1
|
Qiu Q, Yang M, Gong D, Liang H, Chen T. Potassium and calcium channels in different nerve cells act as therapeutic targets in neurological disorders. Neural Regen Res 2025; 20:1258-1276. [PMID: 38845230 PMCID: PMC11624876 DOI: 10.4103/nrr.nrr-d-23-01766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/18/2024] [Accepted: 04/07/2024] [Indexed: 07/31/2024] Open
Abstract
The central nervous system, information integration center of the body, is mainly composed of neurons and glial cells. The neuron is one of the most basic and important structural and functional units of the central nervous system, with sensory stimulation and excitation conduction functions. Astrocytes and microglia belong to the glial cell family, which is the main source of cytokines and represents the main defense system of the central nervous system. Nerve cells undergo neurotransmission or gliotransmission, which regulates neuronal activity via the ion channels, receptors, or transporters expressed on nerve cell membranes. Ion channels, composed of large transmembrane proteins, play crucial roles in maintaining nerve cell homeostasis. These channels are also important for control of the membrane potential and in the secretion of neurotransmitters. A variety of cellular functions and life activities, including functional regulation of the central nervous system, the generation and conduction of nerve excitation, the occurrence of receptor potential, heart pulsation, smooth muscle peristalsis, skeletal muscle contraction, and hormone secretion, are closely related to ion channels associated with passive transmembrane transport. Two types of ion channels in the central nervous system, potassium channels and calcium channels, are closely related to various neurological disorders, including Alzheimer's disease, Parkinson's disease, and epilepsy. Accordingly, various drugs that can affect these ion channels have been explored deeply to provide new directions for the treatment of these neurological disorders. In this review, we focus on the functions of potassium and calcium ion channels in different nerve cells and their involvement in neurological disorders such as Parkinson's disease, Alzheimer's disease, depression, epilepsy, autism, and rare disorders. We also describe several clinical drugs that target potassium or calcium channels in nerve cells and could be used to treat these disorders. We concluded that there are few clinical drugs that can improve the pathology these diseases by acting on potassium or calcium ions. Although a few novel ion-channel-specific modulators have been discovered, meaningful therapies have largely not yet been realized. The lack of target-specific drugs, their requirement to cross the blood-brain barrier, and their exact underlying mechanisms all need further attention. This review aims to explain the urgent problems that need research progress and provide comprehensive information aiming to arouse the research community's interest in the development of ion channel-targeting drugs and the identification of new therapeutic targets for that can increase the cure rate of nervous system diseases and reduce the occurrence of adverse reactions in other systems.
Collapse
Affiliation(s)
- Qing Qiu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Mengting Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Danfeng Gong
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| | - Haiying Liang
- Department of Pharmacy, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian Province, China
| | - Tingting Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu Province, China
| |
Collapse
|
2
|
Koch NA, Corrigan BW, Feyerabend M, Gulli RA, Jimenez-Sosa MS, Abbass M, Sunstrum JK, Matovic S, Roussy M, Luna R, Mestern SA, Mahmoudian B, Vijayraghavan S, Igarashi H, Pradeepan KS, Assis WJ, Pruszynski JA, Tripathy S, Staiger JF, Gonzalez-Burgos G, Neef A, Treue S, Everling S, Inoue W, Khadra A, Martinez-Trujillo JC. Spike frequency adaptation in primate lateral prefrontal cortex neurons results from interplay between intrinsic properties and circuit dynamics. Cell Rep 2025; 44:115159. [PMID: 39772396 DOI: 10.1016/j.celrep.2024.115159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/19/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Cortical neurons in brain slices display intrinsic spike frequency adaptation (I-SFA) to constant current inputs, while extracellular recordings show extrinsic SFA (E-SFA) during sustained visual stimulation. Inferring how I-SFA contributes to E-SFA during behavior is challenging due to the isolated nature of slice recordings. To address this, we recorded macaque lateral prefrontal cortex (LPFC) neurons in vivo during a visually guided saccade task and in vitro in brain slices. Broad-spiking (BS) putative pyramidal cells and narrow-spiking (NS) putative inhibitory interneurons exhibit both E-SFA and I-SFA. Developing a data-driven hybrid circuit model comprising NS model neurons receiving BS input reveals that NS model neurons exhibit longer SFA than observed in vivo; however, adding feedforward inhibition corrects this in a manner dependent on I-SFA. Identification of this circuit motif shaping E-SFA in LPFC highlights the roles of both intrinsic and network mechanisms in neural activity underlying behavior.
Collapse
Affiliation(s)
- Nils A Koch
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Benjamin W Corrigan
- Department of Biology, York University, Toronto, ON, Canada; Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Michael Feyerabend
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada
| | - Roberto A Gulli
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | | | - Mohamad Abbass
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Julia K Sunstrum
- Western Institute for Neuroscience, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada; Neuroscience Graduate Program, Western University, London, ON, Canada
| | - Sara Matovic
- Western Institute for Neuroscience, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Megan Roussy
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Rogelio Luna
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Samuel A Mestern
- Western Institute for Neuroscience, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Borna Mahmoudian
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Susheel Vijayraghavan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Hiroyuki Igarashi
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada; Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Kartik S Pradeepan
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada
| | - William J Assis
- Western Institute for Neuroscience, Western University, London, ON, Canada
| | - J Andrew Pruszynski
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada
| | - Shreejoy Tripathy
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Jochen F Staiger
- Department of Neuroanatomy, University Medical Center, Georg-August-University, Göttingen, Germany
| | | | - Andreas Neef
- Campus Institute for Dynamics of Biological Networks, Göttingen, Germany; Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany; Bernstein Center for Computational Neuroscience, Göttingen, Germany
| | - Stefan Treue
- Cognitive Neuroscience Laboratory, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany; Faculty for Biology and Psychology, University of Göttingen, Göttingen, Germany; Leibniz ScienceCampus, Primate Cognition, Göttingen, Germany
| | - Stefan Everling
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada
| | - Wataru Inoue
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Anmar Khadra
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Department of Physiology, McGill University, Montreal, QC, Canada.
| | - Julio C Martinez-Trujillo
- Department of Clinical Neurological Sciences, London Health Sciences Centre, Western University, London, ON, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada; Western Institute for Neuroscience, Western University, London, ON, Canada
| |
Collapse
|
3
|
Ting JT, Johansen NJ, Kalmbach BE, Taskin N, Lee B, Clark JK, Kendrick R, Ng L, Radaelli C, Weed N, Enstrom R, Ransford S, Redford I, Walling-Bell S, Dalley R, Tieu M, Goldy J, Jorstad N, Smith K, Bakken T, Lein ES, Owen SF. Distinctive physiology of molecularly identified medium spiny neurons in the macaque putamen. Cell Rep 2024; 43:114963. [PMID: 39514389 DOI: 10.1016/j.celrep.2024.114963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/11/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The distinctive physiology of striatal medium spiny neurons (MSNs) underlies their ability to integrate sensory and motor input. In rodents, MSNs have a hyperpolarized resting potential and low input resistance. When activated, they have a delayed onset of spiking and regular spike rate. Here, we show that in the macaque putamen, latency to spike is reduced and spike rate adaptation is increased relative to mouse. We use whole-cell brain slice recordings and recover single-cell gene expression using Patch-seq to distinguish macaque MSN cell types. Species differences in the expression of ion channel genes including the calcium-activated chloride channel, ANO2, and an auxiliary subunit of the A-type potassium channel, DPP10, are correlated with species differences in spike rate adaptation and latency to the first spike, respectively. These surprising divergences in physiology better define the strengths and limitations of mouse models for understanding neuronal and circuit function in the primate basal ganglia.
Collapse
Affiliation(s)
- Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA 98109, USA; The Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | | | - Brian E Kalmbach
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurobiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jason K Clark
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rennie Kendrick
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lindsay Ng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Rachel Enstrom
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Shea Ransford
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ingrid Redford
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Rachel Dalley
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Nik Jorstad
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kimberly Smith
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Trygve Bakken
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Scott F Owen
- Allen Institute for Brain Science, Seattle, WA 98109, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Du Z, Li M, Chen G, Xiang M, Jia D, Cheng JX, Yang C. Mid-Infrared Photoacoustic Stimulation of Neurons through Vibrational Excitation in Polydimethylsiloxane. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405677. [PMID: 38994890 PMCID: PMC11425203 DOI: 10.1002/advs.202405677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/26/2024] [Indexed: 07/13/2024]
Abstract
Photoacoustic (PA) emitters are emerging ultrasound sources offering high spatial resolution and ease of miniaturization. Thus far, PA emitters rely on electronic transitions of absorbers embedded in an expansion matrix such as polydimethylsiloxane (PDMS). Here, it is shown that mid-infrared vibrational excitation of C─H bonds in a transparent PDMS film can lead to efficient mid-infrared photoacoustic conversion (MIPA). MIPA shows 37.5 times more efficient than the commonly used PA emitters based on carbon nanotubes embedded in PDMS. Successful neural stimulation through MIPA both in a wide field with a size up to a 100 µm radius and in single-cell precision is achieved. Owing to the low heat conductivity of PDMS, less than a 0.5 °C temperature increase is found on the surface of a PDMS film during successful neural stimulation, suggesting a non-thermal mechanism. MIPA emitters allow repetitive wide-field neural stimulation, opening up opportunities for high-throughput screening of mechano-sensitive ion channels and regulators.
Collapse
Affiliation(s)
- Zhiyi Du
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
| | - Mingsheng Li
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Guo Chen
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Maijie Xiang
- Division of Materials Science and Engineering, Boston University, Boston, MA, 02215, USA
| | - Danchen Jia
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| | - Ji-Xin Cheng
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Chen Yang
- Department of Chemistry, Boston University, Boston, MA, 02215, USA
- Department of Electrical and Computer Engineering, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
5
|
Yarotskyy V, Nass SR, Hahn YK, Contois L, McQuiston AR, Knapp PE, Hauser KF. Sustained fentanyl exposure inhibits neuronal activity in dissociated striatal neuronal-glial cocultures through actions independent of opioid receptors. J Neurophysiol 2024; 132:1056-1073. [PMID: 39110896 PMCID: PMC11427067 DOI: 10.1152/jn.00444.2023] [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: 11/30/2023] [Revised: 06/26/2024] [Accepted: 07/31/2024] [Indexed: 09/19/2024] Open
Abstract
Besides having high potency and efficacy at the µ-opioid (MOR) and other opioid receptor types, fentanyl has some affinity for some adrenergic receptor types, which may underlie its unique pathophysiological differences from typical opioids. To better understand the unique actions of fentanyl, we assessed the extent to which fentanyl alters striatal medium spiny neuron (MSN) activity via opioid receptors or α1-adrenoceptors in dopamine type 1 or type 2 receptor (D1 or D2)-expressing MSNs. In neuronal and mixed-glial cocultures from the striatum, acute fentanyl (100 nM) exposure decreased the frequency of spontaneous action potentials. Overnight exposure of cocultures to 100 nM fentanyl severely reduced the proportion of MSNs with spontaneous action potentials, which was unaffected by coexposure to the opioid receptor antagonist naloxone (10 µM) but fully negated by coadministering the pan-α1-adrenoceptor inverse agonist prazosin (100 nM) and partially reversed by the selective α1A-adrenoceptor antagonist RS 100329 (300 nM). Acute fentanyl (100 nM) exposure modestly reduced the frequency of action potentials and caused firing rate adaptations in D2, but not D1, MSNs. Prolonged (2-5 h) fentanyl (100 nM) application dramatically attenuated firing rates in both D1 and D2 MSNs. To identify possible cellular sites of α1-adrenoceptor action, α1-adrenoceptors were localized in subpopulations of striatal astroglia and neurons by immunocytochemistry and Adra1a mRNA by in situ hybridization in astrocytes. Thus, sustained fentanyl exposure can inhibit striatal MSN activity via a nonopioid receptor-dependent pathway, which may be modulated via complex actions in α1-adrenoceptor-expressing striatal neurons and/or glia.NEW & NOTEWORTHY Acute fentanyl exposure attenuated the activity of striatal medium spiny neurons (MSNs) in vitro and in dopamine D2, but not D1, receptor-expressing MSNs in ex vivo slices. By contrast, sustained fentanyl exposure suppressed the spontaneous activity of MSNs cocultured with glia through a nonopioid receptor-dependent mechanism modulated, in part, by α1-adrenoceptors. Fentanyl exposure can affect striatal function via a nonopioid receptor mechanism of action that appears mediated by α1-adrenoreceptor-expressing striatal neurons and/or astroglia.
