1
|
Huang C, Englitz B, Reznik A, Zeldenrust F, Celikel T. Information transfer and recovery for the sense of touch. Cereb Cortex 2025; 35:bhaf073. [PMID: 40197640 PMCID: PMC11976729 DOI: 10.1093/cercor/bhaf073] [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: 10/19/2024] [Revised: 11/26/2024] [Accepted: 01/02/2025] [Indexed: 04/10/2025] Open
Abstract
Transformation of postsynaptic potentials into action potentials is the rate-limiting step of communication in neural networks. The efficiency of this intracellular information transfer also powerfully shapes stimulus representations in sensory cortices. Using whole-cell recordings and information-theoretic measures, we show herein that somatic postsynaptic potentials accurately represent stimulus location on a trial-by-trial basis in single neurons, even 4 synapses away from the sensory periphery in the whisker system. This information is largely lost during action potential generation but can be rapidly (<20 ms) recovered using complementary information in local populations in a cell-type-specific manner. These results show that as sensory information is transferred from one neural locus to another, the circuits reconstruct the stimulus with high fidelity so that sensory representations of single neurons faithfully represent the stimulus in the periphery, but only in their postsynaptic potentials, resulting in lossless information processing for the sense of touch in the primary somatosensory cortex.
Collapse
Affiliation(s)
- Chao Huang
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
- Laboratory of Neural Circuits and Plasticity, University of Southern California, 3616 Watt Way, Los Angeles, CA 90089, United States
| | - Bernhard Englitz
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Andrey Reznik
- Laboratory of Neural Circuits and Plasticity, University of Southern California, 3616 Watt Way, Los Angeles, CA 90089, United States
| | - Fleur Zeldenrust
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
| | - Tansu Celikel
- Department of Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands
- School of Psychology, Georgia Institute of Technology, 654 Cherry Street, Atlanta, GA 30332-0170, United States
| |
Collapse
|
2
|
Towner TT, Coleman HJ, Goyden MA, Vore AS, Papastrat KM, Varlinskaya EI, Werner DF. Prelimbic cortex perineuronal net expression and social behavior: Impact of adolescent intermittent ethanol exposure. Neuropharmacology 2025; 262:110195. [PMID: 39437849 DOI: 10.1016/j.neuropharm.2024.110195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/05/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Adolescent intermittent ethanol (AIE) exposure in rats leads to social deficits. Parvalbumin (PV) expressing fast-spiking interneurons in the prelimbic cortex (PrL) contribute to social behavior, and perineuronal nets (PNNs) within the PrL preferentially encompass and regulate PV interneurons. AIE exposure increases PNNs, but it is unknown if this upregulation contributes to AIE-induced social impairments. The current study was designed to determine the effect of AIE exposure on PNN expression in the PrL and to assess whether PNN dysregulation contributes to social deficits elicited by AIE. cFos-LacZ male and female rats were exposed every other day to tap water or ethanol (4 g/kg, 25% w/v) via intragastric gavage between postnatal day (P) 25-45. We evaluated neuronal activation by β-galactosidase expression and PNN levels either at the end of the exposure regimen on P45 and/or in adulthood on P70. In addition, we used Chondroitinase ABC (ChABC) to deplete PNNs following adolescent exposure (P48) and allowed for PNN restoration before social testing in adulthhod. AIE exposure increased PNN expression in the PrL of adult males, but decreased PNNs immediately following AIE. Vesicular glutamate transporter 2 (vGlut2) and vesicular GABA transporter (vGat) near PNNs were downregulated only in AIE-exposed females. Gene expression of PNN components was largely unaffected by AIE exposure. Removal and reestablishment of PrL PNNs by ChABC led to upregulation of PNNs and social impairments in males, regardless of adolescent exposure. These data suggest that AIE exposure in males upregulates PrL PNNs that likely contribute to social impairments induced by AIE.
Collapse
Affiliation(s)
- Trevor T Towner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Harper J Coleman
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Matthew A Goyden
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Andrew S Vore
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Kimberly M Papastrat
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Elena I Varlinskaya
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - David F Werner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA.
| |
Collapse
|
3
|
Farkhondeh Tale Navi F, Heysieattalab S, Raoufy MR, Sabaghypour S, Nazari M, Nazari MA. Adaptive closed-loop modulation of cortical theta oscillations: Insights into the neural dynamics of navigational decision-making. Brain Stimul 2024; 17:1101-1118. [PMID: 39277130 DOI: 10.1016/j.brs.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 08/04/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024] Open
Abstract
Navigational decision-making tasks, such as spatial working memory (SWM), rely highly on information integration from several cortical and sub-cortical regions. Performance in SWM tasks is associated with theta rhythm, including low-frequency oscillations related to movement and memory. The interaction of the ventral hippocampus (vHPC) and medial prefrontal cortex (mPFC), reflected in theta synchrony, is essential in various steps of information processing during SWM. We used a closed-loop neurofeedback (CLNF) system to upregulate theta power in the mPFC and investigate its effects on circuit dynamics and behavior in animal models. Specifically, we hypothesized that enhancing the power of the theta rhythm in the mPFC might improve SWM performance. Animals were divided into three groups: closed-loop (CL), random-loop (RL), and OFF (without stimulation). We recorded local field potential (LFP) in the mPFC while electrical reward stimulation contingent on cortical theta activity was delivered to the lateral hypothalamus (LH), which is considered one of the central reward-associated regions. We also recorded LFP in the vHPC to evaluate the related subcortical neural changes. Results revealed a sustained increase in the theta power in both mPFC and vHPC for the CL group. Our analysis also revealed an increase in mPFC-vHPC synchronization in the theta range over the stimulation sessions in the CL group, as measured by coherence and cross-correlation in the theta frequency band. The reinforcement of this circuit improved spatial decision-making performance in the subsequent behavioral results. Our findings provide direct evidence of the relationship between specific theta upregulation and SWM performance and suggest that theta oscillations are integral to cognitive processes. Overall, this study highlights the potential of adaptive CLNF systems in investigating neural dynamics in various brain circuits.
Collapse
Affiliation(s)
- Farhad Farkhondeh Tale Navi
- Department of Cognitive Neuroscience, Faculty of Education and Psychology, University of Tabriz, Tabriz, Iran
| | - Soomaayeh Heysieattalab
- Department of Cognitive Neuroscience, Faculty of Education and Psychology, University of Tabriz, Tabriz, Iran
| | - Mohammad Reza Raoufy
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Saied Sabaghypour
- Department of Cognitive Neuroscience, Faculty of Education and Psychology, University of Tabriz, Tabriz, Iran
| | - Milad Nazari
- Department of Molecular Biology and Genetics, Aarhus University, Denmark
| | - Mohammad Ali Nazari
- Department of Cognitive Neuroscience, Faculty of Education and Psychology, University of Tabriz, Tabriz, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Vaughn MJ, Laswick Z, Wang H, Haas JS. Functionally Distinct Circuits Are Linked by Heterocellular Electrical Synapses in the Thalamic Reticular Nucleus. eNeuro 2024; 11:ENEURO.0269-23.2023. [PMID: 38164593 PMCID: PMC10849028 DOI: 10.1523/eneuro.0269-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/10/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
The thalamic reticular nucleus (TRN) inhibits sensory thalamocortical relay neurons and is a key regulator of sensory attention as well as sleep and wake states. Recent developments have identified two distinct genetic subtypes of TRN neurons, calbindin-expressing (CB) and somatostatin-expressing (SOM) neurons. These subtypes differ in localization within the TRN, electrophysiological properties, and importantly, targeting of thalamocortical relay channels. CB neurons send inhibition to and receive excitation from first-order thalamic relay nuclei, while SOM neurons send inhibition to and receive excitation from higher-order thalamic areas. These differences create distinct channels of information flow. It is unknown whether TRN neurons form electrical synapses between SOM and CB neurons and consequently bridge first-order and higher-order thalamic channels. Here, we use GFP reporter mice to label and record from CB-expressing and SOM-expressing TRN neurons. We confirm that GFP expression properly differentiates TRN subtypes based on electrophysiological differences, and we identified electrical synapses between pairs of neurons with and without common GFP expression for both CB and SOM types. That is, electrical synapses link both within and across subtypes of neurons in the TRN, forming either homocellular or heterocellular synapses. Therefore, we conclude that electrical synapses within the TRN provide a substrate for functionally linking thalamocortical first-order and higher-order channels within the TRN.
Collapse
Affiliation(s)
- Mitchell J Vaughn
- Department of Biological Sciences, Lehigh University, Bethlehem 18015, Pennsylvania
| | - Zachary Laswick
- Department of Biological Sciences, Lehigh University, Bethlehem 18015, Pennsylvania
| | - Huaixing Wang
- Department of Biological Sciences, Lehigh University, Bethlehem 18015, Pennsylvania
| | - Julie S Haas
- Department of Biological Sciences, Lehigh University, Bethlehem 18015, Pennsylvania
| |
Collapse
|
5
|
Zhang B, Li L, Tang X, Zeng J, Song Y, Hou Z, Ma T, Afewerky HK, Li H, Lu Y, He A, Li X. Distribution Patterns of Subgroups of Inhibitory Neurons Divided by Calbindin 1. Mol Neurobiol 2023; 60:7285-7296. [PMID: 37548854 DOI: 10.1007/s12035-023-03542-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/25/2023] [Indexed: 08/08/2023]
Abstract
The inhibitory neurons in the brain play an essential role in neural network firing patterns by releasing γ-aminobutyric acid (GABA) as the neurotransmitter. In the mouse brain, based on the protein molecular markers, inhibitory neurons are usually to be divided into three non-overlapping groups: parvalbumin (PV), neuropeptide somatostatin (SST), and vasoactive intestinal peptide (VIP)-expressing neurons. Each neuronal group exhibited unique properties in molecule, electrophysiology, circuitry, and function. Calbindin 1 (Calb1), a ubiquitous calcium-binding protein, often acts as a "divider" in excitatory neuronal classification. Based on Calb1 expression, the excitatory neurons from the same brain region can be classified into two subgroups with distinct properties. Besides excitatory neurons, Calb1 also expresses in part of inhibitory neurons. But, to date, little research focused on the intersectional relationship between inhibitory neuronal subtypes and Calb1. In this study, we genetically targeted Calb1-expression (Calb1+) and Calb1-lacking (Calb1-) subgroups of PV and SST neurons throughout the mouse brain by flexibly crossing transgenic mice relying on multi-recombinant systems, and the distribution patterns and electrophysiological properties of each subgroup were further demonstrated. Thus, this study provided novel insights and strategies into inhibitory neuronal classification.
Collapse
Affiliation(s)
- Bing Zhang
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lanfang Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaomei Tang
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinyu Zeng
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yige Song
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhenye Hou
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Henok Kessete Afewerky
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Youming Lu
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Aodi He
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xinyan Li
- Institute for Brain Research, Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
6
|
Jung YJ, Sun SH, Almasi A, Yunzab M, Meffin H, Ibbotson MR. Characterization of extracellular spike waveforms recorded in wallaby primary visual cortex. Front Neurosci 2023; 17:1244952. [PMID: 37746137 PMCID: PMC10517629 DOI: 10.3389/fnins.2023.1244952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/25/2023] [Indexed: 09/26/2023] Open
Abstract
Extracellular recordings were made from 642 units in the primary visual cortex (V1) of a highly visual marsupial, the Tammar wallaby. The receptive field (RF) characteristics of the cells were objectively estimated using the non-linear input model (NIM), and these were correlated with spike shapes. We found that wallaby cortical units had 68% regular spiking (RS), 12% fast spiking (FS), 4% triphasic spiking (TS), 5% compound spiking (CS) and 11% positive spiking (PS). RS waveforms are most often associated with recordings from pyramidal or spiny stellate cell bodies, suggesting that recordings from these cell types dominate in the wallaby cortex. In wallaby, 70-80% of FS and RS cells had orientation selective RFs and had evenly distributed linear and nonlinear RFs. We found that 47% of wallaby PS units were non-orientation selective and they were dominated by linear RFs. Previous studies suggest that the PS units represent recordings from the axon terminals of non-orientation selective cells originating in the lateral geniculate nucleus (LGN). If this is also true in wallaby, as strongly suggested by their low response latencies and bursty spiking properties, the results suggest that significantly more neurons in wallaby LGN are already orientation selective. In wallaby, less than 10% of recorded spikes had triphasic (TS) or sluggish compound spiking (CS) waveforms. These units had a mixture of orientation selective and non-oriented properties, and their cellular origins remain difficult to classify.
