1
|
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
|
2
|
Liu M, Sun X. Temporal integration on the dendrites of fast-spiking basket cells. Sci Rep 2024; 14:30278. [PMID: 39632942 PMCID: PMC11618596 DOI: 10.1038/s41598-024-81655-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
Neurons receive synaptic inputs with diverse temporal patterns in vivo, and their integration of these patterns is critical for understanding information processing mechanisms in the brain. Fast-spiking basket cells, which perform both supralinear and sublinear dendritic integration, are essential for inhibitory control in the hippocampus. However, their responses and the mechanisms underlying different temporal input patterns remain unclear. To address this question, we apply inputs with varying windows of time to a detailed compartmental model of basket cells. Our results reveal that when synaptic inputs are randomly dispersed, temporal integration in FS BCs exhibits a sigmoid-like response within the temporal window. In contrast, synchronous input protocols more effectively elicit action potentials, while asynchronous inputs generate more spikes in response to suprathreshold stimuli. Further analysis shows that the supralinear dendrites of fast-spiking basket cells primarily mediate this nonlinearity to asynchronous inputs, owing to their larger dendritic diameters. Moreover, we discover that delayed rectifier [Formula: see text] channels reduce sensitivity to synchronous inputs, whereas N-type [Formula: see text] channels enhance sensitivity to asynchronous inputs. These results provide insights into the mechanisms underlying the temporal coding of fast-spiking basket cells, which is crucial for understanding their role in neuronal oscillations.
Collapse
Affiliation(s)
- Ming Liu
- School of Science, Beijing University of Posts and Telecommunications, Beijing, 100876, China
| | - Xiaojuan Sun
- School of Science, Beijing University of Posts and Telecommunications, Beijing, 100876, China.
- Key Laboratory of Mathematics and Information Networks (Beijing University of Posts and Telecommunications), Ministry of Education, Beijing, 100876, China.
| |
Collapse
|
3
|
Scheuer KS, Jansson AM, Zhao X, Jackson MB. Inter and intralaminar excitation of parvalbumin interneurons in mouse barrel cortex. PLoS One 2024; 19:e0289901. [PMID: 38870124 PMCID: PMC11175493 DOI: 10.1371/journal.pone.0289901] [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: 07/27/2023] [Accepted: 04/29/2024] [Indexed: 06/15/2024] Open
Abstract
Parvalbumin (PV) interneurons are inhibitory fast-spiking cells with essential roles in directing the flow of information through cortical circuits. These neurons set the balance between excitation and inhibition and control rhythmic activity. PV interneurons differ between cortical layers in their morphology, circuitry, and function, but how their electrophysiological properties vary has received little attention. Here we investigate responses of PV interneurons in different layers of primary somatosensory barrel cortex (BC) to different excitatory inputs. With the genetically-encoded hybrid voltage sensor, hVOS, we recorded voltage changes in many L2/3 and L4 PV interneurons simultaneously, with stimulation applied to either L2/3 or L4. A semi-automated procedure was developed to identify small regions of interest corresponding to single responsive PV interneurons. Amplitude, half-width, and rise-time were greater for PV interneurons residing in L2/3 compared to L4. Stimulation in L2/3 elicited responses in both L2/3 and L4 with longer latency compared to stimulation in L4. These differences in latency between layers could influence their windows for temporal integration. Thus, PV interneurons in different cortical layers of BC respond in a layer specific and input specific manner, and these differences have potential roles in cortical computations.
Collapse
Affiliation(s)
- Katherine S. Scheuer
- Cellular and Molecular Biology PhD Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anna M. Jansson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xinyu Zhao
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Meyer B. Jackson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
4
|
Brünner H, Kim H, Ährlund-Richter S, van Lunteren JA, Crestani AP, Meletis K, Carlén M. Cell-type-specific representation of spatial context in the rat prefrontal cortex. iScience 2024; 27:109743. [PMID: 38711459 PMCID: PMC11070673 DOI: 10.1016/j.isci.2024.109743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/09/2024] [Accepted: 04/11/2024] [Indexed: 05/08/2024] Open
Abstract
The ability to represent one's own position in relation to cues, goals, or threats is crucial to successful goal-directed behavior. Using optotagging in knock-in rats expressing Cre recombinase in parvalbumin (PV) neurons (PV-Cre rats), we demonstrate cell-type-specific encoding of spatial and movement variables in the medial prefrontal cortex (mPFC) during goal-directed reward seeking. Single neurons encoded the conjunction of the animal's spatial position and the run direction, referred to as the spatial context. The spatial context was most prominently represented by the inhibitory PV interneurons. Movement toward the reward was signified by increased local field potential (LFP) oscillations in the gamma band but this LFP signature was not related to the spatial information in the neuronal firing. The results highlight how spatial information is incorporated into cognitive operations in the mPFC. The presented PV-Cre line opens the door for expanded research approaches in rats.
Collapse
Affiliation(s)
- Hans Brünner
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Hoseok Kim
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Ana Paula Crestani
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience and Behavioral Sciences, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
O'Connell MN, Barczak A. Auditory Biomarkers of Neuropsychiatric Disorders in Nonhuman Primates. ADVANCES IN NEUROBIOLOGY 2024; 40:219-234. [PMID: 39562447 DOI: 10.1007/978-3-031-69491-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Animal models of neuropsychiatric disorders with appropriate biomarkers can greatly inform the neurobiological basis of disorder-related deficits of cognitive and/or sensory processes. Given the genetic, physiologic, and behavioral similarities between humans and nonhuman primates (NHPs), NHP studies are monumentally important for preclinical translational research. Capitalizing on the NHP's similarities with human systems provides one of the best opportunities to gain detailed insight into the mechanisms underlying disorder-related symptoms and to accumulate a foundation of information for the development of therapeutic interventions. Here, we discuss how results from NHP studies have provided insight into the generation and modulation of select auditory biomarkers of schizophrenia including auditory steady-state responses and mismatch negativity. Since neuro-oscillatory activity has been shown to be relatively preserved across species, we highlight how incorporating the analysis of local and network-level oscillations from multiple nodes across different pathways involved in auditory processing has been used to further the precision of translational comparisons across species.
Collapse
Affiliation(s)
- Monica N O'Connell
- Translational Neuroscience Division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| | - Annamaria Barczak
- Translational Neuroscience Division, Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
6
|
Milicevic KD, Barbeau BL, Lovic DD, Patel AA, Ivanova VO, Antic SD. Physiological features of parvalbumin-expressing GABAergic interneurons contributing to high-frequency oscillations in the cerebral cortex. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 6:100121. [PMID: 38616956 PMCID: PMC11015061 DOI: 10.1016/j.crneur.2023.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 11/13/2023] [Accepted: 12/01/2023] [Indexed: 04/16/2024] Open
Abstract
Parvalbumin-expressing (PV+) inhibitory interneurons drive gamma oscillations (30-80 Hz), which underlie higher cognitive functions. In this review, we discuss two groups/aspects of fundamental properties of PV+ interneurons. In the first group (dubbed Before Axon), we list properties representing optimal synaptic integration in PV+ interneurons designed to support fast oscillations. For example: [i] Information can neither enter nor leave the neocortex without the engagement of fast PV+ -mediated inhibition; [ii] Voltage responses in PV+ interneuron dendrites integrate linearly to reduce impact of the fluctuations in the afferent drive; and [iii] Reversed somatodendritic Rm gradient accelerates the time courses of synaptic potentials arriving at the soma. In the second group (dubbed After Axon), we list morphological and biophysical properties responsible for (a) short synaptic delays, and (b) efficient postsynaptic outcomes. For example: [i] Fast-spiking ability that allows PV+ interneurons to outpace other cortical neurons (pyramidal neurons). [ii] Myelinated axon (which is only found in the PV+ subclass of interneurons) to secure fast-spiking at the initial axon segment; and [iii] Inhibitory autapses - autoinhibition, which assures brief biphasic voltage transients and supports postinhibitory rebounds. Recent advent of scientific tools, such as viral strategies to target PV cells and the ability to monitor PV cells via in vivo imaging during behavior, will aid in defining the role of PV cells in the CNS. Given the link between PV+ interneurons and cognition, in the future, it would be useful to carry out physiological recordings in the PV+ cell type selectively and characterize if and how psychiatric and neurological diseases affect initiation and propagation of electrical signals in this cortical sub-circuit. Voltage imaging may allow fast recordings of electrical signals from many PV+ interneurons simultaneously.
Collapse
Affiliation(s)
- Katarina D. Milicevic
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
- University of Belgrade, Faculty of Biology, Center for Laser Microscopy, Belgrade, 11000, Serbia
| | - Brianna L. Barbeau
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| | - Darko D. Lovic
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
- University of Belgrade, Faculty of Biology, Center for Laser Microscopy, Belgrade, 11000, Serbia
| | - Aayushi A. Patel
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| | - Violetta O. Ivanova
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| | - Srdjan D. Antic
- University of Connecticut Health, School of Medicine, Institute for Systems Genomics, Farmington, CT, 06030, USA
| |
Collapse
|
7
|
Medalla M, Mo B, Nasar R, Zhou Y, Park J, Luebke JI. Comparative features of calretinin, calbindin, and parvalbumin expressing interneurons in mouse and monkey primary visual and frontal cortices. J Comp Neurol 2023; 531:1934-1962. [PMID: 37357562 PMCID: PMC10749991 DOI: 10.1002/cne.25514] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023]
Abstract
Fundamental differences in excitatory pyramidal cells across cortical areas and species highlight the implausibility of extrapolation from mouse to primate neurons and cortical networks. Far less is known about comparative regional and species-specific features of neurochemically distinct cortical inhibitory interneurons. Here, we quantified the density, laminar distribution, and somatodendritic morphology of inhibitory interneurons expressing one or more of the calcium-binding proteins (CaBPs) (calretinin [CR], calbindin [CB], and/or parvalbumin [PV]) in mouse (Mus musculus) versus rhesus monkey (Macaca mulatta) in two functionally and cytoarchitectonically distinct regions-the primary visual and frontal cortical areas-using immunofluorescent multilabeling, stereological counting, and 3D reconstructions. There were significantly higher densities of CB+ and PV+ neurons in visual compared to frontal areas in both species. The main species difference was the significantly greater density and proportion of CR+ interneurons and lower extent of CaBP coexpression in monkey compared to mouse cortices. Cluster analyses revealed that the somatodendritic morphology of layer 2-3 inhibitory interneurons is more dependent on CaBP expression than on species and area. Only modest effects of species were observed for CB+ and PV+ interneuron morphologies, while CR+ neurons showed no difference. By contrast to pyramidal cells that show highly distinctive area- and species-specific features, here we found more subtle differences in the distribution and features of interneurons across areas and species. These data yield insight into how nuanced differences in the population organization and properties of neurons may underlie specializations in cortical regions to confer species- and area-specific functional capacities.
Collapse
Affiliation(s)
- Maria Medalla
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, 7th Floor, Boston, MA 02215
| | - Bingxin Mo
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Rakin Nasar
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Yuxin Zhou
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Junwoo Park
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, 7th Floor, Boston, MA 02215
| |
Collapse
|
8
|
Druga R, Salaj M, Al-Redouan A. Parvalbumin - Positive Neurons in the Neocortex: A Review. Physiol Res 2023; 72:S173-S191. [PMID: 37565421 PMCID: PMC10660579 DOI: 10.33549/physiolres.935005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/02/2023] [Indexed: 12/01/2023] Open
Abstract
The calcium binding protein parvalbumin (PV) in the mammalian neocortex is expressed in a subpopulation of cortical GABAergic inhibitory interneurons. PV - producing interneurons represent the largest subpopulation of neocortical inhibitory cells, exhibit mutual chemical and electrical synaptic contacts and are well known to generate gamma oscillation. This review summarizes basic data of the distribution, afferent and efferent connections and physiological properties of parvalbumin expressing neurons in the neocortex. Basic data about participation of PV-positive neurons in cortical microcircuits are presented. Autaptic connections, metabolism and perineuronal nets (PNN) of PV positive neurons are also discussed.
