1
|
Oldre EN, Webb BD, Sperringer JE, Maness PF. Regulation of perisomatic synapses from cholecystokinin basket interneurons through NrCAM and Ankyrin B. CURRENT RESEARCH IN NEUROBIOLOGY 2025; 8:100150. [PMID: 40276719 PMCID: PMC12018208 DOI: 10.1016/j.crneur.2025.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/07/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
The perisomatic region of cortical pyramidal neurons (PNs) integrates local and long-range inputs and regulates firing. This domain receives GABAergic inputs from cholecystokinin (CCK)- and Parvalbumin (PV)-expressing basket cells (BCs) but how synaptic contacts are established is unclear. Neuron-glial related cell adhesion molecule (NrCAM) is a homophilic transmembrane protein that binds the scaffold protein Ankyrin B. Here we show that NrCAM and Ankyrin B mediate perisomatic synaptic contact between CCK-BCs and PNs in mouse medial prefrontal cortex (mPFC). Immunolabeling of CCK-BC terminals for vesicular glutamate transporter-3 (VGLUT3) or vesicular GABA transporter (VGAT) revealed a significant decrease in CCK-BC synaptic puncta on PN soma in NrCAM-null mice, however no decrease in PV-BC puncta or cell loss. VGLUT3+ CCK-BC puncta were also decreased by Ankyrin B deletion from PNs in Nex1Cre-ERT2:Ank2flox/flox:EGFP mice. A novel CCK-BC reporter mouse expressing tdTomato (tdT) at the Synuclein-γ (Sncg) locus showed NrCAM localized to Sncg + CCK-BCs, and to postsynaptic PN soma in Nex1Cre-ERT2:Ank2+/+:EGFP mice. Results suggest that NrCAM and Ankyrin B contribute to the establishment of connectivity between CCK-BCs and excitatory neurons of the mPFC.
Collapse
Affiliation(s)
- Erik N. Oldre
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Barrett D. Webb
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Justin E. Sperringer
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Patricia F. Maness
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
2
|
Jézéquel J, Condomitti G, Kroon T, Hamid F, Sanalidou S, Garcés T, Maeso P, Balia M, Hu Z, Sahara S, Rico B. Cadherins orchestrate specific patterns of perisomatic inhibition onto distinct pyramidal cell populations. Nat Commun 2025; 16:4481. [PMID: 40368888 PMCID: PMC12078473 DOI: 10.1038/s41467-025-59635-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
GABAergic interneurons were thought to regulate excitatory networks by establishing unselective connections onto diverse pyramidal cell populations, but recent studies demonstrate the existence of a cell type-specific inhibitory connectome. How and when interneurons establish precise connectivity patterns among intermingled populations of excitatory neurons remains enigmatic. We explore the molecular mechanisms orchestrating the emergence of cell type-specific inhibition in the mouse cerebral cortex. We demonstrate that layer 5 intra- (L5 IT) and extra-telencephalic (L5 ET) neurons express unique transcriptional programs, allowing them to shape parvalbumin- (PV+) and cholecystokinin-positive (CCK+) interneuron wiring. We identified Cdh12 and Cdh13, two cadherin superfamily members, as underpinnings of cell type- and input-specific inhibitory patterns of L5 pyramidal cell populations. Multiplex monosynaptic tracing revealed a minimal overlap between IT and ET presynaptic inhibitory networks and suggests that different PV+ basket cell populations innervate distinct L5 pyramidal cell types. Here, we unravel the contribution of cadherins in shaping cell-type-specific cortical interneuron wiring.
Collapse
Affiliation(s)
- Julie Jézéquel
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Giuseppe Condomitti
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Tim Kroon
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Fursham Hamid
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Stella Sanalidou
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Teresa Garcés
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Patricia Maeso
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Maddalena Balia
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Zhaohui Hu
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Setsuko Sahara
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Beatriz Rico
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom.
| |
Collapse
|
3
|
Malhotra S, Donneger F, Farrell JS, Dudok B, Losonczy A, Soltesz I. Integrating endocannabinoid signaling, CCK interneurons, and hippocampal circuit dynamics in behaving animals. Neuron 2025:S0896-6273(25)00188-6. [PMID: 40267911 DOI: 10.1016/j.neuron.2025.03.016] [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: 07/12/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/25/2025]
Abstract
The brain's endocannabinoid signaling system modulates a diverse range of physiological phenomena and is also involved in various psychiatric and neurological disorders. The basic components of the molecular machinery underlying endocannabinoid-mediated synaptic signaling have been known for decades. However, limitations associated with the short-lived nature of endocannabinoid lipid signals had made it challenging to determine the spatiotemporal specificity and dynamics of endocannabinoid signaling in vivo. Here, we discuss how novel technologies have recently enabled unprecedented insights into endocannabinoid signaling taking place at specific synapses in behaving animals. In this review, we primarily focus on cannabinoid-sensitive inhibition in the hippocampus in relation to place cell properties to illustrate the potential of these novel methodologies. In addition, we highlight implications of these approaches and insights for the unraveling of cannabinoid regulation of synapses in vivo in other brain circuits in both health and disease.
Collapse
Affiliation(s)
- Shreya Malhotra
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
| | - Florian Donneger
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Jordan S Farrell
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Center, Harvard Medical School, Boston, MA, USA
| | - Barna Dudok
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY, USA; Kavli Institute for Brain Sciences, Columbia University, New York, NY, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
4
|
Soto C, Orihuela L, Apostol G, Vivar C. Running Reverses Chronic Stress-Induced Changes in Serotonergic Modulation of Hippocampal Granule Cells and Altered Behavioural Responses. Eur J Neurosci 2025; 61:e70084. [PMID: 40165404 PMCID: PMC11959172 DOI: 10.1111/ejn.70084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/14/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
Chronic stress increases susceptibility to anxiety and depression disorders, recurrent and common psychiatric conditions. Current antidepressant medications have varying degrees of efficacy and often have multiple side effects limiting treatment adherence. Physical exercise has beneficial effects on stress-related mental disorders. However, the underlying mechanisms are unclear. Dentate gyrus granule cells (GCs) excitability may mediate stress resilience. Here, we expose young adult C57Bl6 mice to chronic restraint stress (CRS) for 14 days followed by 30 days of running treatment. Behavioural evaluation before and after treatment showed that the behavioural alterations elicited by CRS were mitigated by running. Next, we evaluated serotonergic modulation of GC excitability, as a potential mechanism underlying running-induced stress resilience. Electrophysiological recordings indicate that CRS alters serotonergic modulation of GC excitability. Utilising (S)-WAY 100135 and Tropisetron, antagonists of 5-HT1A and 5-HT3 receptors respectively, we show that running recovers 5-HT1A receptor activity lost by CRS. Additionally, running promotes the indirect modulation of GCs through 5-HT3 receptor activation. Thus, 5-HT1A and 5-HT3 receptors may be potential targets for the treatment of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Carmen Soto
- Laboratory of Neurogenesis and Neuroplasticity. Department of Physiology, Biophysics and NeuroscienceCentro de Investigacion y de Estudios Avanzados del Instituto Politécnico NacionalMexico CityMexico
| | - Lazaro P. Orihuela
- Laboratory of Neurogenesis and Neuroplasticity. Department of Physiology, Biophysics and NeuroscienceCentro de Investigacion y de Estudios Avanzados del Instituto Politécnico NacionalMexico CityMexico
| | - Grego Apostol
- Laboratory of Neurogenesis and Neuroplasticity. Department of Physiology, Biophysics and NeuroscienceCentro de Investigacion y de Estudios Avanzados del Instituto Politécnico NacionalMexico CityMexico
| | - Carmen Vivar
- Laboratory of Neurogenesis and Neuroplasticity. Department of Physiology, Biophysics and NeuroscienceCentro de Investigacion y de Estudios Avanzados del Instituto Politécnico NacionalMexico CityMexico
| |
Collapse
|
5
|
Caccavano AP, Vlachos A, McLean N, Kimmel S, Kim JH, Vargish G, Mahadevan V, Hewitt L, Rossi AM, Spineux I, Wu SJ, Furlanis E, Dai M, Leyva Garcia B, Wang Y, Chittajallu R, London E, Yuan X, Hunt S, Abebe D, Eldridge MAG, Cummins AC, Hines BE, Plotnikova A, Mohanty A, Averbeck BB, Zaghloul KA, Dimidschstein J, Fishell G, Pelkey KA, McBain CJ. Divergent opioid-mediated suppression of inhibition between hippocampus and neocortex across species and development. Neuron 2025:S0896-6273(25)00177-1. [PMID: 40147437 DOI: 10.1016/j.neuron.2025.03.005] [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: 01/22/2024] [Revised: 11/01/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
Within adult rodent hippocampus (HPC), opioids suppress inhibitory parvalbumin-expressing interneurons (PV-INs), disinhibiting local microcircuits. However, it is unknown whether this disinhibitory motif is conserved across cortical regions, species, or development. We observed that PV-IN-mediated inhibition is robustly suppressed by opioids in HPC proper but not primary neocortex in mice and non-human primates, with spontaneous inhibitory tone in resected human tissue also following a consistent dichotomy. This hippocampal disinhibitory motif is established in early development when PV-INs and opioids regulate early population activity. Morphine pretreatment partially occludes this acute opioid-mediated suppression, with implications for the effects of opioids on hippocampal network activity important for learning and memory. Our findings demonstrate that PV-INs exhibit divergent opioid sensitivity across brain regions, which is remarkably conserved over evolution, and highlight the underappreciated role of opioids acting through immature PV-INs in shaping hippocampal development.
Collapse
Affiliation(s)
- Adam P Caccavano
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Anna Vlachos
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nadiya McLean
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sarah Kimmel
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - June Hoan Kim
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Geoffrey Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Vivek Mahadevan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Lauren Hewitt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anthony M Rossi
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ilona Spineux
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sherry Jingjing Wu
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elisabetta Furlanis
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Min Dai
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brenda Leyva Garcia
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yating Wang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Edra London
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Daniel Abebe
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mark A G Eldridge
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Alex C Cummins
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Brendan E Hines
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Anya Plotnikova
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Arya Mohanty
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Bruno B Averbeck
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Kareem A Zaghloul
- National Institute of Neurological Disorders and Stroke (NINDS) Intramural Research Program, NIH, Bethesda, MD 20892, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Oldre EN, Webb BD, Sperringer JE, Maness PF. Regulation of Perisomatic Synapses from Cholecystokinin Basket Interneurons through NrCAM and Ankyrin B. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.04.621872. [PMID: 39574611 PMCID: PMC11580885 DOI: 10.1101/2024.11.04.621872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The perisomatic region of cortical pyramidal neurons (PNs) integrates local and long-range inputs and regulates firing. This domain receives GABAergic inputs from cholecystokinin (CCK)- and Parvalbumin (PV)-expressing basket cells (BCs) but how synaptic contacts are established is unclear. Neuron-glial related cell adhesion molecule (NrCAM) is a homophilic transmembrane protein that binds the scaffold protein Ankyrin B. Here we show that NrCAM and Ankyrin B mediate perisomatic synaptic contact between CCK-BCs and PNs in mouse medial prefrontal cortex (mPFC). Immunolabeling of CCK-BC terminals for vesicular glutamate transporter-3 (VGLUT3) or vesicular GABA transporter (VGAT) revealed a significant decrease in CCK-BC synaptic puncta on PN soma in NrCAM-null mice, however no decrease in PV-BC puncta or cell loss. VGLUT3+ CCK-BC puncta were also decreased by Ankyrin B deletion from PNs in Nex1Cre-ERT2:Ank2 flox/flox :EGFP mice. A novel CCK-BC reporter mouse expressing tdTomato (tdT) at the Synuclein-γ ( Sncg ) locus showed NrCAM localized to Sncg+ CCK-BCs, and to postsynaptic PN soma in Nex1Cre-ERT2:Ank2 +/+ :EGFP mice. Results suggest that NrCAM and Ankyrin B contribute to the establishment of connectivity between CCK-BCs and excitatory neurons of the mPFC.