Collapse
Affiliation(s)
- Young Jun Jung
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, Australia
- National Vision Research Institute, Australian College of Optometry Carlton, Carlton, VIC, Australia
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Shi H. Sun
- National Vision Research Institute, Australian College of Optometry Carlton, Carlton, VIC, Australia
| | - Ali Almasi
- National Vision Research Institute, Australian College of Optometry Carlton, Carlton, VIC, Australia
| | - Molis Yunzab
- National Vision Research Institute, Australian College of Optometry Carlton, Carlton, VIC, Australia
| | - Hamish Meffin
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, Australia
| | - Michael R. Ibbotson
- Department of Biomedical Engineering, The University of Melbourne, Melbourne, VIC, Australia
- National Vision Research Institute, Australian College of Optometry Carlton, Carlton, VIC, Australia
- Department of Optometry and Vision Sciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Bod R, Tóth K, Essam N, Tóth EZ, Erõss L, Entz L, Bagó AG, Fabó D, Ulbert I, Wittner L. Synaptic alterations and neuronal firing in human epileptic neocortical excitatory networks. Front Synaptic Neurosci 2023; 15:1233569. [PMID: 37635750 PMCID: PMC10450510 DOI: 10.3389/fnsyn.2023.1233569] [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: 06/02/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Epilepsy is a prevalent neurological condition, with underlying neuronal mechanisms involving hyperexcitability and hypersynchrony. Imbalance between excitatory and inhibitory circuits, as well as histological reorganization are relatively well-documented in animal models or even in the human hippocampus, but less is known about human neocortical epileptic activity. Our knowledge about changes in the excitatory signaling is especially scarce, compared to that about the inhibitory cell population. This study investigated the firing properties of single neurons in the human neocortex in vitro, during pharmacological blockade of glutamate receptors, and additionally evaluated anatomical changes in the excitatory circuit in tissue samples from epileptic and non-epileptic patients. Both epileptic and non-epileptic tissues exhibited spontaneous population activity (SPA), NMDA receptor antagonization reduced SPA recurrence only in epileptic tissue, whereas further blockade of AMPA/kainate receptors reversibly abolished SPA emergence regardless of epilepsy. Firing rates did not significantly change in excitatory principal cells and inhibitory interneurons during pharmacological experiments. Granular layer (L4) neurons showed an increased firing rate in epileptic compared to non-epileptic tissue. The burstiness of neurons remained unchanged, except for that of inhibitory cells in epileptic recordings, which decreased during blockade of glutamate receptors. Crosscorrelograms computed from single neuron discharge revealed both mono- and polysynaptic connections, particularly involving intrinsically bursting principal cells. Histological investigations found similar densities of SMI-32-immunopositive long-range projecting pyramidal cells in both groups, and shorter excitatory synaptic active zones with a higher proportion of perforated synapses in the epileptic group. These findings provide insights into epileptic modifications from the perspective of the excitatory system and highlight discrete alterations in firing patterns and synaptic structure. Our data suggest that NMDA-dependent glutamatergic signaling, as well as the excitatory synaptic machinery are perturbed in epilepsy, which might contribute to epileptic activity in the human neocortex.
Collapse
Affiliation(s)
- Réka Bod
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
- Semmelweis University Doctoral School, Budapest, Hungary
| | - Kinga Tóth
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Nour Essam
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Estilla Zsófia Tóth
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
- Semmelweis University Doctoral School, Budapest, Hungary
| | - Loránd Erõss
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| | - László Entz
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| | - Attila G. Bagó
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| | - Dániel Fabó
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| | - István Ulbert
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
- Semmelweis University Doctoral School, Budapest, Hungary
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Lucia Wittner
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
- Semmelweis University Doctoral School, Budapest, Hungary
- National Institute of Mental Health, Neurology and Neurosurgery, Budapest, Hungary
| |
Collapse
|
8
|
Gebicke-Haerter PJ. The computational power of the human brain. Front Cell Neurosci 2023; 17:1220030. [PMID: 37608987 PMCID: PMC10441807 DOI: 10.3389/fncel.2023.1220030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/05/2023] [Indexed: 08/24/2023] Open
Abstract
At the end of the 20th century, analog systems in computer science have been widely replaced by digital systems due to their higher computing power. Nevertheless, the question keeps being intriguing until now: is the brain analog or digital? Initially, the latter has been favored, considering it as a Turing machine that works like a digital computer. However, more recently, digital and analog processes have been combined to implant human behavior in robots, endowing them with artificial intelligence (AI). Therefore, we think it is timely to compare mathematical models with the biology of computation in the brain. To this end, digital and analog processes clearly identified in cellular and molecular interactions in the Central Nervous System are highlighted. But above that, we try to pinpoint reasons distinguishing in silico computation from salient features of biological computation. First, genuinely analog information processing has been observed in electrical synapses and through gap junctions, the latter both in neurons and astrocytes. Apparently opposed to that, neuronal action potentials (APs) or spikes represent clearly digital events, like the yes/no or 1/0 of a Turing machine. However, spikes are rarely uniform, but can vary in amplitude and widths, which has significant, differential effects on transmitter release at the presynaptic terminal, where notwithstanding the quantal (vesicular) release itself is digital. Conversely, at the dendritic site of the postsynaptic neuron, there are numerous analog events of computation. Moreover, synaptic transmission of information is not only neuronal, but heavily influenced by astrocytes tightly ensheathing the majority of synapses in brain (tripartite synapse). At least at this point, LTP and LTD modifying synaptic plasticity and believed to induce short and long-term memory processes including consolidation (equivalent to RAM and ROM in electronic devices) have to be discussed. The present knowledge of how the brain stores and retrieves memories includes a variety of options (e.g., neuronal network oscillations, engram cells, astrocytic syncytium). Also epigenetic features play crucial roles in memory formation and its consolidation, which necessarily guides to molecular events like gene transcription and translation. In conclusion, brain computation is not only digital or analog, or a combination of both, but encompasses features in parallel, and of higher orders of complexity.
Collapse
Affiliation(s)
- Peter J. Gebicke-Haerter
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
9
|
Lee SN, Cho HJ, Jeong H, Ryu B, Lee HJ, Kim M, Yoo J, Woo JS, Lee HH. Cryo-EM structures of human Cx36/GJD2 neuronal gap junction channel. Nat Commun 2023; 14:1347. [PMID: 36906653 PMCID: PMC10008584 DOI: 10.1038/s41467-023-37040-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 02/28/2023] [Indexed: 03/13/2023] Open
Abstract
Connexin 36 (Cx36) is responsible for signal transmission in electrical synapses by forming interneuronal gap junctions. Despite the critical role of Cx36 in normal brain function, the molecular architecture of the Cx36 gap junction channel (GJC) is unknown. Here, we determine cryo-electron microscopy structures of Cx36 GJC at 2.2-3.6 Å resolutions, revealing a dynamic equilibrium between its closed and open states. In the closed state, channel pores are obstructed by lipids, while N-terminal helices (NTHs) are excluded from the pore. In the open state with pore-lining NTHs, the pore is more acidic than those in Cx26 and Cx46/50 GJCs, explaining its strong cation selectivity. The conformational change during channel opening also includes the α-to-π-helix transition of the first transmembrane helix, which weakens the protomer-protomer interaction. Our structural analyses provide high resolution information on the conformational flexibility of Cx36 GJC and suggest a potential role of lipids in the channel gating.
Collapse
Affiliation(s)
- Seu-Na Lee
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Hwa-Jin Cho
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, 08826, Korea
| | - Hyeongseop Jeong
- Center for Research Equipment, Korea Basic Science Institute, Chungcheongbuk-do, 28119, Korea
| | - Bumhan Ryu
- Research Solution Center, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Hyuk-Joon Lee
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Minsoo Kim
- Department of Physics, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Jejoong Yoo
- Department of Physics, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Jae-Sung Woo
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea.
| | - Hyung Ho Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
10
|
Ribarič S. Detecting Early Cognitive Decline in Alzheimer's Disease with Brain Synaptic Structural and Functional Evaluation. Biomedicines 2023; 11:355. [PMID: 36830892 PMCID: PMC9952956 DOI: 10.3390/biomedicines11020355] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Early cognitive decline in patients with Alzheimer's (AD) is associated with quantifiable structural and functional connectivity changes in the brain. AD dysregulation of Aβ and tau metabolism progressively disrupt normal synaptic function, leading to loss of synapses, decreased hippocampal synaptic density and early hippocampal atrophy. Advances in brain imaging techniques in living patients have enabled the transition from clinical signs and symptoms-based AD diagnosis to biomarkers-based diagnosis, with functional brain imaging techniques, quantitative EEG, and body fluids sampling. The hippocampus has a central role in semantic and episodic memory processing. This cognitive function is critically dependent on normal intrahippocampal connections and normal hippocampal functional connectivity with many cortical regions, including the perirhinal and the entorhinal cortex, parahippocampal cortex, association regions in the temporal and parietal lobes, and prefrontal cortex. Therefore, decreased hippocampal synaptic density is reflected in the altered functional connectivity of intrinsic brain networks (aka large-scale networks), including the parietal memory, default mode, and salience networks. This narrative review discusses recent critical issues related to detecting AD-associated early cognitive decline with brain synaptic structural and functional markers in high-risk or neuropsychologically diagnosed patients with subjective cognitive impairment or mild cognitive impairment.
Collapse
Affiliation(s)
- Samo Ribarič
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
11
|
Bhatt M, Gauthier-Manuel L, Lazzarin E, Zerlotti R, Ziegler C, Bazzone A, Stockner T, Bossi E. A comparative review on the well-studied GAT1 and the understudied BGT-1 in the brain. Front Physiol 2023; 14:1145973. [PMID: 37123280 PMCID: PMC10137170 DOI: 10.3389/fphys.2023.1145973] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
γ-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the central nervous system (CNS). Its homeostasis is maintained by neuronal and glial GABA transporters (GATs). The four GATs identified in humans are GAT1 (SLC6A1), GAT2 (SLC6A13), GAT3 (SLC6A11), and betaine/GABA transporter-1 BGT-1 (SLC6A12) which are all members of the solute carrier 6 (SLC6) family of sodium-dependent transporters. While GAT1 has been investigated extensively, the other GABA transporters are less studied and their role in CNS is not clearly defined. Altered GABAergic neurotransmission is involved in different diseases, but the importance of the different transporters remained understudied and limits drug targeting. In this review, the well-studied GABA transporter GAT1 is compared with the less-studied BGT-1 with the aim to leverage the knowledge on GAT1 to shed new light on the open questions concerning BGT-1. The most recent knowledge on transporter structure, functions, expression, and localization is discussed along with their specific role as drug targets for neurological and neurodegenerative disorders. We review and discuss data on the binding sites for Na+, Cl-, substrates, and inhibitors by building on the recent cryo-EM structure of GAT1 to highlight specific molecular determinants of transporter functions. The role of the two proteins in GABA homeostasis is investigated by looking at the transport coupling mechanism, as well as structural and kinetic transport models. Furthermore, we review information on selective inhibitors together with the pharmacophore hypothesis of transporter substrates.