Collapse
Affiliation(s)
- R Druga
- Department of Anatomy, 2nd Medical Faculty, Charles University Prague, Czech Republic.
| | | | | |
Collapse
|
9
|
Scheuer KS, Jansson AM, Zhao X, Jackson MB. Inter and Intralaminar Excitation of Parvalbumin Interneurons in Mouse Barrel Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543448. [PMID: 37398428 PMCID: PMC10312540 DOI: 10.1101/2023.06.02.543448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Parvalbumin (PV) interneurons are inhibitory fast-spiking cells with essential roles in directing the flow of information through cortical circuits. These neurons set the balance between excitation and inhibition, control rhythmic activity, and have been linked to disorders including autism spectrum and schizophrenia. PV interneurons differ between cortical layers in their morphology, circuitry, and function, but how their electrophysiological properties vary has received little attention. Here we investigate responses of PV interneurons in different layers of primary somatosensory barrel cortex (BC) to different excitatory inputs. With the genetically-encoded hybrid voltage sensor, hVOS, we recorded voltage changes simultaneously in many L2/3 and L4 PV interneurons to stimulation in either L2/3 or L4. Decay-times were consistent across L2/3 and L4. Amplitude, half-width, and rise-time were greater for PV interneurons residing in L2/3 compared to L4. Stimulation in L2/3 elicited responses in both L2/3 and L4 with longer latency compared to stimulation in L4. These differences in latency between layers could influence their windows for temporal integration. Thus PV interneurons in different cortical layers of BC show differences in response properties with potential roles in cortical computations.
Collapse
Affiliation(s)
- Kate S Scheuer
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Anna M Jansson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Meyer B Jackson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| |
Collapse
|
10
|
Medalla M, Mo B, Nasar R, Zhou Y, Park J, Luebke JI. Comparative Features of Calretinin, Calbindin and Parvalbumin Expressing Interneurons in Mouse and Monkey Primary Visual and Frontal Cortices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530269. [PMID: 36909556 PMCID: PMC10002648 DOI: 10.1101/2023.02.27.530269] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Much is known about differences in pyramidal cells across cortical areas and species, but studies of interneurons have focused on comparisons within single cortical areas and/or species. Here we quantified the distribution and somato-dendritic morphology of interneurons expressing one or more of the calcium binding proteins (CaBPs) calretinin (CR), calbindin (CB) and/or parvalbumin (PV) in mouse ( Mus musculus ) versus rhesus monkey ( Macaca mulatta ) in two functionally and cytoarchitectonically distinct regions- the primary visual and frontal cortical areas. The density, laminar distribution and morphology of interneurons were assessed in serial brain sections using immunofluorescent multi-labeling, stereological counting and 3D reconstructions. There were significantly higher densities of CB+ and PV+ neurons in visual compared to frontal areas in both species. The main species difference was the significantly greater density and proportion of CR+ interneurons and lower extent of CaBP co-expression in monkey compared to mouse cortices. Cluster analyses revealed that the somato-dendritic morphology of layer 2-3 inhibitory interneurons is more dependent on CaBP expression than on species and area. Only modest effects of species were observed for CB+ and PV+ interneuron morphologies, while CR+ neurons showed no difference. By contrast to pyramidal cells which show highly distinctive area- and species-specific features, here we found more subtle differences in the distribution and features of interneurons across areas and species. These data yield insight into how nuanced differences in the population organization and properties of neurons may underlie specializations in cortical regions to confer species and area-specific functional capacities. Key Points Somato-dendritic morphology of distinct interneurons did not substantially scale and vary across areas and species- differences were mainly dependent on CaBP expression.Cortical diversity in inhibitory function across areas and species is thus likely to be derived from differential laminar distribution and densities of distinct interneuron subclasses.In contrast to pyramidal cells which differ widely in distribution and morphology across areas and species, the features of interneurons appears to be relatively more conserved across areas and species.
Collapse
Affiliation(s)
- Maria Medalla
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, 7th Floor, Boston, MA 02215
| | - Bingxin Mo
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Rakin Nasar
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Yuxin Zhou
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Junwoo Park
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
| | - Jennifer I Luebke
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, 72 East Concord St. L10, Boston MA 02118
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, 7th Floor, Boston, MA 02215
| |
Collapse
|
11
|
Szegedi V, Bakos E, Furdan S, Kovács BH, Varga D, Erdélyi M, Barzó P, Szücs A, Tamás G, Lamsa K. HCN channels at the cell soma ensure the rapid electrical reactivity of fast-spiking interneurons in human neocortex. PLoS Biol 2023; 21:e3002001. [PMID: 36745683 PMCID: PMC9934405 DOI: 10.1371/journal.pbio.3002001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 02/16/2023] [Accepted: 01/17/2023] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence indicates that there are substantial species differences in the properties of mammalian neurons, yet theories on circuit activity and information processing in the human brain are based heavily on results obtained from rodents and other experimental animals. This knowledge gap may be particularly important for understanding the neocortex, the brain area responsible for the most complex neuronal operations and showing the greatest evolutionary divergence. Here, we examined differences in the electrophysiological properties of human and mouse fast-spiking GABAergic basket cells, among the most abundant inhibitory interneurons in cortex. Analyses of membrane potential responses to current input, pharmacologically isolated somatic leak currents, isolated soma outside-out patch recordings, and immunohistochemical staining revealed that human neocortical basket cells abundantly express hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channel isoforms HCN1 and HCN2 at the cell soma membrane, whereas these channels are sparse at the rodent basket cell soma membrane. Antagonist experiments showed that HCN channels in human neurons contribute to the resting membrane potential and cell excitability at the cell soma, accelerate somatic membrane potential kinetics, and shorten the lag between excitatory postsynaptic potentials and action potential generation. These effects are important because the soma of human fast-spiking neurons without HCN channels exhibit low persistent ion leak and slow membrane potential kinetics, compared with mouse fast-spiking neurons. HCN channels speed up human cell membrane potential kinetics and help attain an input-output rate close to that of rodent cells. Computational modeling demonstrated that HCN channel activity at the human fast-spiking cell soma membrane is sufficient to accelerate the input-output function as observed in cell recordings. Thus, human and mouse fast-spiking neurons exhibit functionally significant differences in ion channel composition at the cell soma membrane to set the speed and fidelity of their input-output function. These HCN channels ensure fast electrical reactivity of fast-spiking cells in human neocortex.
Collapse
Affiliation(s)
- Viktor Szegedi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Emőke Bakos
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Szabina Furdan
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Bálint H. Kovács
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Dániel Varga
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Pál Barzó
- Department of Neurosurgery, University of Szeged, Szeged, Hungary
| | - Attila Szücs
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
- Neuronal Cell Biology Research Group, Eötvös Loránd University, Budapest, Budapest, Hungary
| | - Gábor Tamás
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Karri Lamsa
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
- * E-mail: ,
| |
Collapse
|
12
|
Nakamura T, Dinh TH, Asai M, Matsumoto J, Nishimaru H, Setogawa T, Honda S, Yamada H, Mihara T, Nishijo H. Suppressive effects of ketamine on auditory steady-state responses in intact, awake macaques: A non-human primate model of schizophrenia. Brain Res Bull 2023; 193:84-94. [PMID: 36539101 DOI: 10.1016/j.brainresbull.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Auditory steady-state responses (ASSRs) are recurrent neural activities entrained to regular cyclic auditory stimulation. ASSRs are altered in individuals with schizophrenia, and may be related to hypofunction of the N-methyl-D-aspartate (NMDA) glutamate receptor. Noncompetitive NMDA receptor antagonists, including ketamine, have been used in ASSR studies of rodent models of schizophrenia. Although animal studies using non-human primates are required to complement rodent studies, the effects of ketamine on ASSRs are unknown in intact awake non-human primates. In this study, after administration of vehicle or ketamine, click trains at 20-83.3 Hz were presented to elicit ASSRs during recording of electroencephalograms in intact, awake macaque monkeys. The results indicated that ASSRs quantified by event-related spectral perturbation and inter-trial coherence were maximal at 83.3 Hz after vehicle administration, and that ketamine reduced ASSRs at 58.8 and 83.3 Hz, but not at 20 and 40 Hz. The present results demonstrated a reduction of ASSRs by the NMDA receptor antagonist at optimal frequencies with maximal responses in intact, awake macaques, comparable to ASSR reduction in patients with schizophrenia. These findings suggest that ASSR can be used as a neurophysiological biomarker of the disturbance of gamma-oscillatory neural circuits in this ketamine model of schizophrenia using intact, awake macaques. Thus, this model with ASSRs would be useful in the investigation of human brain pathophysiology as well as in preclinical translational research.
Collapse
Affiliation(s)
- Tomoya Nakamura
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Department of Anatomy, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Trong Ha Dinh
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Department of Physiology, Vietnam Military Medical University, Hanoi 100000, Viet Nam
| | - Makoto Asai
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Jumpei Matsumoto
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| | - Hiroshi Nishimaru
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| | - Tsuyoshi Setogawa
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| | - Sokichi Honda
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Hiroshi Yamada
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Takuma Mihara
- Candidate Discovery Science Labs, Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Ibaraki 305-8585, Japan
| | - Hisao Nishijo
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan; Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan.
| |
Collapse
|
13
|
Aussel A, Fiebelkorn IC, Kastner S, Kopell NJ, Pittman-Polletta BR. Interacting rhythms enhance sensitivity of target detection in a fronto-parietal computational model of visual attention. eLife 2023; 12:e67684. [PMID: 36718998 PMCID: PMC10129332 DOI: 10.7554/elife.67684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 01/12/2023] [Indexed: 02/01/2023] Open
Abstract
Even during sustained attention, enhanced processing of attended stimuli waxes and wanes rhythmically, with periods of enhanced and relatively diminished visual processing (and subsequent target detection) alternating at 4 or 8 Hz in a sustained visual attention task. These alternating attentional states occur alongside alternating dynamical states, in which lateral intraparietal cortex (LIP), the frontal eye field (FEF), and the mediodorsal pulvinar (mdPul) exhibit different activity and functional connectivity at α, β, and γ frequencies-rhythms associated with visual processing, working memory, and motor suppression. To assess whether and how these multiple interacting rhythms contribute to periodicity in attention, we propose a detailed computational model of FEF and LIP. When driven by θ-rhythmic inputs simulating experimentally-observed mdPul activity, this model reproduced the rhythmic dynamics and behavioral consequences of observed attentional states, revealing that the frequencies and mechanisms of the observed rhythms allow for peak sensitivity in visual target detection while maintaining functional flexibility.
Collapse
Affiliation(s)
- Amélie Aussel
- Cognitive Rhythms Collaborative, Boston UniversityBostonUnited States
- Department of Mathematics and Statistics, Boston UniversityRochesterUnited States
| | - Ian C Fiebelkorn
- Department of Neuroscience and Del Monte Institute for Neuroscience, University of Rochester Medical Center, University of RochesterRochesterUnited States
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Sabine Kastner
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
- Department of Psychology, Princeton UniversityPrincetonUnited States
| | - Nancy J Kopell
- Cognitive Rhythms Collaborative, Boston UniversityBostonUnited States
- Department of Mathematics and Statistics, Boston UniversityRochesterUnited States
| | - Benjamin Rafael Pittman-Polletta
- Cognitive Rhythms Collaborative, Boston UniversityBostonUnited States
- Department of Mathematics and Statistics, Boston UniversityRochesterUnited States
| |
Collapse
|
14
|
Roussel Y, Verasztó C, Rodarie D, Damart T, Reimann M, Ramaswamy S, Markram H, Keller D. Mapping of morpho-electric features to molecular identity of cortical inhibitory neurons. PLoS Comput Biol 2023; 19:e1010058. [PMID: 36602951 DOI: 10.1371/journal.pcbi.1010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/26/2022] [Indexed: 01/06/2023] Open
Abstract
Knowledge of the cell-type-specific composition of the brain is useful in order to understand the role of each cell type as part of the network. Here, we estimated the composition of the whole cortex in terms of well characterized morphological and electrophysiological inhibitory neuron types (me-types). We derived probabilistic me-type densities from an existing atlas of molecularly defined cell-type densities in the mouse cortex. We used a well-established me-type classification from rat somatosensory cortex to populate the cortex. These me-types were well characterized morphologically and electrophysiologically but they lacked molecular marker identity labels. To extrapolate this missing information, we employed an additional dataset from the Allen Institute for Brain Science containing molecular identity as well as morphological and electrophysiological data for mouse cortical neurons. We first built a latent space based on a number of comparable morphological and electrical features common to both data sources. We then identified 19 morpho-electrical clusters that merged neurons from both datasets while being molecularly homogeneous. The resulting clusters best mirror the molecular identity classification solely using available morpho-electrical features. Finally, we stochastically assigned a molecular identity to a me-type neuron based on the latent space cluster it was assigned to. The resulting mapping was used to derive inhibitory me-types densities in the cortex.