Collapse
|
7
|
Caccavano AP, Vlachos A, McLean N, Kimmel S, Kim JH, Vargish G, Mahadevan V, Hewitt L, Rossi AM, Spineux I, Wu SJ, Furlanis E, Dai M, Garcia BL, Wang Y, Chittajallu R, London E, Yuan X, Hunt S, Abebe D, Eldridge MAG, Cummins AC, Hines BE, Plotnikova A, Mohanty A, Averbeck BB, Zaghloul K, Dimidschstein J, Fishell G, Pelkey KA, McBain CJ. Divergent opioid-mediated suppression of inhibition between hippocampus and neocortex across species and development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.20.576455. [PMID: 38313283 PMCID: PMC10836073 DOI: 10.1101/2024.01.20.576455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Within the adult rodent hippocampus, opioids suppress inhibitory parvalbumin-expressing interneurons (PV-INs), thus disinhibiting local micro-circuits. However, it is unknown if this disinhibitory motif is conserved in other cortical regions, species, or across development. We observed that PV-IN mediated inhibition is robustly suppressed by opioids in hippocampus proper but not primary neocortex in mice and nonhuman primates, with spontaneous inhibitory tone in resected human tissue also following a consistent dichotomy. This hippocampal disinhibitory motif was established in early development when PV-INs and opioids were found to regulate early population activity. Acute opioid-mediated modulation was partially occluded with morphine pretreatment, with implications for the effects of opioids on hippocampal network activity important for learning and memory. Together, these findings demonstrate that PV-INs exhibit a divergence in opioid sensitivity across brain regions that is remarkably conserved across evolution and highlights the underappreciated role of opioids acting through immature PV-INs in shaping hippocampal development.
Collapse
Affiliation(s)
- Adam P Caccavano
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anna Vlachos
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Nadiya McLean
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sarah Kimmel
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - June Hoan Kim
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Geoffrey Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Vivek Mahadevan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Lauren Hewitt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anthony M Rossi
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ilona Spineux
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sherry Jingjing Wu
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elisabetta Furlanis
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Min Dai
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brenda Leyva Garcia
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yating Wang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Edra London
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Daniel Abebe
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mark A G Eldridge
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Alex C Cummins
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Brendan E Hines
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Anya Plotnikova
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Arya Mohanty
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Bruno B Averbeck
- National Institute of Mental Health (NIMH), NIH, Bethesda, MD 20892, USA
| | - Kareem Zaghloul
- National Institute of Neurological Disorders and Stroke (NINDS) Intramural Research Program, NIH, Bethesda, MD 20892, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kenneth A Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Chris J McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Section on Cellular and Synaptic Physiology, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Zhang XH, Anderson KM, Dong HM, Chopra S, Dhamala E, Emani PS, Gerstein MB, Margulies DS, Holmes AJ. The cell-type underpinnings of the human functional cortical connectome. Nat Neurosci 2025; 28:150-160. [PMID: 39572742 DOI: 10.1038/s41593-024-01812-2] [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: 09/26/2024] [Indexed: 11/27/2024]
Abstract
The functional properties of the human brain arise, in part, from the vast assortment of cell types that pattern the cerebral cortex. The cortical sheet can be broadly divided into distinct networks, which are embedded into processing streams, or gradients, that extend from unimodal systems through higher-order association territories. Here using microarray data from the Allen Human Brain Atlas and single-nucleus RNA-sequencing data from multiple cortical territories, we demonstrate that cell-type distributions are spatially coupled to the functional organization of cortex, as estimated through functional magnetic resonance imaging. Differentially enriched cells follow the spatial topography of both functional gradients and associated large-scale networks. Distinct cellular fingerprints were evident across networks, and a classifier trained on postmortem cell-type distributions was able to predict the functional network allegiance of cortical tissue samples. These data indicate that the in vivo organization of the cortical sheet is reflected in the spatial variability of its cellular composition.
Collapse
Affiliation(s)
- Xi-Han Zhang
- Department of Psychology, Yale University, New Haven, CT, USA.
| | | | - Hao-Ming Dong
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Sidhant Chopra
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Elvisha Dhamala
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Prashant S Emani
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Mark B Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
| | - Daniel S Margulies
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Cognitive Neuroanatomy Lab, Université Paris Cité, INCC UMR 8002, CNRS, Paris, France
| | - Avram J Holmes
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
9
|
Somogyi P, Horie S, Lukacs I, Hunter E, Sarkany B, Viney T, Livermore J, Plaha P, Stacey R, Ansorge O, El Mestikawy S, Zhao Q. Synaptic Targets and Cellular Sources of CB1 Cannabinoid Receptor and Vesicular Glutamate Transporter-3 Expressing Nerve Terminals in Relation to GABAergic Neurons in the Human Cerebral Cortex. Eur J Neurosci 2025; 61:e16652. [PMID: 39810425 PMCID: PMC11733414 DOI: 10.1111/ejn.16652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/16/2025]
Abstract
Cannabinoid receptor 1 (CB1) regulates synaptic transmission through presynaptic receptors in nerve terminals, and its physiological roles are of clinical relevance. The cellular sources and synaptic targets of CB1-expressing terminals in the human cerebral cortex are undefined. We demonstrate a variable laminar pattern of CB1-immunoreactive axons and electron microscopically show that CB1-positive GABAergic terminals make type-2 synapses innervating dendritic shafts (69%), dendritic spines (20%) and somata (11%) in neocortical layers 2-3. Of the CB1-immunopositive GABAergic terminals, 25% were vesicular-glutamate-transporter-3 (VGLUT3)-immunoreactive, suggesting GABAergic/glutamatergic co-transmission on dendritic shafts. In vitro recorded and labelled VGLUT3 or CB1-positive GABAergic interneurons expressed cholecystokinin, vasoactive-intestinal-polypeptide and calretinin, had diverse firing, axons and dendrites, and included rosehip, neurogliaform and basket cells, but not double bouquet or axo-axonic cells. CB1-positive interneurons innervated pyramidal cells and GABAergic interneurons. Glutamatergic synaptic terminals formed type-1 synapses and some were positive for CB1 receptor with a distribution that appeared different from that in GABAergic terminals. From the sampled VGLUT3-positive terminals, 60% formed type-1 synapses with dendritic spines (80%) or shafts (20%) and 52% were also positive for VGLUT1, suggesting intracortical origin. Some VGLUT3-positive terminals were immunopositive for vesicular-monoamine-transporter-2, suggesting 5-HT/glutamate co-transmission. Overall, the results show that CB1 regulates GABA release mainly to dendritic shafts of both pyramidal cells and interneurons and predict CB1-regulated co-release of GABA and glutamate from single cortical interneurons. We also demonstrate the co-existence of multiple vesicular glutamate transporters in a select population of terminals probably originating from cortical neurons and innervating dendritic spines in the human cerebral cortex.
Collapse
Affiliation(s)
- Peter Somogyi
- Department of PharmacologyUniversity of OxfordOxfordUK
| | - Sawa Horie
- Department of PharmacologyUniversity of OxfordOxfordUK
- Kawasaki Medical SchoolOkayamaJapan
- Department of Anatomy and NeurobiologyNational Defense Medical CollegeSaitamaJapan
| | - Istvan Lukacs
- Department of PharmacologyUniversity of OxfordOxfordUK
- Institute of Experimental MedicineBudapestHungary
| | - Emily Hunter
- Department of PharmacologyUniversity of OxfordOxfordUK
| | | | | | - James Livermore
- Department of Neurosurgery, John Radcliffe HospitalOUH NHS Foundation TrustOxfordUK
- Department of NeurosurgeryLeeds General InfirmaryLeedsUK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe HospitalOUH NHS Foundation TrustOxfordUK
| | - Richard Stacey
- Department of Neurosurgery, John Radcliffe HospitalOUH NHS Foundation TrustOxfordUK
| | - Olaf Ansorge
- Nuffield Department of Clinical NeurosciencesUniv. OxfordOxfordUK
| | - Salah El Mestikawy
- Douglas Research CentreMcGill University and the Montreal West Island IUHSSCMontréalCanada
| | - Qianru Zhao
- Department of PharmacologyUniversity of OxfordOxfordUK
- Department of Chemical Biology, School of Pharmaceutical SciencesSouth‐Central Minzu UniversityWuhanChina
| |
Collapse
|
10
|
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
|
11
|
Fekete Z, Weisz F, Karlócai MR, Veres JM, Andrási T, Hájos N. Synaptic communication within the microcircuits of pyramidal neurons and basket cells in the mouse prefrontal cortex. J Physiol 2024. [PMID: 39418315 DOI: 10.1113/jp286284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Basket cells are inhibitory interneurons in cortical structures with the potential to efficiently control the activity of their postsynaptic partners. Although their contribution to higher order cognitive functions associated with the medial prefrontal cortex (mPFC) relies on the characteristics of their synaptic connections, the way that they are embedded into local circuits is still not fully uncovered. Here, we determined the synaptic properties of excitatory and inhibitory connections between pyramidal neurons (PNs), cholecystokinin-containing basket cells (CCKBCs) and parvalbumin-containing basket cells (PVBCs) in the mouse mPFC. By performing paired recordings, we revealed that PVBCs receive larger unitary excitatory postsynaptic currents from PNs with shorter latency and faster kinetic properties compared to events evoked in CCKBCs. Also, unitary inhibitory postsynaptic currents in PNs were more reliably evoked by PVBCs than by CCKBCs, yet the former connections showed profound short-term depression. Moreover, we demonstrated that CCKBCs and PVBCs in the mPFC are connected with each other. Because alterations in PVBC function have been linked to neurological and psychiatric conditions such as Alzheimer's disease and schizophrenia and CCKBC vulnerability might play a role in mood disorders, a deeper understanding of the general features of basket cell synapses could serve as a reference point for future investigations with therapeutic objectives. KEY POINTS: Cholecystokinin- (CCKBCs) and parvalbumin-expressing basket cells (PVBCs) have distinct passive and active membrane properties. Pyramidal neurons give rise to larger unitary excitatory postsynaptic currents in PVBCs compared to events in CCKBCs. Unitary inhibitory postsynaptic currents in pyramidal neurons are more reliably evoked by PVBCs than by CCKBCs. Basket cells form chemical synapses and gap junctions with their own cell type. The two basket cell types are connected with each other.
Collapse
Affiliation(s)
- Zsuzsanna Fekete
- Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Filippo Weisz
- Institute of Experimental Medicine, Budapest, Hungary
| | | | - Judit M Veres
- Institute of Experimental Medicine, Budapest, Hungary
| | - Tibor Andrási
- Institute of Experimental Medicine, Budapest, Hungary
| | - Norbert Hájos
- Institute of Experimental Medicine, Budapest, Hungary
- The Linda and Jack Gill Center for Molecular Bioscience, Indiana University Bloomington, Indiana, USA
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, Indiana, USA
| |
Collapse
|
12
|
Fu X, Tasker JG. Neuromodulation of inhibitory synaptic transmission in the basolateral amygdala during fear and anxiety. Front Cell Neurosci 2024; 18:1421617. [PMID: 38994327 PMCID: PMC11236696 DOI: 10.3389/fncel.2024.1421617] [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: 04/22/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
The basolateral amygdala plays pivotal roles in the regulation of fear and anxiety and these processes are profoundly modulated by different neuromodulatory systems that are recruited during emotional arousal. Recent studies suggest activities of BLA interneurons and inhibitory synaptic transmission in BLA principal cells are regulated by neuromodulators to influence the output and oscillatory network states of the BLA, and ultimately the behavioral expression of fear and anxiety. In this review, we first summarize a cellular mechanism of stress-induced anxiogenesis mediated by the interaction of glucocorticoid and endocannabinoid signaling at inhibitory synapses in the BLA. Then we discuss cell type-specific activity patterns induced by neuromodulators converging on the Gq signaling pathway in BLA perisomatic parvalbumin-expressing (PV) and cholecystokinin-expressing (CCK) basket cells and their effects on BLA network oscillations and fear learning.
