of online cognitive remediation. Social functioning, information processing efficiency during cognitive tasks, and theta- and gamma-band E/I balance were measured one day before and after the treatment. RESULTS Compared to sham tDCS, active cathodal tDCS was effective in enhancing overall social functioning [F(1, 58) = 6.79, p = .012, ηp2 = 0.105, 90% CI: (0.013, 0.234)] and information processing efficiency during cognitive tasks [F(1, 58) = 10.07, p = .002, ηp2 = 0.148, 90% CI: (0.034, 0.284)] in these individuals. Electroencephalography data showed that this cathodal tDCS protocol was effective in reducing the theta-band E/I ratio of the cortical midline structures [F(1, 58) = 4.65, p = .035, ηp2 = 0.074, 90% CI: (0.010, 0.150)] and that this reduction significantly predicted information processing efficiency enhancement (b = -2.546, 95% BCa CI: [-4.979, -0.113], p = .041). CONCLUSION Our results support the use of multisession cathodal tDCS over the left dorsolateral prefrontal cortex combined with online cognitive remediation for reducing the elevated theta-band E/I ratio in sociocognitive information processing circuits in ASD patients, resulting in more adaptive regulation of global brain dynamics that is associated with enhanced information processing efficiency after the intervention.
Collapse
Affiliation(s)
- Melody M Y Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region; Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Coco X T Choi
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Tom C W Tsoi
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region
| | - Caroline K S Shea
- Alice Ho Miu Ling Nethersole Hospital, Hospital Authority, Hong Kong Special Administrative Region; Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Klaire W K Yiu
- Alice Ho Miu Ling Nethersole Hospital, Hospital Authority, Hong Kong Special Administrative Region
| | - Yvonne M Y Han
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region; University Research Facility in Behavioral and Systems Neuroscience (UBSN), The Hong Kong Polytechnic University, Hong Kong Special Administrative Region.
| |
Collapse
|
44
|
Ceanga M, Rahmati V, Haselmann H, Schmidl L, Hunter D, Brauer AK, Liebscher S, Kreye J, Prüss H, Groc L, Hallermann S, Dalmau J, Ori A, Heckmann M, Geis C. Human NMDAR autoantibodies disrupt excitatory-inhibitory balance, leading to hippocampal network hypersynchrony. Cell Rep 2023; 42:113166. [PMID: 37768823 DOI: 10.1016/j.celrep.2023.113166] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2023] [Accepted: 09/07/2023] [Indexed: 09/30/2023] Open
Abstract
Anti-NMDA receptor autoantibodies (NMDAR-Abs) in patients with NMDAR encephalitis cause severe disease symptoms resembling psychosis and cause cognitive dysfunction. After passive transfer of patients' cerebrospinal fluid or human monoclonal anti-GluN1-autoantibodies in mice, we find a disrupted excitatory-inhibitory balance resulting from CA1 neuronal hypoexcitability, reduced AMPA receptor (AMPAR) signaling, and faster synaptic inhibition in acute hippocampal slices. Functional alterations are also reflected in widespread remodeling of the hippocampal proteome, including changes in glutamatergic and GABAergic neurotransmission. NMDAR-Abs amplify network γ oscillations and disrupt θ-γ coupling. A data-informed network model reveals that lower AMPAR strength and faster GABAA receptor current kinetics chiefly account for these abnormal oscillations. As predicted in silico and evidenced ex vivo, positive allosteric modulation of AMPARs alleviates aberrant γ activity, reinforcing the causative effects of the excitatory-inhibitory imbalance. Collectively, NMDAR-Ab-induced aberrant synaptic, cellular, and network dynamics provide conceptual insights into NMDAR-Ab-mediated pathomechanisms and reveal promising therapeutic targets that merit future in vivo validation.