Collapse
Affiliation(s)
- Yann Roussel
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Csaba Verasztó
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Dimitri Rodarie
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Tanguy Damart
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Michael Reimann
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Srikanth Ramaswamy
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Henry Markram
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Daniel Keller
- Blue Brain Project, École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| |
Collapse
|
15
|
Proskurina EY, Chizhov AV, Zaitsev AV. Optogenetic Low-Frequency Stimulation of Principal Neurons, but Not Parvalbumin-Positive Interneurons, Prevents Generation of Ictal Discharges in Rodent Entorhinal Cortex in an In Vitro 4-Aminopyridine Model. Int J Mol Sci 2022; 24:ijms24010195. [PMID: 36613660 PMCID: PMC9820186 DOI: 10.3390/ijms24010195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Low-frequency electrical stimulation is used to treat some drug-resistant forms of epilepsy. Despite the effectiveness of the method in suppressing seizures, there is a considerable risk of side effects. An optogenetic approach allows the targeting of specific populations of neurons, which can increase the effectiveness and safety of low-frequency stimulation. In our study, we tested the efficacy of the suppression of ictal activity in entorhinal cortex slices in a 4-aminopyridine model with three variants of low-frequency light stimulation (LFLS): (1) activation of excitatory and inhibitory neurons (on Thy1-ChR2-YFP mice), (2) activation of inhibitory interneurons only (on PV-Cre mice after virus injection with channelrhodopsin2 gene), and (3) hyperpolarization of excitatory neurons (on Wistar rats after virus injection with archaerhodopsin gene). Only in the first variant did simultaneous LFLS of excitatory and inhibitory neurons replace ictal activity with interictal activity. We suggest that LFLS caused changes in the concentration gradients of K+ and Na+ cations across the neuron membrane, which activated Na-K pumping. According to the mathematical modeling, the increase in Na-K pump activity in neurons induced by LFLS led to an antiepileptic effect. Thus, a less specific and generalized optogenetic effect on entorhinal cortex neurons was more effective in suppressing ictal activity in the 4-aminopyridine model.
Collapse
Affiliation(s)
- Elena Y. Proskurina
- Almazov National Medical Research Centre, 2 Akkuratova Street, 197341 St. Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 44 Toreza Prospekt, 194223 St. Petersburg, Russia
| | - Anton V. Chizhov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 44 Toreza Prospekt, 194223 St. Petersburg, Russia
- Computational Physics Laboratory, Ioffe Institute, 26 Polytekhnicheskaya Street, 194021 St. Petersburg, Russia
- MathNeuro Team, Inria Centre at Universite Cote d’Azur, 06902 Sophia Antipolis, France
| | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 44 Toreza Prospekt, 194223 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
16
|
Ostos S, Aparicio G, Fernaud-Espinosa I, DeFelipe J, Muñoz A. Quantitative analysis of the GABAergic innervation of the soma and axon initial segment of pyramidal cells in the human and mouse neocortex. Cereb Cortex 2022; 33:3882-3909. [PMID: 36058205 DOI: 10.1093/cercor/bhac314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 11/13/2022] Open
Abstract
Perisomatic GABAergic innervation in the cerebral cortex is carried out mostly by basket and chandelier cells, which differentially participate in the control of pyramidal cell action potential output and synchronization. These cells establish multiple synapses with the cell body (and proximal dendrites) and the axon initial segment (AIS) of pyramidal neurons, respectively. Using multiple immunofluorescence, confocal microscopy and 3D quantification techniques, we have estimated the number and density of GABAergic boutons on the cell body and AIS of pyramidal neurons located through cortical layers of the human and mouse neocortex. The results revealed, in both species, that there is clear variability across layers regarding the density and number of perisomatic GABAergic boutons. We found a positive linear correlation between the surface area of the soma, or the AIS, and the number of GABAergic terminals in apposition to these 2 neuronal domains. Furthermore, the density of perisomatic GABAergic boutons was higher in the human cortex than in the mouse. These results suggest a selectivity for the GABAergic innervation of the cell body and AIS that might be related to the different functional attributes of the microcircuits in which neurons from different layers are involved in both human and mouse.
Collapse
Affiliation(s)
- Sandra Ostos
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Guillermo Aparicio
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Isabel Fernaud-Espinosa
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Javier DeFelipe
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, 28223, Pozuelo de Alarcón, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Alberto Muñoz
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, 28223, Pozuelo de Alarcón, Madrid, Spain.,Departamento de Biología Celular, Universidad Complutense, José Antonio Novais 12, 28040 Madrid, Spain
| |
Collapse
|
17
|
Gonzalez AE, Jorgensen ET, Ramos JD, Harkness JH, Aadland JA, Brown TE, Sorg BA. Impact of Perineuronal Net Removal in the Rat Medial Prefrontal Cortex on Parvalbumin Interneurons After Reinstatement of Cocaine Conditioned Place Preference. Front Cell Neurosci 2022; 16:932391. [PMID: 35966203 PMCID: PMC9366391 DOI: 10.3389/fncel.2022.932391] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
Parvalbumin (PV)-positive cells are GABAergic fast-spiking interneurons that modulate the activity of pyramidal neurons in the medial prefrontal cortex (mPFC) and their output to brain areas associated with learning and memory. The majority of PV cells within the mPFC are surrounded by a specialized extracellular matrix structure called the perineuronal net (PNN). We have shown that removal of PNNs with the enzyme chondroitinase-ABC (Ch-ABC) in the mPFC prevents the consolidation and reconsolidation of cocaine-associated conditioned place preference (CPP) memories. Here we examined the extent to which retrieval of a CPP memory during cocaine-primed reinstatement altered the levels and function of PV neurons and their surrounding PNNs during the reconsolidation period. We further determined the extent to which PNN removal prior to reinstatement altered PV intensity levels and PV cell function. Male Sprague-Dawley rats were trained for cocaine-induced conditioned place preference (CPP) followed by extinction training, microinjection of Ch-ABC in the prelimbic PFC, and cocaine-induced reinstatement. Rats were sacrificed immediately prior to reinstatement or at 2 h, 6 h, or 48 h after reinstatement for immunohistochemistry or 2 h later for electrophysiology. Our findings indicate that PNN removal only partially diminished reinstatement. Cocaine-primed reinstatement produced only minor changes in PNN or PV intensity in vehicle controls. However, after PNN removal, the intensity of remaining PNN-surrounded PV cells was decreased at all times except at 2 h post-reinstatement, at which time cocaine increased PV intensity. Consistent with this, in vehicle controls, PV neurons naturally devoid of PNNs showed a similar pattern to Ch-ABC-treated rats prior to and after cocaine reinstatement, suggesting a protective effect of PNNs on cocaine-induced changes in PV intensity. Using whole-cell patch-clamp, cocaine-primed reinstatement in Ch-ABC-treated rats decreased the number of elicited action potentials but increased excitatory synaptic transmission, which may have been compensatory. These findings suggest that without PNNs, cocaine-induced reinstatement produces rapid changes in PV intensity and PV cell excitability, which may in turn regulate output of the mPFC post-memory retrieval and diminish the maintenance of cocaine memory during reconsolidation.
Collapse
Affiliation(s)
- Angela E. Gonzalez
- Program in Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, United States
- Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Emily T. Jorgensen
- Neuroscience Graduate Program and School of Pharmacy, University of Wyoming, Laramie, WY, United States
| | - Jonathan D. Ramos
- Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| | | | - Jake A. Aadland
- Neuroscience Graduate Program and School of Pharmacy, University of Wyoming, Laramie, WY, United States
| | - Travis E. Brown
- Neuroscience Graduate Program and School of Pharmacy, University of Wyoming, Laramie, WY, United States
| | - Barbara A. Sorg
- Program in Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, United States
- Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
- *Correspondence: Barbara A. Sorg
| |
Collapse
|
18
|
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
|
19
|
Malwade S, Gasthaus J, Bellardita C, Andelic M, Moric B, Korshunova I, Kiehn O, Vasistha NA, Khodosevich K. Identification of Vulnerable Interneuron Subtypes in 15q13.3 Microdeletion Syndrome Using Single-Cell Transcriptomics. Biol Psychiatry 2022; 91:727-739. [PMID: 34838304 DOI: 10.1016/j.biopsych.2021.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 11/02/2022]
Abstract
BACKGROUND A number of rare copy number variants (CNVs) have been linked to neurodevelopmental disorders. However, because CNVs encompass many genes, it is often difficult to identify the mechanisms that lead to developmental perturbations. METHODS We used 15q13.3 microdeletion to propose and validate a novel strategy to predict the impact of CNV genes on brain development that could further guide functional studies. We analyzed single-cell transcriptomics datasets containing cortical interneurons to identify their developmental vulnerability to 15q13.3 microdeletion, which was validated in mouse models. RESULTS We found that Klf13-but not other 15q13.3 genes-is expressed by precursors and neuroblasts in the medial and caudal ganglionic eminences during development, with a peak of expression at embryonic day (E)13.5 and E18.5, respectively. In contrast, in the adult mouse brain, Klf13 expression is negligible. Using Df(h15q13.3)/+ and Klf13+/- embryos, we observed a precursor subtype-specific impairment in proliferation in the medial ganglionic eminence and caudal ganglionic eminence at E13.5 and E17.5, respectively, corresponding to vulnerability predicted by Klf13 expression patterns. Finally, Klf13+/- mice showed a layer-specific decrease in parvalbumin and somatostatin cortical interneurons accompanied by changes in locomotor and anxiety-related behavior. CONCLUSIONS We show that the impact of 15q13.3 microdeletion on precursor proliferation is grounded in a reduction in Klf13 expression. The lack of Klf13 in Df(h15q13.3)/+ cortex might be the major reason for perturbed density of cortical interneurons. Thus, the behavioral defects seen in 15q13.3 microdeletion could stem from a developmental perturbation owing to selective vulnerability of cortical interneurons during sensitive stages of their development.