Collapse
Affiliation(s)
- Manan Bhatt
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Centre for Neuroscience—University of Insubria, Varese, Italy
| | - Laure Gauthier-Manuel
- Department of Biophysics II/Structural Biology, University of Regensburg, Regensburg, Germany
| | - Erika Lazzarin
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringerstr, Vienna
| | - Rocco Zerlotti
- Department of Biophysics II/Structural Biology, University of Regensburg, Regensburg, Germany
- Nanion Technologies GmbH, Munich, Germany
| | - Christine Ziegler
- Department of Biophysics II/Structural Biology, University of Regensburg, Regensburg, Germany
| | | | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, Waehringerstr, Vienna
- *Correspondence: Thomas Stockner, ; Elena Bossi,
| | - Elena Bossi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Centre for Neuroscience—University of Insubria, Varese, Italy
- *Correspondence: Thomas Stockner, ; Elena Bossi,
| |
Collapse
|
12
|
Herring CA, Simmons RK, Freytag S, Poppe D, Moffet JJD, Pflueger J, Buckberry S, Vargas-Landin DB, Clément O, Echeverría EG, Sutton GJ, Alvarez-Franco A, Hou R, Pflueger C, McDonald K, Polo JM, Forrest ARR, Nowak AK, Voineagu I, Martelotto L, Lister R. Human prefrontal cortex gene regulatory dynamics from gestation to adulthood at single-cell resolution. Cell 2022; 185:4428-4447.e28. [PMID: 36318921 DOI: 10.1016/j.cell.2022.09.039] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 07/19/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Human brain development is underpinned by cellular and molecular reconfigurations continuing into the third decade of life. To reveal cell dynamics orchestrating neural maturation, we profiled human prefrontal cortex gene expression and chromatin accessibility at single-cell resolution from gestation to adulthood. Integrative analyses define the dynamic trajectories of each cell type, revealing major gene expression reconfiguration at the prenatal-to-postnatal transition in all cell types followed by continuous reconfiguration into adulthood and identifying regulatory networks guiding cellular developmental programs, states, and functions. We uncover links between expression dynamics and developmental milestones, characterize the diverse timing of when cells acquire adult-like states, and identify molecular convergence from distinct developmental origins. We further reveal cellular dynamics and their regulators implicated in neurological disorders. Finally, using this reference, we benchmark cell identities and maturation states in organoid models. Together, this captures the dynamic regulatory landscape of human cortical development.
Collapse
Affiliation(s)
- Charles A Herring
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Rebecca K Simmons
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Saskia Freytag
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Daniel Poppe
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Joel J D Moffet
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Jahnvi Pflueger
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Sam Buckberry
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Dulce B Vargas-Landin
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Olivier Clément
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Enrique Goñi Echeverría
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Gavin J Sutton
- School of Biotechnology and Biomolecular Sciences, Cellular Genomics Futures Institute, and the RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| | - Alba Alvarez-Franco
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid 28029, Spain
| | - Rui Hou
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Christian Pflueger
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Kerrie McDonald
- Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jose M Polo
- Adelaide Centre for Epigenetics and the South Australian Immunogenomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3000, Australia
| | - Alistair R R Forrest
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Anna K Nowak
- Medical School, University of Western Australia, Perth, WA 6009, Australia
| | - Irina Voineagu
- School of Biotechnology and Biomolecular Sciences, Cellular Genomics Futures Institute, and the RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| | - Luciano Martelotto
- Adelaide Centre for Epigenetics and the South Australian Immunogenomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; University of Melbourne Centre for Cancer Research, Victoria Comprehensive Cancer Centre, Melbourne, VIC 3000, Australia
| | - Ryan Lister
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia.
| |
Collapse
|
13
|
Le Cong D, Sato D, Ikarashi K, Fujimoto T, Ochi G, Yamashiro K. Effect of whole-hand water flow stimulation on the neural balance between excitation and inhibition in the primary somatosensory cortex. Front Hum Neurosci 2022; 16:962936. [DOI: 10.3389/fnhum.2022.962936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Sustained peripheral somatosensory stimulations, such as high-frequency repetitive somatosensory stimulation (HF-RSS) and vibrated stimulation, are effective in altering the balance between excitation and inhibition in the somatosensory cortex (S1) and motor cortex (M1). A recent study reported that whole-hand water flow (WF) stimulation induced neural disinhibition in the M1. Based on previous results, we hypothesized that whole-hand WF stimulation would lead to neural disinhibition in the S1 because there is a strong neural connection between M1 and S1 and aimed to examine whether whole-hand WF stimulation would change the neural balance between excitation and inhibition in the S1. Nineteen healthy volunteers were studied by measuring excitation and inhibition in the S1 before and after each of the four 15-min interventions. The excitation and inhibition in the S1 were assessed using somatosensory evoked potentials (SEPs) and paired-pulse inhibition (PPI) induced by single- and paired-pulse stimulations, respectively. The four interventions were as follows: control, whole-hand water immersion, whole-hand WF, and HF-RSS. The results showed no significant changes in SEPs and PPI following any intervention. However, changes in PPI with an interstimulus interval (ISI) of 30 ms were significantly correlated with the baseline value before whole-hand WF. Thus, the present findings indicated that the whole-hand WF stimulation had a greater decreased neural inhibition in participants with higher neural inhibition in the S1 at baseline. Considering previous results on M1, the present results possibly show that S1 has lower plasticity than M1 and that the duration (15 min) of each intervention may not have been enough to alter the balance of excitation and inhibition in the S1.
Collapse
|
14
|
Jung K, Choi Y, Kwon HB. Cortical control of chandelier cells in neural codes. Front Cell Neurosci 2022; 16:992409. [PMID: 36299494 PMCID: PMC9588934 DOI: 10.3389/fncel.2022.992409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022] Open
Abstract
Various cortical functions arise from the dynamic interplay of excitation and inhibition. GABAergic interneurons that mediate synaptic inhibition display significant diversity in cell morphology, electrophysiology, plasticity rule, and connectivity. These heterogeneous features are thought to underlie their functional diversity. Emerging attention on specific properties of the various interneuron types has emphasized the crucial role of cell-type specific inhibition in cortical neural processing. However, knowledge is still limited on how each interneuron type forms distinct neural circuits and regulates network activity in health and disease. To dissect interneuron heterogeneity at single cell-type precision, we focus on the chandelier cell (ChC), one of the most distinctive GABAergic interneuron types that exclusively innervate the axon initial segments (AIS) of excitatory pyramidal neurons. Here we review the current understanding of the structural and functional properties of ChCs and their implications in behavioral functions, network activity, and psychiatric disorders. These findings provide insights into the distinctive roles of various single-type interneurons in cortical neural coding and the pathophysiology of cortical dysfunction.
Collapse
|
15
|
Vaughn MJ, Haas JS. On the Diverse Functions of Electrical Synapses. Front Cell Neurosci 2022; 16:910015. [PMID: 35755782 PMCID: PMC9219736 DOI: 10.3389/fncel.2022.910015] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Electrical synapses are the neurophysiological product of gap junctional pores between neurons that allow bidirectional flow of current between neurons. They are expressed throughout the mammalian nervous system, including cortex, hippocampus, thalamus, retina, cerebellum, and inferior olive. Classically, the function of electrical synapses has been associated with synchrony, logically following that continuous conductance provided by gap junctions facilitates the reduction of voltage differences between coupled neurons. Indeed, electrical synapses promote synchrony at many anatomical and frequency ranges across the brain. However, a growing body of literature shows there is greater complexity to the computational function of electrical synapses. The paired membranes that embed electrical synapses act as low-pass filters, and as such, electrical synapses can preferentially transfer spike after hyperpolarizations, effectively providing spike-dependent inhibition. Other functions include driving asynchronous firing, improving signal to noise ratio, aiding in discrimination of dissimilar inputs, or dampening signals by shunting current. The diverse ways by which electrical synapses contribute to neuronal integration merits furthers study. Here we review how functions of electrical synapses vary across circuits and brain regions and depend critically on the context of the neurons and brain circuits involved. Computational modeling of electrical synapses embedded in multi-cellular models and experiments utilizing optical control and measurement of cellular activity will be essential in determining the specific roles performed by electrical synapses in varying contexts.
Collapse
Affiliation(s)
- Mitchell J Vaughn
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| | - Julie S Haas
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, United States
| |
Collapse
|
16
|
Theta and gamma oscillatory dynamics in mouse models of Alzheimer's disease: A path to prospective therapeutic intervention. Neurosci Biobehav Rev 2022; 136:104628. [PMID: 35331816 DOI: 10.1016/j.neubiorev.2022.104628] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 12/26/2022]
Abstract
Understanding the neural basis of cognitive deficits, a key feature of Alzheimer's disease (AD), is imperative for achieving the therapy of the disease. Rhythmic oscillatory activities in neural systems are a fundamental mechanism for diverse brain functions, including cognition. In several neurological conditions like AD, aberrant neural oscillations have been shown to play a central role. Furthermore, manipulation of brain oscillations in animals has confirmed their impact on cognition and disease. In this article, we review the evidence from mouse models that shows how synchronized oscillatory activity is intricately linked to AD machinery. We primarily focus on recent reports showing abnormal oscillatory activities at theta and gamma frequencies in AD condition and their influence on cellular disturbances and cognitive impairments. A thorough comprehension of the role that neuronal oscillations play in AD pathology should pave the way to therapeutic interventions that can curb the disease.
Collapse
|
17
|
Intrinsic Sources and Functional Impacts of Asymmetry at Electrical Synapses. eNeuro 2022; 9:ENEURO.0469-21.2022. [PMID: 35135867 PMCID: PMC8925721 DOI: 10.1523/eneuro.0469-21.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/14/2022] [Accepted: 02/02/2022] [Indexed: 11/21/2022] Open
Abstract
Electrical synapses couple inhibitory neurons across the brain, underlying a variety of functions that are modifiable by activity. Despite recent advances, many functions and contributions of electrical synapses within neural circuitry remain underappreciated. Among these are the sources and impacts of electrical synapse asymmetry. Using multi-compartmental models of neurons coupled through dendritic electrical synapses, we investigated intrinsic factors that contribute to effective synaptic asymmetry and that result in modulation of spike timing and synchrony between coupled cells. We show that electrical synapse location along a dendrite, input resistance, internal dendritic resistance, or directional conduction of the electrical synapse itself each alter asymmetry as measured by coupling between cell somas. Conversely, we note that asymmetrical gap junction (GJ) conductance can be masked by each of these properties. Furthermore, we show that asymmetry modulates spike timing and latency of coupled cells by up to tens of milliseconds, depending on direction of conduction or dendritic location of the electrical synapse. Coordination of rhythmic activity between two cells also depends on asymmetry. These simulations illustrate that causes of asymmetry are diverse, may not be apparent in somatic measurements of electrical coupling, influence dendritic processing, and produce a variety of outcomes on spiking and synchrony of coupled cells. Our findings highlight aspects of electrical synapses that should always be included in experimental demonstrations of coupling, and when assembling simulated networks containing electrical synapses.
Collapse
|
18
|
Herrmann T, Gerth M, Dittmann R, Pensold D, Ungelenk M, Liebmann L, Hübner CA. Disruption of KCC2 in Parvalbumin-Positive Interneurons Is Associated With a Decreased Seizure Threshold and a Progressive Loss of Parvalbumin-Positive Interneurons. Front Mol Neurosci 2022; 14:807090. [PMID: 35185464 PMCID: PMC8850922 DOI: 10.3389/fnmol.2021.807090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023] Open
Abstract
GABAA receptors are ligand-gated ion channels, which are predominantly permeable for chloride. The neuronal K-Cl cotransporter KCC2 lowers the intraneuronal chloride concentration and thus plays an important role for GABA signaling. KCC2 loss-of-function is associated with seizures and epilepsy. Here, we show that KCC2 is expressed in the majority of parvalbumin-positive interneurons (PV-INs) of the mouse brain. PV-INs receive excitatory input from principle cells and in turn control principle cell activity by perisomatic inhibition and inhibitory input from other interneurons. Upon Cre-mediated disruption of KCC2 in mice, the polarity of the GABA response of PV-INs changed from hyperpolarization to depolarization for the majority of PV-INs. Reduced excitatory postsynaptic potential-spike (E-S) coupling and increased spontaneous inhibitory postsynaptic current (sIPSC) frequencies further suggest that PV-INs are disinhibited upon disruption of KCC2. In vivo, PV-IN-specific KCC2 knockout mice display a reduced seizure threshold and develop spontaneous sometimes fatal seizures. We further found a time dependent loss of PV-INs, which was preceded by an up-regulation of pro-apoptotic genes upon disruption of KCC2.