Collapse
Affiliation(s)
- Viktor Yarotskyy
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Yun-Kyung Hahn
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Liangru Contois
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - A Rory McQuiston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
- Institute for Drug and Alcohol Studies, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
6
|
Bader R, Linke S, Gernert S. Nonlinear dynamical social and political prediction algorithm for city planning and public participation using the impulse pattern formulation. CHAOS (WOODBURY, N.Y.) 2024; 34:093130. [PMID: 39298339 DOI: 10.1063/5.0211618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024]
Abstract
A nonlinear-dynamical algorithm for city planning is proposed as an impulse pattern formulation (IPF) for predicting relevant parameters such as health, artistic freedom, or financial developments of different social or political stakeholders over the cause of a planning process. The IPF has already shown high predictive precision at low computational cost in musical instrument simulations, brain dynamics, and human-human interactions. The social and political IPF consists of three basic equations of system state developments, self-adaptation of stakeholders, two adaptive interactions, and external impact terms suitable for respective planning situations. Typical scenarios of stakeholder interactions and developments are modeled by adjusting a set of system parameters. These include stakeholder reaction to external input, enhanced system stability through self-adaptation, stakeholder convergence due to adaptive interaction, as well as complex dynamics in terms of fixed stakeholder impacts. A workflow for implementing the algorithm in real city planning scenarios is outlined. This workflow includes machine learning of a suitable set of parameters suggesting best-practice planning to aim at the desired development of the planning process and its output.
Collapse
Affiliation(s)
- R Bader
- Institute of Systematic Musicology, University of Hamburg, Neue Rabenstr. 13, 20354 Hamburg, Germany
| | - S Linke
- Hamburg University for Applied Sciences, Ligeti Center, Veritaskai, 1, 21079 Hamburg, Germany
| | - S Gernert
- Freie und Hansestadt Hamburg Landesbetrieb Immobilienmanagement und Grundvermögen, Millerntorplatz 1, 20359 Hamburg, Germany
| |
Collapse
|
7
|
Rostami V, Rost T, Schmitt FJ, van Albada SJ, Riehle A, Nawrot MP. Spiking attractor model of motor cortex explains modulation of neural and behavioral variability by prior target information. Nat Commun 2024; 15:6304. [PMID: 39060243 PMCID: PMC11282312 DOI: 10.1038/s41467-024-49889-4] [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: 03/29/2022] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
When preparing a movement, we often rely on partial or incomplete information, which can decrement task performance. In behaving monkeys we show that the degree of cued target information is reflected in both, neural variability in motor cortex and behavioral reaction times. We study the underlying mechanisms in a spiking motor-cortical attractor model. By introducing a biologically realistic network topology where excitatory neuron clusters are locally balanced with inhibitory neuron clusters we robustly achieve metastable network activity across a wide range of network parameters. In application to the monkey task, the model performs target-specific action selection and accurately reproduces the task-epoch dependent reduction of trial-to-trial variability in vivo where the degree of reduction directly reflects the amount of processed target information, while spiking irregularity remained constant throughout the task. In the context of incomplete cue information, the increased target selection time of the model can explain increased behavioral reaction times. We conclude that context-dependent neural and behavioral variability is a signum of attractor computation in the motor cortex.
Collapse
Affiliation(s)
- Vahid Rostami
- Institute of Zoology, University of Cologne, Cologne, Germany
| | - Thomas Rost
- Institute of Zoology, University of Cologne, Cologne, Germany
| | | | - Sacha Jennifer van Albada
- Institute of Zoology, University of Cologne, Cologne, Germany
- Institute for Advanced Simulation (IAS-6), Jülich Research Center, Jülich, Germany
| | - Alexa Riehle
- Institute for Advanced Simulation (IAS-6), Jülich Research Center, Jülich, Germany
- UMR7289 Institut de Neurosciences de la Timone (INT), Centre National de la Recherche Scientifique (CNRS)-Aix-Marseille Université (AMU), Marseille, France
| | | |
Collapse
|
8
|
Mittal D, Narayanan R. Network motifs in cellular neurophysiology. Trends Neurosci 2024; 47:506-521. [PMID: 38806296 DOI: 10.1016/j.tins.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
Concepts from network science and graph theory, including the framework of network motifs, have been frequently applied in studying neuronal networks and other biological complex systems. Network-based approaches can also be used to study the functions of individual neurons, where cellular elements such as ion channels and membrane voltage are conceptualized as nodes within a network, and their interactions are denoted by edges. Network motifs in this context provide functional building blocks that help to illuminate the principles of cellular neurophysiology. In this review we build a case that network motifs operating within neurons provide tools for defining the functional architecture of single-neuron physiology and neuronal adaptations. We highlight the presence of such computational motifs in the cellular mechanisms underlying action potential generation, neuronal oscillations, dendritic integration, and neuronal plasticity. Future work applying the network motifs perspective may help to decipher the functional complexities of neurons and their adaptation during health and disease.
Collapse
Affiliation(s)
- Divyansh Mittal
- Centre for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Rishikesh Narayanan
- Cellular Neurophysiology Laboratory, Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
9
|
Mandracchia B, Zheng C, Rajendran S, Liu W, Forghani P, Xu C, Jia S. High-speed optical imaging with sCMOS pixel reassignment. Nat Commun 2024; 15:4598. [PMID: 38816394 PMCID: PMC11139943 DOI: 10.1038/s41467-024-48987-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Fluorescence microscopy has undergone rapid advancements, offering unprecedented visualization of biological events and shedding light on the intricate mechanisms governing living organisms. However, the exploration of rapid biological dynamics still poses a significant challenge due to the limitations of current digital camera architectures and the inherent compromise between imaging speed and other capabilities. Here, we introduce sHAPR, a high-speed acquisition technique that leverages the operating principles of sCMOS cameras to capture fast cellular and subcellular processes. sHAPR harnesses custom fiber optics to convert microscopy images into one-dimensional recordings, enabling acquisition at the maximum camera readout rate, typically between 25 and 250 kHz. We have demonstrated the utility of sHAPR with a variety of phantom and dynamic systems, including high-throughput flow cytometry, cardiomyocyte contraction, and neuronal calcium waves, using a standard epi-fluorescence microscope. sHAPR is highly adaptable and can be integrated into existing microscopy systems without requiring extensive platform modifications. This method pushes the boundaries of current fluorescence imaging capabilities, opening up new avenues for investigating high-speed biological phenomena.
Collapse
Affiliation(s)
- Biagio Mandracchia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- E.T.S.I. Telecomunicación, Universidad de Valladolid, Valladolid, Spain
| | - Corey Zheng
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Suraj Rajendran
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Wenhao Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Parvin Forghani
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Chunhui Xu
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
10
|
Kim H, Zhong Z, Cui X, Sung H, Agrawal N, Jiang T, Dus M, Yapici N. HisCl1 regulates gustatory habituation in sweet taste neurons and mediates sugar ingestion in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592591. [PMID: 38765964 PMCID: PMC11100615 DOI: 10.1101/2024.05.06.592591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Similar to other animals, the fly, Drosophila melanogaster, reduces its responsiveness to tastants with repeated exposure, a phenomenon called gustatory habituation. Previous studies have focused on the circuit basis of gustatory habituation in the fly chemosensory system1,2. However, gustatory neurons reduce their firing rate during repeated stimulation3, suggesting that cell-autonomous mechanisms also contribute to habituation. Here, we used deep learning-based pose estimation and optogenetic stimulation to demonstrate that continuous activation of sweet taste neurons causes gustatory habituation in flies. We conducted a transgenic RNAi screen to identify genes involved in this process and found that knocking down Histamine-gated chloride channel subunit 1 (HisCl1) in the sweet taste neurons significantly reduced gustatory habituation. Anatomical analysis showed that HisCl1 is expressed in the sweet taste neurons of various chemosensory organs. Using single sensilla electrophysiology, we showed that sweet taste neurons reduced their firing rate with prolonged exposure to sucrose. Knocking down HisCl1 in sweet taste neurons suppressed gustatory habituation by reducing the spike frequency adaptation observed in these neurons during high-concentration sucrose stimulation. Finally, we showed that flies lacking HisCl1 in sweet taste neurons increased their consumption of high-concentration sucrose solution at their first meal bout compared to control flies. Together, our results demonstrate that HisCl1 tunes spike frequency adaptation in sweet taste neurons and contributes to gustatory habituation and food intake regulation in flies. Since HisCl1 is highly conserved across many dipteran and hymenopteran species, our findings open a new direction in studying insect gustatory habituation.
Collapse
Affiliation(s)
- Haein Kim
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Ziqing Zhong
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
- Current address: Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA, USA
| | - Xinyue Cui
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Hayeon Sung
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - Naman Agrawal
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Tianxing Jiang
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | - Monica Dus
- Department of Molecular, Cellular and Developmental Biology, The University of Michigan, Ann Arbor, MI, USA
| | - Nilay Yapici
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| |
Collapse
|
11
|
Kanigowski D, Urban-Ciecko J. Conditioning and pseudoconditioning differently change intrinsic excitability of inhibitory interneurons in the neocortex. Cereb Cortex 2024; 34:bhae109. [PMID: 38572735 PMCID: PMC10993172 DOI: 10.1093/cercor/bhae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Many studies indicate a broad role of various classes of GABAergic interneurons in the processes related to learning. However, little is known about how the learning process affects intrinsic excitability of specific classes of interneurons in the neocortex. To determine this, we employed a simple model of conditional learning in mice where vibrissae stimulation was used as a conditioned stimulus and a tail shock as an unconditioned one. In vitro whole-cell patch-clamp recordings showed an increase in intrinsic excitability of low-threshold spiking somatostatin-expressing interneurons (SST-INs) in layer 4 (L4) of the somatosensory (barrel) cortex after the conditioning paradigm. In contrast, pseudoconditioning reduced intrinsic excitability of SST-LTS, parvalbumin-expressing interneurons (PV-INs), and vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) with accommodating pattern in L4 of the barrel cortex. In general, increased intrinsic excitability was accompanied by narrowing of action potentials (APs), whereas decreased intrinsic excitability coincided with AP broadening. Altogether, these results show that both conditioning and pseudoconditioning lead to plastic changes in intrinsic excitability of GABAergic interneurons in a cell-specific manner. In this way, changes in intrinsic excitability can be perceived as a common mechanism of learning-induced plasticity in the GABAergic system.
Collapse
Affiliation(s)
- Dominik Kanigowski
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Joanna Urban-Ciecko
- Laboratory of Electrophysiology, Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093 Warsaw, Poland
| |
Collapse
|
12
|
Martini L, Amprimo G, Di Carlo S, Olmo G, Ferraris C, Savino A, Bardini R. Neuronal Spike Shapes (NSS): A straightforward approach to investigate heterogeneity in neuronal excitability states. Comput Biol Med 2024; 168:107783. [PMID: 38056213 DOI: 10.1016/j.compbiomed.2023.107783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/23/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023]
Abstract
The mammalian brain exhibits a remarkable diversity of neurons, contributing to its intricate architecture and functional complexity. The analysis of multimodal single-cell datasets enables the investigation of cell types and states heterogeneity. In this study, we introduce the Neuronal Spike Shapes (NSS), a straightforward approach for the exploration of excitability states of neurons based on their Action Potential (AP) waveforms. The NSS method describes the AP waveform based on a triangular representation complemented by a set of derived electrophysiological (EP) features. To support this hypothesis, we validate the proposed approach on two datasets of murine cortical neurons, focusing it on GABAergic neurons. The validation process involves a combination of NSS-based clustering analysis, features exploration, Differential Expression (DE), and Gene Ontology (GO) enrichment analysis. Results show that the NSS-based analysis captures neuronal excitability states that possess biological relevance independently of cell subtype. In particular, Neuronal Spike Shapes (NSS) captures, among others, a well-characterized fast-spiking excitability state, supported by both electrophysiological and transcriptomic validation. Gene Ontology Enrichment Analysis reveals voltage-gated potassium (K+) channels as specific markers of the identified NSS partitions. This finding strongly corroborates the biological relevance of NSS partitions as excitability states, as the expression of voltage-gated K+ channels regulates the hyperpolarization phase of the AP, being directly implicated in the regulation of neuronal excitability.