Collapse
Affiliation(s)
- Susmita Malwade
- Biotech Research and Innovation Center (BRIC), Copenhagen Biocenter, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Janina Gasthaus
- Biotech Research and Innovation Center (BRIC), Copenhagen Biocenter, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carmelo Bellardita
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matej Andelic
- Biotech Research and Innovation Center (BRIC), Copenhagen Biocenter, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Borna Moric
- Biotech Research and Innovation Center (BRIC), Copenhagen Biocenter, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irina Korshunova
- Biotech Research and Innovation Center (BRIC), Copenhagen Biocenter, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Kiehn
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Navneet A Vasistha
- Biotech Research and Innovation Center (BRIC), Copenhagen Biocenter, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Konstantin Khodosevich
- Biotech Research and Innovation Center (BRIC), Copenhagen Biocenter, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
20
|
Differential distribution of inhibitory neuron types in subregions of claustrum and dorsal endopiriform nucleus of the short-tailed fruit bat. Brain Struct Funct 2022; 227:1615-1640. [PMID: 35188589 DOI: 10.1007/s00429-022-02459-0] [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: 09/14/2021] [Accepted: 01/17/2022] [Indexed: 12/22/2022]
Abstract
Few brain regions have such wide-ranging inputs and outputs as the claustrum does, and fewer have posed equivalent challenges in defining their structural boundaries. We studied the distributions of three calcium-binding proteins-calretinin, parvalbumin, and calbindin-in the claustrum and dorsal endopiriform nucleus of the fruit bat, Carollia perspicillata. The proportionately large sizes of claustrum and dorsal endopiriform nucleus in Carollia brain afford unique access to these structures' intrinsic anatomy. Latexin immunoreactivity permits a separation of claustrum into core and shell subregions and an equivalent separation of dorsal endopiriform nucleus. Using latexin labeling, we found that the claustral shell in Carollia brain can be further subdivided into at least four distinct subregions. Calretinin and parvalbumin immunoreactivity reinforced the boundaries of the claustral core and its shell subregions with diametrically opposite distribution patterns. Calretinin, parvalbumin, and calbindin all colocalized with GAD67, indicating that these proteins label inhibitory neurons in both claustrum and dorsal endopiriform nucleus. Calretinin, however, also colocalized with latexin in a subset of neurons. Confocal microscopy revealed appositions that suggest synaptic contacts between cells labeled for each of the three calcium-binding proteins and latexin-immunoreactive somata in claustrum and dorsal endopiriform nucleus. Our results indicate significant subregional differences in the intrinsic inhibitory connectivity within and between claustrum and dorsal endopiriform nucleus. We conclude that the claustrum is structurally more complex than previously appreciated and that claustral and dorsal endopiriform nucleus subregions are differentially modulated by multiple inhibitory systems. These findings can also account for the excitability differences between claustrum and dorsal endopiriform nucleus described previously.
Collapse
|
21
|
Li S, Wang XJ. Hierarchical timescales in the neocortex: Mathematical mechanism and biological insights. Proc Natl Acad Sci U S A 2022; 119:e2110274119. [PMID: 35110401 PMCID: PMC8832993 DOI: 10.1073/pnas.2110274119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/07/2021] [Indexed: 11/18/2022] Open
Abstract
A cardinal feature of the neocortex is the progressive increase of the spatial receptive fields along the cortical hierarchy. Recently, theoretical and experimental findings have shown that the temporal response windows also gradually enlarge, so that early sensory neural circuits operate on short timescales whereas higher-association areas are capable of integrating information over a long period of time. While an increased receptive field is accounted for by spatial summation of inputs from neurons in an upstream area, the emergence of timescale hierarchy cannot be readily explained, especially given the dense interareal cortical connectivity known in the modern connectome. To uncover the required neurobiological properties, we carried out a rigorous analysis of an anatomically based large-scale cortex model of macaque monkeys. Using a perturbation method, we show that the segregation of disparate timescales is defined in terms of the localization of eigenvectors of the connectivity matrix, which depends on three circuit properties: 1) a macroscopic gradient of synaptic excitation, 2) distinct electrophysiological properties between excitatory and inhibitory neuronal populations, and 3) a detailed balance between long-range excitatory inputs and local inhibitory inputs for each area-to-area pathway. Our work thus provides a quantitative understanding of the mechanism underlying the emergence of timescale hierarchy in large-scale primate cortical networks.
Collapse
Affiliation(s)
- Songting Li
- School of Mathematical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China;
- Institute of Natural Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Ministry of Education Key Laboratory of Scientific and Engineering Computing, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiao-Jing Wang
- Center for Neural Science, New York University, New York, NY 10003
| |
Collapse
|
22
|
Miyamae T, Hashimoto T, Abraham M, Kawabata R, Koshikizawa S, Bian Y, Nishihata Y, Kikuchi M, Ermentrout GB, Lewis DA, Gonzalez-Burgos G. Kcns3 deficiency disrupts Parvalbumin neuron physiology in mouse prefrontal cortex: Implications for the pathophysiology of schizophrenia. Neurobiol Dis 2021; 155:105382. [PMID: 33940180 PMCID: PMC8557947 DOI: 10.1016/j.nbd.2021.105382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 01/04/2023] Open
Abstract
The unique fast spiking (FS) phenotype of cortical parvalbumin-positive (PV) neurons depends on the expression of multiple subtypes of voltage-gated potassium channels (Kv). PV neurons selectively express Kcns3, the gene encoding Kv9.3 subunits, suggesting that Kcns3 expression is critical for the FS phenotype. KCNS3 expression is lower in PV neurons in the neocortex of subjects with schizophrenia, but the effects of this alteration are unclear, because Kv9.3 subunit function is poorly understood. Therefore, to assess the role of Kv9.3 subunits in PV neuron function, we combined gene expression analyses, computational modeling, and electrophysiology in acute slices from the cortex of Kcns3-deficient mice. Kcns3 mRNA levels were ~ 50% lower in cortical PV neurons from Kcns3-deficient relative to wildtype mice. While silent per se, Kv9.3 subunits are believed to amplify the Kv2.1 current in Kv2.1-Kv9.3 channel complexes. Hence, to assess the consequences of reducing Kv9.3 levels, we simulated the effects of decreasing the Kv2.1-mediated current in a computational model. The FS cell model with reduced Kv2.1 produced spike trains with irregular inter-spike intervals, or stuttering, and greater Na+ channel inactivation. As in the computational model, PV basket cells (PVBCs) from Kcns3-deficient mice displayed spike trains with strong stuttering, which depressed PVBC firing. Moreover, Kcns3 deficiency impaired the recruitment of PVBC firing at gamma frequency by stimuli mimicking synaptic input observed during cortical UP states. Our data indicate that Kv9.3 subunits are critical for PVBC physiology and suggest that KCNS3 deficiency in schizophrenia could impair PV neuron firing, possibly contributing to deficits in cortical gamma oscillations in the illness.
Collapse
Affiliation(s)
- Takeaki Miyamae
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Takanori Hashimoto
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan.
| | - Monica Abraham
- Department of Mathematics, Faculty of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Rika Kawabata
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Sho Koshikizawa
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Yufan Bian
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan
| | - Yosuke Nishihata
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Mitsuru Kikuchi
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa 920-8640, Japan; Research Center for Child Mental Development, Kanazawa University, Kanazawa 920-8640, Japan
| | - G Bard Ermentrout
- Department of Mathematics, Faculty of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Guillermo Gonzalez-Burgos
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
23
|
Bucher EA, Collins JM, King AE, Vickers JC, Kirkcaldie MTK. Coherence and cognition in the cortex: the fundamental role of parvalbumin, myelin, and the perineuronal net. Brain Struct Funct 2021; 226:2041-2055. [PMID: 34175994 DOI: 10.1007/s00429-021-02327-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/17/2021] [Indexed: 11/28/2022]
Abstract
The calcium binding protein parvalbumin is expressed in interneurons of two main morphologies, the basket and chandelier cells, which target perisomatic domains on principal cells and are extensively interconnected in laminar networks by synapses and gap junctions. Beyond its utility as a convenient cellular marker, parvalbumin is an unambiguous identifier of the key role that these interneurons play in the fundamental functions of the cortex. They provide a temporal framework for principal cell activity by propagating gamma oscillation, providing coherence for cortical information processing and the basis for timing-dependent plasticity processes. As these parvalbumin networks mature, they are physically and functionally stabilised by axonal myelination and development of the extracellular matrix structure termed the perineuronal net. This maturation correlates with the emergence of high-speed, highly energetic activity and provides a coherent foundation for the unique ability of the cortex to cross-correlate activity across sensory modes and internal representations.
Collapse
Affiliation(s)
- Ellie A Bucher
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - Jessica M Collins
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia
| | - Matthew T K Kirkcaldie
- Wicking Dementia Research and Education Centre, University of Tasmania, Private Bag 143, Hobart, TAS, 7001, Australia.
| |
Collapse
|
24
|
Staiger JF, Petersen CCH. Neuronal Circuits in Barrel Cortex for Whisker Sensory Perception. Physiol Rev 2020; 101:353-415. [PMID: 32816652 DOI: 10.1152/physrev.00019.2019] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The array of whiskers on the snout provides rodents with tactile sensory information relating to the size, shape and texture of objects in their immediate environment. Rodents can use their whiskers to detect stimuli, distinguish textures, locate objects and navigate. Important aspects of whisker sensation are thought to result from neuronal computations in the whisker somatosensory cortex (wS1). Each whisker is individually represented in the somatotopic map of wS1 by an anatomical unit named a 'barrel' (hence also called barrel cortex). This allows precise investigation of sensory processing in the context of a well-defined map. Here, we first review the signaling pathways from the whiskers to wS1, and then discuss current understanding of the various types of excitatory and inhibitory neurons present within wS1. Different classes of cells can be defined according to anatomical, electrophysiological and molecular features. The synaptic connectivity of neurons within local wS1 microcircuits, as well as their long-range interactions and the impact of neuromodulators, are beginning to be understood. Recent technological progress has allowed cell-type-specific connectivity to be related to cell-type-specific activity during whisker-related behaviors. An important goal for future research is to obtain a causal and mechanistic understanding of how selected aspects of tactile sensory information are processed by specific types of neurons in the synaptically connected neuronal networks of wS1 and signaled to downstream brain areas, thus contributing to sensory-guided decision-making.
Collapse
Affiliation(s)
- Jochen F Staiger
- University Medical Center Göttingen, Institute for Neuroanatomy, Göttingen, Germany; and Laboratory of Sensory Processing, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carl C H Petersen
- University Medical Center Göttingen, Institute for Neuroanatomy, Göttingen, Germany; and Laboratory of Sensory Processing, Faculty of Life Sciences, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
25
|
Chittajallu R, Auville K, Mahadevan V, Lai M, Hunt S, Calvigioni D, Pelkey KA, Zaghloul KA, McBain CJ. Activity-dependent tuning of intrinsic excitability in mouse and human neurogliaform cells. eLife 2020; 9:57571. [PMID: 32496194 PMCID: PMC7299336 DOI: 10.7554/elife.57571] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022] Open
Abstract
The ability to modulate the efficacy of synaptic communication between neurons constitutes an essential property critical for normal brain function. Animal models have proved invaluable in revealing a wealth of diverse cellular mechanisms underlying varied plasticity modes. However, to what extent these processes are mirrored in humans is largely uncharted thus questioning their relevance in human circuit function. In this study, we focus on neurogliaform cells, that possess specialized physiological features enabling them to impart a widespread inhibitory influence on neural activity. We demonstrate that this prominent neuronal subtype, embedded in both mouse and human neural circuits, undergo remarkably similar activity-dependent modulation manifesting as epochs of enhanced intrinsic excitability. In principle, these evolutionary conserved plasticity routes likely tune the extent of neurogliaform cell mediated inhibition thus constituting canonical circuit mechanisms underlying human cognitive processing and behavior.