Collapse
Affiliation(s)
- Mihai Ceanga
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Vahid Rahmati
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Holger Haselmann
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Lars Schmidl
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747 Jena, Germany
| | - Daniel Hunter
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France
| | - Anna-Katherina Brauer
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Martinsried, Germany; Biomedical Center, Ludwig Maximilians University Munich, Martinsried, Germany
| | - Sabine Liebscher
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig Maximilians University Munich, Martinsried, Germany; Biomedical Center, Ludwig Maximilians University Munich, Martinsried, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jakob Kreye
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Laurent Groc
- Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, 33000 Bordeaux, France
| | - Stefan Hallermann
- Carl Ludwig Institute for Physiology, Faculty of Medicine, Leipzig University, 04103 Leipzig, Germany
| | - Josep Dalmau
- Catalan Institution for Research and Advanced Studies (ICREA) and IDIBAPS-Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Manfred Heckmann
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, 97070 Würzburg, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Department of Neurology, Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|
45
|
Krishnakumaran R, Ray S. Temporal characteristics of gamma rhythm constrain properties of noise in an inhibition-stabilized network model. Cereb Cortex 2023; 33:10108-10121. [PMID: 37492002 PMCID: PMC10502791 DOI: 10.1093/cercor/bhad270] [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: 02/10/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/27/2023] Open
Abstract
Gamma rhythm refers to oscillatory neural activity between 30 and 80 Hz, induced in visual cortex by stimuli such as iso-luminant hues or gratings. The power and peak frequency of gamma depend on the properties of the stimulus such as size and contrast. Gamma waveform is typically arch-shaped, with narrow troughs and broad peaks, and can be replicated in a self-oscillating Wilson-Cowan (WC) model operating in an appropriate regime. However, oscillations in this model are infinitely long, unlike physiological gamma that occurs in short bursts. Further, unlike the model, gamma is faster after stimulus onset and slows down over time. Here, we first characterized gamma burst duration in local field potential data recorded from two monkeys as they viewed full screen iso-luminant hues. We then added different types of noise in the inputs to the WC model and tested how that affected duration and temporal dynamics of gamma. While the model failed with the often-used Poisson noise, Ornstein-Uhlenbeck noise applied to both the excitatory and the inhibitory populations replicated the duration and slowing of gamma and replicated the shape and stimulus dependencies. Thus, the temporal dynamics of gamma oscillations put constraints on the type and properties of underlying neural noise.
Collapse
Affiliation(s)
- R Krishnakumaran
- IISc Mathematics Initiative, Department of Mathematics, Indian Institute of Science, C V Raman road, Bangalore 560012, Karnataka, India
| | - Supratim Ray
- IISc Mathematics Initiative, Department of Mathematics, Indian Institute of Science, C V Raman road, Bangalore 560012, Karnataka, India
- Centre for Neuroscience, Indian Institute of Science, C V Raman road, Bangalore 560012, Karnataka, India
| |
Collapse
|
46
|
Jacob MS, Sargent K, Roach BJ, Shamshiri EA, Mathalon DH, Ford JM. The Scanner as the Stimulus: Deficient Gamma-BOLD Coupling in Schizophrenia at Rest. Schizophr Bull 2023; 49:1364-1374. [PMID: 37098100 PMCID: PMC10483456 DOI: 10.1093/schbul/sbad014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Functional magnetic resonance imaging (fMRI) scanners are unavoidably loud and uncomfortable experimental tools that are necessary for schizophrenia (SZ) neuroscience research. The validity of fMRI paradigms might be undermined by well-known sensory processing abnormalities in SZ that could exert distinct effects on neural activity in the presence of scanner background sound. Given the ubiquity of resting-state fMRI (rs-fMRI) paradigms in SZ research, elucidating the relationship between neural, hemodynamic, and sensory processing deficits during scanning is necessary to refine the construct validity of the MR neuroimaging environment. We recorded simultaneous electroencephalography (EEG)-fMRI at rest in people with SZ (n = 57) and healthy control participants without a psychiatric diagnosis (n = 46) and identified gamma EEG activity in the same frequency range as the background sounds emitted from our scanner during a resting-state sequence. In participants with SZ, gamma coupling to the hemodynamic signal was reduced in bilateral auditory regions of the superior temporal gyri. Impaired gamma-hemodynamic coupling was associated with sensory gating deficits and worse symptom severity. Fundamental sensory-neural processing deficits in SZ are present at rest when considering scanner background sound as a "stimulus." This finding may impact the interpretation of rs-fMRI activity in studies of people with SZ. Future neuroimaging research in SZ might consider background sound as a confounding variable, potentially related to fluctuations in neural excitability and arousal.