Collapse
Affiliation(s)
- Lorenzo Martini
- Politecnico di Torino - Control and Computer Engineering Department, Corso Duca degli Abruzzi, 24, Turin, 10129, Italy.
| | - Gianluca Amprimo
- Politecnico di Torino - Control and Computer Engineering Department, Corso Duca degli Abruzzi, 24, Turin, 10129, Italy; Institute of Electronics, Information Engineering and Telecommunications, National Research Council, Corso Duca degli Abruzzi, 24, Turin, 10029, Italy.
| | - Stefano Di Carlo
- Politecnico di Torino - Control and Computer Engineering Department, Corso Duca degli Abruzzi, 24, Turin, 10129, Italy. https://www.smilies.polito.it
| | - Gabriella Olmo
- Politecnico di Torino - Control and Computer Engineering Department, Corso Duca degli Abruzzi, 24, Turin, 10129, Italy. https://www.sysbio.polito.it/analytics-technologies-health/
| | - Claudia Ferraris
- Institute of Electronics, Information Engineering and Telecommunications, National Research Council, Corso Duca degli Abruzzi, 24, Turin, 10029, Italy. https://www.ieiit.cnr.it/people/Ferraris-Claudia
| | - Alessandro Savino
- Politecnico di Torino - Control and Computer Engineering Department, Corso Duca degli Abruzzi, 24, Turin, 10129, Italy. https://www.smilies.polito.it
| | - Roberta Bardini
- Politecnico di Torino - Control and Computer Engineering Department, Corso Duca degli Abruzzi, 24, Turin, 10129, Italy.
| |
Collapse
|
13
|
Naderi S, Motamedi F, Pourbadie HG, Rafiei S, Khodagholi F, Naderi N, Janahmadi M. Neuroprotective Effects of Ferrostatin and Necrostatin Against Entorhinal Amyloidopathy-Induced Electrophysiological Alterations Mediated by voltage-gated Ca 2+ Channels in the Dentate Gyrus Granular Cells. Neurochem Res 2024; 49:99-116. [PMID: 37615884 DOI: 10.1007/s11064-023-04006-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/07/2023] [Accepted: 07/29/2023] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is the main form of dementia. Abnormal deposition of amyloid-beta (Aβ) peptides in neurons and synapses cause neuronal loss and cognitive deficits. We have previously reported that ferroptosis and necroptosis were implicated in Aβ25-35 neurotoxicity, and their specific inhibitors had attenuating effects on cognitive impairment induced by Aβ25-35 neurotoxicity. Here, we aimed to examine the impact of ferroptosis and necroptosis inhibition following the Aβ25-35 neurotoxicity on the neuronal excitability of dentate gyrus (DG) and the possible involvement of voltage-gated Ca2+ channels in their effects. After inducing Aβ25-35 neurotoxicity, electrophysiological alterations in the intrinsic properties and excitability were recorded by the whole-cell patch-clamp under current-clamp condition. Voltage-clamp recordings were also performed to shed light on the involvement of calcium channel currents. Aβ25-35 neurotoxicity induced a considerable reduction in input resistance (Rin), accompanied by a profoundly decreased excitability and a reduction in the amplitude of voltage-gated calcium channel currents in the DG granule cells. However, three days of administration of either ferrostatin-1 (Fer-1), a ferroptosis inhibitor, or Necrostatin-1 (Nec-1), a necroptosis inhibitor, in the entorhinal cortex could almost preserve the normal excitability and the Ca2+ currents. In conclusion, these findings suggest that ferroptosis and necroptosis involvement in EC amyloidopathy could be a potential candidate to prevent the suppressive effect of Aβ on the Ca2+ channel current and neuronal function, which might take place in neurons during the development of AD.
Collapse
Affiliation(s)
- Soudabeh Naderi
- School of Medicine, Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shahrbanoo Rafiei
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Naderi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Studtmann C, Ladislav M, Safari M, Khondaker R, Chen Y, Vaughan GA, Topolski MA, Tomović E, Balík A, Swanger SA. Ventral posterolateral and ventral posteromedial thalamocortical neurons have distinct physiological properties. J Neurophysiol 2023; 130:1492-1507. [PMID: 37937368 PMCID: PMC11068404 DOI: 10.1152/jn.00525.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/09/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023] Open
Abstract
Somatosensory information is propagated from the periphery to the cerebral cortex by two parallel pathways through the ventral posterolateral (VPL) and ventral posteromedial (VPM) thalamus. VPL and VPM neurons receive somatosensory signals from the body and head, respectively. VPL and VPM neurons may also receive cell type-specific GABAergic input from the reticular nucleus of the thalamus. Although VPL and VPM neurons have distinct connectivity and physiological roles, differences in their functional properties remain unclear as they are often studied as one ventrobasal thalamus neuron population. Here, we directly compared synaptic and intrinsic properties of VPL and VPM neurons in C57Bl/6J mice of both sexes aged P25-P32. VPL neurons showed greater depolarization-induced spike firing and spike frequency adaptation than VPM neurons. VPL and VPM neurons fired similar numbers of spikes during hyperpolarization rebound bursts, but VPM neurons exhibited shorter burst latency compared with VPL neurons, which correlated with larger sag potential. VPM neurons had larger membrane capacitance and more complex dendritic arbors. Recordings of spontaneous and evoked synaptic transmission suggested that VPL neurons receive stronger excitatory synaptic input, whereas inhibitory synapse strength was stronger in VPM neurons. This work indicates that VPL and VPM thalamocortical neurons have distinct intrinsic and synaptic properties. The observed functional differences could have important implications for their specific physiological and pathophysiological roles within the somatosensory thalamocortical network.NEW & NOTEWORTHY This study revealed that somatosensory thalamocortical neurons in the VPL and VPM have substantial differences in excitatory synaptic input and intrinsic firing properties. The distinct properties suggest that VPL and VPM neurons could process somatosensory information differently and have selective vulnerability to disease. This work improves our understanding of nucleus-specific neuron function in the thalamus and demonstrates the critical importance of studying these parallel somatosensory pathways separately.
Collapse
Affiliation(s)
- Carleigh Studtmann
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Marek Ladislav
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
| | - Mona Safari
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Rabeya Khondaker
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Yang Chen
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia, United States
| | - Grace A Vaughan
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia, United States
| | - Mackenzie A Topolski
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
| | - Eni Tomović
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Aleš Balík
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Sharon A Swanger
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, Virginia, United States
| |
Collapse
|
15
|
Nanclares C, Noriega-Prieto JA, Labrada-Moncada FE, Cvetanovic M, Araque A, Kofuji P. Altered calcium signaling in Bergmann glia contributes to spinocerebellar ataxia type-1 in a mouse model of SCA1. Neurobiol Dis 2023; 187:106318. [PMID: 37802154 PMCID: PMC10624966 DOI: 10.1016/j.nbd.2023.106318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by an abnormal expansion of glutamine (Q) encoding CAG repeats in the ATAXIN1 (ATXN1) gene and characterized by progressive cerebellar ataxia, dysarthria, and eventual deterioration of bulbar functions. SCA1 shows severe degeneration of cerebellar Purkinje cells (PCs) and activation of Bergmann glia (BG), a type of cerebellar astroglia closely associated with PCs. Combining electrophysiological recordings, calcium imaging techniques, and chemogenetic approaches, we have investigated the electrical intrinsic and synaptic properties of PCs and the physiological properties of BG in SCA1 mouse model expressing mutant ATXN1 only in PCs. PCs of SCA1 mice displayed lower spontaneous firing rate and larger slow afterhyperpolarization currents (sIAHP) than wildtype mice, whereas the properties of the synaptic inputs were unaffected. BG of SCA1 mice showed higher calcium hyperactivity and gliotransmission, manifested by higher frequency of NMDAR-mediated slow inward currents (SICs) in PC. Preventing the BG calcium hyperexcitability of SCA1 mice by loading BG with the calcium chelator BAPTA restored sIAHP and spontaneous firing rate of PCs to similar levels of wildtype mice. Moreover, mimicking the BG hyperactivity by activating BG expressing Gq-DREADDs in wildtype mice reproduced the SCA1 pathological phenotype of PCs, i.e., enhancement of sIAHP and decrease of spontaneous firing rate. These results indicate that the intrinsic electrical properties of PCs, but not their synaptic properties, were altered in SCA1 mice and that these alterations were associated with the hyperexcitability of BG. Moreover, preventing BG hyperexcitability in SCA1 mice and promoting BG hyperexcitability in wildtype mice prevented and mimicked, respectively, the pathological electrophysiological phenotype of PCs. Therefore, BG plays a relevant role in the dysfunction of the electrical intrinsic properties of PCs in SCA1 mice, suggesting that they may serve as potential targets for therapeutic approaches to treat the spinocerebellar ataxia type 1.
Collapse
Affiliation(s)
- Carmen Nanclares
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
16
|
Kern FB, Chao ZC. Short-term neuronal and synaptic plasticity act in synergy for deviance detection in spiking networks. PLoS Comput Biol 2023; 19:e1011554. [PMID: 37831721 PMCID: PMC10599548 DOI: 10.1371/journal.pcbi.1011554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/25/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Sensory areas of cortex respond more strongly to infrequent stimuli when these violate previously established regularities, a phenomenon known as deviance detection (DD). Previous modeling work has mainly attempted to explain DD on the basis of synaptic plasticity. However, a large fraction of cortical neurons also exhibit firing rate adaptation, an underexplored potential mechanism. Here, we investigate DD in a spiking neuronal network model with two types of short-term plasticity, fast synaptic short-term depression (STD) and slower threshold adaptation (TA). We probe the model with an oddball stimulation paradigm and assess DD by evaluating the network responses. We find that TA is sufficient to elicit DD. It achieves this by habituating neurons near the stimulation site that respond earliest to the frequently presented standard stimulus (local fatigue), which diminishes the response and promotes the recovery (global fatigue) of the wider network. Further, we find a synergy effect between STD and TA, where they interact with each other to achieve greater DD than the sum of their individual effects. We show that this synergy is caused by the local fatigue added by STD, which inhibits the global response to the frequently presented stimulus, allowing greater recovery of TA-mediated global fatigue and making the network more responsive to the deviant stimulus. Finally, we show that the magnitude of DD strongly depends on the timescale of stimulation. We conclude that highly predictable information can be encoded in strong local fatigue, which allows greater global recovery and subsequent heightened sensitivity for DD.
Collapse
Affiliation(s)
- Felix Benjamin Kern
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| | - Zenas C. Chao
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Borges FS, Protachevicz PR, Souza DLM, Bittencourt CF, Gabrick EC, Bentivoglio LE, Szezech JD, Batista AM, Caldas IL, Dura-Bernal S, Pena RFO. The Roles of Potassium and Calcium Currents in the Bistable Firing Transition. Brain Sci 2023; 13:1347. [PMID: 37759949 PMCID: PMC10527161 DOI: 10.3390/brainsci13091347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Healthy brains display a wide range of firing patterns, from synchronized oscillations during slow-wave sleep to desynchronized firing during movement. These physiological activities coexist with periods of pathological hyperactivity in the epileptic brain, where neurons can fire in synchronized bursts. Most cortical neurons are pyramidal regular spiking (RS) cells with frequency adaptation and do not exhibit bursts in current-clamp experiments (in vitro). In this work, we investigate the transition mechanism of spike-to-burst patterns due to slow potassium and calcium currents, considering a conductance-based model of a cortical RS cell. The joint influence of potassium and calcium ion channels on high synchronous patterns is investigated for different synaptic couplings (gsyn) and external current inputs (I). Our results suggest that slow potassium currents play an important role in the emergence of high-synchronous activities, as well as in the spike-to-burst firing pattern transitions. This transition is related to the bistable dynamics of the neuronal network, where physiological asynchronous states coexist with pathological burst synchronization. The hysteresis curve of the coefficient of variation of the inter-spike interval demonstrates that a burst can be initiated by firing states with neuronal synchronization. Furthermore, we notice that high-threshold (IL) and low-threshold (IT) ion channels play a role in increasing and decreasing the parameter conditions (gsyn and I) in which bistable dynamics occur, respectively. For high values of IL conductance, a synchronous burst appears when neurons are weakly coupled and receive more external input. On the other hand, when the conductance IT increases, higher coupling and lower I are necessary to produce burst synchronization. In light of our results, we suggest that channel subtype-specific pharmacological interactions can be useful to induce transitions from pathological high bursting states to healthy states.
Collapse
Affiliation(s)
- Fernando S. Borges
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Center for Mathematics, Computation and Cognition, Federal University of ABC, São Bernardo do Campo 09606-045, Brazil
| | | | - Diogo L. M. Souza
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - Conrado F. Bittencourt
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - Enrique C. Gabrick
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - Lucas E. Bentivoglio
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
| | - José D. Szezech
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa 84030-900, Brazil
| | - Antonio M. Batista
- Graduate Program in Science, State University of Ponta Grossa, Ponta Grossa 84010-330, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa 84030-900, Brazil
| | - Iberê L. Caldas
- Institute of Physics, University of São Paulo, São Paulo 05508-090, Brazil
| | - Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
- Center for Biomedical Imaging and Neuromodulation, The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - Rodrigo F. O. Pena
- Department of Biological Sciences, Florida Atlantic University, Jupiter, FL 33458, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
18
|
Xiao K, Li Y, Chitwood RA, Magee JC. A critical role for CaMKII in behavioral timescale synaptic plasticity in hippocampal CA1 pyramidal neurons. SCIENCE ADVANCES 2023; 9:eadi3088. [PMID: 37672577 PMCID: PMC10482326 DOI: 10.1126/sciadv.adi3088] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/03/2023] [Indexed: 09/08/2023]
Abstract
Behavioral timescale synaptic plasticity (BTSP) is a type of non-Hebbian synaptic plasticity reported to underlie place field formation. Despite this important function, the molecular mechanisms underlying BTSP are poorly understood. The α-calcium-calmodulin-dependent protein kinase II (αCaMKII) is activated by synaptic transmission-mediated calcium influx, and its subsequent phosphorylation is central to synaptic plasticity. Because the activity of αCaMKII is known to outlast the event triggering phosphorylation, we hypothesized that it could mediate the extended timescale of BTSP. To examine the role of αCaMKII in BTSP, we performed whole-cell in vivo and in vitro recordings in CA1 pyramidal neurons from mice engineered with a point mutation at the autophosphorylation site (T286A) causing accelerated signaling kinetics. Here, we demonstrate a profound deficit in synaptic plasticity, strongly suggesting that αCaMKII signaling is required for BTSP. This study elucidates part of the molecular mechanism of BTSP and provides insight into the function of αCaMKII in place cell formation and ultimately learning and memory.