Collapse
Affiliation(s)
- Ramesh Chittajallu
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Kurt Auville
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Vivek Mahadevan
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Mandy Lai
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Steven Hunt
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Daniela Calvigioni
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Kenneth A Pelkey
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Kareem A Zaghloul
- Surgical Neurology Branch, National Institutes of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Chris J McBain
- Laboratory of Cellular and Synaptic Physiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| |
Collapse
|
26
|
Vanni S, Hokkanen H, Werner F, Angelucci A. Anatomy and Physiology of Macaque Visual Cortical Areas V1, V2, and V5/MT: Bases for Biologically Realistic Models. Cereb Cortex 2020; 30:3483-3517. [PMID: 31897474 PMCID: PMC7233004 DOI: 10.1093/cercor/bhz322] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/02/2019] [Indexed: 12/22/2022] Open
Abstract
The cerebral cortex of primates encompasses multiple anatomically and physiologically distinct areas processing visual information. Areas V1, V2, and V5/MT are conserved across mammals and are central for visual behavior. To facilitate the generation of biologically accurate computational models of primate early visual processing, here we provide an overview of over 350 published studies of these three areas in the genus Macaca, whose visual system provides the closest model for human vision. The literature reports 14 anatomical connection types from the lateral geniculate nucleus of the thalamus to V1 having distinct layers of origin or termination, and 194 connection types between V1, V2, and V5, forming multiple parallel and interacting visual processing streams. Moreover, within V1, there are reports of 286 and 120 types of intrinsic excitatory and inhibitory connections, respectively. Physiologically, tuning of neuronal responses to 11 types of visual stimulus parameters has been consistently reported. Overall, the optimal spatial frequency (SF) of constituent neurons decreases with cortical hierarchy. Moreover, V5 neurons are distinct from neurons in other areas for their higher direction selectivity, higher contrast sensitivity, higher temporal frequency tuning, and wider SF bandwidth. We also discuss currently unavailable data that could be useful for biologically accurate models.
Collapse
Affiliation(s)
- Simo Vanni
- HUS Neurocenter, Department of Neurology, Helsinki University Hospital, 00290 Helsinki, Finland
- Department of Neurosciences, University of Helsinki, 00100 Helsinki, Finland
| | - Henri Hokkanen
- HUS Neurocenter, Department of Neurology, Helsinki University Hospital, 00290 Helsinki, Finland
- Department of Neurosciences, University of Helsinki, 00100 Helsinki, Finland
| | - Francesca Werner
- HUS Neurocenter, Department of Neurology, Helsinki University Hospital, 00290 Helsinki, Finland
- Department of Neurosciences, University of Helsinki, 00100 Helsinki, Finland
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Alessandra Angelucci
- Department of Ophthalmology and Visual Sciences, Moran Eye Institute, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
27
|
Kelly JG, Hawken MJ. GABAergic and non-GABAergic subpopulations of Kv3.1b-expressing neurons in macaque V2 and MT: laminar distributions and proportion of total neuronal population. Brain Struct Funct 2020; 225:1135-1152. [PMID: 32266458 DOI: 10.1007/s00429-020-02065-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 03/27/2020] [Indexed: 11/26/2022]
Abstract
The Kv3.1b potassium channel subunit, which facilitates the fast-spiking phenotype characteristic of parvalbumin (PV)-expressing inhibitory interneurons, is also expressed by subpopulations of excitatory neurons in macaque cortex. We have previously shown that V1 neurons expressing Kv3.1b but not PV or GABA were largely concentrated within layers 4Cα and 4B of V1, suggesting laminar or pathway specificity. In the current study, the distribution and pattern of co-immunoreactivity of GABA, PV, and Kv3.1b across layers in extrastriate cortical areas V2 and MT of the macaque monkey were measured using the same triple immunofluorescence labeling, confocal microscopy, and partially automated cell-counting strategies used in V1. For comparison, densities of the overall cell and neuronal populations were also measured for each layer of V2 and MT using tissue sections immunofluorescence labeled for the pan-neuronal marker NeuN. GABAergic neurons accounted for 14% of the total neuronal population in V2 and 25% in MT. Neurons expressing Kv3.1b but neither GABA nor PV were present in both areas. This subpopulation was most prevalent in the lowest subcompartment of layer 3, comprising 5% of the total neuronal population in layer 3C of both areas, and 41% and 36% of all Kv3.1b+ neurons in this layer in V2 and MT, respectively. The prevalence and laminar distribution of this subpopulation were remarkably consistent between V2 and MT and showed a striking similarity to the patterns observed previously in V1, suggesting a common contribution to the cortical circuit across areas.
Collapse
Affiliation(s)
- Jenna G Kelly
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, 10003, USA
| | - Michael J Hawken
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, 10003, USA.
| |
Collapse
|
28
|
Bensaid S, Modolo J, Merlet I, Wendling F, Benquet P. COALIA: A Computational Model of Human EEG for Consciousness Research. Front Syst Neurosci 2019; 13:59. [PMID: 31798421 PMCID: PMC6863981 DOI: 10.3389/fnsys.2019.00059] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/07/2019] [Indexed: 01/27/2023] Open
Abstract
Understanding the origin of the main physiological processes involved in consciousness is a major challenge of contemporary neuroscience, with crucial implications for the study of Disorders of Consciousness (DOC). The difficulties in achieving this task include the considerable quantity of experimental data in this field, along with the non-intuitive, nonlinear nature of neuronal dynamics. One possibility of integrating the main results from the experimental literature into a cohesive framework, while accounting for nonlinear brain dynamics, is the use of physiologically-inspired computational models. In this study, we present a physiologically-grounded computational model, attempting to account for the main micro-circuits identified in the human cortex, while including the specificities of each neuronal type. More specifically, the model accounts for thalamo-cortical (vertical) regulation of cortico-cortical (horizontal) connectivity, which is a central mechanism for brain information integration and processing. The distinct neuronal assemblies communicate through feedforward and feedback excitatory and inhibitory synaptic connections implemented in a template brain accounting for long-range connectome. The EEG generated by this physiologically-based simulated brain is validated through comparison with brain rhythms recorded in humans in two states of consciousness (wakefulness, sleep). Using the model, it is possible to reproduce the local disynaptic disinhibition of basket cells (fast GABAergic inhibition) and glutamatergic pyramidal neurons through long-range activation of vasoactive intestinal-peptide (VIP) interneurons that induced inhibition of somatostatin positive (SST) interneurons. The model (COALIA) predicts that the strength and dynamics of the thalamic output on the cortex control the local and long-range cortical processing of information. Furthermore, the model reproduces and explains clinical results regarding the complexity of transcranial magnetic stimulation TMS-evoked EEG responses in DOC patients and healthy volunteers, through a modulation of thalamo-cortical connectivity that governs the level of cortico-cortical communication. This new model provides a quantitative framework to accelerate the study of the physiological mechanisms involved in the emergence, maintenance and disruption (sleep, anesthesia, DOC) of consciousness.
Collapse
Affiliation(s)
| | | | | | - Fabrice Wendling
- INSERM, Laboratoire Traitement du Signal et de l’Image (LTSI)—U1099, University of Rennes, Rennes, France
| | | |
Collapse
|
29
|
Tzilivaki A, Kastellakis G, Poirazi P. Challenging the point neuron dogma: FS basket cells as 2-stage nonlinear integrators. Nat Commun 2019; 10:3664. [PMID: 31413258 PMCID: PMC6694133 DOI: 10.1038/s41467-019-11537-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022] Open
Abstract
Interneurons are critical for the proper functioning of neural circuits. While often morphologically complex, their dendrites have been ignored for decades, treating them as linear point neurons. Exciting new findings reveal complex, non-linear dendritic computations that call for a new theory of interneuron arithmetic. Using detailed biophysical models, we predict that dendrites of FS basket cells in both hippocampus and prefrontal cortex come in two flavors: supralinear, supporting local sodium spikes within large-volume branches and sublinear, in small-volume branches. Synaptic activation of varying sets of these dendrites leads to somatic firing variability that cannot be fully explained by the point neuron reduction. Instead, a 2-stage artificial neural network (ANN), with sub- and supralinear hidden nodes, captures most of the variance. Reduced neuronal circuit modeling suggest that this bi-modal, 2-stage integration in FS basket cells confers substantial resource savings in memory encoding as well as the linking of memories across time.
Collapse
Affiliation(s)
- Alexandra Tzilivaki
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, 70013, Greece
- Department of Biology, University of Crete, Heraklion, 70013, Greece
- Einstein Center for Neurosciences Berlin, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117, Berlin, Germany
| | - George Kastellakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, 70013, Greece
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, 70013, Greece.
| |
Collapse
|
30
|
Povysheva N, Nigam A, Brisbin AK, Johnson JW, Barrionuevo G. Oxygen-Glucose Deprivation Differentially Affects Neocortical Pyramidal Neurons and Parvalbumin-Positive Interneurons. Neuroscience 2019; 412:72-82. [PMID: 31152933 DOI: 10.1016/j.neuroscience.2019.05.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 05/02/2019] [Accepted: 05/21/2019] [Indexed: 11/28/2022]
Abstract
Stroke is a devastating brain disorder. The pathophysiology of stroke is associated with an impaired excitation-inhibition balance in the area that surrounds the infarct core after the insult, the peri-infarct zone. Here we exposed slices from adult mouse prefrontal cortex to oxygen-glucose deprivation and reoxygenation (OGD-RO) to study ischemia-induced changes in the activity of excitatory pyramidal neurons and inhibitory parvalbumin (PV)-positive interneurons. We found that during current-clamp recordings, PV-positive interneurons were more vulnerable to OGD-RO than pyramidal neurons as indicated by the lower percentage of recovery of PV-positive interneurons. However, neither the amplitude of OGD-induced depolarization observed in current-clamp mode nor the OGD-associated current observed in voltage-clamp mode differed between the two cell types. Large amplitude, presumably action-potential dependent, spontaneous postsynaptic inhibitory currents recorded from pyramidal neurons were less frequent after OGD-RO than in control condition. Disynaptic inhibitory postsynaptic currents (dIPSCs) in pyramidal neurons produced predominantly by PV-positive interneurons were reduced by OGD-RO. Following OGD-RO, dendrites of PV-positive interneurons exhibited more pathological beading than those of pyramidal neurons. Our data support the hypothesis that the differential vulnerability to ischemia-like conditions of excitatory and inhibitory neurons leads to the altered excitation-inhibition balance associated with stroke pathophysiology.
Collapse
Affiliation(s)
- Nadya Povysheva
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| | - Aparna Nigam
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Alyssa K Brisbin
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Jon W Johnson
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Germán Barrionuevo
- Department of Neuroscience and Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, 15260, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
31
|
Kelly JG, García-Marín V, Rudy B, Hawken MJ. Densities and Laminar Distributions of Kv3.1b-, PV-, GABA-, and SMI-32-Immunoreactive Neurons in Macaque Area V1. Cereb Cortex 2019; 29:1921-1937. [PMID: 29668858 PMCID: PMC6458914 DOI: 10.1093/cercor/bhy072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 03/06/2018] [Indexed: 12/11/2022] Open
Abstract
The Kv3.1b potassium channel subunit is associated with narrow spike widths and fast-spiking properties. In macaque primary visual cortex (V1), subsets of neurons have previously been found to be Kv3.1b-immunoreactive (ir) but not parvalbumin (PV)-ir or not GABA-ir, suggesting that they may be both fast-spiking and excitatory. This population includes Meynert cells, the large layer 5/6 pyramidal neurons that are also labeled by the neurofilament antibody SMI-32. In the present study, triple immunofluorescence labeling and confocal microscopy were used to measure the distribution of Kv3.1b-ir, non-PV-ir, non-GABA-ir neurons across cortical depth in V1, and to determine whether, like the Meynert cells, other Kv3.1b-ir excitatory neurons were also SMI-32-ir pyramidal neurons. We found that Kv3.1b-ir, non-PV-ir, non-GABA-ir neurons were most prevalent in the M pathway-associated layers 4 Cα and 4B. GABAergic neurons accounted for a smaller fraction (11%) of the total neuronal population across layers 1-6 than has previously been reported. Of Kv3.1b-ir neurons, PV expression reliably indicated GABA expression. Kv3.1b-ir, non-PV-ir neurons varied in SMI-32 coimmunoreactivity. The results suggest the existence of a heterogeneous population of excitatory neurons in macaque V1 with the potential for sustained high firing rates, and these neurons were particularly abundant in layers 4B and 4 Cα.