Collapse
Affiliation(s)
- Michael S Jacob
- Mental Health Service, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121, USA
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Kaia Sargent
- Mental Health Service, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121, USA
| | - Brian J Roach
- Mental Health Service, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121, USA
| | - Elhum A Shamshiri
- Mental Health Service, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121, USA
| | - Daniel H Mathalon
- Mental Health Service, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121, USA
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Judith M Ford
- Mental Health Service, San Francisco VA Medical Center, 4150 Clement St, San Francisco, CA 94121, USA
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
47
|
Menétrey MQ, Herzog MH, Pascucci D. Pre-stimulus alpha activity modulates long-lasting unconscious feature integration. Neuroimage 2023; 278:120298. [PMID: 37517573 DOI: 10.1016/j.neuroimage.2023.120298] [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: 03/28/2023] [Revised: 06/28/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023] Open
Abstract
Pre-stimulus alpha (α) activity can influence perception of shortly presented, low-contrast stimuli. The underlying mechanisms are often thought to affect perception exactly at the time of presentation. In addition, it is suggested that α cycles determine temporal windows of integration. However, in everyday situations, stimuli are usually presented for periods longer than ∼100 ms and perception is often an integration of information across space and time. Moving objects are just one example. Hence, the question is whether α activity plays a role also in temporal integration, especially when stimuli are integrated over several α cycles. Using electroencephalography (EEG), we investigated the relationship between pre-stimulus brain activity and long-lasting integration in the sequential metacontrast paradigm (SQM), where two opposite vernier offsets, embedded in a stream of lines, are unconsciously integrated into a single percept. We show that increases in α power, even 300 ms before the stimulus, affected the probability of reporting the first offset, shown at the very beginning of the SQM. This effect was mediated by the systematic slowing of the α rhythm that followed the peak in α power. No phase effects were found. Together, our results demonstrate a cascade of neural changes, following spontaneous bursts of α activity and extending beyond a single moment, which influences the sensory representation of visual features for hundreds of milliseconds. Crucially, as feature integration in the SQM occurs before a conscious percept is elicited, this also provides evidence that α activity is linked to mechanisms regulating unconscious processing.
Collapse
Affiliation(s)
- Maëlan Q Menétrey
- Laboratory of Psychophysics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Michael H Herzog
- Laboratory of Psychophysics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - David Pascucci
- Laboratory of Psychophysics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
48
|
Comeaux P, Clark K, Noudoost B. A recruitment through coherence theory of working memory. Prog Neurobiol 2023; 228:102491. [PMID: 37393039 PMCID: PMC10530428 DOI: 10.1016/j.pneurobio.2023.102491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
The interactions between prefrontal cortex and other areas during working memory have been studied for decades. Here we outline a conceptual framework describing interactions between these areas during working memory, and review evidence for key elements of this model. We specifically suggest that a top-down signal sent from prefrontal to sensory areas drives oscillations in these areas. Spike timing within sensory areas becomes locked to these working-memory-driven oscillations, and the phase of spiking conveys information about the representation available within these areas. Downstream areas receiving these phase-locked spikes from sensory areas can recover this information via a combination of coherent oscillations and gating of input efficacy based on the phase of their local oscillations. Although the conceptual framework is based on prefrontal interactions with sensory areas during working memory, we also discuss the broader implications of this framework for flexible communication between brain areas in general.