Collapse
Affiliation(s)
- Kuo Xiao
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Yiding Li
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Raymond A. Chitwood
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey C. Magee
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Borges FS, Protachevicz PR, Souza DLM, Bittencourt CF, Gabrick EC, Bentivoglio LE, Szezech JD, Batista AM, Caldas IL, Dura-Bernal S, Pena RFO. The Role of Potassium and Calcium Currents in the Bistable Firing Transition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553625. [PMID: 37645875 PMCID: PMC10462112 DOI: 10.1101/2023.08.16.553625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Healthy brains display a wide range of firing patterns, from synchronized oscillations during slowwave sleep to desynchronized firing during movement. These physiological activities coexist with periods of pathological hyperactivity in the epileptic brain, where neurons can fire in synchronized bursts. Most cortical neurons are pyramidal regular spiking cells (RS) with frequency adaptation and do not exhibit bursts in current-clamp experiments ( in vitro ). In this work, we investigate the transition mechanism of spike-to-burst patterns due to slow potassium and calcium currents, considering a conductance-based model of a cortical RS cell. The joint influence of potassium and calcium ion channels on high synchronous patterns is investigated for different synaptic couplings ( g syn ) and external current inputs ( I ). Our results suggest that slow potassium currents play an important role in the emergence of high-synchronous activities, as well as in the spike-to-burst firing pattern transitions. This transition is related to bistable dynamics of the neuronal network, where physiological asynchronous states coexist with pathological burst synchronization. The hysteresis curve of the coefficient of variation of the inter-spike interval demonstrates that a burst can be initiated by firing states with neuronal synchronization. Furthermore, we notice that high-threshold ( I L ) and low-threshold ( I T ) ion channels play a role in increasing and decreasing the parameter conditions ( g syn and I ) in which bistable dynamics occur, respectively. For high values of I L conductance, a synchronous burst appears when neurons are weakly coupled and receive more external input. On the other hand, when the conductance I T increases, higher coupling and lower I are necessary to produce burst synchronization. In light of our results, we suggest that channel subtype-specific pharmacological interactions can be useful to induce transitions from pathological high bursting states to healthy states.
Collapse
Affiliation(s)
- Fernando S Borges
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
- Center for Mathematics, Computation, and Cognition, Federal University of ABC, 09606-045 São Bernardo do Campo, SP, Brazil
| | | | - Diogo L M Souza
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Conrado F Bittencourt
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Enrique C Gabrick
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Lucas E Bentivoglio
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - José D Szezech
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Antonio M Batista
- Graduate Program in Science, State University of Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Iberê L Caldas
- Institute of Physics, University of São Paulo, 05508-090 São Paulo, SP, Brazil
| | - Salvador Dura-Bernal
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
- Center for Biomedical Imaging and Neuromodulation, The Nathan S. Kline Institute for Psychiatric Research, New York, USA
| | - Rodrigo F O Pena
- Department of Biological Sciences, Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
20
|
Wang J, Deng B, Gao T, Wang J, Tan H. Spike-frequency adaptation inhibits the pairwise spike correlation. Front Neurosci 2023; 17:1193930. [PMID: 37378017 PMCID: PMC10291049 DOI: 10.3389/fnins.2023.1193930] [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: 03/27/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction The spike train output correlation with pairwise neurons determines the neural population coding, which depends on the average firing rate of individual neurons. Spike frequency adaptation (SFA), which serves as an essential cellular encoding strategy, modulates the firing rates of individual neurons. However, the mechanism by which the SFA modulates the output correlation of the spike trains remains unclear. Methods We introduce a pairwise neuron model that receives correlated inputs to generate spike trains, and the output correlation is qualified using Pearson correlation coefficient. The SFA is modeled using adaptation currents to examine its effect on the output correlation. Moreover, we use dynamic thresholds to explore the effect of SFA on output correlation. Furthermore, a simple phenomenological neuron model with a threshold-linear transfer function is utilized to confirm the effect of SFA on decreasing the output correlation. Results The results show that the adaptation currents decreased the output correlation by reducing the firing rate of a single neuron. At the onset of a correlated input, a transient process shows a decrease in interspike intervals (ISIs), resulting in a temporary increase in the correlation. When the adaptation current is sufficiently activated, the correlation reached a steady state, and the ISIs are maintained at higher values. The enhanced adaptation current achieved by increasing the adaptation conductance further reduces the pairwise correlation. While the time and slide windows influence the correlation, they make no difference in the effect of SFA on decreasing the output correlation. Moreover, SFA simulated by dynamic thresholds also decreases the output correlation. Furthermore, the simple phenomenological neuron model with a threshold-linear transfer function confirms the effect of SFA on decreasing the output correlation. The strength of the signal input and the slope of the linear component of the transfer function, the latter of which can be decreased by SFA, could together modulate the strength of the output correlation. Stronger SFA will decrease the slope and hence decrease the output correlation. Conclusions The results reveal that the SFA reduces the output correlation with pairwise neurons in the network by reducing the firing rate of individual neurons. This study provides a link between cellular non-linear mechanisms and network coding strategies.
Collapse
Affiliation(s)
- Jixuan Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Bin Deng
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Tianshi Gao
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Jiang Wang
- School of Electrical and Information Engineering, Tianjin University, Tianjin, China
| | - Hong Tan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Salners T, Avila KE, Nicholson B, Myers CR, Beggs J, Dahmen KA. Recurrent activity in neuronal avalanches. Sci Rep 2023; 13:4871. [PMID: 36964158 PMCID: PMC10039060 DOI: 10.1038/s41598-023-31851-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/17/2023] [Indexed: 03/26/2023] Open
Abstract
A new statistical analysis of large neuronal avalanches observed in mouse and rat brain tissues reveals a substantial degree of recurrent activity and cyclic patterns of activation not seen in smaller avalanches. To explain these observations, we adapted a model of structural weakening in materials. In this model, dynamical weakening of neuron firing thresholds closely replicates experimental avalanche size distributions, firing number distributions, and patterns of cyclic activity. This agreement between model and data suggests that a mechanism like dynamical weakening plays a key role in recurrent activity found in large neuronal avalanches. We expect these results to illuminate the causes and dynamics of large avalanches, like those seen in seizures.
Collapse
Affiliation(s)
- Tyler Salners
- Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, IL, 61801, USA.
| | - Karina E Avila
- Physics Department, University Kaiserslautern, Erwin-Schrödinger-Straße, 67663, Kaiserslautern, Germany
| | - Benjamin Nicholson
- Laboratory of Atomic and Solid State Physics, Clark Hall, Cornell University, Ithaca, NY, 14853-2501, USA
| | - Christopher R Myers
- Laboratory of Atomic and Solid State Physics, Clark Hall, Cornell University, Ithaca, NY, 14853-2501, USA
- Center for Advanced Computing, Cornell University, Ithaca, NY, 14853, USA
| | - John Beggs
- Department of Physics, Indiana University, Bloomington, IN, 47405, USA
| | - Karin A Dahmen
- Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, IL, 61801, USA
| |
Collapse
|
22
|
Mechanism of kisspeptin neuron synchronization for pulsatile hormone secretion in male mice. Cell Rep 2023; 42:111914. [PMID: 36640343 DOI: 10.1016/j.celrep.2022.111914] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/31/2022] [Accepted: 12/13/2022] [Indexed: 01/04/2023] Open
Abstract
The mechanism by which arcuate nucleus kisspeptin (ARNKISS) neurons co-expressing glutamate, neurokinin B, and dynorphin intermittently synchronize their activity to generate pulsatile hormone secretion remains unknown. An acute brain slice preparation maintaining synchronized ARNKISS neuron burst firing was used alongside in vivo GCaMP GRIN lens microendoscope and fiber photometry imaging coupled with intra-ARN microinfusion. Studies in intact and gonadectomized male mice revealed that ARNKISS neuron synchronizations result from near-random emergent network activity within the population and that this was critically dependent on local glutamate-AMPA signaling. Whereas neurokinin B operated to potentiate glutamate-generated synchronizations, dynorphin-kappa opioid tone within the network served as a gate for synchronization initiation. These observations force a departure from the existing "KNDy hypothesis" for ARNKISS neuron synchronization. A "glutamate two-transition" mechanism is proposed to underlie synchronizations in this key hypothalamic central pattern generator driving mammalian fertility.
Collapse
|
23
|
Martin L, Jaime K, Ramos F, Robles F. Bio-inspired cognitive architecture of episodic memory. COGN SYST RES 2022. [DOI: 10.1016/j.cogsys.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
24
|
Snyder RR, Blitz DM. Multiple intrinsic membrane properties are modulated in a switch from single- to dual-network activity. J Neurophysiol 2022; 128:1181-1198. [PMID: 36197020 PMCID: PMC9621714 DOI: 10.1152/jn.00337.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/14/2022] [Accepted: 10/01/2022] [Indexed: 11/22/2022] Open
Abstract
Neural network flexibility includes changes in neuronal participation between networks, such as the switching of neurons between single- and dual-network activity. We previously identified a neuron that is recruited to burst in time with an additional network via modulation of its intrinsic membrane properties, instead of being recruited synaptically into the second network. However, the modulated intrinsic properties were not determined. Here, we use small networks in the Jonah crab (Cancer borealis) stomatogastric nervous system (STNS) to examine modulation of intrinsic properties underlying neuropeptide (Gly1-SIFamide)-elicited neuronal switching. The lateral posterior gastric neuron (LPG) switches from exclusive participation in the fast pyloric (∼1 Hz) network, due to electrical coupling, to dual-network activity that includes periodic escapes from the fast rhythm via intrinsically generated oscillations at the slower gastric mill network frequency (∼0.1 Hz). We isolated LPG from both networks by pharmacology and hyperpolarizing current injection. Gly1-SIFamide increased LPG intrinsic excitability and rebound from inhibition and decreased spike frequency adaptation, which can all contribute to intrinsic bursting. Using ion substitution and channel blockers, we found that a hyperpolarization-activated current, a persistent sodium current, and calcium or calcium-related current(s) appear to be primary contributors to Gly1-SIFamide-elicited LPG intrinsic bursting. However, this intrinsic bursting was more sensitive to blocking currents when LPG received rhythmic electrical coupling input from the fast network than in the isolated condition. Overall, a switch from single- to dual-network activity can involve modulation of multiple intrinsic properties, while synaptic input from a second network can shape the contributions of these properties.NEW & NOTEWORTHY Neuropeptide-elicited intrinsic bursting was recently determined to switch a neuron from single- to dual-network participation. Here we identified multiple intrinsic properties modulated in the dual-network state and candidate ion channels underlying the intrinsic bursting. Bursting at the second network frequency was more sensitive to blocking currents in the dual-network state than when neurons were synaptically isolated from their home network. Thus, synaptic input can shape the contributions of modulated intrinsic properties underlying dual-network activity.
Collapse
Affiliation(s)
- Ryan R Snyder
- Department of Biology and Center for Neuroscience, Miami University, Oxford, Ohio
| | - Dawn M Blitz
- Department of Biology and Center for Neuroscience, Miami University, Oxford, Ohio
| |
Collapse
|
25
|
Shen K, Duan Q, Duan W, Xu S, An N, Ke Y, Wang L, Liu S, Yang H, Zhang C. Vascular endothelial growth factor-C modulates cortical NMDA receptor activity in cortical lesions of young patients and rat model with focal cortical dysplasia. Brain Pathol 2022; 32:e13065. [PMID: 35259773 PMCID: PMC9425019 DOI: 10.1111/bpa.13065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/20/2022] [Accepted: 02/25/2022] [Indexed: 12/01/2022] Open
Abstract
Emergence of dysmorphic neurons is the primary pathology in focal cortical dysplasia (FCD) associated pediatric intractable epilepsy; however, the etiologies related to the development and function of dysmorphic neurons are not fully understood. Our previous studies revealed that the expression of vascular endothelial growth factor-C (VEGF-C) and corresponding receptors VEGFR-2, VEGFR-3 was increased in the epileptic lesions of patients with tuberous sclerosis complex or mesial temporal lobe epilepsy. Here, we showed that the expression of VEGF-C, VEGFR-2, and VEGFR-3 was increased at both mRNA and protein levels in patients with cortical lesions of type I, IIa, and IIb FCD. The immunoreactivity of VEGF-C, VEGFR-2 and VEGFR-3 was located in the micro-columnar neurons in FCD type I lesions, dysplastic neurons (DNs) in FCD type IIa lesions, balloon cells (BCs) and astrocytes in FCD type IIb lesions. Additionally, the amplitude of evoked-EPSCs (eEPSC) mediated by NMDA receptor, the ratio of NMDA receptor- and AMPA receptor-mediated eEPSC were increased in the dysmorphic neurons of FCD rats established by prenatal X-ray radiation. Furthermore, NMDA receptor mediated current in dysmorphic neurons was further potentiated by exogenous administration of VEGF-C, however, could be antagonized by ki8751, the blocker of VEGFR-2. These results suggest that VEGF-C system participate in the pathogenesis of cortical lesions in patients with FCD in association with modulating NMDA receptor-mediated currents.