Collapse
Affiliation(s)
- Jenna G Kelly
- Center for Neural Science, New York University, New York, NY, USA
| | | | - Bernardo Rudy
- New York University Neuroscience Institute, New York University School of Medicine, Smilow Research Building Sixth Floor, 522 First Ave., New York, NY, USA
| | - Michael J Hawken
- Center for Neural Science, New York University, New York, NY, USA
| |
Collapse
|
32
|
Petanjek Z, Sedmak D, Džaja D, Hladnik A, Rašin MR, Jovanov-Milosevic N. The Protracted Maturation of Associative Layer IIIC Pyramidal Neurons in the Human Prefrontal Cortex During Childhood: A Major Role in Cognitive Development and Selective Alteration in Autism. Front Psychiatry 2019; 10:122. [PMID: 30923504 PMCID: PMC6426783 DOI: 10.3389/fpsyt.2019.00122] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 02/18/2019] [Indexed: 12/12/2022] Open
Abstract
The human specific cognitive shift starts around the age of 2 years with the onset of self-awareness, and continues with extraordinary increase in cognitive capacities during early childhood. Diffuse changes in functional connectivity in children aged 2-6 years indicate an increase in the capacity of cortical network. Interestingly, structural network complexity does not increase during this time and, thus, it is likely to be induced by selective maturation of a specific neuronal subclass. Here, we provide an overview of a subclass of cortico-cortical neurons, the associative layer IIIC pyramids of the human prefrontal cortex. Their local axonal collaterals are in control of the prefrontal cortico-cortical output, while their long projections modulate inter-areal processing. In this way, layer IIIC pyramids are the major integrative element of cortical processing, and changes in their connectivity patterns will affect global cortical functioning. Layer IIIC neurons have a unique pattern of dendritic maturation. In contrast to other classes of principal neurons, they undergo an additional phase of extensive dendritic growth during early childhood, and show characteristic molecular changes. Taken together, circuits associated with layer IIIC neurons have the most protracted period of developmental plasticity. This unique feature is advanced but also provides a window of opportunity for pathological events to disrupt normal formation of cognitive circuits involving layer IIIC neurons. In this manuscript, we discuss how disrupted dendritic and axonal maturation of layer IIIC neurons may lead into global cortical disconnectivity, affecting development of complex communication and social abilities. We also propose a model that developmentally dictated incorporation of layer IIIC neurons into maturing cortico-cortical circuits between 2 to 6 years will reveal a previous (perinatal) lesion affecting other classes of principal neurons. This "disclosure" of pre-existing functionally silent lesions of other neuronal classes induced by development of layer IIIC associative neurons, or their direct alteration, could be found in different forms of autism spectrum disorders. Understanding the gene-environment interaction in shaping cognitive microcircuitries may be fundamental for developing rehabilitation and prevention strategies in autism spectrum and other cognitive disorders.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Domagoj Džaja
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mladen Roko Rašin
- Department of Neuroscience and Cell Biology, Rutgers University, Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Nataša Jovanov-Milosevic
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Medical Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
33
|
Changes in Functional Properties of Rat Hippocampal Neurons Following Pentylenetetrazole-induced Status Epilepticus. Neuroscience 2018; 399:103-116. [PMID: 30593922 DOI: 10.1016/j.neuroscience.2018.12.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 11/13/2018] [Accepted: 12/17/2018] [Indexed: 12/28/2022]
Abstract
Pathophysiological remodeling processes following status epilepticus (SE) play a critical role in the pathophysiology of epilepsy but have not yet been not fully investigated. In the present study, we examined changes in intrinsic properties of pyramidal neurons, basal excitatory synaptic transmission, and short-term synaptic plasticity in hippocampal slices of rats after SE. Seizures were induced in 3-week-old rats by an intraperitoneal pentylenetetrazole (PTZ) injection. Only animals with generalized seizures lasting more than 30 min were included in the experiments. We found that CA1 pyramidal neurons became more excitable and started firing at a lower excitatory input due to a significant increase in input resistance. However, basal excitatory synaptic transmission was reduced in CA3-CA1 synapses, thus preventing the propagation of excitation through neural networks. A significant increase in paired-pulse facilitation 1 d after SE pointed to a decrease in the probability of glutamate release. Increased intrinsic excitability of neurons and decreased synaptic transmission differentially affected the excitability of a neural network. In terms of changes in seizure susceptibility after SE, we observed a significant increase in the maximal electroshock threshold 1 day after SE, suggesting a decrease in seizure susceptibility. However, after 1 week, there was no difference in seizure susceptibility between control and post-SE rats. The effects of SE on functional properties of hippocampal neurons were transient in the PTZ model, and most of them had recovered 1 week after SE. However, some minor alterations, such as smaller amplitude field potentials, were observed 1 month after SE.
Collapse
|
34
|
Jarovi J, Volle J, Yu X, Guan L, Takehara-Nishiuchi K. Prefrontal Theta Oscillations Promote Selective Encoding of Behaviorally Relevant Events. eNeuro 2018; 5:ENEURO.0407-18.2018. [PMID: 30693310 PMCID: PMC6348453 DOI: 10.1523/eneuro.0407-18.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 11/21/2022] Open
Abstract
The ability to capture the most relevant information from everyday experiences without constantly learning unimportant details is vital to survival and mental health. While decreased activity of the medial prefrontal cortex (mPFC) is associated with failed or inflexible encoding of relevant events in the hippocampus, mechanisms used by the mPFC to discern behavioral relevance of events are not clear. To address this question, we chemogenetically activated excitatory neurons in the mPFC of male rats and examined its impact on local network activity and differential associative learning dependent on the hippocampus. Rats were exposed to two neutral stimuli in two environments whose contingency with an aversive stimulus changed systematically across days. Over 2 weeks of differential and reversal learning, theta band activity began to ramp up toward the expected onset of the aversive stimulus, and this ramping activity tracked the subsequent shift of the set (stimulus modality to environment) predictive of the aversive stimulus. With chemogenetic mPFC activation, the ramping activity emerged within a few sessions of differential learning, which paralleled faster learning and stronger correlations between the ramping activity and conditioned responses. Chemogenetic mPFC activity, however, did not affect the adjustment of ramping activity or behavior during reversal learning or set-shifting, suggesting that the faster learning was not because of a general enhancement of attention, sensory, or motor processing. Thus, the dynamics of the mPFC network activation during events provide a relevance-signaling mechanism through which the mPFC exerts executive control over the encoding of those events in the hippocampus.
Collapse
Affiliation(s)
| | | | | | | | - Kaori Takehara-Nishiuchi
- Department of Cell and Systems Biology
- Department of Psychology
- Neuroscience Program, University of Toronto, Toronto M5S 3G3, Canada
| |
Collapse
|
35
|
Coppola JJ, Disney AA. Most calbindin-immunoreactive neurons, but few calretinin-immunoreactive neurons, express the m1 acetylcholine receptor in the middle temporal visual area of the macaque monkey. Brain Behav 2018; 8:e01071. [PMID: 30094962 PMCID: PMC6160643 DOI: 10.1002/brb3.1071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/10/2018] [Accepted: 04/15/2018] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Release of the neuromodulator acetylcholine into cortical circuits supports cognition, although its precise role and mechanisms of action are not well understood. Little is known about functional differences in cholinergic modulatory effects across cortical model systems, but anatomical evidence suggests that such differences likely exist because, for example, the expression of cholinergic receptors differs profoundly both within and between species. METHODS In the primary visual cortex (V1) of macaque monkeys, cholinergic receptors are strongly expressed by inhibitory interneurons. Using dual-immunofluorescence confocal microscopy, we examine m1 muscarinic acetylcholine receptor expression by two subclasses of inhibitory interneurons-identified by their expression of the calcium-binding proteins calbindin and calretinin-in the middle temporal extrastriate area (MT) of the macaque. RESULTS AND CONCLUSIONS We find that the majority of calbindin-immunoreactive neurons (55%) and only few calretinin-immunoreactive neurons (10%) express the m1 acetylcholine receptor. These results differ from the pattern observed in V1 of the same species, lending further support to the notion that cholinergic modulation in the cortex is tuned such that different cortical compartments will respond to acetylcholine release in different ways.
Collapse
Affiliation(s)
| | - Anita A. Disney
- Department of PsychologyVanderbilt UniversityNashvilleTennessee
| |
Collapse
|
36
|
Stedehouder J, Couey JJ, Brizee D, Hosseini B, Slotman JA, Dirven CMF, Shpak G, Houtsmuller AB, Kushner SA. Fast-spiking Parvalbumin Interneurons are Frequently Myelinated in the Cerebral Cortex of Mice and Humans. Cereb Cortex 2018; 27:5001-5013. [PMID: 28922832 DOI: 10.1093/cercor/bhx203] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Indexed: 12/29/2022] Open
Abstract
Myelination, the insulating ensheathment of axons by oligodendrocytes, is thought to both optimize signal propagation and provide metabolic support. Despite the well-established physiological importance of myelination to neuronal function, relatively little is known about the myelination of GABAergic interneurons in the cerebral cortex. Here, we report that a large fraction of myelin in mouse cerebral cortex ensheaths GABAergic interneurons, reaching up to 80% in hippocampal subregions. Moreover, we find that a very high proportion of neocortical and hippocampal parvalbumin (PV) interneurons exhibit axonal myelination. Using a combination of intracellular recordings and biocytin labeling of ex vivo human neocortex, we also confirm that axons of fast-spiking PV interneurons are extensively myelinated in the human brain. PV interneuron myelination in both mice and humans exhibits a stereotyped topography with a bias towards proximal axonal segments and relatively short internodes (~27 μm) interspersed with branch points. Interestingly, myelin-deficient Shiverer mice exhibit an increased density and more proximal location of en passant boutons, suggesting that myelination might function in part to regulate synapse formation along PV interneuron axons. Taken together, fast-spiking interneuron myelination is likely to have broad implications for cerebral cortex function in health and disease.
Collapse
Affiliation(s)
- J Stedehouder
- Department of Psychiatry, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - J J Couey
- Department of Psychiatry, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - D Brizee
- Department of Psychiatry, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - B Hosseini
- Department of Psychiatry, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - J A Slotman
- Erasmus Optical Imaging Centre, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.,Department of Pathology, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - C M F Dirven
- Department of Neurosurgery, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - G Shpak
- Department of Psychiatry, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - A B Houtsmuller
- Erasmus Optical Imaging Centre, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands.,Department of Pathology, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - S A Kushner
- Department of Psychiatry, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
37
|
Expression and localization of CB1R, NAPE-PLD, and FAAH in the vervet monkey nucleus accumbens. Sci Rep 2018; 8:8689. [PMID: 29875385 PMCID: PMC5989267 DOI: 10.1038/s41598-018-26826-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/15/2018] [Indexed: 11/14/2022] Open
Abstract
Extensive rodent literature suggests that the endocannabinoid (eCB) system present in the nucleus accumbens (NAc) modulates dopamine (DA) release in this area. However, expression patterns of the cannabinoid receptor type 1 (CB1R), the synthesizing enzyme N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD), and the degradation enzyme fatty acid amide hydrolase (FAAH) in the NAc have not yet been described in non-human primates. The goal of this study is therefore to characterize the expression and localization of the eCB system within the NAc of vervet monkeys (Chlorocebus sabaeus) using Western blots and immunohistochemistry. Results show that CB1R, NAPE-PLD, and FAAH are expressed across the NAc rostrocaudal axis, both in the core and shell. CB1R, NAPE-PLD, and FAAH are localized in medium spiny neurons (MSNs) and fast-spiking GABAergic interneurons (FSIs). Dopaminergic projections and astrocytes did not express CB1R, NAPE-PLD, or FAAH. These data show that the eCB system is present in the vervet monkey NAc and supports its role in the primate brain reward circuit.