Collapse
|
19
|
Huang C, Zeldenrust F, Celikel T. Cortical Representation of Touch in Silico. Neuroinformatics 2022; 20:1013-1039. [PMID: 35486347 PMCID: PMC9588483 DOI: 10.1007/s12021-022-09576-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2022] [Indexed: 12/31/2022]
Abstract
With its six layers and ~ 12,000 neurons, a cortical column is a complex network whose function is plausibly greater than the sum of its constituents'. Functional characterization of its network components will require going beyond the brute-force modulation of the neural activity of a small group of neurons. Here we introduce an open-source, biologically inspired, computationally efficient network model of the somatosensory cortex's granular and supragranular layers after reconstructing the barrel cortex in soma resolution. Comparisons of the network activity to empirical observations showed that the in silico network replicates the known properties of touch representations and whisker deprivation-induced changes in synaptic strength induced in vivo. Simulations show that the history of the membrane potential acts as a spatial filter that determines the presynaptic population of neurons contributing to a post-synaptic action potential; this spatial filtering might be critical for synaptic integration of top-down and bottom-up information.
Collapse
Affiliation(s)
- Chao Huang
- grid.9647.c0000 0004 7669 9786Department of Biology, University of Leipzig, Leipzig, Germany
| | - Fleur Zeldenrust
- grid.5590.90000000122931605Donders Institute for Brain, Cognition, and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Tansu Celikel
- grid.5590.90000000122931605Donders Institute for Brain, Cognition, and Behaviour, Radboud University, Nijmegen, the Netherlands ,grid.213917.f0000 0001 2097 4943School of Psychology, Georgia Institute of Technology, Atlanta, GA USA
| |
Collapse
|
20
|
Effects of Ketamine and Midazolam on Simultaneous EEG/fMRI Data During Working Memory Processes. Brain Topogr 2021; 34:863-880. [PMID: 34642836 DOI: 10.1007/s10548-021-00876-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/20/2021] [Indexed: 10/20/2022]
Abstract
Reliable measures of cognitive brain activity from functional neuroimaging techniques may provide early indications of efficacy in clinical trials. Functional magnetic resonance imaging and electroencephalography provide complementary spatiotemporal information and simultaneous recording of these two modalities can remove inter-session drug response and environment variability. We sought to assess the effects of ketamine and midazolam on simultaneous electrophysiological and hemodynamic recordings during working memory (WM) processes. Thirty participants were included in a placebo-controlled, three-way crossover design with ketamine and midazolam. Compared to placebo, ketamine administration attenuated theta power increases and alpha power decreases and midazolam attenuated low beta band decreases to increasing WM load. Additionally, ketamine caused larger blood-oxygen-dependent (BOLD) signal increases in the supplementary motor area and angular gyrus, and weaker deactivations of the default mode network (DMN), whereas no difference was found between midazolam and placebo. Ketamine administration caused positive temporal correlations between frontal-midline theta (fm-theta) power and the BOLD signal to disappear and attenuated negative correlations. However, the relationship between fm-theta and the BOLD signal from DMN areas was maintained in some participants during ketamine administration, as increasing theta strength was associated with stronger BOLD signal reductions in these areas. The presence of, and ability to manipulate, both positive and negative associations between the BOLD signal and fm-theta suggest the presence of multiple fm-theta components involved in WM processes, with ketamine administration disrupting one or more of these theta-linked WM strategies.
Collapse
|
21
|
Gutman-Wei AY, Brown SP. Mechanisms Underlying Target Selectivity for Cell Types and Subcellular Domains in Developing Neocortical Circuits. Front Neural Circuits 2021; 15:728832. [PMID: 34630048 PMCID: PMC8497978 DOI: 10.3389/fncir.2021.728832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/25/2021] [Indexed: 11/25/2022] Open
Abstract
The cerebral cortex contains numerous neuronal cell types, distinguished by their molecular identity as well as their electrophysiological and morphological properties. Cortical function is reliant on stereotyped patterns of synaptic connectivity and synaptic function among these neuron types, but how these patterns are established during development remains poorly understood. Selective targeting not only of different cell types but also of distinct postsynaptic neuronal domains occurs in many brain circuits and is directed by multiple mechanisms. These mechanisms include the regulation of axonal and dendritic guidance and fine-scale morphogenesis of pre- and postsynaptic processes, lineage relationships, activity dependent mechanisms and intercellular molecular determinants such as transmembrane and secreted molecules, many of which have also been implicated in neurodevelopmental disorders. However, many studies of synaptic targeting have focused on circuits in which neuronal processes target different lamina, such that cell-type-biased connectivity may be confounded with mechanisms of laminar specificity. In the cerebral cortex, each cortical layer contains cell bodies and processes from intermingled neuronal cell types, an arrangement that presents a challenge for the development of target-selective synapse formation. Here, we address progress and future directions in the study of cell-type-biased synaptic targeting in the cerebral cortex. We highlight challenges to identifying developmental mechanisms generating stereotyped patterns of intracortical connectivity, recent developments in uncovering the determinants of synaptic target selection during cortical synapse formation, and current gaps in the understanding of cortical synapse specificity.
Collapse
Affiliation(s)
- Alan Y. Gutman-Wei
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Solange P. Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
22
|
Lee E, Lee S, Shin JJ, Choi W, Chung C, Lee S, Kim J, Ha S, Kim R, Yoo T, Yoo YE, Kim J, Noh YW, Rhim I, Lee SY, Kim W, Lee T, Shin H, Cho IJ, Deisseroth K, Kim SJ, Park JM, Jung MW, Paik SB, Kim E. Excitatory synapses and gap junctions cooperate to improve Pv neuronal burst firing and cortical social cognition in Shank2-mutant mice. Nat Commun 2021; 12:5116. [PMID: 34433814 PMCID: PMC8387434 DOI: 10.1038/s41467-021-25356-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
NMDA receptor (NMDAR) and GABA neuronal dysfunctions are observed in animal models of autism spectrum disorders, but how these dysfunctions impair social cognition and behavior remains unclear. We report here that NMDARs in cortical parvalbumin (Pv)-positive interneurons cooperate with gap junctions to promote high-frequency (>80 Hz) Pv neuronal burst firing and social cognition. Shank2–/– mice, displaying improved sociability upon NMDAR activation, show impaired cortical social representation and inhibitory neuronal burst firing. Cortical Shank2–/– Pv neurons show decreased NMDAR activity, which suppresses the cooperation between NMDARs and gap junctions (GJs) for normal burst firing. Shank2–/– Pv neurons show compensatory increases in GJ activity that are not sufficient for social rescue. However, optogenetic boosting of Pv neuronal bursts, requiring GJs, rescues cortical social cognition in Shank2–/– mice, similar to the NMDAR-dependent social rescue. Therefore, NMDARs and gap junctions cooperate to promote cortical Pv neuronal bursts and social cognition. How NMDAR and GABA neuronal dysfunctions result in impaired social behaviour is unclear. Here, the authors show that NMDARs and gap junctions in cortical PV interneurons modulate burst firing, affecting social behaviour.
Collapse
Affiliation(s)
- Eunee Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea.,Department of Anatomy, College of Medicine, Yonsei University, Seoul, Korea
| | - Seungjoon Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Jae Jin Shin
- Department of Brain and Cognitive Science, College of Natural Science, Seoul National University, Seoul, Korea.,Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Korea
| | - Woochul Choi
- Program of Brain and Cognitive Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Korea
| | - Changuk Chung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Suho Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Jihye Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Seungmin Ha
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Ryunhee Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Taesun Yoo
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Ye-Eun Yoo
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Jisoo Kim
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Young Woo Noh
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Issac Rhim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Soo Yeon Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Woohyun Kim
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Taekyung Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Karl Deisseroth
- Department of Bioengineering, Department of Psychiatry and Behavioral Sciences, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Sang Jeong Kim
- Department of Physiology, College of Medicine, Seoul National University, Seoul, Korea
| | - Joo Min Park
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Korea.
| | - Min Whan Jung
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea. .,Department of Biological Sciences, KAIST, Daejeon, Korea.
| | - Se-Bum Paik
- Program of Brain and Cognitive Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute for Science and Technology (KAIST), Daejeon, Korea.
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea. .,Department of Biological Sciences, KAIST, Daejeon, Korea.
| |
Collapse
|
23
|
Choi U, Wang H, Hu M, Kim S, Sieburth D. Presynaptic coupling by electrical synapses coordinates a rhythmic behavior by synchronizing the activities of a neuron pair. Proc Natl Acad Sci U S A 2021; 118:e2022599118. [PMID: 33972428 PMCID: PMC8157971 DOI: 10.1073/pnas.2022599118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Electrical synapses are specialized structures that mediate the flow of electrical currents between neurons and have well known roles in synchronizing the activities of neuronal populations, both by mediating the current transfer from more active to less active neurons and by shunting currents from active neurons to their less active neighbors. However, how these positive and negative functions of electrical synapses are coordinated to shape rhythmic synaptic outputs and behavior is not well understood. Here, using a combination of genetics, behavioral analysis, and live calcium imaging in Caenorhabditis elegans, we show that electrical synapses formed by the gap junction protein INX-1/innexin couple the presynaptic terminals of a pair of motor neurons (AVL and DVB) to synchronize their activation in response to a pacemaker signal. Live calcium imaging reveals that inx-1/innexin mutations lead to asynchronous activation of AVL and DVB, due, in part, to loss of AVL-mediated activation of DVB by the pacemaker. In addition, loss of inx-1 leads to the ectopic activation of DVB at inappropriate times during the cycle through the activation of the L-type voltage-gated calcium channel EGL-19. We propose that electrical synapses between AVL and DVB presynaptic terminals function to ensure the precise and robust execution of a specific step in a rhythmic behavior by both synchronizing the activities of presynaptic terminals in response to pacemaker signaling and by inhibiting their activation in between cycles when pacemaker signaling is low.
Collapse
Affiliation(s)
- Ukjin Choi
- Development, Stem Cell, and Regenerative Medicine Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Han Wang
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Mingxi Hu
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Sungjin Kim
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033;
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
24
|
Matsuda YT, Miyamoto H, Joho RH, Hensch TK. K v3.1 channels regulate the rate of critical period plasticity. Neurosci Res 2021; 167:3-10. [PMID: 33872635 DOI: 10.1016/j.neures.2021.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/18/2022]
Abstract
Experience-dependent plasticity within visual cortex is controlled by postnatal maturation of inhibitory circuits, which are both morphologically diverse and precisely connected. Gene-targeted disruption of the voltage-dependent potassium channel Kv3.1 broadens action potentials and reduces net inhibitory function of parvalbumin (PV)-positive GABA subtypes within the neocortex. In mice lacking Kv3.1, the rate of input loss from an eye deprived of vision was slowed two-fold, despite otherwise normal critical period timecourse and receptive field properties. Rapid ocular dominance plasticity was restored by local or systemic enhancement of GABAergic transmission with acute benzodiazepine infusion. Diazepam instead exacerbated a global suppression of slow-wave oscillations during sleep described previously in these mutant mice, which therefore did not account for the rescued plasticity. Rapid ocular dominance shifts closely reflected Kv3.1 gene dosage that prevented prolonged spike discharge of their target pyramidal cells in vivo or the spike amplitude decrement of fast-spiking cells during bouts of high-frequency firing in vitro. Late postnatal expression of this unique channel in fast-spiking interneurons thus subtly regulates the speed of critical period plasticity with implications for mental illnesses.