Collapse
Affiliation(s)
- Xin Fu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Jeffrey G. Tasker
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
13
|
Kamalova A, Manoocheri K, Liu X, Casello SM, Huang M, Baimel C, Jang EV, Anastasiades PG, Collins DP, Carter AG. CCK+ Interneurons Contribute to Thalamus-Evoked Feed-Forward Inhibition in the Prelimbic Prefrontal Cortex. J Neurosci 2024; 44:e0957232024. [PMID: 38697841 PMCID: PMC11154858 DOI: 10.1523/jneurosci.0957-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Interneurons in the medial prefrontal cortex (PFC) regulate local neural activity to influence cognitive, motivated, and emotional behaviors. Parvalbumin-expressing (PV+) interneurons are the primary mediators of thalamus-evoked feed-forward inhibition across the mouse cortex, including the anterior cingulate cortex, where they are engaged by inputs from the mediodorsal (MD) thalamus. In contrast, in the adjacent prelimbic (PL) cortex, we find that PV+ interneurons are scarce in the principal thalamorecipient layer 3 (L3), suggesting distinct mechanisms of inhibition. To identify the interneurons that mediate MD-evoked inhibition in PL, we combine slice physiology, optogenetics, and intersectional genetic tools in mice of both sexes. We find interneurons expressing cholecystokinin (CCK+) are abundant in L3 of PL, with cells exhibiting fast-spiking (fs) or non-fast-spiking (nfs) properties. MD inputs make stronger connections onto fs-CCK+ interneurons, driving them to fire more readily than nearby L3 pyramidal cells and other interneurons. CCK+ interneurons in turn make inhibitory, perisomatic connections onto L3 pyramidal cells, where they exhibit cannabinoid 1 receptor (CB1R) mediated modulation. Moreover, MD-evoked feed-forward inhibition, but not direct excitation, is also sensitive to CB1R modulation. Our findings indicate that CCK+ interneurons contribute to MD-evoked inhibition in PL, revealing a mechanism by which cannabinoids can modulate MD-PFC communication.
Collapse
Affiliation(s)
- Aichurok Kamalova
- Center for Neural Science, New York University, New York, New York 10003
| | - Kasra Manoocheri
- Center for Neural Science, New York University, New York, New York 10003
| | - Xingchen Liu
- Center for Neural Science, New York University, New York, New York 10003
| | - Sanne M Casello
- Center for Neural Science, New York University, New York, New York 10003
| | - Matthew Huang
- Center for Neural Science, New York University, New York, New York 10003
| | - Corey Baimel
- Center for Neural Science, New York University, New York, New York 10003
| | - Emily V Jang
- Center for Neural Science, New York University, New York, New York 10003
| | | | - David P Collins
- Center for Neural Science, New York University, New York, New York 10003
| | - Adam G Carter
- Center for Neural Science, New York University, New York, New York 10003
| |
Collapse
|
14
|
Leitch B. Parvalbumin Interneuron Dysfunction in Neurological Disorders: Focus on Epilepsy and Alzheimer's Disease. Int J Mol Sci 2024; 25:5549. [PMID: 38791587 PMCID: PMC11122153 DOI: 10.3390/ijms25105549] [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: 04/29/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Parvalbumin expressing (PV+) GABAergic interneurons are fast spiking neurons that provide powerful but relatively short-lived inhibition to principal excitatory cells in the brain. They play a vital role in feedforward and feedback synaptic inhibition, preventing run away excitation in neural networks. Hence, their dysfunction can lead to hyperexcitability and increased susceptibility to seizures. PV+ interneurons are also key players in generating gamma oscillations, which are synchronized neural oscillations associated with various cognitive functions. PV+ interneuron are particularly vulnerable to aging and their degeneration has been associated with cognitive decline and memory impairment in dementia and Alzheimer's disease (AD). Overall, dysfunction of PV+ interneurons disrupts the normal excitatory/inhibitory balance within specific neurocircuits in the brain and thus has been linked to a wide range of neurodevelopmental and neuropsychiatric disorders. This review focuses on the role of dysfunctional PV+ inhibitory interneurons in the generation of epileptic seizures and cognitive impairment and their potential as targets in the design of future therapeutic strategies to treat these disorders. Recent research using cutting-edge optogenetic and chemogenetic technologies has demonstrated that they can be selectively manipulated to control seizures and restore the balance of neural activity in the brains of animal models. This suggests that PV+ interneurons could be important targets in developing future treatments for patients with epilepsy and comorbid disorders, such as AD, where seizures and cognitive decline are directly linked to specific PV+ interneuron deficits.
Collapse
Affiliation(s)
- Beulah Leitch
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
15
|
Bratsch-Prince JX, Warren JW, Jones GC, McDonald AJ, Mott DD. Acetylcholine Engages Distinct Amygdala Microcircuits to Gate Internal Theta Rhythm. J Neurosci 2024; 44:e1568232024. [PMID: 38438258 PMCID: PMC11055655 DOI: 10.1523/jneurosci.1568-23.2024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024] Open
Abstract
Acetylcholine (ACh) is released from basal forebrain cholinergic neurons in response to salient stimuli and engages brain states supporting attention and memory. These high ACh states are associated with theta oscillations, which synchronize neuronal ensembles. Theta oscillations in the basolateral amygdala (BLA) in both humans and rodents have been shown to underlie emotional memory, yet their mechanism remains unclear. Here, using brain slice electrophysiology in male and female mice, we show large ACh stimuli evoke prolonged theta oscillations in BLA local field potentials that depend upon M3 muscarinic receptor activation of cholecystokinin (CCK) interneurons (INs) without the need for external glutamate signaling. Somatostatin (SOM) INs inhibit CCK INs and are themselves inhibited by ACh, providing a functional SOM→CCK IN circuit connection gating BLA theta. Parvalbumin (PV) INs, which can drive BLA oscillations in baseline states, are not involved in the generation of ACh-induced theta, highlighting that ACh induces a cellular switch in the control of BLA oscillatory activity and establishes an internally BLA-driven theta oscillation through CCK INs. Theta activity is more readily evoked in BLA over the cortex or hippocampus, suggesting preferential activation of the BLA during high ACh states. These data reveal a SOM→CCK IN circuit in the BLA that gates internal theta oscillations and suggest a mechanism by which salient stimuli acting through ACh switch the BLA into a network state enabling emotional memory.
Collapse
Affiliation(s)
- Joshua X Bratsch-Prince
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - James W Warren
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - Grace C Jones
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - Alexander J McDonald
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| | - David D Mott
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina 29208
| |
Collapse
|
16
|
Reich N, Hölscher C. Cholecystokinin (CCK): a neuromodulator with therapeutic potential in Alzheimer's and Parkinson's disease. Front Neuroendocrinol 2024; 73:101122. [PMID: 38346453 DOI: 10.1016/j.yfrne.2024.101122] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/04/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Cholecystokinin (CCK) is a neuropeptide modulating digestion, glucose levels, neurotransmitters and memory. Recent studies suggest that CCK exhibits neuroprotective effects in Alzheimer's disease (AD) and Parkinson's disease (PD). Thus, we review the physiological function and therapeutic potential of CCK. The neuropeptide facilitates hippocampal glutamate release and gates GABAergic basket cell activity, which improves declarative memory acquisition, but inhibits consolidation. Cortical CCK alters recognition memory and enhances audio-visual processing. By stimulating CCK-1 receptors (CCK-1Rs), sulphated CCK-8 elicits dopamine release in the substantia nigra and striatum. In the mesolimbic pathway, CCK release is triggered by dopamine and terminates reward responses via CCK-2Rs. Importantly, activation of hippocampal and nigral CCK-2Rs is neuroprotective by evoking AMPK activation, expression of mitochondrial fusion modulators and autophagy. Other benefits include vagus nerve/CCK-1R-mediated expression of brain-derived neurotrophic factor, intestinal protection and suppression of inflammation. We also discuss caveats and the therapeutic combination of CCK with other peptide hormones.
Collapse
Affiliation(s)
- Niklas Reich
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK; Faculty of Health and Medicine, Biomedical & Life Sciences Division, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Christian Hölscher
- Second associated Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, China; Henan Academy of Innovations in Medical Science, Neurodegeneration research group, Xinzhen, Henan province, China
| |
Collapse
|
17
|
Nguyen R, Sivakumaran S, Lambe EK, Kim JC. Ventral hippocampal cholecystokinin interneurons gate contextual reward memory. iScience 2024; 27:108824. [PMID: 38303709 PMCID: PMC10831933 DOI: 10.1016/j.isci.2024.108824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/06/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Associating contexts with rewards depends on hippocampal circuits, with local inhibitory interneurons positioned to play an important role in shaping activity. Here, we demonstrate that the encoding of context-reward memory requires a ventral hippocampus (vHPC) to nucleus accumbens (NAc) circuit that is gated by cholecystokinin (CCK) interneurons. In a sucrose conditioned place preference (CPP) task, optogenetically inhibiting vHPC-NAc terminals impaired the acquisition of place preference. Transsynaptic rabies tracing revealed vHPC-NAc neurons were monosynaptically innervated by CCK interneurons. Using intersectional genetic targeting of CCK interneurons, ex vivo optogenetic activation of CCK interneurons increased GABAergic transmission onto vHPC-NAc neurons, while in vivo optogenetic inhibition of CCK interneurons increased cFos in these projection neurons. Notably, CCK interneuron inhibition during sucrose CPP learning increased time spent in the sucrose-associated location, suggesting enhanced place-reward memory. Our findings reveal a previously unknown hippocampal microcircuit crucial for modulating the strength of contextual reward learning.
Collapse
Affiliation(s)
- Robin Nguyen
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | | | - Evelyn K. Lambe
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of OBGYN, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Jun Chul Kim
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Machold R, Dellal S, Valero M, Zurita H, Kruglikov I, Meng JH, Hanson JL, Hashikawa Y, Schuman B, Buzsáki G, Rudy B. Id2 GABAergic interneurons comprise a neglected fourth major group of cortical inhibitory cells. eLife 2023; 12:e85893. [PMID: 37665123 PMCID: PMC10581691 DOI: 10.7554/elife.85893] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Cortical GABAergic interneurons (INs) represent a diverse population of mainly locally projecting cells that provide specialized forms of inhibition to pyramidal neurons and other INs. Most recent work on INs has focused on subtypes distinguished by expression of Parvalbumin (PV), Somatostatin (SST), or Vasoactive Intestinal Peptide (VIP). However, a fourth group that includes neurogliaform cells (NGFCs) has been less well characterized due to a lack of genetic tools. Here, we show that these INs can be accessed experimentally using intersectional genetics with the gene Id2. We find that outside of layer 1 (L1), the majority of Id2 INs are NGFCs that express high levels of neuropeptide Y (NPY) and exhibit a late-spiking firing pattern, with extensive local connectivity. While much sparser, non-NGFC Id2 INs had more variable properties, with most cells corresponding to a diverse group of INs that strongly expresses the neuropeptide CCK. In vivo, using silicon probe recordings, we observed several distinguishing aspects of NGFC activity, including a strong rebound in activity immediately following the cortical down state during NREM sleep. Our study provides insights into IN diversity and NGFC distribution and properties, and outlines an intersectional genetics approach for further study of this underappreciated group of INs.