Collapse
|
38
|
Psychiatry in a Dish: Stem Cells and Brain Organoids Modeling Autism Spectrum Disorders. Biol Psychiatry 2018; 83:558-568. [PMID: 29295738 DOI: 10.1016/j.biopsych.2017.11.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 12/23/2022]
Abstract
Autism spectrum disorders are a group of pervasive neurodevelopmental conditions with heterogeneous etiology, characterized by deficits in social cognition, communication, and behavioral flexibility. Despite an increasing scientific effort to find the pathophysiological explanations for the disease, the neurobiological links remain unclear. A large amount of evidence suggests that pathological processes taking place in early embryonic neurodevelopment might be responsible for later manifestation of autistic symptoms. This dysfunctional development includes altered maturation/differentiation processes, disturbances in cell-cell communication, and an unbalanced ratio between certain neuronal populations. All those processes are highly dependent on the interconnectivity and three-dimensional organizations of the brain. Moreover, in order to gain a deeper understanding of the complex neurobiology of autism spectrum disorders, valid disease models are pivotal. Induced pluripotent stem cells could potentially help to elucidate the complex mechanisms of the disease and lead to the development of more effective individualized treatment. The induced pluripotent stem cells approach allows comparison between the development of various cellular phenotypes generated from cell lines of patients and healthy individuals. A newly advanced organoid technology makes it possible to create three-dimensional in vitro models of brain development and structural interconnectivity, based on induced pluripotent stem cells derived from the respective individuals. The biggest challenge for modeling psychiatric diseases in vitro is finding and establishing the link between cellular and molecular findings with the clinical symptoms, and this review aims to give an overview over the feasibility and applicability of this new tissue engineering tool in psychiatry.
Collapse
|
39
|
Manning EE, Ahmari SE. How can preclinical mouse models be used to gain insight into prefrontal cortex dysfunction in obsessive-compulsive disorder? Brain Neurosci Adv 2018; 2:2398212818783896. [PMID: 32166143 PMCID: PMC7058260 DOI: 10.1177/2398212818783896] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/18/2018] [Indexed: 01/09/2023] Open
Abstract
Obsessive-compulsive disorder is a debilitating psychiatric disorder that is characterised by perseverative thoughts and behaviours. Cognitive and affective disturbances play a central role in this illness, and it is therefore not surprising that clinical neuroimaging studies have demonstrated widespread alterations in prefrontal cortex functioning in patients. Preclinical mouse experimental systems provide the opportunity to gain mechanistic insight into the neurobiological changes underlying prefrontal cortex dysfunction through new technologies that allow measurement and manipulation of activity in discrete neural populations in awake, behaving mice. However, recent preclinical research has focused on striatal dysfunction, and has therefore provided relatively little insight regarding the role of the prefrontal cortex in obsessive-compulsive disorder-relevant behaviours. Here, we will discuss a number of translational prefrontal cortex-dependent paradigms, including obsessive-compulsive disorder-relevant tasks that produce compulsive responding, and how they can be leveraged in this context. Drawing on recent examples that have led to mechanistic insight about specific genes, cell types and circuits that mediate prefrontal cortex contributions to distinct aspects of cognition, we will provide a framework for applying similar strategies to identify neural mechanisms underlying obsessive-compulsive disorder-relevant behavioural domains. We propose that research using clinically relevant paradigms will accelerate translation of findings from preclinical mouse models, thus supporting the development of novel therapeutics targeted to specific pathophysiological mechanisms.
Collapse
Affiliation(s)
| | - Susanne E. Ahmari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
40
|
Rich S, Zochowski M, Booth V. Dichotomous Dynamics in E-I Networks with Strongly and Weakly Intra-connected Inhibitory Neurons. Front Neural Circuits 2017; 11:104. [PMID: 29326558 PMCID: PMC5733501 DOI: 10.3389/fncir.2017.00104] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/04/2017] [Indexed: 11/13/2022] Open
Abstract
The interconnectivity between excitatory and inhibitory neural networks informs mechanisms by which rhythmic bursts of excitatory activity can be produced in the brain. One such mechanism, Pyramidal Interneuron Network Gamma (PING), relies primarily upon reciprocal connectivity between the excitatory and inhibitory networks, while also including intra-connectivity of inhibitory cells. The causal relationship between excitatory activity and the subsequent burst of inhibitory activity is of paramount importance to the mechanism and has been well studied. However, the role of the intra-connectivity of the inhibitory network, while important for PING, has not been studied in detail, as most analyses of PING simply assume that inhibitory intra-connectivity is strong enough to suppress subsequent firing following the initial inhibitory burst. In this paper we investigate the role that the strength of inhibitory intra-connectivity plays in determining the dynamics of PING-style networks. We show that networks with weak inhibitory intra-connectivity exhibit variations in burst dynamics of both the excitatory and inhibitory cells that are not obtained with strong inhibitory intra-connectivity. Networks with weak inhibitory intra-connectivity exhibit excitatory rhythmic bursts with weak excitatory-to-inhibitory synapses for which classical PING networks would show no rhythmic activity. Additionally, variations in dynamics of these networks as the excitatory-to-inhibitory synaptic weight increases illustrates the important role that consistent pattern formation in the inhibitory cells serves in maintaining organized and periodic excitatory bursts. Finally, motivated by these results and the known diversity of interneurons, we show that a PING-style network with two inhibitory subnetworks, one strongly intra-connected and one weakly intra-connected, exhibits organized and periodic excitatory activity over a larger parameter regime than networks with a homogeneous inhibitory population. Taken together, these results serve to better articulate the role of inhibitory intra-connectivity in generating PING-like rhythms, while also revealing how heterogeneity amongst inhibitory synapses might make such rhythms more robust to a variety of network parameters.
Collapse
Affiliation(s)
- Scott Rich
- Applied and Interdisciplinary Mathematics, University of Michigan, Ann Arbor, MI, United States
| | - Michal Zochowski
- Department of Physics and Biophysics, University of Michigan, Ann Arbor, MI, United States
| | - Victoria Booth
- Department of Mathematics and Anesthesiology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
41
|
McGregor C, Riordan A, Thornton J. Estrogens and the cognitive symptoms of schizophrenia: Possible neuroprotective mechanisms. Front Neuroendocrinol 2017; 47:19-33. [PMID: 28673758 DOI: 10.1016/j.yfrne.2017.06.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 06/25/2017] [Accepted: 06/27/2017] [Indexed: 02/07/2023]
Abstract
Schizophrenia is a complex neuropsychiatric illness with marked sex differences. Women have later onset and lesser symptoms, which has led to the hypothesis that estrogens are protective in schizophrenia. Cognitive dysfunction is a hallmark of the disease and the symptom most correlated with functional outcome. Here we describe a number of mechanisms by which estrogens may be therapeutic in schizophrenia, with a focus on cognitive symptoms. We review the relationship between estrogens and brain derived neurotrophic factor, neuroinflammation, NMDA receptors, GABA receptors, and luteinizing hormone. Exploring these pathways may enable novel treatments for schizophrenia and a greater understanding of this devastating disease.
Collapse
Affiliation(s)
- Claire McGregor
- Department of Neuroscience, Oberlin College, 119 Woodland St, Oberlin, OH 44074, USA.
| | - Alexander Riordan
- Department of Neuroscience, Oberlin College, 119 Woodland St, Oberlin, OH 44074, USA
| | - Janice Thornton
- Department of Neuroscience, Oberlin College, 119 Woodland St, Oberlin, OH 44074, USA
| |
Collapse
|
42
|
Abstract
Cortical networks are composed of glutamatergic excitatory projection neurons and local GABAergic inhibitory interneurons that gate signal flow and sculpt network dynamics. Although they represent a minority of the total neocortical neuronal population, GABAergic interneurons are highly heterogeneous, forming functional classes based on their morphological, electrophysiological, and molecular features, as well as connectivity and in vivo patterns of activity. Here we review our current understanding of neocortical interneuron diversity and the properties that distinguish cell types. We then discuss how the involvement of multiple cell types, each with a specific set of cellular properties, plays a crucial role in diversifying and increasing the computational power of a relatively small number of simple circuit motifs forming cortical networks. We illustrate how recent advances in the field have shed light onto the mechanisms by which GABAergic inhibition contributes to network operations.
Collapse
|
43
|
Wang B, Ke W, Guang J, Chen G, Yin L, Deng S, He Q, Liu Y, He T, Zheng R, Jiang Y, Zhang X, Li T, Luan G, Lu HD, Zhang M, Zhang X, Shu Y. Firing Frequency Maxima of Fast-Spiking Neurons in Human, Monkey, and Mouse Neocortex. Front Cell Neurosci 2016; 10:239. [PMID: 27803650 PMCID: PMC5067378 DOI: 10.3389/fncel.2016.00239] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/30/2016] [Indexed: 12/13/2022] Open
Abstract
Cortical fast-spiking (FS) neurons generate high-frequency action potentials (APs) without apparent frequency accommodation, thus providing fast and precise inhibition. However, the maximal firing frequency that they can reach, particularly in primate neocortex, remains unclear. Here, by recording in human, monkey, and mouse neocortical slices, we revealed that FS neurons in human association cortices (mostly temporal) could generate APs at a maximal mean frequency (Fmean) of 338 Hz and a maximal instantaneous frequency (Finst) of 453 Hz, and they increase with age. The maximal firing frequency of FS neurons in the association cortices (frontal and temporal) of monkey was even higher (Fmean 450 Hz, Finst 611 Hz), whereas in the association cortex (entorhinal) of mouse it was much lower (Fmean 215 Hz, Finst 342 Hz). Moreover, FS neurons in mouse primary visual cortex (V1) could fire at higher frequencies (Fmean 415 Hz, Finst 582 Hz) than those in association cortex. We further validated our in vitro data by examining spikes of putative FS neurons in behaving monkey and mouse. Together, our results demonstrate that the maximal firing frequency of FS neurons varies between species and cortical areas.
Collapse
Affiliation(s)
- Bo Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal UniversityBeijing, China; Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and University of Chinese Academy of SciencesShanghai, China
| | - Wei Ke
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Jing Guang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and University of Chinese Academy of Sciences Shanghai, China
| | - Guang Chen
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and University of Chinese Academy of Sciences Shanghai, China
| | - Luping Yin
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and University of Chinese Academy of Sciences Shanghai, China
| | - Suixin Deng
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Quansheng He
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Yaping Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Ting He
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Rui Zheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and University of Chinese Academy of Sciences Shanghai, China
| | - Yanbo Jiang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and University of Chinese Academy of Sciences Shanghai, China
| | - Xiaoxue Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Tianfu Li
- Department of Neurosurgery, Brain Institute, and Department of Neurology, Epilepsy Center, Beijing Sanbo Brain Hospital, Capital Medical University Beijing, China
| | - Guoming Luan
- Department of Neurosurgery, Brain Institute, and Department of Neurology, Epilepsy Center, Beijing Sanbo Brain Hospital, Capital Medical University Beijing, China
| | - Haidong D Lu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Mingsha Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| | - Yousheng Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, The Collaborative Innovation Center for Brain Science, Beijing Normal University Beijing, China
| |
Collapse
|
44
|
Hawasli AH, Kim D, Ledbetter NM, Dahiya S, Barbour DL, Leuthardt EC. Influence of White and Gray Matter Connections on Endogenous Human Cortical Oscillations. Front Hum Neurosci 2016; 10:330. [PMID: 27445767 PMCID: PMC4923146 DOI: 10.3389/fnhum.2016.00330] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/15/2016] [Indexed: 12/03/2022] Open
Abstract
Brain oscillations reflect changes in electrical potentials summated across neuronal populations. Low- and high-frequency rhythms have different modulation patterns. Slower rhythms are spatially broad, while faster rhythms are more local. From this observation, we hypothesized that low- and high-frequency oscillations reflect white- and gray-matter communications, respectively, and synchronization between low-frequency phase with high-frequency amplitude represents a mechanism enabling distributed brain-networks to coordinate local processing. Testing this common understanding, we selectively disrupted white or gray matter connections to human cortex while recording surface field potentials. Counter to our original hypotheses, we found that cortex consists of independent oscillatory-units (IOUs) that maintain their own complex endogenous rhythm structure. IOUs are differentially modulated by white and gray matter connections. White-matter connections maintain topographical anatomic heterogeneity (i.e., separable processing in cortical space) and gray-matter connections segregate cortical synchronization patterns (i.e., separable temporal processing through phase-power coupling). Modulation of distinct oscillatory modules enables the functional diversity necessary for complex processing in the human brain.