Collapse
Affiliation(s)
- Yoshi-Taka Matsuda
- Laboratory for Neuronal Circuit Development, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan; Department of Child Studies, Shiraume Gakuen University, 1-830 Kodaira-shi, Tokyo, 187-8570 Japan
| | - Hiroyuki Miyamoto
- Laboratory for Neuronal Circuit Development, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan; International Research Center for Neurointelligence, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Rolf H Joho
- Center for Basic Neuroscience, Univ. Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Takao K Hensch
- Laboratory for Neuronal Circuit Development, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan; International Research Center for Neurointelligence, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan; Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA, 02138, USA.
| |
Collapse
|
25
|
Song CG, Kang X, Yang F, Du WQ, Zhang JJ, Liu L, Kang JJ, Jia N, Yue H, Fan LY, Wu SX, Jiang W, Gao F. Endocannabinoid system in the neurodevelopment of GABAergic interneurons: implications for neurological and psychiatric disorders. Rev Neurosci 2021; 32:803-831. [PMID: 33781002 DOI: 10.1515/revneuro-2020-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023]
Abstract
In mature mammalian brains, the endocannabinoid system (ECS) plays an important role in the regulation of synaptic plasticity and the functioning of neural networks. Besides, the ECS also contributes to the neurodevelopment of the central nervous system. Due to the increase in the medical and recreational use of cannabis, it is inevitable and essential to elaborate the roles of the ECS on neurodevelopment. GABAergic interneurons represent a group of inhibitory neurons that are vital in controlling neural network activity. However, the role of the ECS in the neurodevelopment of GABAergic interneurons remains to be fully elucidated. In this review, we provide a brief introduction of the ECS and interneuron diversity. We focus on the process of interneuron development and the role of ECS in the modulation of interneuron development, from the expansion of the neural stem/progenitor cells to the migration, specification and maturation of interneurons. We further discuss the potential implications of the ECS and interneurons in the pathogenesis of neurological and psychiatric disorders, including epilepsy, schizophrenia, major depressive disorder and autism spectrum disorder.
Collapse
Affiliation(s)
- Chang-Geng Song
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China.,Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Xin Kang
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Fang Yang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Wan-Qing Du
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Jia-Jia Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Long Liu
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Jun-Jun Kang
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Ning Jia
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Hui Yue
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Lu-Yu Fan
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Sheng-Xi Wu
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Fang Gao
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| |
Collapse
|
26
|
Burns TF, Rajan R. Sensing and processing whisker deflections in rodents. PeerJ 2021; 9:e10730. [PMID: 33665005 PMCID: PMC7906041 DOI: 10.7717/peerj.10730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/17/2020] [Indexed: 11/20/2022] Open
Abstract
The classical view of sensory information mainly flowing into barrel cortex at layer IV, moving up for complex feature processing and lateral interactions in layers II and III, then down to layers V and VI for output and corticothalamic feedback is becoming increasingly undermined by new evidence. We review the neurophysiology of sensing and processing whisker deflections, emphasizing the general processing and organisational principles present along the entire sensory pathway—from the site of physical deflection at the whiskers to the encoding of deflections in the barrel cortex. Many of these principles support the classical view. However, we also highlight the growing number of exceptions to these general principles, which complexify the system and which investigators should be mindful of when interpreting their results. We identify gaps in the literature for experimentalists and theorists to investigate, not just to better understand whisker sensation but also to better understand sensory and cortical processing.
Collapse
Affiliation(s)
- Thomas F Burns
- Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Ramesh Rajan
- Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Miczán V, Kelemen K, Glavinics JR, László ZI, Barti B, Kenesei K, Kisfali M, Katona I. NECAB1 and NECAB2 are Prevalent Calcium-Binding Proteins of CB1/CCK-Positive GABAergic Interneurons. Cereb Cortex 2021; 31:1786-1806. [PMID: 33230531 PMCID: PMC7869086 DOI: 10.1093/cercor/bhaa326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
The molecular repertoire of the "Ca2+-signaling toolkit" supports the specific kinetic requirements of Ca2+-dependent processes in different neuronal types. A well-known example is the unique expression pattern of calcium-binding proteins, such as parvalbumin, calbindin, and calretinin. These cytosolic Ca2+-buffers control presynaptic and somatodendritic processes in a cell-type-specific manner and have been used as neurochemical markers of GABAergic interneuron types for decades. Surprisingly, to date no typifying calcium-binding proteins have been found in CB1 cannabinoid receptor/cholecystokinin (CB1/CCK)-positive interneurons that represent a large population of GABAergic cells in cortical circuits. Because CB1/CCK-positive interneurons display disparate presynaptic and somatodendritic Ca2+-transients compared with other interneurons, we tested the hypothesis that they express alternative calcium-binding proteins. By in silico data mining in mouse single-cell RNA-seq databases, we identified high expression of Necab1 and Necab2 genes encoding N-terminal EF-hand calcium-binding proteins 1 and 2, respectively, in CB1/CCK-positive interneurons. Fluorescent in situ hybridization and immunostaining revealed cell-type-specific distribution of NECAB1 and NECAB2 throughout the isocortex, hippocampal formation, and basolateral amygdala complex. Combination of patch-clamp electrophysiology, confocal, and STORM super-resolution microscopy uncovered subcellular nanoscale differences indicating functional division of labor between the two calcium-binding proteins. These findings highlight NECAB1 and NECAB2 as predominant calcium-binding proteins in CB1/CCK-positive interneurons.
Collapse
Affiliation(s)
- Vivien Miczán
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Roska Tamás Doctoral School of Sciences and Technology, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest 1083, Hungary
| | - Krisztina Kelemen
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș 540142, Romania
| | - Judit R Glavinics
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Zsófia I László
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Benjámin Barti
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Kata Kenesei
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Máté Kisfali
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
28
|
Regulatory Elements Inserted into AAVs Confer Preferential Activity in Cortical Interneurons. eNeuro 2020; 7:ENEURO.0211-20.2020. [PMID: 33199411 PMCID: PMC7768279 DOI: 10.1523/eneuro.0211-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 11/21/2022] Open
Abstract
Cortical interneuron (CIN) dysfunction is thought to play a major role in neuropsychiatric conditions like epilepsy, schizophrenia and autism. It is therefore essential to understand how the development, physiology, and functions of CINs influence cortical circuit activity and behavior in model organisms such as mice and primates. While transgenic driver lines are powerful tools for studying CINs in mice, this technology is limited in other species. An alternative approach is to use viral vectors such as AAV, which can be used in multiple species including primates and also have potential for therapeutic use in humans. Thus, we sought to discover gene regulatory enhancer elements (REs) that can be used in viral vectors to drive expression in specific cell types. The present study describes the systematic genome-wide identification of putative REs (pREs) that are preferentially active in immature CINs by histone modification chromatin immunoprecipitation and sequencing (ChIP-seq). We evaluated two novel pREs in AAV vectors, alongside the well-established Dlx I12b enhancer, and found that they drove CIN-specific reporter expression in adult mice. We also showed that the identified Arl4d pRE could drive sufficient expression of channelrhodopsin for optogenetic rescue of behavioral deficits in the Dlx5/6+/- mouse model of fast-spiking CIN dysfunction.
Collapse
|
29
|
Chiarelli AM, Croce P, Assenza G, Merla A, Granata G, Giannantoni NM, Pizzella V, Tecchio F, Zappasodi F. Electroencephalography-Derived Prognosis of Functional Recovery in Acute Stroke Through Machine Learning Approaches. Int J Neural Syst 2020; 30:2050067. [PMID: 33236654 DOI: 10.1142/s0129065720500677] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Stroke, if not lethal, is a primary cause of disability. Early assessment of markers of recovery can allow personalized interventions; however, it is difficult to deliver indexes in the acute phase able to predict recovery. In this perspective, evaluation of electrical brain activity may provide useful information. A machine learning approach was explored here to predict post-stroke recovery relying on multi-channel electroencephalographic (EEG) recordings of few minutes performed at rest. A data-driven model, based on partial least square (PLS) regression, was trained on 19-channel EEG recordings performed within 10 days after mono-hemispheric stroke in 101 patients. The band-wise (delta: 1-4[Formula: see text]Hz, theta: 4-7[Formula: see text]Hz, alpha: 8-14[Formula: see text]Hz and beta: 15-30[Formula: see text]Hz) EEG effective powers were used as features to predict the recovery at 6 months (based on clinical status evaluated through the NIH Stroke Scale, NIHSS) in an optimized and cross-validated framework. In order to exploit the multimodal contribution to prognosis, the EEG-based prediction of recovery was combined with NIHSS scores in the acute phase and both were fed to a nonlinear support vector regressor (SVR). The prediction performance of EEG was at least as good as that of the acute clinical status scores. A posteriori evaluation of the features exploited by the analysis highlighted a lower delta and higher alpha activity in patients showing a positive outcome, independently of the affected hemisphere. The multimodal approach showed better prediction capabilities compared to the acute NIHSS scores alone ([Formula: see text] versus [Formula: see text], AUC = 0.80 versus AUC = 0.70, [Formula: see text]). The multimodal and multivariate model can be used in acute phase to infer recovery relying on standard EEG recordings of few minutes performed at rest together with clinical assessment, to be exploited for early and personalized therapies. The easiness of performing EEG may allow such an approach to become a standard-of-care and, thanks to the increasing number of labeled samples, further improving the model predictive power.
Collapse
Affiliation(s)
- Antonio Maria Chiarelli
- Department of Neuroscience, Imaging and Clinical Sciences and the Institute for Advanced Biomedical Technologies, Università G. d'Annunzio, Chieti, 66100, Italy
| | - Pierpaolo Croce
- Department of Neuroscience, Imaging and Clinical Sciences and the Institute for Advanced Biomedical Technologies, Università G. d'Annunzio, Chieti, 66100, Italy
| | - Giovanni Assenza
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
| | - Arcangelo Merla
- Department of Neuroscience, Imaging and Clinical Sciences and the Institute for Advanced Biomedical Technologies, Università G. d'Annunzio, Chieti, 66100, Italy
| | - Giuseppe Granata
- Fondazione Policlinico A. Gemelli IRCCS, Catholic University of Sacred Heart, Rome, Italy
| | | | - Vittorio Pizzella
- Department of Neuroscience, Imaging and Clinical Sciences and the Institute for Advanced Biomedical Technologies, Università G. d'Annunzio, Chieti, 66100, Italy
| | - Franca Tecchio
- Laboratory of Electrophysiology for Translational NeuroScience (LET'S), Istituto di Scienze e Teconologie della Cognizione (ISTC) - Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Filippo Zappasodi
- Department of Neuroscience, Imaging and Clinical Sciences and the Institute for Advanced Biomedical Technologies, Università G. d'Annunzio, Chieti, 66100, Italy
| |
Collapse
|
30
|
Chronic Restraint Stress Affects Network Oscillations in the Anterior Cingulate Cortex in Mice. Neuroscience 2020; 437:172-183. [PMID: 32335214 DOI: 10.1016/j.neuroscience.2020.04.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/21/2022]
Abstract
The anterior cingulate cortex (ACC) is vulnerable to stress. Its dysfunction is observed in psychiatric disorders manifested as alterations in network oscillations. Mechanisms linking stress load to disturbed emotional-cognitive behaviors are of essential importance to further elucidate therapeutic strategies for psychiatric diseases. Here, we analyzed the effects of chronic restraint stress (CRS) load in juvenile mice on kainic acid (KA)-induced network oscillations in ACC slice preparations and on the forced swim test (FST). The immobility time (IT) was shortened at the beginning of the FST in CRS mice. Power spectral density (PSD) obtained from KA-induced oscillations in field potentials in the superficial layers of the ACC were altered in slices from the CRS mice. The PSD was decreased in CRS mice at the alpha (8-12 Hz), beta (13-30 Hz), low gamma (30-50 Hz), and high gamma (50-80 Hz) components. Noradrenaline increased the PSD of the theta (3-8 Hz) components in both the control and CRS groups, and also in alpha components only in the CRS group. Dopamine did not modulate the PSD of any frequency components in the control mice, whereas it enhanced the PSD of theta and alpha components in CRS mice. It was suggested that chronic stress load affects the dynamics of the network oscillations in the ACC with enhanced cathecolaminergic modulation.