Collapse
Affiliation(s)
- Robert Machold
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Shlomo Dellal
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Manuel Valero
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Hector Zurita
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Ilya Kruglikov
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - John Hongyu Meng
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Center for Neural Science, New York UniversityNew YorkUnited States
| | - Jessica L Hanson
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Yoshiko Hashikawa
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Benjamin Schuman
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - György Buzsáki
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
| | - Bernardo Rudy
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Department of Neuroscience and Physiology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Anesthesiology, Perioperative Care and Pain Medicine, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
19
|
Jung K, Chang M, Steinecke A, Burke B, Choi Y, Oisi Y, Fitzpatrick D, Taniguchi H, Kwon HB. An adaptive behavioral control motif mediated by cortical axo-axonic inhibition. Nat Neurosci 2023; 26:1379-1393. [PMID: 37474640 PMCID: PMC10400431 DOI: 10.1038/s41593-023-01380-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/13/2023] [Indexed: 07/22/2023]
Abstract
Genetically defined subgroups of inhibitory interneurons are thought to play distinct roles in learning, but heterogeneity within these subgroups has limited our understanding of the scope and nature of their specific contributions. Here we reveal that the chandelier cell (ChC), an interneuron type that specializes in inhibiting the axon-initial segment (AIS) of pyramidal neurons, establishes cortical microcircuits for organizing neural coding through selective axo-axonic synaptic plasticity. We found that organized motor control is mediated by enhanced population coding of direction-tuned premotor neurons, with tuning refined through suppression of irrelevant neuronal activity. ChCs contribute to learning-dependent refinements by providing selective inhibitory control over individual pyramidal neurons rather than global suppression. Quantitative analysis of structural plasticity across axo-axonic synapses revealed that ChCs redistributed inhibitory weights to individual pyramidal neurons during learning. These results demonstrate an adaptive logic of the inhibitory circuit motif responsible for organizing distributed neural representations. Thus, ChCs permit efficient cortical computation in a targeted cell-specific manner.
Collapse
Affiliation(s)
- Kanghoon Jung
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Minhyeok Chang
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - André Steinecke
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Benjamin Burke
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Youngjin Choi
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yasuhiro Oisi
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | | | - Hiroki Taniguchi
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- Department of Pathology, Chronic Brain Injury program, Ohio State University, Columbus, OH, USA
| | - Hyung-Bae Kwon
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
- Max Planck Institute of Neurobiology, Martinsried, Germany.
| |
Collapse
|
20
|
Zhang XH, Anderson KM, Dong HM, Chopra S, Dhamala E, Emani PS, Margulies D, Holmes AJ. The Cellular Underpinnings of the Human Cortical Connectome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547828. [PMID: 37461642 PMCID: PMC10349999 DOI: 10.1101/2023.07.05.547828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
The functional properties of the human brain arise, in part, from the vast assortment of cell types that pattern the cortex. The cortical sheet can be broadly divided into distinct networks, which are further embedded into processing streams, or gradients, that extend from unimodal systems through higher-order association territories. Here, using transcriptional data from the Allen Human Brain Atlas, we demonstrate that imputed cell type distributions are spatially coupled to the functional organization of cortex, as estimated through fMRI. Cortical cellular profiles follow the macro-scale organization of the functional gradients as well as the associated large-scale networks. Distinct cellular fingerprints were evident across networks, and a classifier trained on post-mortem cell-type distributions was able to predict the functional network allegiance of cortical tissue samples. These data indicate that the in vivo organization of the cortical sheet is reflected in the spatial variability of its cellular composition.
Collapse
Affiliation(s)
- Xi-Han Zhang
- Department of Psychology, Yale University, New Haven, CT, USA
| | | | - Hao-Ming Dong
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Sidhant Chopra
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Elvisha Dhamala
- Department of Psychology, Yale University, New Haven, CT, USA
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Prashant S. Emani
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Daniel Margulies
- CNRS, Integrative Neuroscience and Cognition Center (UMR 8002), Université de Paris, Paris, France
| | - Avram J. Holmes
- Department of Psychology, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
21
|
Jung K, Chang M, Steinecke A, Berke B, Choi Y, Oisi Y, Fitzpatrick D, Taniguchi H, Kwon HB. An adaptive behavioral control motif mediated by cortical axo-axonic inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.10.531767. [PMID: 36945592 PMCID: PMC10029003 DOI: 10.1101/2023.03.10.531767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Neural circuits are reorganized with specificity during learning. Genetically-defined subgroups of inhibitory interneurons are thought to play distinct roles in learning, but heterogeneity within these subgroups has limited our understanding of the scope and nature of their specific contributions to learning. Here we reveal that the chandelier cell (ChC), an interneuron type that specializes in inhibiting the axon-initial segment (AIS) of pyramidal neurons, establishes cortical microcircuits for organizing neural coding through selective axo-axonic synaptic plasticity. We find that organized motor control is mediated by enhanced population coding of direction-tuned premotor neurons, whose tuning is refined through suppression of irrelevant neuronal activity. ChCs are required for learning-dependent refinements via providing selective inhibitory control over pyramidal neurons rather than global suppression. Quantitative analysis on structural plasticity of axo-axonic synapses revealed that ChCs redistributed inhibitory weights to individual pyramidal neurons during learning. These results demonstrate an adaptive logic of the inhibitory circuit motif responsible for organizing distributed neural representations. Thus, ChCs permit efficient cortical computation in a target cell specific manner, which highlights the significance of interneuron diversity.
Collapse
Affiliation(s)
- Kanghoon Jung
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
- Current address: Allen Institute for Neural Dynamics, Seattle, WA 98109, USA
| | - Minhyeok Chang
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- equally contributed
| | - André Steinecke
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
- equally contributed
| | - Benjamin Berke
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Youngjin Choi
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Yasuhiro Oisi
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
| | - David Fitzpatrick
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
| | - Hiroki Taniguchi
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
- Department of Pathology, Chronic Brain Injury program, Ohio State University, Columbus, Ohio 43210, USA
| | - Hyung-Bae Kwon
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
- Max Planck Institute of Neurobiology, Martinsried 82152, Germany
| |
Collapse
|
22
|
Lukacs IP, Francavilla R, Field M, Hunter E, Howarth M, Horie S, Plaha P, Stacey R, Livermore L, Ansorge O, Tamas G, Somogyi P. Differential effects of group III metabotropic glutamate receptors on spontaneous inhibitory synaptic currents in spine-innervating double bouquet and parvalbumin-expressing dendrite-targeting GABAergic interneurons in human neocortex. Cereb Cortex 2023; 33:2101-2142. [PMID: 35667019 PMCID: PMC9977385 DOI: 10.1093/cercor/bhac195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/12/2022] Open
Abstract
Diverse neocortical GABAergic neurons specialize in synaptic targeting and their effects are modulated by presynaptic metabotropic glutamate receptors (mGluRs) suppressing neurotransmitter release in rodents, but their effects in human neocortex are unknown. We tested whether activation of group III mGluRs by L-AP4 changes GABAA receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in 2 distinct dendritic spine-innervating GABAergic interneurons recorded in vitro in human neocortex. Calbindin-positive double bouquet cells (DBCs) had columnar "horsetail" axons descending through layers II-V innervating dendritic spines (48%) and shafts, but not somata of pyramidal and nonpyramidal neurons. Parvalbumin-expressing dendrite-targeting cell (PV-DTC) axons extended in all directions innervating dendritic spines (22%), shafts (65%), and somata (13%). As measured, 20% of GABAergic neuropil synapses innervate spines, hence DBCs, but not PV-DTCs, preferentially select spine targets. Group III mGluR activation paradoxically increased the frequency of sIPSCs in DBCs (to median 137% of baseline) but suppressed it in PV-DTCs (median 92%), leaving the amplitude unchanged. The facilitation of sIPSCs in DBCs may result from their unique GABAergic input being disinhibited via network effect. We conclude that dendritic spines receive specialized, diverse GABAergic inputs, and group III mGluRs differentially regulate GABAergic synaptic transmission to distinct GABAergic cell types in human cortex.
Collapse
Affiliation(s)
- Istvan P Lukacs
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | | | - Martin Field
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Emily Hunter
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Michael Howarth
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Sawa Horie
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Richard Stacey
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Laurent Livermore
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Gabor Tamas
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | - Peter Somogyi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
23
|
Stieve BJ, Smith MM, Krook-Magnuson E. LINCs Are Vulnerable to Epileptic Insult and Fail to Provide Seizure Control via On-Demand Activation. eNeuro 2023; 10:ENEURO.0195-22.2022. [PMID: 36725340 PMCID: PMC9933934 DOI: 10.1523/eneuro.0195-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 02/03/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is notoriously pharmacoresistant, and identifying novel therapeutic targets for controlling seizures is crucial. Long-range inhibitory neuronal nitric oxide synthase-expressing cells (LINCs), a population of hippocampal neurons, were recently identified as a unique source of widespread inhibition in CA1, able to elicit both GABAA-mediated and GABAB-mediated postsynaptic inhibition. We therefore hypothesized that LINCs could be an effective target for seizure control. LINCs were optogenetically activated for on-demand seizure intervention in the intrahippocampal kainate (KA) mouse model of chronic TLE. Unexpectedly, LINC activation at 1 month post-KA did not substantially reduce seizure duration in either male or female mice. We tested two different sets of stimulation parameters, both previously found to be effective with on-demand optogenetic approaches, but neither was successful. Quantification of LINCs following intervention revealed a substantial reduction of LINC numbers compared with saline-injected controls. We also observed a decreased number of LINCs when the site of initial insult (i.e., KA injection) was moved to the amygdala [basolateral amygdala (BLA)-KA], and correspondingly, no effect of light delivery on BLA-KA seizures. This indicates that LINCs may be a vulnerable population in TLE, regardless of the site of initial insult. To determine whether long-term circuitry changes could influence outcomes, we continued testing once a month for up to 6 months post-KA. However, at no time point did LINC activation provide meaningful seizure suppression. Altogether, our results suggest that LINCs are not a promising target for seizure inhibition in TLE.
Collapse
Affiliation(s)
- Bethany J Stieve
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Madison M Smith
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Esther Krook-Magnuson
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
24
|
The potential role of the cholecystokinin system in declarative memory. Neurochem Int 2023; 162:105440. [PMID: 36375634 DOI: 10.1016/j.neuint.2022.105440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/24/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
As one of the most abundant neuropeptides in the central nervous system, cholecystokinin (CCK) has been suggested to be associated with higher brain functions, including learning and memory. In this review, we examined the potential role of the CCK system in declarative memory. First, we summarized behavioral studies that provide evidence for an important role of CCK in two forms of declarative memory-fear memory and spatial memory. Subsequently, we examined the electrophysiological studies that support the diverse roles of CCK-2 receptor activation in neocortical and hippocampal synaptic plasticity, and discussed the potential mechanisms that may be involved. Last but not least, we discussed whether the reported CCK-mediated synaptic plasticity can explain the strong influence of the CCK signaling system in neocortex and hippocampus dependent declarative memory. The available research supports the role of CCK-mediated synaptic plasticity in neocortex dependent declarative memory acquisition, but further study on the association between CCK-mediated synaptic plasticity and neocortex dependent declarative memory consolidation and retrieval is necessary. Although a direct link between CCK-mediated synaptic plasticity and hippocampus dependent declarative memory is missing, noticeable evidence from morphological, behavioral, and electrophysiological studies encourages further investigation regarding the potential role of CCK-dependent synaptic plasticity in hippocampus dependent declarative memory.