Collapse
Affiliation(s)
- Phillip Comeaux
- Dept. of Biomedical Engineering, University of Utah, 36 S. Wasatch Drive, Salt Lake City, UT 84112, USA; Dept. of Ophthalmology and Visual Sciences, University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Kelsey Clark
- Dept. of Ophthalmology and Visual Sciences, University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA
| | - Behrad Noudoost
- Dept. of Ophthalmology and Visual Sciences, University of Utah, 65 Mario Capecchi Drive, Salt Lake City, UT 84132, USA.
| |
Collapse
|
49
|
Kumar M, Handy G, Kouvaros S, Zhao Y, Brinson LL, Wei E, Bizup B, Doiron B, Tzounopoulos T. Cell-type-specific plasticity of inhibitory interneurons in the rehabilitation of auditory cortex after peripheral damage. Nat Commun 2023; 14:4170. [PMID: 37443148 PMCID: PMC10345144 DOI: 10.1038/s41467-023-39732-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Peripheral sensory organ damage leads to compensatory cortical plasticity that is associated with a remarkable recovery of cortical responses to sound. The precise mechanisms that explain how this plasticity is implemented and distributed over a diverse collection of excitatory and inhibitory cortical neurons remain unknown. After noise trauma and persistent peripheral deficits, we found recovered sound-evoked activity in mouse A1 excitatory principal neurons (PNs), parvalbumin- and vasoactive intestinal peptide-expressing neurons (PVs and VIPs), but reduced activity in somatostatin-expressing neurons (SOMs). This cell-type-specific recovery was also associated with cell-type-specific intrinsic plasticity. These findings, along with our computational modelling results, are consistent with the notion that PV plasticity contributes to PN stability, SOM plasticity allows for increased PN and PV activity, and VIP plasticity enables PN and PV recovery by inhibiting SOMs.
Collapse
Affiliation(s)
- Manoj Kumar
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Gregory Handy
- Departments of Neurobiology and Statistics, University of Chicago, Chicago, IL, 60637, USA
| | - Stylianos Kouvaros
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yanjun Zhao
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Lovisa Ljungqvist Brinson
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Eric Wei
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Brandon Bizup
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Brent Doiron
- Departments of Neurobiology and Statistics, University of Chicago, Chicago, IL, 60637, USA
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
50
|
Bang JW, Parra C, Yu K, Wollstein G, Schuman JS, Chan KC. GABA decrease is associated with degraded neural specificity in the visual cortex of glaucoma patients. Commun Biol 2023; 6:679. [PMID: 37386293 PMCID: PMC10310759 DOI: 10.1038/s42003-023-04918-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/05/2023] [Indexed: 07/01/2023] Open
Abstract
Glaucoma is an age-related neurodegenerative disease of the visual system, affecting both the eye and the brain. Yet its underlying metabolic mechanisms and neurobehavioral relevance remain largely unclear. Here, using proton magnetic resonance spectroscopy and functional magnetic resonance imaging, we investigated the GABAergic and glutamatergic systems in the visual cortex of glaucoma patients, as well as neural specificity, which is shaped by GABA and glutamate signals and underlies efficient sensory and cognitive functions. Our study shows that among the older adults, both GABA and glutamate levels decrease with increasing glaucoma severity regardless of age. Further, our study shows that the reduction of GABA but not glutamate predicts the neural specificity. This association is independent of the impairments on the retina structure, age, and the gray matter volume of the visual cortex. Our results suggest that glaucoma-specific decline of GABA undermines neural specificity in the visual cortex and that targeting GABA could improve the neural specificity in glaucoma.
Collapse
Affiliation(s)
- Ji Won Bang
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10017, USA.
| | - Carlos Parra
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10017, USA
| | - Kevin Yu
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10017, USA
| | - Gadi Wollstein
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10017, USA
- Center for Neural Science, College of Arts and Science, New York University, New York, New York, 10003, USA
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, New York, 11201, USA
| | - Joel S Schuman
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10017, USA
- Center for Neural Science, College of Arts and Science, New York University, New York, New York, 10003, USA
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, New York, 11201, USA
- Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10016, USA
| | - Kevin C Chan
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10017, USA.
- Center for Neural Science, College of Arts and Science, New York University, New York, New York, 10003, USA.
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, New York, 11201, USA.
- Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10016, USA.
- Department of Radiology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, 10016, USA.
| |
Collapse
|