Collapse
|
31
|
Guillem K, Ahmed SH. Reorganization of theta phase-locking in the orbitofrontal cortex drives cocaine choice under the influence. Sci Rep 2020; 10:8041. [PMID: 32415278 PMCID: PMC7228935 DOI: 10.1038/s41598-020-64962-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/22/2020] [Indexed: 12/30/2022] Open
Abstract
Cortical theta oscillations of neuronal activity are a fundamental mechanism driving goal-directed behavior. We previously identified in the rat orbitofrontal cortex (OFC) a neuronal correlate of individual preferences between cocaine use and an alternative nondrug reward (i.e. saccharin). Whether theta oscillations are also associated with choice behavior between a drug and a nondrug reward remains unknown. Here we investigated the temporal structure between single unit activity and theta band oscillations (4-12 Hz) in the OFC of rats choosing between cocaine and saccharin. First, we found that the relative amplitude of theta oscillations is associated with subjective value and preference between two rewards. Second, OFC phase-locked neurons fired on opposite phase of the theta oscillation during saccharin and cocaine rewards, suggesting the existence of two separable neuronal assemblies. Finally, the pharmacological influence of cocaine at the moment of choice altered both theta band power and theta phase-locking in the OFC. That is, this drug influence shifted spike-phase relative to theta cycle and decreased the synchronization of OFC neurons relative to the theta oscillation. Overall, this study indicates that the reorganization of theta phase-locking under the influence of cocaine biases OFC neuronal assemblies in favor of cocaine choice and at the expense of a normally preferred alternative, a neuronal change that may contribute to drug preference in cocaine addiction.
Collapse
Affiliation(s)
- Karine Guillem
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 146 rue Léo-Saignat, F-33000, Bordeaux, France. .,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 146 rue Léo-Saignat, F-33000, Bordeaux, France.
| | - Serge H Ahmed
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 146 rue Léo-Saignat, F-33000, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 146 rue Léo-Saignat, F-33000, Bordeaux, France
| |
Collapse
|
32
|
Sullivan BJ, Ammanuel S, Kipnis PA, Araki Y, Huganir RL, Kadam SD. Low-Dose Perampanel Rescues Cortical Gamma Dysregulation Associated With Parvalbumin Interneuron GluA2 Upregulation in Epileptic Syngap1 +/- Mice. Biol Psychiatry 2020; 87:829-842. [PMID: 32107006 PMCID: PMC7166168 DOI: 10.1016/j.biopsych.2019.12.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Loss-of-function SYNGAP1 mutations cause a neurodevelopmental disorder characterized by intellectual disability and epilepsy. SYNGAP1 is a Ras GTPase-activating protein that underlies the formation and experience-dependent regulation of postsynaptic densities. The mechanisms that contribute to this proposed monogenic cause of intellectual disability and epilepsy remain unresolved. METHODS We established the phenotype of the epileptogenesis in a Syngap1+/- mouse model using 24-hour video electroencephalography (vEEG)/electromyography recordings at advancing ages. We administered an acute low dose of perampanel, a Food and Drug Administration-approved AMPA receptor (AMPAR) antagonist, during a follow-on 24-hour vEEG to investigate the role of AMPARs in Syngap1 haploinsufficiency. Immunohistochemistry was performed to determine the region- and location-specific differences in the expression of the GluA2 AMPAR subunit. RESULTS A progressive worsening of the epilepsy with emergence of multiple seizure phenotypes, interictal spike frequency, sleep dysfunction, and hyperactivity was identified in Syngap1+/- mice. Interictal spikes emerged predominantly during non-rapid eye movement sleep in 24-hour vEEG of Syngap1+/- mice. Myoclonic seizures occurred at behavioral-state transitions both in Syngap1+/- mice and during an overnight EEG from a child with SYNGAP1 haploinsufficiency. In Syngap1+/- mice, EEG spectral power analyses identified a significant loss of gamma power modulation during behavioral-state transitions. A significant region-specific increase of GluA2 AMPAR subunit expression in the somas of parvalbumin-positive interneurons was identified. CONCLUSIONS Acute dosing with perampanel significantly rescued behavioral state-dependent cortical gamma homeostasis, identifying a novel mechanism implicating Ca2+-impermeable AMPARs on parvalbumin-positive interneurons underlying circuit dysfunction in SYNGAP1 haploinsufficiency.
Collapse
Affiliation(s)
- Brennan J Sullivan
- Neuroscience Laboratory, Hugo Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland
| | - Simon Ammanuel
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Pavel A Kipnis
- Neuroscience Laboratory, Hugo Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland
| | - Yoichi Araki
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard L Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shilpa D Kadam
- Neuroscience Laboratory, Hugo Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
33
|
Murphy N, Ramakrishnan N, Walker CP, Polizzotto NR, Cho RY. Intact Auditory Cortical Cross-Frequency Coupling in Early and Chronic Schizophrenia. Front Psychiatry 2020; 11:507. [PMID: 32581881 PMCID: PMC7287164 DOI: 10.3389/fpsyt.2020.00507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/18/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Previous work has identified a hierarchical organization of neural oscillations that supports performance of complex cognitive and perceptual tasks, and can be indexed with phase-amplitude coupling (PAC) between low- and high-frequency oscillations. Our aim was to employ enhanced source localization afforded by magnetoencephalography (MEG) to expand on earlier reports of intact auditory cortical PAC in schizophrenia and to investigate how PAC may evolve over the early and chronic phases of the illness. METHODS Individuals with early schizophrenia (n=12) (≤5 years of illness duration), chronic schizophrenia (n=16) (>5 years of illness duration) and healthy comparators (n = 17) performed the auditory steady state response (ASSR) to 40, 30, and 20 Hz stimuli during MEG recordings. We estimated amplitude and PAC on the MEG ASSR source localized to the auditory cortices. RESULTS Gamma amplitude during 40-Hz ASSR exhibited a significant group by hemisphere interaction, with both patient groups showing reduced right hemisphere amplitude and no overall lateralization in contrast to the right hemisphere lateralization demonstrated in controls. We found significant PAC in the right auditory cortex during the 40-Hz entrainment condition relative to baseline, however, PAC did not differ significantly between groups. CONCLUSIONS In the current study, we demonstrated an apparent sparing of ASSR-related PAC across phases of the illness, in contrast with impaired cortical gamma oscillation amplitudes. The distinction between our PAC and evoked ASSR findings supports the notion of separate but interacting circuits for the generation and maintenance of sensory gamma oscillations. The apparent sparing of PAC in both early and chronic schizophrenia patients could imply that the neuropathology of schizophrenia differentially affects these mechanisms across different stages of the disease. Future studies should investigate the distinction between PAC during passive tasks and more cognitively demanding task such as working memory so that we can begin to understand the influence of schizophrenia neuropathology on the larger framework for modulating neurocomputational capacity.
Collapse
Affiliation(s)
- Nicholas Murphy
- Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States.,Research Service Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, United States
| | - Nithya Ramakrishnan
- Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States.,Research Service Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, United States
| | - Christopher P Walker
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nicola R Polizzotto
- Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Raymond Y Cho
- Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States.,Research Service Line, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, United States.,Menninger Clinic, Houston, TX, United States
| |
Collapse
|
34
|
Kreuzer M, García PS, Brucklacher-Waldert V, Claassen R, Schneider G, Antkowiak B, Drexler B. Diazepam and ethanol differently modulate neuronal activity in organotypic cortical cultures. BMC Neurosci 2019; 20:58. [PMID: 31823754 PMCID: PMC6902402 DOI: 10.1186/s12868-019-0540-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/24/2019] [Indexed: 12/17/2022] Open
Abstract
Background The pharmacodynamic results of diazepam and ethanol administration are similar, in that each can mediate amnestic and sedative-hypnotic effects. Although each of these molecules effectively reduce the activity of central neurons, diazepam does so through modulation of a more specific set of receptor targets (GABAA receptors containing a γ-subunit), while alcohol is less selective in its receptor bioactivity. Our investigation focuses on divergent actions of diazepam and ethanol on the firing patterns of cultured cortical neurons. Method We used electrophysiological recordings from organotypic slice cultures derived from Sprague–Dawley rat neocortex. We exposed these cultures to either diazepam (15 and 30 µM, n = 7) or ethanol (30 and 60 mM, n = 11) and recorded the electrical activity at baseline and experimental conditions. For analysis, we extracted the episodes of spontaneous activity, i.e., cortical up-states. After separation of action potential and local field potential (LFP) activity, we looked at differences in the number of action potentials, in the spectral power of the LFP, as well as in the coupling between action potential and LFP phase. Results While both substances seem to decrease neocortical action potential firing in a not significantly different (p = 0.659, Mann–Whitney U) fashion, diazepam increases the spectral power of the up-state without significantly impacting the spectral composition, whereas ethanol does not significantly change the spectral power but the oscillatory architecture of the up-state as revealed by the Friedman test with Bonferroni correction (p < 0.05). Further, the action potential to LFP-phase coupling reveals a synchronizing effect of diazepam for a wide frequency range and a narrow-band de-synchronizing effect for ethanol (p < 0.05, Kolmogorov–Smirnov test). Conclusion Diazepam and ethanol, induce specific patterns of network depressant actions. Diazepam induces cortical network inhibition and increased synchronicity via gamma subunit containing GABAA receptors. Ethanol also induces cortical network inhibition, but without an increase in synchronicity via a wider span of molecular targets.
Collapse
Affiliation(s)
- Matthias Kreuzer
- Department of Anesthesiology and Intensive Care, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Paul S García
- Department of Anesthesiology, Neuroanesthesia Division, Columbia University Medical Center, New York Presbyterian Hospital, New York, USA
| | - Verena Brucklacher-Waldert
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.,Horizon Discovery, 8100 Cambridge Research Park, Waterbeach, Cambridge, CB25 9TL, UK
| | - Rebecca Claassen
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.,Psychiatrie-Zentrum Linthgebiet, Standort Rapperswil, Untere Bahnhofstrasse 11, 8640, Rapperswil, Switzerland
| | - Gerhard Schneider
- Department of Anesthesiology and Intensive Care, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany
| | - Bernd Antkowiak
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.,Werner Reichardt Center for Integrative Neuroscience, Tübingen, Germany
| | - Berthold Drexler
- Dept. of Anesthesiology and Intensive Care, Experimental Anesthesiology Section, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
35
|
Γ-Aminobutyric acid in adult brain: an update. Behav Brain Res 2019; 376:112224. [DOI: 10.1016/j.bbr.2019.112224] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 01/21/2023]
|
36
|
Plumbly W, Brandon N, Deeb TZ, Hall J, Harwood AJ. L-type voltage-gated calcium channel regulation of in vitro human cortical neuronal networks. Sci Rep 2019; 9:13810. [PMID: 31554851 PMCID: PMC6761148 DOI: 10.1038/s41598-019-50226-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/02/2019] [Indexed: 11/09/2022] Open
Abstract
The combination of in vitro multi-electrode arrays (MEAs) and the neuronal differentiation of stem cells offers the capability to study human neuronal networks from patient or engineered human cell lines. Here, we use MEA-based assays to probe synaptic function and network interactions of hiPSC-derived neurons. Neuronal network behaviour first emerges at approximately 30 days of culture and is driven by glutamate neurotransmission. Over a further 30 days, inhibitory GABAergic signalling shapes network behaviour into a synchronous regular pattern of burst firing activity and low activity periods. Gene mutations in L-type voltage gated calcium channel subunit genes are strongly implicated as genetic risk factors for the development of schizophrenia and bipolar disorder. We find that, although basal neuronal firing rate is unaffected, there is a dose-dependent effect of L-type voltage gated calcium channel inhibitors on synchronous firing patterns of our hiPSC-derived neural networks. This demonstrates that MEA assays have sufficient sensitivity to detect changes in patterns of neuronal interaction that may arise from hypo-function of psychiatric risk genes. Our study highlights the utility of in vitro MEA based platforms for the study of hiPSC neural network activity and their potential use in novel compound screening.