Collapse
|
25
|
Boxer EE, Aoto J. Neurexins and their ligands at inhibitory synapses. Front Synaptic Neurosci 2022; 14:1087238. [PMID: 36618530 PMCID: PMC9812575 DOI: 10.3389/fnsyn.2022.1087238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022] Open
Abstract
Since the discovery of neurexins (Nrxns) as essential and evolutionarily conserved synaptic adhesion molecules, focus has largely centered on their functional contributions to glutamatergic synapses. Recently, significant advances to our understanding of neurexin function at GABAergic synapses have revealed that neurexins can play pleiotropic roles in regulating inhibitory synapse maintenance and function in a brain-region and synapse-specific manner. GABAergic neurons are incredibly diverse, exhibiting distinct synaptic properties, sites of innervation, neuromodulation, and plasticity. Different classes of GABAergic neurons often express distinct repertoires of Nrxn isoforms that exhibit differential alternative exon usage. Further, Nrxn ligands can be differentially expressed and can display synapse-specific localization patterns, which may contribute to the formation of a complex trans-synaptic molecular code that establishes the properties of inhibitory synapse function and properties of local circuitry. In this review, we will discuss how Nrxns and their ligands sculpt synaptic inhibition in a brain-region, cell-type and synapse-specific manner.
Collapse
Affiliation(s)
| | - Jason Aoto
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| |
Collapse
|
26
|
Chemogenetic activation of VGLUT3-expressing neurons decreases movement. Eur J Pharmacol 2022; 935:175298. [PMID: 36198338 DOI: 10.1016/j.ejphar.2022.175298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) are responsible for the storage of glutamate into secretory vesicles. The VGLUT3 isoform is mainly expressed in neurons that secrete other classical neurotransmitters, including the cholinergic interneurons in the striatum, and VGLUT3-expressing neurons often secrete two distinct neurotransmitters. VGLUT3 is discretely distributed throughout the brain and is found in subpopulations of spinal cord interneurons, in subset of neurons in the dorsal root ganglion, and in Merkel cells. Mice with a global loss of VGLUT3 are hyperactive and the modulation of specific VGLUT3-expressing circuits can lead to changes in movement. In this study, we tested the hypothesis that increased activity of VGLUT3-expressing neurons is associated with decreased movement. Using a mouse line expressing excitatory designer receptor exclusively activated by designer drugs (hM3Dq-DREADD) on VGLUT3-expressing neurons, we showed that activation of hM3Dq signalling acutely decreased locomotor activity. This decreased locomotion was likely not due to circuit changes mediated by glutamate nor acetylcholine released from VGLUT3-expressing neurons, as activation of hM3Dq signalling in mice that do not release glutamate or acetylcholine from VGLUT3-expressing neurons also decreased locomotor activity. This suggests that other neurotransmitters are likely driving this hypoactive phenotype. We used these mouse lines to compare the effects of DREADD agonists in vivo. We observed that clozapine-N-oxide (CNO), clozapine, compound 21 and perlapine show small differences in the speed at which they prompt behavioural responses but the four of them are selective DREADD ligands.
Collapse
|
27
|
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
|
28
|
Koukouli F, Montmerle M, Aguirre A, De Brito Van Velze M, Peixoto J, Choudhary V, Varilh M, Julio-Kalajzic F, Allene C, Mendéz P, Zerlaut Y, Marsicano G, Schlüter OM, Rebola N, Bacci A, Lourenço J. Visual-area-specific tonic modulation of GABA release by endocannabinoids sets the activity and coordination of neocortical principal neurons. Cell Rep 2022; 40:111202. [PMID: 36001978 PMCID: PMC9433882 DOI: 10.1016/j.celrep.2022.111202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 05/24/2022] [Accepted: 07/21/2022] [Indexed: 12/01/2022] Open
Abstract
Perisomatic inhibition of pyramidal neurons (PNs) coordinates cortical network activity during sensory processing, and this role is mainly attributed to parvalbumin-expressing basket cells (BCs). However, cannabinoid receptor type 1 (CB1)-expressing interneurons are also BCs, but the connectivity and function of these elusive but prominent neocortical inhibitory neurons are unclear. We find that their connectivity pattern is visual area specific. Persistently active CB1 signaling suppresses GABA release from CB1 BCs in the medial secondary visual cortex (V2M), but not in the primary visual cortex (V1). Accordingly, in vivo, tonic CB1 signaling is responsible for higher but less coordinated PN activity in the V2M than in the V1. These differential firing dynamics in the V1 and V2M can be captured by a computational network model that incorporates visual-area-specific properties. Our results indicate a differential CB1-mediated mechanism controlling PN activity, suggesting an alternative connectivity scheme of a specific GABAergic circuit in different cortical areas. CB1+ basket cells exhibit visual-area-specific morphology and connectivity patterns Tonic CB1 signaling underlies high pyramidal neurons (PN) activity in V2M but not V1 Tonic CB1 signaling differentially modulates PN-correlated activity in V1 and V2M Numerical simulations capture specific CB1-dependent firing dynamics of V1 and V2M
Collapse
Affiliation(s)
- Fani Koukouli
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Martin Montmerle
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Andrea Aguirre
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Jérémy Peixoto
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Vikash Choudhary
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Marjorie Varilh
- INSERM, U1215 NeuroCentre Magendie, University of Bordeaux, 33077 Bordeaux, France
| | | | - Camille Allene
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Yann Zerlaut
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, University of Bordeaux, 33077 Bordeaux, France
| | - Oliver M Schlüter
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany; Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nelson Rebola
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Alberto Bacci
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France.
| | - Joana Lourenço
- ICM - Institut du Cerveau, Sorbonne Université, INSERM, CNRS, Paris, France.
| |
Collapse
|
29
|
The role of inhibitory circuits in hippocampal memory processing. Nat Rev Neurosci 2022; 23:476-492. [DOI: 10.1038/s41583-022-00599-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 11/08/2022]
|
30
|
Tian D, Izumi SI. Transcranial Magnetic Stimulation and Neocortical Neurons: The Micro-Macro Connection. Front Neurosci 2022; 16:866245. [PMID: 35495053 PMCID: PMC9039343 DOI: 10.3389/fnins.2022.866245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 12/20/2022] Open
Abstract
Understanding the operation of cortical circuits is an important and necessary task in both neuroscience and neurorehabilitation. The functioning of the neocortex results from integrative neuronal activity, which can be probed non-invasively by transcranial magnetic stimulation (TMS). Despite a clear indication of the direct involvement of cortical neurons in TMS, no explicit connection model has been made between the microscopic neuronal landscape and the macroscopic TMS outcome. Here we have performed an integrative review of multidisciplinary evidence regarding motor cortex neurocytology and TMS-related neurophysiology with the aim of elucidating the micro–macro connections underlying TMS. Neurocytological evidence from animal and human studies has been reviewed to describe the landscape of the cortical neurons covering the taxonomy, morphology, circuit wiring, and excitatory–inhibitory balance. Evidence from TMS studies in healthy humans is discussed, with emphasis on the TMS pulse and paradigm selectivity that reflect the underlying neural circuitry constitution. As a result, we propose a preliminary neuronal model of the human motor cortex and then link the TMS mechanisms with the neuronal model by stimulus intensity, direction of induced current, and paired-pulse timing. As TMS bears great developmental potential for both a probe and modulator of neural network activity and neurotransmission, the connection model will act as a foundation for future combined studies of neurocytology and neurophysiology, as well as the technical advances and application of TMS.
Collapse
Affiliation(s)
- Dongting Tian
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- *Correspondence: Dongting Tian,
| | - Shin-Ichi Izumi
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduates School of Medicine, Sendai, Japan
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
- Shin-Ichi Izumi,
| |
Collapse
|
31
|
Tukker JJ, Beed P, Brecht M, Kempter R, Moser EI, Schmitz D. Microcircuits for spatial coding in the medial entorhinal cortex. Physiol Rev 2022; 102:653-688. [PMID: 34254836 PMCID: PMC8759973 DOI: 10.1152/physrev.00042.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The hippocampal formation is critically involved in learning and memory and contains a large proportion of neurons encoding aspects of the organism's spatial surroundings. In the medial entorhinal cortex (MEC), this includes grid cells with their distinctive hexagonal firing fields as well as a host of other functionally defined cell types including head direction cells, speed cells, border cells, and object-vector cells. Such spatial coding emerges from the processing of external inputs by local microcircuits. However, it remains unclear exactly how local microcircuits and their dynamics within the MEC contribute to spatial discharge patterns. In this review we focus on recent investigations of intrinsic MEC connectivity, which have started to describe and quantify both excitatory and inhibitory wiring in the superficial layers of the MEC. Although the picture is far from complete, it appears that these layers contain robust recurrent connectivity that could sustain the attractor dynamics posited to underlie grid pattern formation. These findings pave the way to a deeper understanding of the mechanisms underlying spatial navigation and memory.
Collapse
Affiliation(s)
- John J Tukker
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Prateep Beed
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humbold-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Brecht
- Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Richard Kempter
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Edvard I Moser
- Einstein Center for Neurosciences Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Neuroscience Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humbold-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
32
|
Guzman SJ, Schlögl A, Espinoza C, Zhang X, Suter BA, Jonas P. How connectivity rules and synaptic properties shape the efficacy of pattern separation in the entorhinal cortex-dentate gyrus-CA3 network. NATURE COMPUTATIONAL SCIENCE 2021; 1:830-842. [PMID: 38217181 DOI: 10.1038/s43588-021-00157-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/12/2021] [Indexed: 01/15/2024]
Abstract
Pattern separation is a fundamental brain computation that converts small differences in input patterns into large differences in output patterns. Several synaptic mechanisms of pattern separation have been proposed, including code expansion, inhibition and plasticity; however, which of these mechanisms play a role in the entorhinal cortex (EC)-dentate gyrus (DG)-CA3 circuit, a classical pattern separation circuit, remains unclear. Here we show that a biologically realistic, full-scale EC-DG-CA3 circuit model, including granule cells (GCs) and parvalbumin-positive inhibitory interneurons (PV+-INs) in the DG, is an efficient pattern separator. Both external gamma-modulated inhibition and internal lateral inhibition mediated by PV+-INs substantially contributed to pattern separation. Both local connectivity and fast signaling at GC-PV+-IN synapses were important for maximum effectiveness. Similarly, mossy fiber synapses with conditional detonator properties contributed to pattern separation. By contrast, perforant path synapses with Hebbian synaptic plasticity and direct EC-CA3 connection shifted the network towards pattern completion. Our results demonstrate that the specific properties of cells and synapses optimize higher-order computations in biological networks and might be useful to improve the deep learning capabilities of technical networks.
