1
|
Tzilivaki A, Tukker JJ, Maier N, Poirazi P, Sammons RP, Schmitz D. Hippocampal GABAergic interneurons and memory. Neuron 2023; 111:3154-3175. [PMID: 37467748 PMCID: PMC10593603 DOI: 10.1016/j.neuron.2023.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/04/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
One of the most captivating questions in neuroscience revolves around the brain's ability to efficiently and durably capture and store information. It must process continuous input from sensory organs while also encoding memories that can persist throughout a lifetime. What are the cellular-, subcellular-, and network-level mechanisms that underlie this remarkable capacity for long-term information storage? Furthermore, what contributions do distinct types of GABAergic interneurons make to this process? As the hippocampus plays a pivotal role in memory, our review focuses on three aspects: (1) delineation of hippocampal interneuron types and their connectivity, (2) interneuron plasticity, and (3) activity patterns of interneurons during memory-related rhythms, including the role of long-range interneurons and disinhibition. We explore how these three elements, together showcasing the remarkable diversity of inhibitory circuits, shape the processing of memories in the hippocampus.
Collapse
Affiliation(s)
- Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany; NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany
| | - John J Tukker
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
| | - Nikolaus Maier
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
| | - Panayiota Poirazi
- Foundation for Research and Technology Hellas (FORTH), Institute of Molecular Biology and Biotechnology (IMBB), N. Plastira 100, Heraklion, Crete, Greece
| | - Rosanna P Sammons
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
| | - Dietmar Schmitz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany; Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany; NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany; Bernstein Center for Computational Neuroscience, Humboldt-Universität zu Berlin, Philippstrasse. 13, 10115 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany.
| |
Collapse
|
2
|
Kodirov SA, Bonni K, Wehrmeister M, Lutz B. Depolarization-initiated endogenous cannabinoid release and underlying retrograde neurotransmission in interneurons of amygdala. ACTA ACUST UNITED AC 2021; 28:44-52. [PMID: 33452114 PMCID: PMC7812861 DOI: 10.1101/lm.052555.120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022]
Abstract
The depolarization is also important for the short-term synaptic plasticity, known as depolarization-induced suppression of excitation (DSE). The two major types of neurons and their synapses in the lateral nucleus of amygdala (LA) are prone to plasticity. However, DSE in interneurons has not been reported in amygdala in general and in LA in particular. Therefore, we conducted the patch-clamp experiments with LA interneurons. These neurons were identified by lack of adaptation in firing rate of action potentials. In this study, we show for the first time a transient suppression of neurotransmission at synapses both within the local network and between cortical inputs and interneurons of the LA. The retrograde neurotransmission from GABAergic interneurons were comparable with that of glutamatergic pyramidal cells. That is the axonal terminals of cortical inputs do not posses selectivity toward two neuronal subtypes. However, the DSE of both types of neurons involve an increase in intracellular Ca2+ and the release of endogenous cannabinoids (eCB) and activation of presynaptic CB1 receptors. The magnitude of DSE was significantly higher in interneurons compared with pyramidal cells, though developed with some latency.
Collapse
Affiliation(s)
- Sodikdjon A Kodirov
- Institute of Physiological Chemistry and Pathobiochemistry, University Medical Center Mainz, 55099 Mainz, Germany.,Center for Biomedical Studies, Department of Biological Sciences, University of Texas at Brownsville, Texas 78520, USA
| | - Kathrin Bonni
- Institute of Physiological Chemistry and Pathobiochemistry, University Medical Center Mainz, 55099 Mainz, Germany
| | - Michael Wehrmeister
- Institute of Physiological Chemistry and Pathobiochemistry, University Medical Center Mainz, 55099 Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry and Pathobiochemistry, University Medical Center Mainz, 55099 Mainz, Germany
| |
Collapse
|
3
|
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: 9] [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
|
4
|
Petrache AL, Khan AA, Nicholson MW, Monaco A, Kuta-Siejkowska M, Haider S, Hilton S, Jovanovic JN, Ali AB. Selective Modulation of α5 GABA A Receptors Exacerbates Aberrant Inhibition at Key Hippocampal Neuronal Circuits in APP Mouse Model of Alzheimer's Disease. Front Cell Neurosci 2020; 14:568194. [PMID: 33262690 PMCID: PMC7686552 DOI: 10.3389/fncel.2020.568194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/28/2020] [Indexed: 11/13/2022] Open