Collapse
Affiliation(s)
- William Plumbly
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Nick Brandon
- Neuroscience, IMED Biotech Unit, AstraZeneca, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Tarek Z Deeb
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Adrian J Harwood
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
37
|
Parker DA, Hamm JP, McDowell JE, Keedy SK, Gershon ES, Ivleva EI, Pearlson GD, Keshavan MS, Tamminga CA, Sweeney JA, Clementz BA. Auditory steady-state EEG response across the schizo-bipolar spectrum. Schizophr Res 2019; 209:218-226. [PMID: 31080153 PMCID: PMC6661193 DOI: 10.1016/j.schres.2019.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 11/29/2022]
Abstract
UNLABELLED Deviant auditory steady-state responses (aSSRs) in the gamma range (30-90 Hz) may be translational biomarkers for schizophrenia (SZ). This study tests whether aSSR deviations are (i) specific to SZ across the psychosis dimension, (ii) specific to particular frequency bands, and (iii) present in bipolar I disorder without psychosis (BDNP). METHODS Beta (20-), low- (40-), and high-gamma (80-Hz) aSSRs were measured with EEG and compared across 113 SZ, 105 schizoaffective disorder (SAD), 99 bipolar disorder with psychosis (BDP), 68 BDNP, and 137 healthy comparison subjects (HC). Standard aSSR measures (single-trial power [STP] and inter-trial phase coherence [ITC]), as well as evoked responses to stimulus onsets/offsets and pre-stimulus power, were quantified. Multivariate canonical discriminant analysis was used to summarize variables that efficiently and maximally differentiated groups. RESULTS (i) Psychosis groups showed reduced responses on ITC 20 Hz, STP/ITC 40 Hz, STP/ITC 80 Hz, indicating dimensional reductions in aSSR across the psychosis spectrum not specific to aSSR frequency. For the 40- and 80-Hz ITCs there was greater reduction in SZ compared to SAD, possibly indexing cortical disruptions linked to psychosis without mood symptoms. (ii) All probands had elevated pre-stimulus power, possibly compromising neural entrainment to the steady-state stimuli. (iii) Onset/Offset and 80 Hz ITC responses were most important for group discrimination and showed dimensional reduction across the schizo-bipolar spectrum. CONCLUSIONS Deviant aSSRs were found across the schizo-bipolar spectrum at multiple frequencies with psychosis status and severity linked to greatest reductions at low and high gamma.
Collapse
Affiliation(s)
- David A. Parker
- Departments of Psychology and Neuroscience, Bio-Imaging Research Center, University of Georgia, Athens, GA, United States
| | - Jordan P. Hamm
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Jennifer E. McDowell
- Departments of Psychology and Neuroscience, Bio-Imaging Research Center, University of Georgia, Athens, GA, United States
| | - Sarah K. Keedy
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - Elliot S. Gershon
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, United States
| | - Elena I. Ivleva
- Department of Psychiatry, UT-Southwestern Medical Center, Dallas, TX, United States
| | - Godfrey D. Pearlson
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, CT, United States,Institute of Living, Hartford Hospital, Hartford, CT, United States
| | - Matcheri S. Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Carol A. Tamminga
- Department of Psychiatry, UT-Southwestern Medical Center, Dallas, TX, United States
| | - John A. Sweeney
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, United States
| | - Brett A. Clementz
- Departments of Psychology and Neuroscience, Bio-Imaging Research Center, University of Georgia, Athens, GA, United States
| |
Collapse
|
38
|
Phase Coherent Currents Underlying Neocortical Seizure-Like State Transitions. eNeuro 2019; 6:eN-NWR-0426-18. [PMID: 30923739 PMCID: PMC6437657 DOI: 10.1523/eneuro.0426-18.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
In the epileptic brain, phase amplitude cross-frequency coupling (CFC) features have been used to objectively classify seizure-related states, and the inter-seizure state has been demonstrated as being random, in contrast to the seizure state being predictable; however, the excitatory and inhibitory networks underlying their dynamics remain unclear. Therefore, the objectives of this study are to classify the dynamics of seizure sub-states labeling seizure-like event (SLE) onset and termination intervals using CFC features and to obtain their underlying excitatory/inhibitory cellular correlates. SLEs were induced in mouse neocortical brain slices using a low-magnesium perfusate, and were recorded in Layer II/III using simultaneous local field potential (LFP) and whole-cell voltage clamp electrodes. Classification of onset and termination of SLE transitions was investigated using CFC features in conjunction with an unsupervised two-state hidden Markov model (HMM). γ-Distributions of their durations indicated that both are predictable. Furthermore, omitting 4 Hz from the HMM classifier switched both SLE sub-states from statistically deterministic to random without changing the dynamics of the SLE state. These results were generalized to 4-aminopyridine (4-AP)-induced SLEs and human seizure traces. Only during these sub-states, both excitatory and inhibitory currents coupled with the field. Where excitatory currents phase locked to a broad range of frequencies between 1 and 12 Hz, inhibitory currents dominantly phase locked at 4 Hz. We conclude that inhibition underlies the predictability of neocortical CFC-defined SLE transition sub-states.
Collapse
|
39
|
Xiong YB, Bo QJ, Wang CM, Tian Q, Liu Y, Wang CY. Differential of Frequency and Duration Mismatch Negativity and Theta Power Deficits in First-Episode and Chronic Schizophrenia. Front Behav Neurosci 2019; 13:37. [PMID: 30894804 PMCID: PMC6414796 DOI: 10.3389/fnbeh.2019.00037] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 02/13/2019] [Indexed: 01/10/2023] Open
Abstract
Background: Due to its impairment in patients with schizophrenia, mismatch negativity (MMN) generation has been identified as a potential biomarker for identifying primary impairments in auditory sensory processing. This study aimed to investigate the dysfunctional differences in different MMN deviants and evoked theta power in patients with first-episode schizophrenia (FES) and chronic schizophrenia (CS). Methods: We measured frequency and duration MMN from 40 FES, 40 CS, and 40 healthy controls (HC). Evoked theta power was analyzed by event-related spectral perturbation (ERSP) approaches. Results: Deficits in duration MMN were observed in both FES (p = 0.048, Bonferroni-adjusted) and CS (p < 0.001, Bonferroni-adjusted). However, deficits in frequency MMN were restricted to the CS (p < 0.001, Bonferroni-adjusted). Evoked theta power deficits were observed in both patient groups when compared with the HC (p FES = 0.001, p CS < 0.001, Bonferroni-adjusted), yet no significant differences were found between FES and CS. Frequency MMN was correlated with the MATRICS consensus cognitive battery (MCCB) combined score (r = -0.327, p < 0.05) and MCCB verbal learning (r = -0.328, p < 0.05) in FES. Evoked theta power was correlated with MCCB working memory in both FES (r = 0.347, p < 0.05) and CS (r = 0.408, p < 0.01). Conclusion: These findings suggest that duration MMN and evoked theta power deficits may be more sensitive for detection of schizophrenia during its early stages. Moreover, frequency MMN and theta power could potentially linked to poor cognitive functioning in schizophrenic patients. The findings mentioned above indicated that the neural mechanisms of the three indexes may vary between people.
Collapse
Affiliation(s)
- Yan-Bing Xiong
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing, China.,Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing, China.,The National Clinical Research Center for Mental Disorders, Beijing, China
| | - Qi-Jing Bo
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing, China.,Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing, China.,The National Clinical Research Center for Mental Disorders, Beijing, China
| | - Chang-Ming Wang
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing, China.,Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing, China.,The National Clinical Research Center for Mental Disorders, Beijing, China
| | - Qing Tian
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing, China.,Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing, China.,The National Clinical Research Center for Mental Disorders, Beijing, China
| | - Yi Liu
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing, China.,Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing, China.,The National Clinical Research Center for Mental Disorders, Beijing, China
| | - Chuan-Yue Wang
- Department of Psychiatry, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing, China.,Beijing Institute for Brain Disorders Center of Schizophrenia, Beijing, China.,The National Clinical Research Center for Mental Disorders, Beijing, China
| |
Collapse
|
40
|
Gamma oscillations in somatosensory cortex recruit prefrontal and descending serotonergic pathways in aversion and nociception. Nat Commun 2019; 10:983. [PMID: 30816113 PMCID: PMC6395755 DOI: 10.1038/s41467-019-08873-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/04/2019] [Indexed: 12/21/2022] Open
Abstract
In humans, gamma-band oscillations in the primary somatosensory cortex (S1) correlate with subjective pain perception. However, functional contributions to pain and the nature of underlying circuits are unclear. Here we report that gamma oscillations, but not other rhythms, are specifically strengthened independently of any motor component in the S1 cortex of mice during nociception. Moreover, mice with inflammatory pain show elevated resting gamma and alpha activity and increased gamma power in response to sub-threshold stimuli, in association with behavioral nociceptive hypersensitivity. Inducing gamma oscillations via optogenetic activation of parvalbumin-expressing inhibitory interneurons in the S1 cortex enhances nociceptive sensitivity and induces aversive avoidance behavior. Activity mapping identified a network of prefrontal cortical and subcortical centers whilst morphological tracing and pharmacological studies demonstrate the requirement of descending serotonergic facilitatory pathways in these pain-related behaviors. This study thus describes a mechanistic framework for modulation of pain by specific activity patterns in the S1 cortex. Gamma oscillations in somatosensory areas in humans correlate with pain perception and pain stimulus intensity, but could also reflect cognitive processes such as attention. Here the authors provide evidence in mice that these oscillations causally contribute to pain perception.
Collapse
|
41
|
Pham T, Haas JS. Electrical synapses regulate both subthreshold integration and population activity of principal cells in response to transient inputs within canonical feedforward circuits. PLoS Comput Biol 2019; 15:e1006440. [PMID: 30802238 PMCID: PMC6405166 DOI: 10.1371/journal.pcbi.1006440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 03/07/2019] [Accepted: 01/29/2019] [Indexed: 12/05/2022] Open
Abstract
As information about the world traverses the brain, the signals exchanged between neurons are passed and modulated by synapses, or specialized contacts between neurons. While neurotransmitter-based synapses tend to exert either excitatory or inhibitory pulses of influence on the postsynaptic neuron, electrical synapses, composed of plaques of gap junction channels, continuously transmit signals that can either excite or inhibit a coupled neighbor. A growing body of evidence indicates that electrical synapses, similar to their chemical counterparts, are modified in strength during physiological neuronal activity. The synchronizing role of electrical synapses in neuronal oscillations has been well established, but their impact on transient signal processing in the brain is much less understood. Here we constructed computational models based on the canonical feedforward neuronal circuit and included electrical synapses between inhibitory interneurons. We provided discrete closely-timed inputs to the circuits, and characterize the influence of electrical synapse strength on both subthreshold summation and spike trains in the output neuron. Our simulations highlight the diverse and powerful roles that electrical synapses play even in simple circuits. Because these canonical circuits are represented widely throughout the brain, we expect that these are general principles for the influence of electrical synapses on transient signal processing across the brain. The roles that electrical synapses play in neural oscillations, network synchronization and rhythmicity are well established, but their roles in neuronal processing of transient inputs are much less understood. Here, we used computational models of canonical feedforward circuits and networks to investigate how electrical synapses regulate the flow of transient signals passing through those circuits. We show that because the influence of electrical synapses on coupled neighbors can be either inhibitory or excitatory, their role in network information processing is heterogeneous, and powerful. Because electrical synapses between interneurons are widespread across the brain, and in addition to a growing body of evidence for their activity-dependent plasticity, we expect the effects we describe here to play a substantial role in how the brain processes incoming sensory inputs.