Collapse
Affiliation(s)
- S Jose Guzman
- IST Austria, Klosterneuburg, Austria
- Institute of Molecular Biotechnology, Vienna, Austria
| | | | - Claudia Espinoza
- IST Austria, Klosterneuburg, Austria
- Medical University of Austria, Division of Cognitive Neurobiology, Vienna, Austria
| | - Xiaomin Zhang
- IST Austria, Klosterneuburg, Austria
- Brain Research Institute, University of Zürich, Zurich, Switzerland
| | | | | |
Collapse
|
33
|
Poleksic J, Aksic M, Kapor S, Aleksic D, Stojkovic T, Radovic M, Djulejic V, Markovic B, Stamatakis A. Effects of Maternal Deprivation on the Prefrontal Cortex of Male Rats: Cellular, Neurochemical, and Behavioral Outcomes. Front Behav Neurosci 2021; 15:666547. [PMID: 34819843 PMCID: PMC8606589 DOI: 10.3389/fnbeh.2021.666547] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/08/2021] [Indexed: 01/13/2023] Open
Abstract
Stressful events experienced during early life are associated with increased vulnerability of developing psychopathology in adulthood. In the present study, we exposed 9-day-old Wistar rats to 24 h maternal deprivation (MD) with the aim to investigate the impact of early life stress (ELS) on morphological, biochemical, and functional aspects of the prefrontal cortex (PFC), a brain region particularly sensitive to stress. We found that in the superficial medial orbital cortex (MO), young adult male rats had reduced density of GAD67 and CCK immunopositive cells, while the rostral part of the ventral lateral orbital cortex (roVLO) showed a decrease in the density of GAD67 immunopositive cells in both superficial and deep layers. In addition, the superficial rostral part of area 1 of the cingulate cortex (roCg1) and deep prelimbic cortex (PrL) was also affected by MD indicated by the reduction in PV immunopositive cellular density. Furthermore, MD induced upregulation of brain-derived neurotrophic factor (BDNF), while it did not affect the overall expression of Iba1 in neonatal or young adult PFC as measured by Western blot, however, microglial activation in young adult MD rats was detected immunohistochemically in deep layers of MO and infralimbic cortex (IL). Interestingly, when young adult male rats were subjected to a behavioral flexibility test in a T-maze, MD rats showed a subtle impairment in T-maze reversal learning indicating a mildly affected PFC function. Taken together, our findings demonstrated that MD reduced the density of interneurons and induced microglial activation, in particular, PFC areas at young adulthood, and could alter synaptic plasticity accompanied by PFC dysfunction.
Collapse
Affiliation(s)
- Joko Poleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milan Aksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Slobodan Kapor
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dubravka Aleksic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tihomir Stojkovic
- Institute of Clinical and Medical Biochemistry, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marina Radovic
- Institute of Physiology and Biochemistry "Ivan Djaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vuk Djulejic
- Institute of Anatomy "Niko Miljanic", School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branka Markovic
- Faculty of Sport and Physical Education, University of Belgrade, Belgrade, Serbia
| | - Antonios Stamatakis
- Biology-Biochemistry Lab, School of Health Sciences, Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
34
|
Wallace MN, Shackleton TM, Thompson Z, Palmer AR. Juxtacellular Labeling of Stellate, Disk and Basket Neurons in the Central Nucleus of the Guinea Pig Inferior Colliculus. Front Neural Circuits 2021; 15:721015. [PMID: 34790099 PMCID: PMC8592287 DOI: 10.3389/fncir.2021.721015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022] Open
Abstract
We reconstructed the intrinsic axons of 32 neurons in the guinea pig inferior colliculus (IC) following juxtacellular labeling. Biocytin was injected into cells in vivo, after first analyzing physiological response properties. Based on axonal morphology there were two classes of neuron: (1) laminar cells (14/32, 44%) with an intrinsic axon and flattened dendrites confined to a single fibrodendritic lamina and (2) translaminar cells (18/32, 56%) with axons that terminated in two or more laminae in the central nucleus (ICc) or the surrounding cortex. There was also one small, low-frequency cell with bushy-like dendrites that was very sensitive to interaural timing differences. The translaminar cells were subdivided into three groups of cells with: (a) stellate dendrites that crossed at least two laminae (8/32, 25%); (b) flattened dendrites confined to one lamina and that had mainly en passant axonal swellings (7/32, 22%) and (c) short, flattened dendrites and axons with distinctive clusters of large terminal boutons in the ICc (3/32, 9%). These terminal clusters were similar to those of cortical basket cells. The 14 laminar cells all had sustained responses apart from one offset response. Almost half the non-basket type translaminar cells (7/15) had onset responses while the others had sustained responses. The basket cells were the only ones to have short-latency (7–9 ms), chopper responses and this distinctive temporal response should allow them to be studied in more detail in future. This is the first description of basket cells in the auditory brainstem, but more work is required to confirm their neurotransmitter and precise post-synaptic targets.
Collapse
Affiliation(s)
- Mark N Wallace
- Hearing Sciences, Mental Health and Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom.,Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Trevor M Shackleton
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Zoe Thompson
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Alan R Palmer
- Medical Research Council Institute of Hearing Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
35
|
Liu YJ, Liu TT, Jiang LH, Liu Q, Ma ZL, Xia TJ, Gu XP. Identification of hub genes associated with cognition in the hippocampus of Alzheimer's Disease. Bioengineered 2021; 12:9598-9609. [PMID: 34719328 PMCID: PMC8810106 DOI: 10.1080/21655979.2021.1999549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s Disease (AD) is a neurodegenerative disease featured by cognitive impairment. This bioinformatic analysis was used to identify hub genes related to cognitive dysfunction in AD. The gene expression profile GSE48350 in the hippocampus of AD patients aged >70 years was obtained from the Gene Expression Omnibus (GEO) database. A total of 96 differentially expressed genes (DEGs) were identified, and subjected to Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses; a protein–protein interaction (PPI) network was constructed. The DEGs were enriched in synapse-related changes. A protein cluster was teased out of PPI. Furthermore, the cognition ranked the first among all the terms of biological process (BP). Next, 4 of 10 hub genes enriched in cognition were identified. The function of these genes was validated using APP/PS1 mice. Cognitive performance was validated by Morris Water Maze (MWM), and gene expression by RT-qPCR, Cholecystokinin (CCK), Tachykinin precursor 1 (TAC1), Calbindin 1 (CALB1) were downregulated in the hippocampus. These genes can provide new directions in the research of the molecular mechanism of AD.
Collapse
Affiliation(s)
- Yu-Jia Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.,Medical School of Nanjing University, Nanjing, Jiangsu Province, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tian-Tian Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.,Medical School of Nanjing University, Nanjing, Jiangsu Province, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Lin-Hao Jiang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.,Medical School of Nanjing University, Nanjing, Jiangsu Province, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qian Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.,Medical School of Nanjing University, Nanjing, Jiangsu Province, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zheng-Liang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Tian-Jiao Xia
- Medical School of Nanjing University, Nanjing, Jiangsu Province, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiao-Ping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| |
Collapse
|
36
|
Senft RA, Dymecki SM. Neuronal pericellular baskets: neurotransmitter convergence and regulation of network excitability. Trends Neurosci 2021; 44:915-924. [PMID: 34565612 PMCID: PMC8551026 DOI: 10.1016/j.tins.2021.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/29/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022]
Abstract
A pericellular basket is a presynaptic configuration of numerous axonal boutons outlining a target neuron soma and its proximal dendrites. Recent studies show neurochemical diversity of pericellular baskets and suggest that neurotransmitter usage together with the dense, soma-proximal boutons may permit strong input effects on different timescales. Here we review the development, distribution, neurochemical phenotypes, and possible functions of pericellular baskets. As an example, we highlight pericellular baskets formed by projections of certain Pet1/Fev neurons of the serotonergic raphe nuclei. We propose that pericellular baskets represent convergence sites of competition or facilitation between neurotransmitter systems on downstream circuitry, especially in limbic brain regions, where pericellular baskets are widespread. Study of these baskets may enhance our understanding of monoamine regulation of memory, social behavior, and brain oscillations.
Collapse
Affiliation(s)
- Rebecca A Senft
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Susan M Dymecki
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Dudok B, Klein PM, Soltesz I. Toward Understanding the Diverse Roles of Perisomatic Interneurons in Epilepsy. Epilepsy Curr 2021; 22:54-60. [PMID: 35233202 PMCID: PMC8832350 DOI: 10.1177/15357597211053687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Epileptic seizures are associated with excessive neuronal spiking. Perisomatic
γ-aminobutyric acid (GABA)ergic interneurons specifically innervate the subcellular
domains of postsynaptic excitatory cells that are critical for spike generation. With a
revolution in transcriptomics-based cell taxonomy driving the development of novel
transgenic mouse lines, selectively monitoring and modulating previously elusive
interneuron types is becoming increasingly feasible. Emerging evidence suggests that the
three types of hippocampal perisomatic interneurons, axo-axonic cells, along with
parvalbumin- and cholecystokinin-expressing basket cells, each follow unique activity
patterns in vivo, suggesting distinctive roles in regulating epileptic networks.
Collapse
Affiliation(s)
- Barna Dudok
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Peter M. Klein
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
38
|
Kuljis DA, Micheva KD, Ray A, Wegner W, Bowman R, Madison DV, Willig KI, Barth AL. Gephyrin-Lacking PV Synapses on Neocortical Pyramidal Neurons. Int J Mol Sci 2021; 22:ijms221810032. [PMID: 34576197 PMCID: PMC8467468 DOI: 10.3390/ijms221810032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/26/2022] Open
Abstract
Gephyrin has long been thought of as a master regulator for inhibitory synapses, acting as a scaffold to organize γ-aminobutyric acid type A receptors (GABAARs) at the post-synaptic density. Accordingly, gephyrin immunostaining has been used as an indicator of inhibitory synapses; despite this, the pan-synaptic localization of gephyrin to specific classes of inhibitory synapses has not been demonstrated. Genetically encoded fibronectin intrabodies generated with mRNA display (FingRs) against gephyrin (Gephyrin.FingR) reliably label endogenous gephyrin, and can be tagged with fluorophores for comprehensive synaptic quantitation and monitoring. Here we investigated input- and target-specific localization of gephyrin at a defined class of inhibitory synapse, using Gephyrin.FingR proteins tagged with EGFP in brain tissue from transgenic mice. Parvalbumin-expressing (PV) neuron presynaptic boutons labeled using Cre- dependent synaptophysin-tdTomato were aligned with postsynaptic Gephyrin.FingR puncta. We discovered that more than one-third of PV boutons adjacent to neocortical pyramidal (Pyr) cell somas lack postsynaptic gephyrin labeling. This finding was confirmed using correlative fluorescence and electron microscopy. Our findings suggest some inhibitory synapses may lack gephyrin. Gephyrin-lacking synapses may play an important role in dynamically regulating cell activity under different physiological conditions.
Collapse
Affiliation(s)
- Dika A. Kuljis
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Kristina D. Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Ajit Ray
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Waja Wegner
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ryan Bowman
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Daniel V. Madison
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Katrin I. Willig
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Alison L. Barth
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
- Correspondence: ; Tel.: +1-412-268-1198
| |
Collapse
|
39
|
Yang Y, Paul A, Bach TN, Huang ZJ, Zhang MQ. Single-cell alternative polyadenylation analysis delineates GABAergic neuron types. BMC Biol 2021; 19:144. [PMID: 34301239 PMCID: PMC8299648 DOI: 10.1186/s12915-021-01076-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 06/17/2021] [Indexed: 01/10/2023] Open
Abstract
Background Alternative polyadenylation (APA) is emerging as an important mechanism in the post-transcriptional regulation of gene expression across eukaryotic species. Recent studies have shown that APA plays key roles in biological processes, such as cell proliferation and differentiation. Single-cell RNA-seq technologies are widely used in gene expression heterogeneity studies; however, systematic studies of APA at the single-cell level are still lacking. Results Here, we described a novel computational framework, SAPAS, that utilizes 3′-tag-based scRNA-seq data to identify novel poly(A) sites and quantify APA at the single-cell level. Applying SAPAS to the scRNA-seq data of phenotype characterized GABAergic interneurons, we identified cell type-specific APA events for different GABAergic neuron types. Genes with cell type-specific APA events are enriched for synaptic architecture and communications. In further, we observed a strong enrichment of heritability for several psychiatric disorders and brain traits in altered 3′ UTRs and coding sequences of cell type-specific APA events. Finally, by exploring the modalities of APA, we discovered that the bimodal APA pattern of Pak3 could classify chandelier cells into different subpopulations that are from different laminar positions. Conclusions We established a method to characterize APA at the single-cell level. When applied to a scRNA-seq dataset of GABAergic interneurons, the single-cell APA analysis not only identified cell type-specific APA events but also revealed that the modality of APA could classify cell subpopulations. Thus, SAPAS will expand our understanding of cellular heterogeneity. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01076-3.