Collapse
Affiliation(s)
- Kai‐Feng Shen
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Qing‐Tian Duan
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Wei Duan
- Department of NeurologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Sen‐Lin Xu
- Institute of PathologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Ning An
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Yan‐Yan Ke
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Li‐Ting Wang
- Biomedical Analysis CenterArmy Medical UniversityChongqingChina
| | - Shi‐Yong Liu
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Hui Yang
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
| | - Chun‐Qing Zhang
- Department of NeurosurgeryEpilepsy Research Center of PLAXinqiao HospitalArmy Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
| |
Collapse
|
26
|
Rahdar M, Hajisoltani R, Davoudi S, Karimi SA, Borjkhani M, Khatibi VA, Hosseinmardi N, Behzadi G, Janahmadi M. Alterations in the intrinsic discharge activity of CA1 pyramidal neurons associated with possible changes in the NADPH diaphorase activity in a rat model of autism induced by prenatal exposure to valproic acid. Brain Res 2022; 1792:148013. [PMID: 35841982 DOI: 10.1016/j.brainres.2022.148013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 07/10/2022] [Indexed: 11/02/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by sensory abnormalities, social skills impairment and cognitive deficits. Although recent evidence indicated that induction of autism-like behavior in animal models causes abnormal neuronal excitability, the impact of autism on neuronal properties is still an important issue. Thus, new findings at the cellular level may shed light on the pathophysiology of autism and may help to find effective treatment strategies. Here, we investigated the behavioral, electrophysiological and histochemical impacts of prenatal exposure to valproic acid (VPA) in rats. Findings revealed that VPA exposure caused a significant increase in the hot plate response latency. The novel object recognition ability was also impaired in VPA-exposed rats. Along with these behavioral alterations, neurons from VPA-exposed animals exhibited altered excitability features in response to depolarizing current injections relative to control neurons. In the VPA-exposed group, these changes consisted of a significant increase in the amplitude, evoked firing frequency and the steady-state standard deviation of spike timing of action potentials (APs). Moreover, the half-width, the AHP amplitude and the decay time constant of APs were significantly decreased in this group. These changes in the evoked electrophysiological properties were accompanied by intrinsic hyperexcitability and lower spike-frequency adaptation and also a significant increase in the number of NADPH-diaphorase stained neurons in the hippocampal CA1 area of the VPA-exposed rats. Taken together, findings demonstrate that abnormal nociception and recognition memory is associated with alterations in the neuronal responsiveness and nitrergic system in a rat model of autism-like.
Collapse
Affiliation(s)
- Mona Rahdar
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Razieh Hajisoltani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Davoudi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Asaad Karimi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Tehran, Iran
| | - Mehdi Borjkhani
- Department of Electrical Engineering, Urmia University of Technology, Urmia, Iran
| | - Vahid Ahli Khatibi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narges Hosseinmardi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gila Behzadi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Burelo K, Sharifshazileh M, Indiveri G, Sarnthein J. Automatic Detection of High-Frequency Oscillations With Neuromorphic Spiking Neural Networks. Front Neurosci 2022; 16:861480. [PMID: 35720714 PMCID: PMC9205405 DOI: 10.3389/fnins.2022.861480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Interictal high-frequency oscillations (HFO) detected in electroencephalography recordings have been proposed as biomarkers of epileptogenesis, seizure propensity, disease severity, and treatment response. Automatic HFO detectors typically analyze the data offline using complex time-consuming algorithms, which limits their clinical application. Neuromorphic circuits offer the possibility of building compact and low-power processing systems that can analyze data on-line and in real time. In this review, we describe a fully automated detection pipeline for HFO that uses, for the first time, spiking neural networks and neuromorphic technology. We demonstrated that our HFO detection pipeline can be applied to recordings from different modalities (intracranial electroencephalography, electrocorticography, and scalp electroencephalography) and validated its operation in a custom-designed neuromorphic processor. Our HFO detection approach resulted in high accuracy and specificity in the prediction of seizure outcome in patients implanted with intracranial electroencephalography and electrocorticography, and in the prediction of epilepsy severity in patients recorded with scalp electroencephalography. Our research provides a further step toward the real-time detection of HFO using compact and low-power neuromorphic devices. The real-time detection of HFO in the operation room may improve the seizure outcome of epilepsy surgery, while the use of our neuromorphic processor for non-invasive therapy monitoring might allow for more effective medication strategies to achieve seizure control. Therefore, this work has the potential to improve the quality of life in patients with epilepsy by improving epilepsy diagnostics and treatment.
Collapse
Affiliation(s)
- Karla Burelo
- Klinik für Neurochirurgie, UniversitätsSpital Zürich, Universität Zürich, Zurich, Switzerland
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich, Switzerland
| | | | - Giacomo Indiveri
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zurich, ETH und Universität Zürich, Zurich, Switzerland
| | - Johannes Sarnthein
- Klinik für Neurochirurgie, UniversitätsSpital Zürich, Universität Zürich, Zurich, Switzerland
- Zentrum für Neurowissenschaften Zurich, ETH und Universität Zürich, Zurich, Switzerland
| |
Collapse
|
28
|
Medalla M, Chang W, Ibañez S, Guillamon-Vivancos T, Nittmann M, Kapitonava A, Busch SE, Moore TL, Rosene DL, Luebke JI. Layer-specific pyramidal neuron properties underlie diverse anterior cingulate cortical motor and limbic networks. Cereb Cortex 2022; 32:2170-2196. [PMID: 34613380 PMCID: PMC9113240 DOI: 10.1093/cercor/bhab347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/13/2022] Open
Abstract
The laminar cellular and circuit mechanisms by which the anterior cingulate cortex (ACC) exerts flexible control of motor and affective information for goal-directed behavior have not been elucidated. Using multimodal tract-tracing, in vitro patch-clamp recording and computational approaches in rhesus monkeys (M. mulatta), we provide evidence that specialized motor and affective network dynamics can be conferred by layer-specific biophysical and structural properties of ACC pyramidal neurons targeting two key downstream structures -the dorsal premotor cortex (PMd) and the amygdala (AMY). AMY-targeting neurons exhibited significant laminar differences, with L5 more excitable (higher input resistance and action potential firing rates) than L3 neurons. Between-pathway differences were found within L5, with AMY-targeting neurons exhibiting greater excitability, apical dendritic complexity, spine densities, and diversity of inhibitory inputs than PMd-targeting neurons. Simulations using a pyramidal-interneuron network model predict that these layer- and pathway-specific single-cell differences contribute to distinct network oscillatory dynamics. L5 AMY-targeting networks are more tuned to slow oscillations well-suited for affective and contextual processing timescales, while PMd-targeting networks showed strong beta/gamma synchrony implicated in rapid sensorimotor processing. These findings are fundamental to our broad understanding of how layer-specific cellular and circuit properties can drive diverse laminar activity found in flexible behavior.
Collapse
Affiliation(s)
- Maria Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Wayne Chang
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Sara Ibañez
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Teresa Guillamon-Vivancos
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Instituto de Neurociencias de Alicante, Alicante, Spain
| | - Mathias Nittmann
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- University of South Florida, Morsani College of Medicine, Tampa, FL, 33612, USA
| | - Anastasia Kapitonava
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Silas E Busch
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Department of Neurobiology, University of Chicago, Chicago, IL, 60637, USA
| | - Tara L Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Douglas L Rosene
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
29
|
Zhao S, Liu D, Liu M, Luo X, Yuan Y. Theoretical analysis of effects of transcranial magneto-acoustical stimulation on neuronal spike-frequency adaptation. BMC Neurosci 2022; 23:26. [PMID: 35501687 PMCID: PMC9063290 DOI: 10.1186/s12868-022-00709-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/04/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Transcranial magneto-acoustical stimulation (TMAS) is a noninvasive technique that has advantages in spatial resolution and penetration depth. It changes the firing properties of neurons through the current generated by focused ultrasound and a static magnetic field. Spike-frequency adaptation is an important dynamic characteristic of neural information processing. METHODS To address the effects of TMAS on neural spike-frequency adaptation, this study employs some ultrasound and magnetic field parameters, such as magnetic flux density, ultrasonic intensity, fundamental ultrasonic frequency, modulation frequency, and duty cycle. Using these different ultrasound and magnetic field parameters, membrane potential curves, spike-frequency curves, and adapted onset spike-frequency curves are exhibited and analyzed. RESULTS The results show that spike-frequency adaptation is strongly dependent on ultrasonic intensity and magnetic flux density and is rarely affected by other parameters. However, modulation frequency and duty cycle influence membrane potentials and spike frequencies to some degree. CONCLUSIONS This study reveals the mechanism of the effects of TMAS on neural spike-frequency adaptation and serves as theoretical guidance for TMAS experiments.
Collapse
Affiliation(s)
- Song Zhao
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Dan Liu
- Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Minzhuang Liu
- Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaoyuan Luo
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, China
| | - Yi Yuan
- Institute of Electrical Engineering, Yanshan University, Qinhuangdao, China
| |
Collapse
|
30
|
Franović I, Eydam S, Yanchuk S, Berner R. Collective Activity Bursting in a Population of Excitable Units Adaptively Coupled to a Pool of Resources. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 2:841829. [PMID: 36926089 PMCID: PMC10013072 DOI: 10.3389/fnetp.2022.841829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/16/2022] [Indexed: 06/18/2023]
Abstract
We study the collective dynamics in a population of excitable units (neurons) adaptively interacting with a pool of resources. The resource pool is influenced by the average activity of the population, whereas the feedback from the resources to the population is comprised of components acting homogeneously or inhomogeneously on individual units of the population. Moreover, the resource pool dynamics is assumed to be slow and has an oscillatory degree of freedom. We show that the feedback loop between the population and the resources can give rise to collective activity bursting in the population. To explain the mechanisms behind this emergent phenomenon, we combine the Ott-Antonsen reduction for the collective dynamics of the population and singular perturbation theory to obtain a reduced system describing the interaction between the population mean field and the resources.
Collapse
Affiliation(s)
- Igor Franović
- Scientific Computing Laboratory, Center for the Study of Complex Systems, Institute of Physics Belgrade, University of Belgrade, Belgrade, Serbia
| | - Sebastian Eydam
- Neural Circuits and Computations Unit, RIKEN Center for Brain Science, Wako, Japan
| | - Serhiy Yanchuk
- Institut für Mathematik, Technische Universität Berlin, Berlin, Germany
- Potsdam Institute for Climate Impact Research, Potsdam, Germany
- Institut für Mathematik, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rico Berner
- Institut für Physik, Humboldt-Universität zu Berlin, Berlin, Germany
- Institut für Theoretische Physik, Technische Universität Berlin, Berlin, Germany
| |
Collapse
|
31
|
Zheng N, Fitzpatrick V, Cheng R, Shi L, Kaplan DL, Yang C. Photoacoustic Carbon Nanotubes Embedded Silk Scaffolds for Neural Stimulation and Regeneration. ACS NANO 2022; 16:2292-2305. [PMID: 35098714 DOI: 10.1021/acsnano.1c08491] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Neural interfaces using biocompatible scaffolds provide crucial properties, such as cell adhesion, structural support, and mass transport, for the functional repair of nerve injuries and neurodegenerative diseases. Neural stimulation has also been found to be effective in promoting neural regeneration. This work provides a generalized strategy to integrate photoacoustic (PA) neural stimulation into hydrogel scaffolds using a nanocomposite hydrogel approach. Specifically, polyethylene glycol (PEG)-functionalized carbon nanotubes (CNT), highly efficient photoacoustic agents, are embedded into silk fibroin to form biocompatible and soft photoacoustic materials. We show that these photoacoustic functional scaffolds enable nongenetic activation of neurons with a spatial precision defined by the area of light illumination, promoting neuron regeneration. These CNT/silk scaffolds offered reliable and repeatable photoacoustic neural stimulation, and 94% of photoacoustic-stimulated neurons exhibit a fluorescence change larger than 10% in calcium imaging in the light-illuminated area. The on-demand photoacoustic stimulation increased neurite outgrowth by 1.74-fold in a rat dorsal root ganglion model, when compared to the unstimulated group. We also confirmed that promoted neurite outgrowth by photoacoustic stimulation is associated with an increased concentration of neurotrophic factor (BDNF). As a multifunctional neural scaffold, CNT/silk scaffolds demonstrated nongenetic PA neural stimulation functions and promoted neurite outgrowth, providing an additional method for nonpharmacological neural regeneration.