Collapse
Affiliation(s)
- Tuan Pham
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
| | - Julie S. Haas
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
42
|
Zappasodi F, Tecchio F, Marzetti L, Pizzella V, Di Lazzaro V, Assenza G. Longitudinal quantitative electroencephalographic study in mono-hemispheric stroke patients. Neural Regen Res 2019; 14:1237-1246. [PMID: 30804255 PMCID: PMC6425833 DOI: 10.4103/1673-5374.251331] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The identification of individual factors modulating clinical recovery after a stroke is fundamental to personalize the therapeutic intervention to enhance the final clinical outcome. In this framework, electrophysiological factors are promising since are more directly related to neuroplasticity, which supports recovery in stroke patients, than neurovascular factors. In this retrospective observational study, we investigated brain neuronal activity assessed via spectral features and Higuchi’s fractal dimension (HFD) of electroencephalographic signals in acute phase (2–10 days from symptom onset, T0) and sub-acute phase (2.5 months, T1) in 24 patients affected by unilateral middle cerebral artery stroke. Longitudinal assessment of the clinical deficits was performed using the National Institutes of Health Stroke Scale (NIHSS), together with the effective recovery calculated as the ratio between difference of NIHSS at T0 and T1 over the NIHSS value at T0. We observed that delta and alpha band electroencephalographic signal power changed between the two phases in both the hemispheres ipsilateral (ILH) and contralateral (CHL) to the lesion. Moreover, at T0, bilateral higher delta band power correlated with worse clinical conditions (Spearman’s rs = 0.460, P = 0.027 for ILH and rs = 0.508, P = 0.013 for CLH), whereas at T1 this occurred only for delta power in ILH (rs = 0.411, P = 0.046) and not for CHL. Inter-hemispheric difference (ILH vs. CLH) of alpha power in patients was lower at T0 than at T1 (P = 0.020). HFD at T0 was lower than at T1 (P = 0.005), and at both phases, ILH HFD was lower than CLH HFD (P = 0.020). These data suggest that inter-hemispheric low band asymmetry and fractal dimension changes from the acute to the sub-acute phase are sensitive to neuroplasticity processes which subtend clinical recovery. The study protocol was approved by the Bioethical Committee of Ospedale San Giovanni Calibita Fatebenefretelli (No. 40/2011) on July 14, 2011.
Collapse
Affiliation(s)
- Filippo Zappasodi
- Department of Neuroscience, Imaging and Clinical Sciences and Institute for Advanced Biomedical Imaging, "G. D'Annunzio" University, Chieti, Italy
| | - Franca Tecchio
- Laboratory of Electrophysiology for Translational NeuroScience (LET'S), ISTC-CNR, and Fondazione Policlinico Gemelli IRCCS, Rome, Italy
| | - Laura Marzetti
- Department of Neuroscience, Imaging and Clinical Sciences and Institute for Advanced Biomedical Imaging, "G. D'Annunzio" University, Chieti, Italy
| | - Vittorio Pizzella
- Department of Neuroscience, Imaging and Clinical Sciences and Institute for Advanced Biomedical Imaging, "G. D'Annunzio" University, Chieti, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Giovanni Assenza
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
43
|
Cardin JA. Inhibitory Interneurons Regulate Temporal Precision and Correlations in Cortical Circuits. Trends Neurosci 2018; 41:689-700. [PMID: 30274604 PMCID: PMC6173199 DOI: 10.1016/j.tins.2018.07.015] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/24/2018] [Accepted: 07/31/2018] [Indexed: 01/16/2023]
Abstract
GABAergic interneurons, which are highly diverse, have long been thought to contribute to the timing of neural activity as well as to the generation and shaping of brain rhythms. GABAergic activity is crucial not only for entrainment of oscillatory activity across a neural population, but also for precise regulation of the timing of action potentials and the suppression of slow-timescale correlations. The diversity of inhibition provides the potential for flexible regulation of patterned activity, but also poses a challenge to identifying the elements of excitatory-inhibitory interactions underlying network engagement. This review highlights the key roles of inhibitory interneurons in spike correlations and brain rhythms, describes several scales on which GABAergic inhibition regulates timing in neural networks, and identifies potential consequences of inhibitory dysfunction.
Collapse
Affiliation(s)
- Jessica A Cardin
- Department of Neuroscience, Yale University, New Haven, CT 06520, USA; Kavli Institute for Neuroscience, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
44
|
Preferential inputs from cholecystokinin-positive neurons to the somatic compartment of parvalbumin-expressing neurons in the mouse primary somatosensory cortex. Brain Res 2018; 1695:18-30. [DOI: 10.1016/j.brainres.2018.05.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 05/10/2018] [Accepted: 05/19/2018] [Indexed: 12/22/2022]
|
45
|
Miriyala A, Kessler S, Rind FC, Wright GA. Burst Firing in Bee Gustatory Neurons Prevents Adaptation. Curr Biol 2018; 28:1585-1594.e3. [PMID: 29754900 DOI: 10.1016/j.cub.2018.03.070] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 02/20/2018] [Accepted: 03/29/2018] [Indexed: 10/16/2022]
Abstract
Animals detect changes in the environment using modality-specific, peripheral sensory neurons. The insect gustatory system encodes tastant identity and concentration through the independent firing of gustatory receptor neurons (GRNs) that spike rapidly at stimulus onset and quickly adapt. Here, we show the first evidence that concentrated sugar evokes a temporally structured burst pattern of spiking involving two GRNs within the gustatory sensilla of bumblebees. Bursts of spikes resulted when a sucrose-activated GRN was inhibited by another GRN at a frequency of ∼22 Hz during the first 1 s of stimulation. Pharmacological blockade of gap junctions abolished bursting, indicating that bee GRNs have electrical synapses that produce a temporal pattern of spikes when one GRN is activated by a sugar ligand. Bursting permitted bee GRNs to maintain a high rate of spiking and to exhibit the slowest rate of adaptation of any insect species. Feeding bout duration correlated with coherent bursting; only sugar concentrations that produced bursting evoked the bumblebee's feeding reflex. Volume of solution imbibed was a direct function of time in contact with food. We propose that gap junctions among GRNs enable a sustained rate of GRN spiking that is necessary to drive continuous feeding by the bee proboscis.
Collapse
Affiliation(s)
- Ashwin Miriyala
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Sébastien Kessler
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - F Claire Rind
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Geraldine A Wright
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
46
|
Abstract
The muscarinic receptor agonist carbachol (CCh) can induce activity in the theta range (4-15 Hz) in the entorhinal cortex (EC), but the underlying network mechanisms remain unclear. Here, we investigated the interplay between interneurons and principal cells in the EC during CCh-induced theta-like field oscillations in an in vitro brain slice preparation using tetrodes. Field oscillations at 10.1 Hz (IQR = 9.5-10.9 Hz) occurred during bath application of CCh (100 μM; n = 32 experiments) and were associated with single-unit (n = 189) firing. Interneuron activity increased before principal cell activity at the onset of the oscillations and both interneurons and principal cells fired at specific oscillation phases with interneurons preceding principal cells, suggesting that interneurons modulate principal cell activity during such oscillations. The regularity of occurrence of CCh-induced oscillations was abolished by applying the GABAA receptor antagonist picrotoxin (100 μM; n = 13). These effects were accompanied by changes in firing with principal cells discharging action potentials before interneurons, along with a loss of preferred firing phase for principal cells in relation to the oscillation peaks. Blocking ionotropic glutamatergic transmission abolished CCh-induced field oscillations (n = 6), suggesting that ionotropic glutamatergic receptor signaling is necessary for their generation. Our results show that neuronal network interactions leading to CCh-induced theta-like field oscillations rest on the close interplay between interneurons and principal cells and that interneurons modulate principal cell activity during such oscillatory activity. Moreover, they underscore the role of ionotropic glutamatergic transmission in this type of oscillations.
Collapse
|
47
|
Sultan KT, Shi SH. Generation of diverse cortical inhibitory interneurons. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:10.1002/wdev.306. [PMID: 29115042 PMCID: PMC5814332 DOI: 10.1002/wdev.306] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/14/2017] [Accepted: 09/19/2017] [Indexed: 12/16/2022]
Abstract
First described by Ramon y Cajal as 'short-axon' cells over a century ago, inhibitory interneurons in the cerebral cortex make up ~20-30% of the neuronal milieu. A key feature of these interneurons is the striking structural and functional diversity, which allows them to modulate neural activity in diverse ways and ultimately endow neural circuits with remarkable computational power. Here, we review our current understanding of the generation of cortical interneurons, with a focus on recent efforts to bridge the gap between progenitor behavior and interneuron production, and how these aspects influence interneuron diversity and organization. WIREs Dev Biol 2018, 7:e306. doi: 10.1002/wdev.306 This article is categorized under: Nervous System Development > Vertebrates: General Principles.
Collapse
Affiliation(s)
- Khadeejah T Sultan
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Medical College, New York, NY, USA
| | - Song-Hai Shi
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
48
|
Nagy JI, Pereda AE, Rash JE. Electrical synapses in mammalian CNS: Past eras, present focus and future directions. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2018; 1860:102-123. [PMID: 28577972 PMCID: PMC5705454 DOI: 10.1016/j.bbamem.2017.05.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 12/19/2022]
Abstract
Gap junctions provide the basis for electrical synapses between neurons. Early studies in well-defined circuits in lower vertebrates laid the foundation for understanding various properties conferred by electrical synaptic transmission. Knowledge surrounding electrical synapses in mammalian systems unfolded first with evidence indicating the presence of gap junctions between neurons in various brain regions, but with little appreciation of their functional roles. Beginning at about the turn of this century, new approaches were applied to scrutinize electrical synapses, revealing the prevalence of neuronal gap junctions, the connexin protein composition of many of those junctions, and the myriad diverse neural systems in which they occur in the mammalian CNS. Subsequent progress indicated that electrical synapses constitute key elements in synaptic circuitry, govern the collective activity of ensembles of electrically coupled neurons, and in part orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie fundamental integrative processes. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- James I Nagy
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - John E Rash
- Department of Biomedical Sciences, and Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
49
|
Nelli S, Itthipuripat S, Srinivasan R, Serences JT. Fluctuations in instantaneous frequency predict alpha amplitude during visual perception. Nat Commun 2017; 8:2071. [PMID: 29234068 PMCID: PMC5727061 DOI: 10.1038/s41467-017-02176-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 11/10/2017] [Indexed: 11/20/2022] Open
Abstract
Rhythmic neural activity in the alpha band (8-13 Hz) is thought to have an important role in the selective processing of visual information. Typically, modulations in alpha amplitude and instantaneous frequency are thought to reflect independent mechanisms impacting dissociable aspects of visual information processing. However, in complex systems with interacting oscillators such as the brain, amplitude and frequency are mathematically dependent. Here, we record electroencephalography in human subjects and show that both alpha amplitude and instantaneous frequency predict behavioral performance in the same visual discrimination task. Consistent with a model of coupled oscillators, we show that fluctuations in instantaneous frequency predict alpha amplitude on a single trial basis, empirically demonstrating that these metrics are not independent. This interdependence suggests that changes in amplitude and instantaneous frequency reflect a common change in the excitatory and inhibitory neural activity that regulates alpha oscillations and visual information processing.
Collapse
Affiliation(s)
- Stephanie Nelli
- Neurosciences Graduate Program, University of California, San Diego, CA, USA.
| | - Sirawaj Itthipuripat
- Neurosciences Graduate Program, University of California, San Diego, CA, USA
- Learning Institute, King Mongkut's University of Technology Thonburi, 10140, Bangkok, Thailand
| | - Ramesh Srinivasan
- Department of Cognitive Sciences, University of California, Irvine, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - John T Serences
- Neurosciences Graduate Program, University of California, San Diego, CA, USA.
- Department of Psychology, University of California, San Diego, CA, USA.
- Kavli Institute for Brain and Mind, University of California, San Diego, CA, USA.
| |
Collapse
|
50
|
de Jong IE, Mørk A. Antagonism of the 5-HT 6 receptor – Preclinical rationale for the treatment of Alzheimer's disease. Neuropharmacology 2017; 125:50-63. [DOI: 10.1016/j.neuropharm.2017.07.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 06/30/2017] [Accepted: 07/11/2017] [Indexed: 11/29/2022]
|