Collapse
Affiliation(s)
- Yang Yang
- Present Address: Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.,Department of Biological Sciences, Center for Systems Biology, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Anirban Paul
- Cold Spring Harbor Laboratory, Harbor, Cold Spring, NY, 11724, USA.,Deparment of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Thao Nguyen Bach
- Department of Biological Sciences, Center for Systems Biology, The University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Z Josh Huang
- Cold Spring Harbor Laboratory, Harbor, Cold Spring, NY, 11724, USA.,Deparment of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Michael Q Zhang
- Department of Biological Sciences, Center for Systems Biology, The University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
40
|
Umschweif G, Medrihan L, McCabe KA, Sagi Y, Greengard P. Activation of the p11/SMARCA3/Neurensin-2 pathway in parvalbumin interneurons mediates the response to chronic antidepressants. Mol Psychiatry 2021; 26:3350-3362. [PMID: 33723417 PMCID: PMC8505248 DOI: 10.1038/s41380-021-01059-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 02/02/2021] [Accepted: 02/18/2021] [Indexed: 12/26/2022]
Abstract
The delayed behavioral response to chronic antidepressants depends on dynamic changes in the hippocampus. It was suggested that the antidepressant protein p11 and the chromatin remodeling factor SMARCA3 mediate this delayed response by inducing transcriptional changes in hippocampal neurons. However, what target genes are regulated by the p11/SMARCA3 complex to mediate the behavioral response to antidepressants, and what cell type mediates these molecular changes remain unknown. Here we report that the p11/SMARCA3 complex represses Neurensin-2 transcription in hippocampal parvalbumin-expressing interneurons after chronic treatment with Selective Serotonin Reuptake Inhibitors (SSRI). The behavioral response to antidepressants requires upregulation of p11, accumulation of SMARCA3 in the cell nucleus, and a consequent repression of Neurensin-2 transcription in these interneurons. We elucidate a functional role for p11/SMARCA3/Neurensin-2 pathway in regulating AMPA-receptor signaling in parvalbumin-expressing interneurons, a function that is enhanced by chronic treatment with SSRIs. These results link SSRIs to dynamic glutamatergic changes and implicate p11/SMARCA3/Neurensin-2 pathway in the development of more specific and efficient therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gali Umschweif
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Lucian Medrihan
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Kathryn A McCabe
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Yotam Sagi
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA.
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| |
Collapse
|
41
|
Rahmati N, Normoyle KP, Glykys J, Dzhala VI, Lillis KP, Kahle KT, Raiyyani R, Jacob T, Staley KJ. Unique Actions of GABA Arising from Cytoplasmic Chloride Microdomains. J Neurosci 2021; 41:4957-4975. [PMID: 33903223 PMCID: PMC8197632 DOI: 10.1523/jneurosci.3175-20.2021] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/10/2021] [Accepted: 04/10/2021] [Indexed: 12/21/2022] Open
Abstract
Developmental, cellular, and subcellular variations in the direction of neuronal Cl- currents elicited by GABAA receptor activation have been frequently reported. We found a corresponding variance in the GABAA receptor reversal potential (EGABA) for synapses originating from individual interneurons onto a single pyramidal cell. These findings suggest a similar heterogeneity in the cytoplasmic intracellular concentration of chloride ([Cl-]i) in individual dendrites. We determined [Cl-]i in the murine hippocampus and cerebral cortex of both sexes by (1) two-photon imaging of the Cl--sensitive, ratiometric fluorescent protein SuperClomeleon; (2) Fluorescence Lifetime IMaging (FLIM) of the Cl--sensitive fluorophore MEQ (6-methoxy-N-ethylquinolinium); and (3) electrophysiological measurements of EGABA by pressure application of GABA and RuBi-GABA uncaging. Fluorometric and electrophysiological estimates of local [Cl-]i were highly correlated. [Cl-]i microdomains persisted after pharmacological inhibition of cation-chloride cotransporters, but were progressively modified after inhibiting the polymerization of the anionic biopolymer actin. These methods collectively demonstrated stable [Cl-]i microdomains in individual neurons in vitro and in vivo and the role of immobile anions in its stability. Our results highlight the existence of functionally significant neuronal Cl- microdomains that modify the impact of GABAergic inputs.SIGNIFICANCE STATEMENT Microdomains of varying chloride concentrations in the neuronal cytoplasm are a predictable consequence of the inhomogeneous distribution of anionic polymers such as actin, tubulin, and nucleic acids. Here, we demonstrate the existence and stability of these microdomains, as well as the consequence for GABAergic synaptic signaling: each interneuron produces a postsynaptic GABAA response with a unique reversal potential. In individual hippocampal pyramidal cells, the range of GABAA reversal potentials evoked by stimulating different interneurons was >20 mV. Some interneurons generated postsynaptic responses in pyramidal cells that reversed at potentials beyond what would be considered purely inhibitory. Cytoplasmic chloride microdomains enable each pyramidal cell to maintain a compendium of unique postsynaptic responses to the activity of individual interneurons.
Collapse
Affiliation(s)
- Negah Rahmati
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kieran P Normoyle
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Joseph Glykys
- Department of Pediatrics and Neurology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Volodymyr I Dzhala
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kyle P Lillis
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kristopher T Kahle
- Departments of Neurosurgery, Pediatrics, and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut 06510
| | - Rehan Raiyyani
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Theju Jacob
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Kevin J Staley
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts 02114
| |
Collapse
|
42
|
Dudok B, Klein PM, Hwaun E, Lee BR, Yao Z, Fong O, Bowler JC, Terada S, Sparks FT, Szabo GG, Farrell JS, Berg J, Daigle TL, Tasic B, Dimidschstein J, Fishell G, Losonczy A, Zeng H, Soltesz I. Alternating sources of perisomatic inhibition during behavior. Neuron 2021; 109:997-1012.e9. [PMID: 33529646 PMCID: PMC7979482 DOI: 10.1016/j.neuron.2021.01.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/02/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Interneurons expressing cholecystokinin (CCK) and parvalbumin (PV) constitute two key GABAergic controllers of hippocampal pyramidal cell output. Although the temporally precise and millisecond-scale inhibitory regulation of neuronal ensembles delivered by PV interneurons is well established, the in vivo recruitment patterns of CCK-expressing basket cell (BC) populations has remained unknown. We show in the CA1 of the mouse hippocampus that the activity of CCK BCs inversely scales with both PV and pyramidal cell activity at the behaviorally relevant timescales of seconds. Intervention experiments indicated that the inverse coupling of CCK and PV GABAergic systems arises through a mechanism involving powerful inhibitory control of CCK BCs by PV cells. The tightly coupled complementarity of two key microcircuit regulatory modules demonstrates a novel form of brain-state-specific segregation of inhibition during spontaneous behavior.
Collapse
Affiliation(s)
- Barna Dudok
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| | - Peter M Klein
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Ernie Hwaun
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Brian R Lee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Olivia Fong
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - John C Bowler
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Satoshi Terada
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Fraser T Sparks
- Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Gergely G Szabo
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jordan S Farrell
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Jim Berg
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tanya L Daigle
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gord Fishell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
43
|
Miczán V, Kelemen K, Glavinics JR, László ZI, Barti B, Kenesei K, Kisfali M, Katona I. NECAB1 and NECAB2 are Prevalent Calcium-Binding Proteins of CB1/CCK-Positive GABAergic Interneurons. Cereb Cortex 2021; 31:1786-1806. [PMID: 33230531 PMCID: PMC7869086 DOI: 10.1093/cercor/bhaa326] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
The molecular repertoire of the "Ca2+-signaling toolkit" supports the specific kinetic requirements of Ca2+-dependent processes in different neuronal types. A well-known example is the unique expression pattern of calcium-binding proteins, such as parvalbumin, calbindin, and calretinin. These cytosolic Ca2+-buffers control presynaptic and somatodendritic processes in a cell-type-specific manner and have been used as neurochemical markers of GABAergic interneuron types for decades. Surprisingly, to date no typifying calcium-binding proteins have been found in CB1 cannabinoid receptor/cholecystokinin (CB1/CCK)-positive interneurons that represent a large population of GABAergic cells in cortical circuits. Because CB1/CCK-positive interneurons display disparate presynaptic and somatodendritic Ca2+-transients compared with other interneurons, we tested the hypothesis that they express alternative calcium-binding proteins. By in silico data mining in mouse single-cell RNA-seq databases, we identified high expression of Necab1 and Necab2 genes encoding N-terminal EF-hand calcium-binding proteins 1 and 2, respectively, in CB1/CCK-positive interneurons. Fluorescent in situ hybridization and immunostaining revealed cell-type-specific distribution of NECAB1 and NECAB2 throughout the isocortex, hippocampal formation, and basolateral amygdala complex. Combination of patch-clamp electrophysiology, confocal, and STORM super-resolution microscopy uncovered subcellular nanoscale differences indicating functional division of labor between the two calcium-binding proteins. These findings highlight NECAB1 and NECAB2 as predominant calcium-binding proteins in CB1/CCK-positive interneurons.
Collapse
Affiliation(s)
- Vivien Miczán
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Roska Tamás Doctoral School of Sciences and Technology, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest 1083, Hungary
| | - Krisztina Kelemen
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Physiology, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, Târgu Mureș 540142, Romania
| | - Judit R Glavinics
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Zsófia I László
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Benjámin Barti
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest 1083, Hungary
| | - Kata Kenesei
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Máté Kisfali
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest 1083, Hungary
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
44
|
Ballaz SJ, Bourin M. Cholecystokinin-Mediated Neuromodulation of Anxiety and Schizophrenia: A "Dimmer-Switch" Hypothesis. Curr Neuropharmacol 2021; 19:925-938. [PMID: 33185164 PMCID: PMC8686311 DOI: 10.2174/1570159x18666201113145143] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/08/2020] [Accepted: 11/10/2020] [Indexed: 11/22/2022] Open
Abstract
Cholecystokinin (CCK), the most abundant brain neuropeptide, is involved in relevant behavioral functions like memory, cognition, and reward through its interactions with the opioid and dopaminergic systems in the limbic system. CCK excites neurons by binding two receptors, CCK1 and CCK2, expressed at low and high levels in the brain, respectively. Historically, CCK2 receptors have been related to the induction of panic attacks in humans. Disturbances in brain CCK expression also underlie the physiopathology of schizophrenia, which is attributed to the modulation by CCK1 receptors of the dopamine flux in the basal striatum. Despite this evidence, neither CCK2 receptor antagonists ameliorate human anxiety nor CCK agonists have consistently shown neuroleptic effects in clinical trials. A neglected aspect of the function of brain CCK is its neuromodulatory role in mental disorders. Interestingly, CCK is expressed in pivotal inhibitory interneurons that sculpt cortical dynamics and the flux of nerve impulses across corticolimbic areas and the excitatory projections to mesolimbic pathways. At the basal striatum, CCK modulates the excitability of glutamate, the release of inhibitory GABA, and the discharge of dopamine. Here we focus on how CCK may reduce rather than trigger anxiety by regulating its cognitive component. Adequate levels of CCK release in the basal striatum may control the interplay between cognition and reward circuitry, which is critical in schizophrenia. Hence, it is proposed that disturbances in the excitatory/ inhibitory interplay modulated by CCK may contribute to the imbalanced interaction between corticolimbic and mesolimbic neural activity found in anxiety and schizophrenia.