Collapse
Affiliation(s)
| | - Vincent Fitzpatrick
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | | | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | | |
Collapse
|
32
|
Momohara Y, Neveu CL, Chen HM, Baxter DA, Byrne JH. Specific Plasticity Loci and Their Synergism Mediate Operant Conditioning. J Neurosci 2022; 42:1211-1223. [PMID: 34992131 PMCID: PMC8883845 DOI: 10.1523/jneurosci.1722-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/26/2021] [Accepted: 12/03/2021] [Indexed: 11/21/2022] Open
Abstract
Despite numerous studies examining the mechanisms of operant conditioning (OC), the diversity of OC plasticity loci and their synergism have not been examined sufficiently. In the well-characterized feeding neural circuit of Aplysia, in vivo and in vitro appetitive OC increases neuronal excitability and electrical coupling among several neurons leading to an increase in expression of ingestive behavior. Here, we used the in vitro analog of OC to investigate whether OC reduces the excitability of a neuron, B4, whose inhibitory connections decrease expression of ingestive behavior. We found OC decreased the excitability of B4. This change appeared intrinsic to B4 because it could be replicated with an analog of OC in isolated cultures of B4 neurons. In addition to changes in B4 excitability, OC decreased the strength of B4's inhibitory connection to a key decision-making neuron, B51. The OC-induced changes were specific without affecting the excitability of another neuron critical for feeding behavior, B8, or the B4-to-B8 inhibitory connection. A conductance-based circuit model indicated that reducing the B4-to-B51 synapse, or increasing B51 excitability, mediated the OC phenotype more effectively than did decreasing B4 excitability. We combined these modifications to examine whether they could act synergistically. Combinations including B51 synergistically enhanced feeding. Taken together, these results suggest modifications of diverse loci work synergistically to mediate OC and that some neurons are well suited to work synergistically with plasticity in other loci.SIGNIFICANCE STATEMENT The ways in which synergism of diverse plasticity loci mediate the change in motor patterns in operant conditioning (OC) are poorly understood. Here, we found that OC was in part mediated by decreasing the intrinsic excitability of a critical neuron of Aplysia feeding behavior, and specifically reducing the strength of one of its inhibitory connections that targets a key decision-making neuron. A conductance-based computational model indicated that the known plasticity loci showed a surprising level of synergism to mediate the behavioral changes associated with OC. These results highlight the importance of understanding the diversity, specificity and synergy among different types of plasticity that encode memory. Also, because OC in Aplysia is mediated by dopamine (DA), the present study provides insights into specific and synergistic mechanisms of DA-mediated reinforcement of behaviors.
Collapse
Affiliation(s)
- Yuto Momohara
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the, University of Texas Health Science Center, Houston, Texas 77030
| | - Curtis L Neveu
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the, University of Texas Health Science Center, Houston, Texas 77030
| | - Hsin-Mei Chen
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the, University of Texas Health Science Center, Houston, Texas 77030
- Center for Nursing Research, Education and Practice, Houston Methodist Academic Institute, Houston, Texas 77030
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the, University of Texas Health Science Center, Houston, Texas 77030
- Engineering Medicine (ENMED), Texas A&M University College of Medicine, Houston, Texas 77030
| | - John H Byrne
- Department of Neurobiology and Anatomy, W.M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School at the, University of Texas Health Science Center, Houston, Texas 77030
| |
Collapse
|
33
|
Abstract
Understanding how the brain learns may lead to machines with human-like intellectual capacities. It was previously proposed that the brain may operate on the principle of predictive coding. However, it is still not well understood how a predictive system could be implemented in the brain. Here we demonstrate that the ability of a single neuron to predict its future activity may provide an effective learning mechanism. Interestingly, this predictive learning rule can be derived from a metabolic principle, where neurons need to minimize their own synaptic activity (cost), while maximizing their impact on local blood supply by recruiting other neurons. We show how this mathematically derived learning rule can provide a theoretical connection between diverse types of brain-inspired algorithms, thus, offering a step toward development of a general theory of neuronal learning. We tested this predictive learning rule in neural network simulations and in data recorded from awake animals. Our results also suggest that spontaneous brain activity provides “training data” for neurons to learn to predict cortical dynamics. Thus, the ability of a single neuron to minimize surprise: i.e. the difference between actual and expected activity, could be an important missing element to understand computation in the brain.
Collapse
|
34
|
Luczak A, Kubo Y. Predictive Neuronal Adaptation as a Basis for Consciousness. Front Syst Neurosci 2022; 15:767461. [PMID: 35087383 PMCID: PMC8789243 DOI: 10.3389/fnsys.2021.767461] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/29/2021] [Indexed: 01/07/2023] Open
Abstract
Being able to correctly predict the future and to adjust own actions accordingly can offer a great survival advantage. In fact, this could be the main reason why brains evolved. Consciousness, the most mysterious feature of brain activity, also seems to be related to predicting the future and detecting surprise: a mismatch between actual and predicted situation. Similarly at a single neuron level, predicting future activity and adapting synaptic inputs accordingly was shown to be the best strategy to maximize the metabolic energy for a neuron. Following on these ideas, here we examined if surprise minimization by single neurons could be a basis for consciousness. First, we showed in simulations that as a neural network learns a new task, then the surprise within neurons (defined as the difference between actual and expected activity) changes similarly to the consciousness of skills in humans. Moreover, implementing adaptation of neuronal activity to minimize surprise at fast time scales (tens of milliseconds) resulted in improved network performance. This improvement is likely because adapting activity based on the internal predictive model allows each neuron to make a more "educated" response to stimuli. Based on those results, we propose that the neuronal predictive adaptation to minimize surprise could be a basic building block of conscious processing. Such adaptation allows neurons to exchange information about own predictions and thus to build more complex predictive models. To be precise, we provide an equation to quantify consciousness as the amount of surprise minus the size of the adaptation error. Since neuronal adaptation can be studied experimentally, this can allow testing directly our hypothesis. Specifically, we postulate that any substance affecting neuronal adaptation will also affect consciousness. Interestingly, our predictive adaptation hypothesis is consistent with multiple ideas presented previously in diverse theories of consciousness, such as global workspace theory, integrated information, attention schema theory, and predictive processing framework. In summary, we present a theoretical, computational, and experimental support for the hypothesis that neuronal adaptation is a possible biological mechanism of conscious processing, and we discuss how this could provide a step toward a unified theory of consciousness.
Collapse
Affiliation(s)
- Artur Luczak
- Canadian Center for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | | |
Collapse
|
35
|
Ho YY, Roeser A, Law G, Johnson BR. Pandemic Teaching: Using the Allen Cell Types Database for Final Semester Projects in an Undergraduate Neurophysiology Lab Course. JOURNAL OF UNDERGRADUATE NEUROSCIENCE EDUCATION : JUNE : A PUBLICATION OF FUN, FACULTY FOR UNDERGRADUATE NEUROSCIENCE 2021; 20:A100-A110. [PMID: 35540944 PMCID: PMC9053425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 06/14/2023]
Abstract
We designed a final semester research project that allowed students to apply the electrophysiological concepts they learned in a lab course to propose and answer experimental questions without access to laboratory equipment. We created the activity based on lesson plans from Ashley Juavinett and the Allen Institute for Brain Science (AIBS) Allen SDK online examples. An interactive graphic interface was added for students to explore and easily quantify subtle neuronal voltage changes. Before starting the final project, students had experience with conventional extracellular and intracellular recording techniques to record and analyze extracellular action potential firing patterns and intracellular resting, action, and synaptic potentials. They demonstrated their understanding of neural signal transmission in required lab reports using data they gathered before the pandemic shutdown. After students left campus, they continued to analyze data and write lab reports focused on neuronal excitability in snail and fly neurons with data supplied by the instructors. For their final project, students were challenged to answer questions addressing neuronal excitability at both the single neuron and neuronal population level by analyzing and interpreting the open-access, patch clamp recording data from the Allen Cell Types Database using code we provided (Python/Jupyter Notebook). This virtual final semester project allowed students to ask real-world medical and scientific questions from "start to end". Through this project, students developed skills to navigate an extensive online database and gained experience with coding-based data analysis. They chose neuronal populations from human and mouse brains to compare passive properties and neuronal excitability between and within brain areas and across different species and disease states. Additionally, students learned to do simple manipulations of Python code, work remotely in teams, and polish their written scientific presentation skills. This activity could complement other remote learning options such as neuronal simulations. Few online sources offer such a wealth of neuroscience data that students can use for class assignments, and even for research and keystone projects. The activity extends the traditional material often taught in upper-level neuroscience courses, with or without a laboratory section, providing a deeper understanding of the range of excitability properties that neurons express.
Collapse
Affiliation(s)
- Yi-Yun Ho
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14853
| | - Andrea Roeser
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14853
| | - Gwenda Law
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Bruce R. Johnson
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14853
| |
Collapse
|
36
|
Qin X, Liu XX, Wang Y, Wang D, Song Y, Zou JX, Pan HQ, Zhai XZ, Zhang YM, Zhang YB, Hu P, Zhang WH. Early life stress induces anxiety-like behavior during adulthood through dysregulation of neuronal plasticity in the basolateral amygdala. Life Sci 2021; 285:119959. [PMID: 34536496 DOI: 10.1016/j.lfs.2021.119959] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
AIMS Early life stress (ELS) increases the risk of psychiatric diseases such as anxiety disorders and depression in later life. Hyperactivation of the basolateral amygdala (BLA) neurons plays a pivotal role in the pathogenesis of stress-related diseases. However, the functional roles of BLA neurons in ELS-induced anxiety disorders are not completely understood. MAIN METHODS Mice were subjected to maternal separation (MS) during postnatal days 3 to 21 to mimic ELS. Anxiety-like behavior was tested by open field test (OFT), elevated plus maze (EPM), and novelty suppressed feeding (NSF). Then, c-fos expression, a proxy for neuronal activity, was evaluated by immunofluorescence. Finally, synaptic transmission and intrinsic excitability were measured by whole-cell patch-clamp recordings. KEY FINDINGS MS significantly increased anxiety-like behavior in adulthood, as indicated by less time spent in the center area of the OFT, less time spent in and fewer entries to the open arms of the EPM, and increased latency to feed in NSF. Mechanistically, MS increased the expression of c-fos in BLA. MS enhanced the excitatory, but not inhibitory, synaptic transmission onto BLA projection neurons (PNs), which was caused by enhanced presynaptic glutamate release. Moreover, MS also markedly increased the intrinsic neuronal excitability of BLA PNs, probably due to the reduced medium afterhyperpolarization (mAHP) in BLA PNs. SIGNIFICANCE Our results suggest that the changes of neuronal activity and synaptic transmission in the BLA PNs may play a crucial role in ELS-induced anxiety-like behavior, and these findings provide new insights into the pathological mechanisms of stress-related anxiety disorders.
Collapse
Affiliation(s)
- Xia Qin
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Xiao-Xuan Liu
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, China; Neurology Department, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yu Wang
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Dan Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ying Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jia-Xin Zou
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Han-Qing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Xiao-Zhou Zhai
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Yong-Mei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yang-Bo Zhang
- Department of Neurology, the Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ping Hu
- Institute of Translational Medicine, Nanchang University, Nanchang 330001, China.
| | - Wen-Hua Zhang
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
37
|
de Pins B, Mendes T, Giralt A, Girault JA. The Non-receptor Tyrosine Kinase Pyk2 in Brain Function and Neurological and Psychiatric Diseases. Front Synaptic Neurosci 2021; 13:749001. [PMID: 34690733 PMCID: PMC8527176 DOI: 10.3389/fnsyn.2021.749001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pyk2 is a non-receptor tyrosine kinase highly enriched in forebrain neurons. Pyk2 is closely related to focal adhesion kinase (FAK), which plays an important role in sensing cell contacts with extracellular matrix and other extracellular signals controlling adhesion and survival. Pyk2 shares some of FAK’s characteristics including recruitment of Src-family kinases after autophosphorylation, scaffolding by interacting with multiple partners, and activation of downstream signaling pathways. Pyk2, however, has the unique property to respond to increases in intracellular free Ca2+, which triggers its autophosphorylation following stimulation of various receptors including glutamate NMDA receptors. Pyk2 is dephosphorylated by the striatal-enriched phosphatase (STEP) that is highly expressed in the same neuronal populations. Pyk2 localization in neurons is dynamic, and altered following stimulation, with post-synaptic and nuclear enrichment. As a signaling protein Pyk2 is involved in multiple pathways resulting in sometimes opposing functions depending on experimental models. Thus Pyk2 has a dual role on neurites and dendritic spines. With Src family kinases Pyk2 participates in postsynaptic regulations including of NMDA receptors and is necessary for specific types of synaptic plasticity and spatial memory tasks. The diverse functions of Pyk2 are also illustrated by its role in pathology. Pyk2 is activated following epileptic seizures or ischemia-reperfusion and may contribute to the consequences of these insults whereas Pyk2 deficit may contribute to the hippocampal phenotype of Huntington’s disease. Pyk2 gene, PTK2B, is associated with the risk for late-onset Alzheimer’s disease. Studies of underlying mechanisms indicate a complex contribution with involvement in amyloid toxicity and tauopathy, combined with possible functional deficits in neurons and contribution in microglia. A role of Pyk2 has also been proposed in stress-induced depression and cocaine addiction. Pyk2 is also important for the mobility of astrocytes and glioblastoma cells. The implication of Pyk2 in various pathological conditions supports its potential interest for therapeutic interventions. This is possible through molecules inhibiting its activity or increasing it through inhibition of STEP or other means, depending on a precise evaluation of the balance between positive and negative consequences of Pyk2 actions.