Collapse
Affiliation(s)
- Ammar H Hawasli
- Department of Neurological Surgery, Washington University School of Medicine Saint Louis, MO, USA
| | - DoHyun Kim
- Department of Biomedical Engineering, Washington University School of Medicine Saint Louis, MO, USA
| | - Noah M Ledbetter
- Department of Biomedical Engineering, Washington University School of Medicine Saint Louis, MO, USA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine Saint Louis, MO, USA
| | - Dennis L Barbour
- Department of Biomedical Engineering, Washington University School of Medicine Saint Louis, MO, USA
| | - Eric C Leuthardt
- Department of Neurological Surgery, Washington University School of MedicineSaint Louis, MO, USA; Department of Biomedical Engineering, Washington University School of MedicineSaint Louis, MO, USA
| |
Collapse
|
45
|
GABAergic Function as a Limiting Factor for Prefrontal Maturation during Adolescence. Trends Neurosci 2016; 39:441-448. [PMID: 27233681 DOI: 10.1016/j.tins.2016.04.010] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/22/2016] [Accepted: 04/25/2016] [Indexed: 11/24/2022]
Abstract
Adolescence is a vulnerable period for the onset of mental illnesses including schizophrenia and affective disorders, yet the neurodevelopmental processes underlying this vulnerability remain poorly understood. The prefrontal cortex (PFC) and its local GABAergic system are thought to contribute to the core of cognitive deficits associated with such disorders. However, clinical and preclinical end-point analyses performed in adults are likely to give limited insight into the cellular mechanisms that are altered during adolescence but are only manifested in adulthood. This perspective summarizes work regarding the developmental trajectories of the GABAergic system in the PFC during adolescence to provide an insight into the increased susceptibility to psychiatric disorders during this critical developmental period.
Collapse
|
46
|
Giustino TF, Maren S. The Role of the Medial Prefrontal Cortex in the Conditioning and Extinction of Fear. Front Behav Neurosci 2015; 9:298. [PMID: 26617500 PMCID: PMC4637424 DOI: 10.3389/fnbeh.2015.00298] [Citation(s) in RCA: 376] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022] Open
Abstract
Once acquired, a fearful memory can persist for a lifetime. Although learned fear can be extinguished, extinction memories are fragile. The resilience of fear memories to extinction may contribute to the maintenance of disorders of fear and anxiety, including post-traumatic stress disorder (PTSD). As such, considerable effort has been placed on understanding the neural circuitry underlying the acquisition, expression, and extinction of emotional memories in rodent models as well as in humans. A triad of brain regions, including the prefrontal cortex, hippocampus, and amygdala, form an essential brain circuit involved in fear conditioning and extinction. Within this circuit, the prefrontal cortex is thought to exert top-down control over subcortical structures to regulate appropriate behavioral responses. Importantly, a division of labor has been proposed in which the prelimbic (PL) and infralimbic (IL) subdivisions of the medial prefrontal cortex (mPFC) regulate the expression and suppression of fear in rodents, respectively. Here, we critically review the anatomical and physiological evidence that has led to this proposed dichotomy of function within mPFC. We propose that under some conditions, the PL and IL act in concert, exhibiting similar patterns of neural activity in response to aversive conditioned stimuli and during the expression or inhibition of conditioned fear. This may stem from common synaptic inputs, parallel downstream outputs, or cortico-cortical interactions. Despite this functional covariation, these mPFC subdivisions may still be coding for largely opposing behavioral outcomes, with PL biased towards fear expression and IL towards suppression.
Collapse
Affiliation(s)
- Thomas F Giustino
- Department of Psychology and Institute for Neuroscience, Texas A&M University College Station, TX, USA
| | - Stephen Maren
- Department of Psychology and Institute for Neuroscience, Texas A&M University College Station, TX, USA
| |
Collapse
|
47
|
Malkin S, Kim K, Tikhonov D, Magazanik L, Zaitsev A. Statistical models suggest presence of two distinct subpopulations of miniature EPSCs in fast-spiking interneurons of rat prefrontal cortex. Neuroscience 2015; 301:508-19. [DOI: 10.1016/j.neuroscience.2015.06.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/18/2015] [Accepted: 06/18/2015] [Indexed: 01/30/2023]
|
48
|
Glykos V, Whittington MA, LeBeau FEN. Subregional differences in the generation of fast network oscillations in the rat medial prefrontal cortex (mPFC) in vitro. J Physiol 2015; 593:3597-615. [PMID: 26041504 PMCID: PMC4560586 DOI: 10.1113/jp270811] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Fast network oscillations in the beta (20-30 Hz) frequency range can be evoked with combined activation of muscarinic and kainate receptors in different subregions of the medial prefrontal cortex (mPFC). Subregional differences were observed as the oscillations in the dorsal prelimbic cortex (PrL) were smaller in magnitude than those in the ventral dorsopeduncular (DP) region, and these differences persisted in trimmed slices containing only PrL and DP regions. Oscillations in both regions were dependent upon GABAA and AMPA receptor activation but NMDA receptor blockade decreased oscillations only in the DP region. Subregional differences in neuronal properties of the presumed pyramidal cells were found between PrL and DP, with many more cells in DP firing rhythmically compared to the PrL region. Presumed inhibitory synaptic potentials (IPSPs) recorded from principal cells were more rhythmic and coherent, and significantly larger in amplitude, in the DP region; the data suggest that variation in the patterns of activity between subregions may reflect distinct functional roles. ABSTRACT Fast network oscillations in the beta (20-30 Hz) and low gamma (30-80 Hz) range underlie higher cognitive functions associated with the medial prefrontal cortex (mPFC) including attention and working memory. Using a combination of kainate (KA, 200 nm) and the cholinergic agonist carbachol (Cb, 10 μm) fast network oscillations, in the beta frequency range, were evoked in the rat mPFC in vitro. Oscillations were elicited in the prelimbic (PrL), infralimbic (IL) and the dorsopeduncular (DP) cortex, with the largest oscillations observed in DP cortex. Oscillations in both the PrL and DP were dependent, with slightly different sensitivities, on γ-aminobutyric acid (GABA)A , α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and kainate receptors, but only oscillations in the DP were significantly reduced by N-methyl-d-aspartate (NMDA) receptor blockade. Intracellular recordings showed that 9/20 regular spiking (RS) cells in the PrL exhibited a notable cAMP-dependent hyperpolarisation activated current (Ih ) in contrast to 16/17 in the DP cortex. Extracellular single unit recordings showed that the majority of cells in the PrL, and DP regions had interspike firing frequencies (IFFs) at beta (20-30 Hz) frequencies and fired at the peak negativity of the field oscillation. Recordings in DP revealed presumed inhibitory postsynaptic potentials (IPSPs) that were larger in amplitude and more rhythmic than those in the PrL region. Our data suggest that each PFC subregion may be capable of generating distinct patterns of network activity with different cell types involved. Variation in the properties of oscillations evoked in the PrL and DP probably reflects the distinct functional roles of these different PFC regions.
Collapse
Affiliation(s)
- Vasileios Glykos
- Institute of Neuroscience, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Miles A Whittington
- York-Hull Medical School, F1- Department of Biology, York University, Heslington, YO10 5DD, UK
| | - Fiona E N LeBeau
- Institute of Neuroscience, Newcastle University, Medical School, Framlington Place, Newcastle-upon-Tyne, NE2 4HH, UK
| |
Collapse
|
49
|
Tse MT, Piantadosi PT, Floresco SB. Prefrontal cortical gamma-aminobutyric acid transmission and cognitive function: drawing links to schizophrenia from preclinical research. Biol Psychiatry 2015; 77:929-39. [PMID: 25442792 DOI: 10.1016/j.biopsych.2014.09.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 08/22/2014] [Accepted: 09/15/2014] [Indexed: 12/28/2022]
Abstract
Cognitive dysfunction in schizophrenia is one of the most pervasive and debilitating aspects of the disorder. Among the numerous neural abnormalities that may contribute to schizophrenia symptoms, perturbations in markers for the inhibitory neurotransmitter gamma-aminobutyric acid (GABA), particularly within the frontal lobes, are some of the most reliable alterations observed at postmortem examination. However, how prefrontal GABA dysfunction contributes to cognitive impairment in schizophrenia remains unclear. We provide an overview of postmortem GABAergic perturbations in the brain affected by schizophrenia and describe circumstantial evidence linking these alterations to cognitive dysfunction. In addition, we conduct a survey of studies using neurodevelopmental, genetic, and pharmacologic rodent models that induce schizophrenia-like cognitive impairments, highlighting the convergence of these mechanistically distinct approaches to prefrontal GABAergic disruption. We review preclinical studies that have directly targeted prefrontal cortical GABAergic transmission using local application of GABAA receptor antagonists. These studies have provided an important link between GABA transmission and cognitive dysfunction in schizophrenia because they show that reducing prefrontal inhibitory transmission induces various cognitive, emotional, and dopaminergic abnormalities that resemble aspects of the disorder. These converging clinical and preclinical findings provide strong support for the idea that perturbations in GABA signaling drive certain forms of cognitive dysfunction in schizophrenia. Future studies using this approach will yield information to refine further a putative "GABA hypothesis" of schizophrenia.
Collapse
Affiliation(s)
- Maric T Tse
- Department of Psychology and Brain Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Patrick T Piantadosi
- Department of Psychology and Brain Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stan B Floresco
- Department of Psychology and Brain Research Centre, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
50
|
Di Miceli M, Gronier B. Psychostimulants and atomoxetine alter the electrophysiological activity of prefrontal cortex neurons, interaction with catecholamine and glutamate NMDA receptors. Psychopharmacology (Berl) 2015; 232:2191-205. [PMID: 25572531 DOI: 10.1007/s00213-014-3849-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/12/2014] [Indexed: 12/29/2022]
Abstract
RATIONALE Attention-deficit hyperactivity disorder (ADHD) is the most frequently diagnosed neuropsychiatric disorder in childhood. Currently available ADHD drugs include the psychostimulants methylphenidate (MPH) and D-amphetamine (D-AMP), acting on norepinephrine and dopamine transporters/release, and atomoxetine (ATX), a selective norepinephrine uptake inhibitor. Recent evidence suggests an involvement of glutamate neurotransmission in the pathology and treatment of ADHD, via mechanisms to be clarified. OBJECTIVE We have investigated how ADHD drugs could modulate, through interaction with catecholamine receptors, basal and glutamate-induced excitability of pyramidal neurons in the prefrontal cortex (PFC), a region which plays a major role in control of attention and impulsivity. METHODS We have used the technique of extracellular single-unit recording in anaesthetised rats coupled with microiontophoresis. RESULTS Both MPH (1-3 mg/kg) and D-AMP (1-9 mg/kg) increased the firing activity of PFC neurons in a dopamine D1 receptor-dependent manner. ATX administration (1-6 mg/kg) also increased the firing of neurons, but this effect is not significantly reversed by D1 (SCH 23390) or alpha1 (prazosin) receptor antagonists but potentiated by alpha2 antagonist (yohimbine). All drugs induced a clear potentiation of the excitatory response of PFC neurons to the microiontophoretic application of the glutamate agonist N-methyl-D-aspartate (NMDA), but not to the glutamate agonist α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA). The potentiating effect of D-AMP on NMDA-induced activation of PFC neurons was partially reversed or prevented by dopamine D1 receptor blockade. CONCLUSION Our data shows that increase in excitability of PFC neurons in basal conditions and via NMDA receptor activation may be involved in the therapeutic response to ADHD drugs.
Collapse
Affiliation(s)
- Mathieu Di Miceli
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, LE1 9BH, UK
| | | |
Collapse
|