Collapse
Affiliation(s)
- Santiago J. Ballaz
- Address correspondence to this author at the School of Biological Sciences & Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí, Ecuador; Tel: 593 (06) 299 9100, ext. 2626; E-mail:
| | | |
Collapse
|
45
|
Perisomatic innervation and neurochemical features of giant pyramidal neurons in both hemispheres of the human primary motor cortex. Brain Struct Funct 2020; 226:281-296. [PMID: 33355694 PMCID: PMC7817582 DOI: 10.1007/s00429-020-02182-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
Betz cells-the gigantopyramidal neurons found in high amount in the primary motor cortex-are among of the most characteristic neuronal cells. A part of them contains the calcium-binding protein parvalbumin (PV) in primates. However, less is known about these cells in the human motor cortex despite their important role in different neurological disorders. Therefore, the aim of our study was to investigate the neurochemical features and perisomatic input properties of Betz cells in control human samples with short post-mortem interval. We used different microscopic techniques to investigate the primary motor cortex of both hemispheres. The soma size and density, and expression of PV of the Betz cells were investigated. Furthermore, we used confocal fluorescent and electron microscopy to examine their perisomatic input. The soma size and density showed moderate variability among samples and hemispheres. Post-mortem interval and hemispherical localization did not influence these features. Around 70% of Betz cells expressed PV, but in less intensity than the cortical interneurons. Betz neurons receive dense perisomatic input, which are mostly VIAAT- (vesicular inhibitory amino acid transporter) and PV immunopositive. In the electron microscope, we found PV-immunolabelled terminals with asymmetric-like synaptic structure, too. Terminals with morphologically similar synaptic specialisation were also found among vGluT2- (vesicular glutamate transporter type 2) immunostained terminals contacting Betz cells. Our data suggest that Betz cells' morphological properties showed less variability among subjects and hemispheres than the density of them. Their neurochemical and perisomatic input characteristics support their role in execution of fast and precise movements.
Collapse
|
46
|
Koster AK, Reese AL, Kuryshev Y, Wen X, McKiernan KA, Gray EE, Wu C, Huguenard JR, Maduke M, Du Bois J. Development and validation of a potent and specific inhibitor for the CLC-2 chloride channel. Proc Natl Acad Sci U S A 2020; 117:32711-32721. [PMID: 33277431 PMCID: PMC7768775 DOI: 10.1073/pnas.2009977117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
CLC-2 is a voltage-gated chloride channel that is widely expressed in mammalian tissues. In the central nervous system, CLC-2 appears in neurons and glia. Studies to define how this channel contributes to normal and pathophysiological function in the central nervous system raise questions that remain unresolved, in part due to the absence of precise pharmacological tools for modulating CLC-2 activity. Herein, we describe the development and optimization of AK-42, a specific small-molecule inhibitor of CLC-2 with nanomolar potency (IC50 = 17 ± 1 nM). AK-42 displays unprecedented selectivity (>1,000-fold) over CLC-1, the closest CLC-2 homolog, and exhibits no off-target engagement against a panel of 61 common channels, receptors, and transporters expressed in brain tissue. Computational docking, validated by mutagenesis and kinetic studies, indicates that AK-42 binds to an extracellular vestibule above the channel pore. In electrophysiological recordings of mouse CA1 hippocampal pyramidal neurons, AK-42 acutely and reversibly inhibits CLC-2 currents; no effect on current is observed on brain slices taken from CLC-2 knockout mice. These results establish AK-42 as a powerful tool for investigating CLC-2 neurophysiology.
Collapse
Affiliation(s)
- Anna K Koster
- Department of Chemistry, Stanford University, Stanford, CA 94305
- Department of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Austin L Reese
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Yuri Kuryshev
- Charles River Laboratories Cleveland, Inc., Cleveland, OH 44128
| | - Xianlan Wen
- Department of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Keri A McKiernan
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Erin E Gray
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Caiyun Wu
- Charles River Laboratories Cleveland, Inc., Cleveland, OH 44128
| | - John R Huguenard
- Department of Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305;
| | - Merritt Maduke
- Department of Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305;
| | - J Du Bois
- Department of Chemistry, Stanford University, Stanford, CA 94305;
| |
Collapse
|
47
|
Traumatic Brain Injury Diminishes Feedforward Activation of Parvalbumin-Expressing Interneurons in the Dentate Gyrus. eNeuro 2020; 7:ENEURO.0195-19.2020. [PMID: 33106385 PMCID: PMC7675145 DOI: 10.1523/eneuro.0195-19.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/14/2020] [Accepted: 09/20/2020] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with aberrant network hyperexcitability in the dentate gyrus (DG). GABAAergic parvalbumin-expressing interneurons (PV-INs) in the DG regulate network excitability with strong, perisomatic inhibition, although the posttraumatic effects on PV-IN function after TBI are not well understood. In this study, we investigated physiological alterations in PV-INs one week after mild lateral fluid percussion injury (LFPI) in mice. PV-IN cell loss was observed in the dentate hilus after LFPI, with surviving PV-INs showing no change in intrinsic membrane properties. Whole-cell voltage clamp recordings in PV-INs revealed alterations in both EPSCs and IPSCs (EPSCs/IPSCs). Evoked EPSCs (eEPSCs) in PV-INs from perforant path electrical stimulation were diminished after injury but could be recovered with application of a GABAA-receptor antagonist. Furthermore, current-clamp recordings using minimal perforant path stimulation demonstrated a decrease in evoked PV-IN action potentials (APs) after LFPI, which could be restored by blocking GABAAergic inhibition. Together, these findings suggest that injury alters synaptic input onto PV-INs, resulting in a net inhibitory effect that reduces feedforward PV-IN activation in the DG. Decreased PV-IN activation suggests a potential mechanism of DG network hyperexcitability contributing to hippocampal dysfunction after TBI.
Collapse
|
48
|
Liu X, Dimidschstein J, Fishell G, Carter AG. Hippocampal inputs engage CCK+ interneurons to mediate endocannabinoid-modulated feed-forward inhibition in the prefrontal cortex. eLife 2020; 9:e55267. [PMID: 33034285 PMCID: PMC7609047 DOI: 10.7554/elife.55267] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022] Open
Abstract
Connections from the ventral hippocampus (vHPC) to the prefrontal cortex (PFC) regulate cognition, emotion, and memory. These functions are also tightly controlled by inhibitory networks in the PFC, whose disruption is thought to contribute to mental health disorders. However, relatively little is known about how the vHPC engages different populations of interneurons in the PFC. Here we use slice physiology and optogenetics to study vHPC-evoked feed-forward inhibition in the mouse PFC. We first show that cholecystokinin (CCK+), parvalbumin (PV+), and somatostatin (SOM+) expressing interneurons are prominent in layer 5 (L5) of infralimbic PFC. We then show that vHPC inputs primarily activate CCK+ and PV+ interneurons, with weaker connections onto SOM+ interneurons. CCK+ interneurons make stronger synapses onto pyramidal tract (PT) cells over nearby intratelencephalic (IT) cells. However, CCK+ inputs undergo depolarization-induced suppression of inhibition (DSI) and CB1 receptor modulation only at IT cells. Moreover, vHPC-evoked feed-forward inhibition undergoes DSI only at IT cells, confirming a central role for CCK+ interneurons. Together, our findings show how vHPC directly engages multiple populations of inhibitory cells in deep layers of the infralimbic PFC, highlighting unexpected roles for both CCK+ interneurons and endocannabinoid modulation in hippocampal-prefrontal communication.
Collapse
Affiliation(s)
- Xingchen Liu
- Center for Neural Science, New York University, New York, United States
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Boston, United States
| | - Gordon Fishell
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Boston, United States
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Adam G Carter
- Center for Neural Science, New York University, New York, United States
| |
Collapse
|
49
|
Sullivan CS, Mohan V, Manis PB, Moy SS, Truong Y, Duncan BW, Maness PF. Developmental Regulation of Basket Interneuron Synapses and Behavior through NCAM in Mouse Prefrontal Cortex. Cereb Cortex 2020; 30:4689-4707. [PMID: 32249896 PMCID: PMC7325800 DOI: 10.1093/cercor/bhaa074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/07/2019] [Indexed: 02/07/2023] Open
Abstract
Parvalbumin (PV)-expressing basket interneurons in the prefrontal cortex (PFC) regulate pyramidal cell firing, synchrony, and network oscillations. Yet, it is unclear how their perisomatic inputs to pyramidal neurons are integrated into neural circuitry and adjusted postnatally. Neural cell adhesion molecule NCAM is expressed in a variety of cells in the PFC and cooperates with EphrinA/EphAs to regulate inhibitory synapse density. Here, analysis of a novel parvalbumin (PV)-Cre: NCAM F/F mouse mutant revealed that NCAM functions presynaptically in PV+ basket interneurons to regulate postnatal elimination of perisomatic synapses. Mutant mice exhibited an increased density of PV+ perisomatic puncta in PFC layer 2/3, while live imaging in mutant brain slices revealed fewer puncta that were dynamically eliminated. Furthermore, EphrinA5-induced growth cone collapse in PV+ interneurons in culture depended on NCAM expression. Electrophysiological recording from layer 2/3 pyramidal cells in mutant PFC slices showed a slower rise time of inhibitory synaptic currents. PV-Cre: NCAM F/F mice exhibited impairments in working memory and social behavior that may be impacted by altered PFC circuitry. These findings suggest that the density of perisomatic synapses of PV+ basket interneurons is regulated postnatally by NCAM, likely through EphrinA-dependent elimination, which is important for appropriate PFC network function and behavior.
Collapse
Affiliation(s)
- Chelsea S Sullivan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Vishwa Mohan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Paul B Manis
- Department of Otolaryngology/Head and Neck Surgery, and Cell Biology and Physiology, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sheryl S Moy
- Department of Psychiatry, Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Young Truong
- Department of Biostatistics, School of Global Public Health, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bryce W Duncan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patricia F Maness
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
50
|
D'Amour JA, Ekins T, Ganatra S, Yuan X, McBain CJ. Aberrant sorting of hippocampal complex pyramidal cells in type I lissencephaly alters topological innervation. eLife 2020; 9:55173. [PMID: 32558643 PMCID: PMC7340499 DOI: 10.7554/elife.55173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/19/2020] [Indexed: 11/30/2022] Open
Abstract
Layering has been a long-appreciated feature of higher order mammalian brain structures but the extent to which it plays an instructive role in synaptic specification remains unknown. Here we examine the formation of synaptic circuitry under cellular heterotopia in hippocampal CA1, using a mouse model of the human neurodevelopmental disorder Type I Lissencephaly. We identify calbindin-expressing principal cells which are mispositioned under cellular heterotopia. Ectopic calbindin-expressing principal cells develop relatively normal morphological features and stunted intrinsic physiological features. Regarding network development, a connectivity preference for cholecystokinin-expressing interneurons to target calbindin-expressing principal cells is diminished. Moreover, in vitro gamma oscillatory activity is less synchronous across heterotopic bands and mutants are less responsive to pharmacological inhibition of cholecystokinin-containing interneurons. This study will aid not only in our understanding of how cellular networks form but highlight vulnerable cellular circuit motifs that might be generalized across disease states.
Collapse
Affiliation(s)
- James A D'Amour
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States.,Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, Bethesda, United States
| | - Tyler Ekins
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States.,Brown University, Department of Neuroscience, Providence, United States
| | - Stuti Ganatra
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Xiaoqing Yuan
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Chris J McBain
- Program in Developmental Neurobiology, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| |
Collapse
|