Collapse
Affiliation(s)
- Benoit de Pins
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Tiago Mendes
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| |
Collapse
|
38
|
Westlund K, Montera M, Goins A, Alles S, Afaghpour-Becklund M, Bartel R, Durvasula R, Kunamneni A. Single-chain Fragment variable antibody targeting cholecystokinin-B receptor for pain reduction. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100067. [PMID: 34458647 PMCID: PMC8378781 DOI: 10.1016/j.ynpai.2021.100067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 05/13/2023]
Abstract
The cholecystokinin B receptor and its neuropeptide ligand are upregulated in chronic neuropathic pain models. Single-chain Fragment variable antibodies were generated as preferred non-opioid targeting therapy blocking the cholecystokinin B receptor to inhibit chronic neuropathic pain models in vivo and in vitro. Engineered antibodies of this type feature binding activity similar to monoclonal antibodies but with stronger affinity and increased tissue penetrability due to their smaller size. More importantly, single-chain Fragment variable antibodies have promising biotherapeutic applications for both nervous and immune systems, now recognized as interactive in chronic pain. A mouse single-chain Fragment variable antibody library recognizing a fifteen amino acid extracellular peptide fragment of the cholecystokinin B receptor was generated from immunized spleens. Ribosome display, a powerful cell-free technology, was applied for recombinant antibody selection. Antibodies with higher affinity, stability, solubility, and binding specificity for cholecystokinin B not A receptor were selected and optimized for in vivo and in vitro efficacy. A single dose of the lead candidate reduced mechanical and cold hypersensitivity in two rodent models of neuropathic pain for at least seven weeks. Continuing efficacy was evident with either intraperitoneal or intranasal dosing. Likewise, the lead single-chain Fragment variable antibody totally prevented development of anxiety- and depression-like behaviors and cognitive deficits typical in the models. Reduction of neuronal firing frequency was evident in trigeminal ganglia primary neuronal cultures treated in vitro with the cholecystokinin B receptor antibody. Immunofluorescent staining intensity in the trigeminal neuron primary cultures was significantly reduced incrementally after overnight binding with increasingly higher dilutions of the single-chain Fragment variable antibody. While it is reported that single-chain Fragment variable antibodies are removed systemically within 2-6 h, Western blot evidence indicates the His-tag marker remained after 7 weeks in the trigeminal ganglia and in the dorsolateral medulla, providing evidence of brain and ganglia penetrance known to be compromised in overactivated states. This project showcases the in vivo efficacy of our lead single-chain Fragment variable antibody indicating its potential for development as a non-opioid, non-addictive therapeutic intervention for chronic pain. Importantly, studies by others have indicated treatments with cholecystokinin B receptor antagonists suppress maintenance and reactivation of morphine dependence in place preference tests while lowering tolerance and dose requirements. Our future studies remain to address these potential benefits that may accompany the cholecystokinin B receptor biological therapy. Both chronic sciatic and orofacial pain can be unrelenting and excruciating, reducing quality of life as well as diminishing physical and mental function. An effective non-opiate, non-addictive therapy with potential to significantly reduce chronic neuropathic pain long term is greatly needed.
Collapse
Key Words
- ANOVA, analysis of variance
- ARM, antibody ribosome mRNA
- Anxiety
- BBB, blood–brain barrier
- CCK-8, cholecystokinin octapeptide
- CCK-BR, cholecystokinin B receptor
- CPP, conditioned place preference
- Chronic pain
- DRG, dorsal root ganglia
- Depression
- Eukaryotic ribosome display
- FRICT-ION, foramen rotundum inflammatory compression trigeminal infraorbital nerve model
- GPCR, G-protein-coupled receptor
- IACUC, Institutional Animal Care and Use Committee
- ION, infraorbital nerve
- MΩ, megaOhms
- PBS, phosphate buffered saline
- SEM, standard error of the mean
- TG, trigeminal ganglia
- ms, milliseconds
- pA, picoAmps
- scFv
- scFv, single-chain Fragment variable antibody
Collapse
Affiliation(s)
- K.N. Westlund
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
- Biomedical Laboratory Research & Development (121F), New Mexico VA
Health Care System, Albuquerque, NM, USA
| | - M.A. Montera
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - A.E. Goins
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - S.R.A. Alles
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - M. Afaghpour-Becklund
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - R. Bartel
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - R. Durvasula
- Division of Infectious Diseases, Department of Internal Medicine, Mayo
Clinic, Jacksonville, FL, USA
- Department of Medicine, Loyola University Medical Center, Maywood, IL
60153-3328, USA
| | - A. Kunamneni
- Division of Infectious Diseases, Department of Internal Medicine, Mayo
Clinic, Jacksonville, FL, USA
- Department of Medicine, Loyola University Medical Center, Maywood, IL
60153-3328, USA
| |
Collapse
|
39
|
Bayasgalan T, Stupniki S, Kovács A, Csemer A, Szentesi P, Pocsai K, Dionisio L, Spitzmaul G, Pál B. Alteration of Mesopontine Cholinergic Function by the Lack of KCNQ4 Subunit. Front Cell Neurosci 2021; 15:707789. [PMID: 34381336 PMCID: PMC8352570 DOI: 10.3389/fncel.2021.707789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
The pedunculopontine nucleus (PPN), a structure known as a cholinergic member of the reticular activating system (RAS), is source and target of cholinergic neuromodulation and contributes to the regulation of the sleep–wakefulness cycle. The M-current is a voltage-gated potassium current modulated mainly by cholinergic signaling. KCNQ subunits ensemble into ion channels responsible for the M-current. In the central nervous system, KCNQ4 expression is restricted to certain brainstem structures such as the RAS nuclei. Here, we investigated the presence and functional significance of KCNQ4 in the PPN by behavioral studies and the gene and protein expressions and slice electrophysiology using a mouse model lacking KCNQ4 expression. We found that this mouse has alterations in the adaptation to changes in light–darkness cycles, representing the potential role of KCNQ4 in the regulation of the sleep–wakefulness cycle. As cholinergic neurons from the PPN participate in the regulation of this cycle, we investigated whether the cholinergic PPN might also possess functional KCNQ4 subunits. Although the M-current is an electrophysiological hallmark of cholinergic neurons, only a subpopulation of them had KCNQ4-dependent M-current. Interestingly, the absence of the KCNQ4 subunit altered the expression patterns of the other KCNQ subunits in the PPN. We also determined that, in wild-type animals, the cholinergic inputs of the PPN modulated the M-current, and these in turn can modulate the level of synchronization between neighboring PPN neurons. Taken together, the KCNQ4 subunit is present in a subpopulation of PPN cholinergic neurons, and it may contribute to the regulation of the sleep–wakefulness cycle.
Collapse
Affiliation(s)
- T Bayasgalan
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - S Stupniki
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - A Kovács
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - A Csemer
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - P Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - K Pocsai
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - L Dionisio
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - G Spitzmaul
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional Del Sur (UNS), Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - B Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
40
|
Shi L, Jiang Y, Fernandez FR, Chen G, Lan L, Man HY, White JA, Cheng JX, Yang C. Non-genetic photoacoustic stimulation of single neurons by a tapered fiber optoacoustic emitter. LIGHT, SCIENCE & APPLICATIONS 2021; 10:143. [PMID: 34257273 PMCID: PMC8277806 DOI: 10.1038/s41377-021-00580-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/09/2021] [Accepted: 06/21/2021] [Indexed: 05/19/2023]
Abstract
Neuromodulation at high spatial resolution poses great significance in advancing fundamental knowledge in the field of neuroscience and offering novel clinical treatments. Here, we developed a tapered fiber optoacoustic emitter (TFOE) generating an ultrasound field with a high spatial precision of 39.6 µm, enabling optoacoustic activation of single neurons or subcellular structures, such as axons and dendrites. Temporally, a single acoustic pulse of sub-microsecond converted by the TFOE from a single laser pulse of 3 ns is shown as the shortest acoustic stimuli so far for successful neuron activation. The precise ultrasound generated by the TFOE enabled the integration of the optoacoustic stimulation with highly stable patch-clamp recording on single neurons. Direct measurements of the electrical response of single neurons to acoustic stimulation, which is difficult for conventional ultrasound stimulation, have been demonstrated. By coupling TFOE with ex vivo brain slice electrophysiology, we unveil cell-type-specific responses of excitatory and inhibitory neurons to acoustic stimulation. These results demonstrate that TFOE is a non-genetic single-cell and sub-cellular modulation technology, which could shed new insights into the mechanism of ultrasound neurostimulation.
Collapse
Affiliation(s)
- Linli Shi
- Department of Chemistry, Boston University, 580 Commonwealth Avenue, Boston, MA, 02215, USA
| | - Ying Jiang
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA
| | - Fernando R Fernandez
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA, 02215, USA
- Neurophotonics Center, Photonics Center, Boston University, 8 St. Mary's Street, Boston, MA, 02215, USA
| | - Guo Chen
- Department of Electrical and Computer Engineering, 8 St. Mary's Street, Boston, MA, 02215, USA
| | - Lu Lan
- Department of Electrical and Computer Engineering, 8 St. Mary's Street, Boston, MA, 02215, USA
| | - Heng-Ye Man
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA, 02215, USA
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA, 02215, USA
| | - John A White
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA, 02215, USA
- Neurophotonics Center, Photonics Center, Boston University, 8 St. Mary's Street, Boston, MA, 02215, USA
| | - Ji-Xin Cheng
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA.
- Department of Electrical and Computer Engineering, 8 St. Mary's Street, Boston, MA, 02215, USA.
| | - Chen Yang
- Department of Chemistry, Boston University, 580 Commonwealth Avenue, Boston, MA, 02215, USA.
- Department of Electrical and Computer Engineering, 8 St. Mary's Street, Boston, MA, 02215, USA.
| |
Collapse
|
41
|
Dwivedi D, Bhalla US. Physiology and Therapeutic Potential of SK, H, and M Medium AfterHyperPolarization Ion Channels. Front Mol Neurosci 2021; 14:658435. [PMID: 34149352 PMCID: PMC8209339 DOI: 10.3389/fnmol.2021.658435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022] Open
Abstract
SK, HCN, and M channels are medium afterhyperpolarization (mAHP)-mediating ion channels. The three channels co-express in various brain regions, and their collective action strongly influences cellular excitability. However, significant diversity exists in the expression of channel isoforms in distinct brain regions and various subcellular compartments, which contributes to an equally diverse set of specific neuronal functions. The current review emphasizes the collective behavior of the three classes of mAHP channels and discusses how these channels function together although they play specialized roles. We discuss the biophysical properties of these channels, signaling pathways that influence the activity of the three mAHP channels, various chemical modulators that alter channel activity and their therapeutic potential in treating various neurological anomalies. Additionally, we discuss the role of mAHP channels in the pathophysiology of various neurological diseases and how their modulation can alleviate some of the symptoms.
Collapse
Affiliation(s)
- Deepanjali Dwivedi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States.,Stanley Center at the Broad, Cambridge, MA, United States
| | - Upinder S Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, GKVK Campus, Bengaluru, India
| |
Collapse
|
42
|
Gao M, Noguchi A, Ikegaya Y. The subiculum sensitizes retrosplenial cortex layer 2/3 pyramidal neurons. J Physiol 2021; 599:3151-3167. [PMID: 33878801 DOI: 10.1113/jp281152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/13/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Neurons in the retrosplenial cortex (RSC), a cerebral region that connects synaptically with various brain regions, are known to increase neuronal activity in accordance with hippocampal sharp wave-ripples. Pyramidal cells in granular RSC (gRSC) layer 2/3, but not layer 5, exhibit slowly ramping depolarization and considerably delayed spikes in response to a step-pulse current injection. The latencies of delayed spikes in RSC layer 2/3 pyramidal neurons were shortened by a preceding current injection. This effect was mimicked by activation of axonal afferents from the subiculum, but not of neocortical afferents. The subiculum is likely to facilitate information processing and flow in the RSC. ABSTRACT The retrosplenial cortex (RSC), a cerebral region involved in diverse cognitive functions, is an anatomical hub that forms monosynaptic connections with various brain areas. Here, we report a unique form of short-term intrinsic plasticity in mouse granular RSC layer 2/3 pyramidal cells. These cells exhibited delayed spikes in response to somatic current injection, but the spike latencies were shortened by a preceding brief depolarization (priming). This priming-induced sensitization is distinct from desensitization, which is commonly observed in other cortical neurons. The facilitatory priming effect lasted for more than 3 s, providing a time window for increased sensitivity to RSC inputs. Based on in vitro and in vivo patch-clamp recordings following optogenetic stimulation of axonal fibres, we found that preactivation of subicular afferents replicated the facilitatory priming effect. The results suggest that subicular inputs to RSC layer 2/3 neurons may modulate subsequent information integration in the RSC layer 2/3 circuits.
Collapse
Affiliation(s)
- Mengxuan Gao
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Asako Noguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan.,Institute of AI and Beyond, The University of Tokyo, Tokyo, 113-0033, Japan.,Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| |
Collapse
|
43
|
Sharifshazileh M, Burelo K, Sarnthein J, Indiveri G. An electronic neuromorphic system for real-time detection of high frequency oscillations (HFO) in intracranial EEG. Nat Commun 2021; 12:3095. [PMID: 34035249 PMCID: PMC8149394 DOI: 10.1038/s41467-021-23342-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 04/20/2021] [Indexed: 02/04/2023] Open
Abstract
The analysis of biomedical signals for clinical studies and therapeutic applications can benefit from embedded devices that can process these signals locally and in real-time. An example is the analysis of intracranial EEG (iEEG) from epilepsy patients for the detection of High Frequency Oscillations (HFO), which are a biomarker for epileptogenic brain tissue. Mixed-signal neuromorphic circuits offer the possibility of building compact and low-power neural network processing systems that can analyze data on-line in real-time. Here we present a neuromorphic system that combines a neural recording headstage with a spiking neural network (SNN) processing core on the same die for processing iEEG, and show how it can reliably detect HFO, thereby achieving state-of-the-art accuracy, sensitivity, and specificity. This is a first feasibility study towards identifying relevant features in iEEG in real-time using mixed-signal neuromorphic computing technologies.
Collapse
Affiliation(s)
- Mohammadali Sharifshazileh
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Karla Burelo
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich, Switzerland
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Johannes Sarnthein
- Department of Neurosurgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Giacomo Indiveri
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
44
|
Neudorfer C, Chow CT, Boutet A, Loh A, Germann J, Elias GJ, Hutchison WD, Lozano AM. Kilohertz-frequency stimulation of the nervous system: A review of underlying mechanisms. Brain Stimul 2021; 14:513-530. [PMID: 33757930 DOI: 10.1016/j.brs.2021.03.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Electrical stimulation in the kilohertz-frequency range has gained interest in the field of neuroscience. The mechanisms underlying stimulation in this frequency range, however, are poorly characterized to date. OBJECTIVE/HYPOTHESIS To summarize the manifold biological effects elicited by kilohertz-frequency stimulation in the context of the currently existing literature and provide a mechanistic framework for the neural responses observed in this frequency range. METHODS A comprehensive search of the peer-reviewed literature was conducted across electronic databases. Relevant computational, clinical, and mechanistic studies were selected for review. RESULTS The effects of kilohertz-frequency stimulation on neural tissue are diverse and yield effects that are distinct from conventional stimulation. Broadly, these can be divided into 1) subthreshold, 2) suprathreshold, 3) synaptic and 4) thermal effects. While facilitation is the dominating mechanism at the subthreshold level, desynchronization, spike-rate adaptation, conduction block, and non-monotonic activation can be observed during suprathreshold kilohertz-frequency stimulation. At the synaptic level, kilohertz-frequency stimulation has been associated with the transient depletion of the available neurotransmitter pool - also known as synaptic fatigue. Finally, thermal effects associated with extrinsic (environmental) and intrinsic (associated with kilohertz-frequency stimulation) temperature changes have been suggested to alter the neural response to stimulation paradigms. CONCLUSION The diverse spectrum of neural responses to stimulation in the kilohertz-frequency range is distinct from that associated with conventional stimulation. This offers the potential for new therapeutic avenues across stimulation modalities. However, stimulation in the kilohertz-frequency range is associated with distinct challenges and caveats that need to be considered in experimental paradigms.
Collapse
Affiliation(s)
- Clemens Neudorfer
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Canada
| | - Clement T Chow
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Canada
| | - Alexandre Boutet
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Canada
| | - Aaron Loh
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Canada
| | - Jürgen Germann
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Canada
| | - Gavin Jb Elias
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Canada
| | - William D Hutchison
- Krembil Research Institute, University of Toronto, Ontario, Canada; Department of Physiology, Toronto Western Hospital and University of Toronto, Ontario, Canada
| | - Andres M Lozano
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Canada; Krembil Research Institute, University of Toronto, Ontario, Canada.
| |
Collapse
|
45
|
Martin L, Jaime K, Ramos F, Robles F. Declarative working memory: A bio-inspired cognitive architecture proposal. COGN SYST RES 2021. [DOI: 10.1016/j.cogsys.2020.10.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Barta T, Kostal L. Regular spiking in high-conductance states: The essential role of inhibition. Phys Rev E 2021; 103:022408. [PMID: 33736083 DOI: 10.1103/physreve.103.022408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 02/03/2021] [Indexed: 06/12/2023]
Abstract
Strong inhibitory input to neurons, which occurs in balanced states of neural networks, increases synaptic current fluctuations. This has led to the assumption that inhibition contributes to the high spike-firing irregularity observed in vivo. We used single compartment neuronal models with time-correlated (due to synaptic filtering) and state-dependent (due to reversal potentials) input to demonstrate that inhibitory input acts to decrease membrane potential fluctuations, a result that cannot be achieved with simplified neural input models. To clarify the effects on spike-firing regularity, we used models with different spike-firing adaptation mechanisms, and we observed that the addition of inhibition increased firing regularity in models with dynamic firing thresholds and decreased firing regularity if spike-firing adaptation was implemented through ionic currents or not at all. This fluctuation-stabilization mechanism provides an alternative perspective on the importance of strong inhibitory inputs observed in balanced states of neural networks, and it highlights the key roles of biologically plausible inputs and specific adaptation mechanisms in neuronal modeling.
Collapse
Affiliation(s)
- Tomas Barta
- Institute of Physiology of the Czech Academy of Sciences, 14220 Prague, Czech Republic; Charles University, First Medical Faculty, 12108 Prague, Czech Republic; and Institute of Ecology and Environmental Sciences, INRAE, 78026 Versailles, France
| | - Lubomir Kostal
- Institute of Physiology of the Czech Academy of Sciences, 14220 Prague, Czech Republic
| |
Collapse
|
47
|
Rayi PR, Bagrov AY, Kaphzan H. Chronic α1-Na/K-ATPase inhibition reverses the elongation of the axon initial segment of the hippocampal CA1 pyramidal neurons in Angelman syndrome model mice. Neuropsychopharmacology 2021; 46:654-664. [PMID: 33214655 PMCID: PMC8027375 DOI: 10.1038/s41386-020-00907-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/01/2020] [Accepted: 10/26/2020] [Indexed: 12/28/2022]
Abstract
Angelman syndrome (AS) is a neurodevelopmental disorder caused by the loss of function of the maternal UBE3A gene. The hippocampus is one of the most prominently affected brain regions in AS model mice, manifesting in severe hippocampal-dependent memory and plasticity deficits. Previous studies in AS mice reported an elongated axon initial segment (AIS) in pyramidal neurons (PNs) of the hippocampal CA1 region. These were the first reports in mammals to show AIS elongation in vivo. Correspondingly, this AIS elongation was linked to enhanced expression of the α1 subunit of Na+/K+-ATPase (α1-NaKA). Recently, it was shown that selective pharmacological inhibition of α1-NaKA by marinobufagenin (MBG) in adult AS mice rescued the hippocampal-dependent deficits via normalizing their compromised activity-dependent calcium (Ca+2) dynamics. In the herein study, we showed that a chronic selective α1-NaKA inhibition reversed the AIS elongation in hippocampal CA1 PNs of adult AS mice, and differentially altered their excitability and intrinsic properties. Taken together, our study is the first to demonstrate in vivo structural plasticity of the AIS in a mammalian model, and further elaborates on the modulatory effects of elevated α1-NaKA levels in the hippocampus of AS mice.
Collapse
Affiliation(s)
- Prudhvi Raj Rayi
- grid.18098.380000 0004 1937 0562Sagol Department of Neurobiology, The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, 3498838 Israel
| | - Alexei Y. Bagrov
- grid.419730.80000 0004 0440 2269Sechenov Institute of Evolutionary Physiology and Biochemistry, 194223 St. Petersburg, Russian Federation
| | - Hanoch Kaphzan
- Sagol Department of Neurobiology, The Integrated Brain and Behavior Research Center, University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
48
|
Somatostatin expressing GABAergic interneurons in the medial entorhinal cortex preferentially inhibit layer III-V pyramidal cells. Commun Biol 2020; 3:754. [PMID: 33303963 PMCID: PMC7728756 DOI: 10.1038/s42003-020-01496-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/13/2020] [Indexed: 12/31/2022] Open
Abstract
GABA released from heterogeneous types of interneurons acts in a complex spatio-temporal manner on postsynaptic targets in the networks. In addition to GABA, a large fraction of GABAergic cells also express neuromodulator peptides. Somatostatin (SOM) containing interneurons, in particular, have been recognized as key players in several brain circuits, however, the action of SOM and its downstream network effects remain largely unknown. Here, we used optogenetics, electrophysiologic, anatomical and behavioral experiments to reveal that the dendrite-targeting, SOM+ GABAergic interneurons demonstrate a unique layer-specific action in the medial entorhinal cortex (MEC) both in terms of GABAergic and SOM-related properties. We show that GABAergic and somatostatinergic neurotransmission originating from SOM+ local interneurons preferentially inhibit layerIII-V pyramidal cells, known to be involved in memory formation. We propose that this dendritic GABA–SOM dual inhibitory network motif within the MEC serves to selectively modulate working-memory formation without affecting the retrieval of already learned spatial navigation tasks. Miklós Kecskés et al. show that somatostatin-expressing interneurons in the medial entorhinal cortex regulate deep-layer pyramidal neurons and impact short-term memory in mice.
Collapse
|
49
|
Seseña E, Soto E, Bueno J, Vega R. Nociceptin/orphanin FQ peptide receptor mediates inhibition of N-type calcium currents in vestibular afferent neurons of the rat. J Neurophysiol 2020; 124:1605-1614. [PMID: 32966754 DOI: 10.1152/jn.00269.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The vestibular system is modulated by various neuromodulators including opioid peptides. The current study was conducted to determine whether activation of nociceptin/orphanin FQ peptide (NOP) receptors modulates voltage-gated calcium currents and action potential discharge of rat vestibular afferent neurons. We performed whole cell patch-clamp recordings on cultured vestibular afferent neurons from P7-P10 Long-Evans rats. Application of nociceptin/orphanin FQ (N/OFQ), a 17-amino acid neuropeptide that is the endogenous ligand for NOP receptor, inhibits the high-voltage activated (HVA) component of the calcium current in a concentration-dependent manner with a half inhibitory concentration of 26 nM. Said inhibitory action on the calcium current is voltage-dependent, which was made clear by the fact that it was reverted in 80% by a depolarizing prepulse. Furthermore, the effect of N/OFQ was blocked by application of the specific NOP-antagonist UFP101, by preincubation with G-protein blocker pertussis toxin, and by coapplication of the specific N-type calcium-current blocker ω-conotoxin-MVIIA. N/OFQ application causes an increase in the duration and maximum rate of repolarization of action potentials. It also decreases repetitive discharge and discharge elicited by sinusoidal stimulation. These results show that in vestibular afferents, NOP receptor activation inhibits N-type calcium current by activating G proteins, mostly through the Gβγ subunit. This suggests that NOP activation produces a presynaptic modulation of primary vestibular afferent neurons' output into the vestibular nuclei, thus taking part in the integration and gain setting of vestibular information in second-order vestibular nucleus neurons.NEW & NOTEWORTHY Our results show that in primary vestibular afferent neurons, activation of the nociceptin/orphanin FQ peptide receptor inhibits the N-type calcium current by a mechanism mediated by G proteins. We propose that calcium current inhibition modulates neurotransmitter release from vestibular afferents, producing a presynaptic modulation of vestibular input to vestibular nuclei, thus contributing to gain control in the vestibular afferent input.
Collapse
Affiliation(s)
- Emmanuel Seseña
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Pue, México
| | - Enrique Soto
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Pue, México
| | - Jesua Bueno
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Pue, México
| | - Rosario Vega
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Pue, México
| |
Collapse
|
50
|
Generation of Sharp Wave-Ripple Events by Disinhibition. J Neurosci 2020; 40:7811-7836. [PMID: 32913107 PMCID: PMC7548694 DOI: 10.1523/jneurosci.2174-19.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 06/29/2020] [Accepted: 07/17/2020] [Indexed: 11/21/2022] Open
Abstract
Sharp wave-ripple complexes (SWRs) are hippocampal network phenomena involved in memory consolidation. To date, the mechanisms underlying their occurrence remain obscure. Here, we show how the interactions between pyramidal cells, parvalbumin-positive (PV+) basket cells, and an unidentified class of anti-SWR interneurons can contribute to the initiation and termination of SWRs. Using a biophysically constrained model of a network of spiking neurons and a rate-model approximation, we demonstrate that SWRs emerge as a result of the competition between two interneuron populations and the resulting disinhibition of pyramidal cells. Our models explain how the activation of pyramidal cells or PV+ cells can trigger SWRs, as shown in vitro, and suggests that PV+ cell-mediated short-term synaptic depression influences the experimentally reported dynamics of SWR events. Furthermore, we predict that the silencing of anti-SWR interneurons can trigger SWRs. These results broaden our understanding of the microcircuits supporting the generation of memory-related network dynamics. SIGNIFICANCE STATEMENT The hippocampus is a part of the mammalian brain that is crucial for episodic memories. During periods of sleep and inactive waking, the extracellular activity of the hippocampus is dominated by sharp wave-ripple events (SWRs), which have been shown to be important for memory consolidation. The mechanisms regulating the emergence of these events are still unclear. We developed a computational model to study the emergence of SWRs and to explain the roles of different cell types in regulating them. The model accounts for several previously unexplained features of SWRs and thus advances the understanding of memory-related dynamics.
Collapse
|