Abstract
Selective negative allosteric modulators (NAMs), targeting α5 subunit-containing GABAA receptors (GABAARs) as potential therapeutic targets for disorders associated with cognitive deficits, including Alzheimer's disease (AD), continually fail clinical trials. We investigated whether this was due to the change in the expression of α5 GABAARs, consequently altering synaptic function during AD pathogenesis. Using medicinal chemistry and computational modeling, we developed aqueous soluble hybrids of 6,6-dimethyl-3-(2-hydroxyethyl) thio-1-(thiazol-2-yl)-6,7-dihydro-2-benzothiophene-4(5H)-one, that demonstrated selective binding and high negative allosteric modulation, specifically for the α5 GABAAR subtypes in constructed HEK293 stable cell-lines. Using a knock-in mouse model of AD (APP NL-F/NL-F), which expresses a mutant form of human amyloid-β (Aβ), we performed immunofluorescence studies combined with electrophysiological whole-cell recordings to investigate the effects of our key molecule, α5-SOP002 in the hippocampal CA1 region. In aged APP NL-F/NL-F mice, selective preservation of α5 GABAARs was observed in, calretinin- (CR), cholecystokinin- (CCK), somatostatin- (SST) expressing interneurons, and pyramidal cells. Previously, we reported that CR dis-inhibitory interneurons, specialized in regulating other interneurons displayed abnormally high levels of synaptic inhibition in the APP NL-F/NL-F mouse model, here we show that this excessive inhibition was "normalized" to control values with bath-applied α5-SOP002 (1 μM). However, α5-SOP002, further impaired inhibition onto CCK and pyramidal cells that were already largely compromised by exhibiting a deficit of inhibition in the AD model. In summary, using a multi-disciplinary approach, we show that exposure to α5 GABAAR NAMs may further compromise aberrant synapses in AD. We, therefore, suggest that the α5 GABAAR is not a suitable therapeutic target for the treatment of AD or other cognitive deficits due to the widespread neuronal-networks that use α5 GABAARs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Afia B Ali
- UCL School of Pharmacy, London, United Kingdom
| |
Collapse
|
5
|
Ecker A, Romani A, Sáray S, Káli S, Migliore M, Falck J, Lange S, Mercer A, Thomson AM, Muller E, Reimann MW, Ramaswamy S. Data-driven integration of hippocampal CA1 synaptic physiology in silico. Hippocampus 2020; 30:1129-1145. [PMID: 32520422 PMCID: PMC7687201 DOI: 10.1002/hipo.23220] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 04/20/2020] [Accepted: 05/07/2020] [Indexed: 12/31/2022]
Abstract
The anatomy and physiology of monosynaptic connections in rodent hippocampal CA1 have been extensively studied in recent decades. Yet, the resulting knowledge remains disparate and difficult to reconcile. Here, we present a data‐driven approach to integrate the current state‐of‐the‐art knowledge on the synaptic anatomy and physiology of rodent hippocampal CA1, including axo‐dendritic innervation patterns, number of synapses per connection, quantal conductances, neurotransmitter release probability, and short‐term plasticity into a single coherent resource. First, we undertook an extensive literature review of paired recordings of hippocampal neurons and compiled experimental data on their synaptic anatomy and physiology. The data collected in this manner is sparse and inhomogeneous due to the diversity of experimental techniques used by different groups, which necessitates the need for an integrative framework to unify these data. To this end, we extended a previously developed workflow for the neocortex to constrain a unifying in silico reconstruction of the synaptic physiology of CA1 connections. Our work identifies gaps in the existing knowledge and provides a complementary resource toward a more complete quantification of synaptic anatomy and physiology in the rodent hippocampal CA1 region.
Collapse
Affiliation(s)
- András Ecker
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Campus Biotech, Geneva, Switzerland
| | - Armando Romani
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Campus Biotech, Geneva, Switzerland
| | - Sára Sáray
- Institute of Experimental Medicine, Budapest, Hungary.,Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Szabolcs Káli
- Institute of Experimental Medicine, Budapest, Hungary.,Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Michele Migliore
- Institute of Biophysics, National Research Council, Palermo, Italy
| | - Joanne Falck
- UCL School of Pharmacy, University College London, London, UK
| | - Sigrun Lange
- UCL School of Pharmacy, University College London, London, UK.,School of Life Sciences, University of Westminster, London, UK
| | - Audrey Mercer
- UCL School of Pharmacy, University College London, London, UK
| | - Alex M Thomson
- UCL School of Pharmacy, University College London, London, UK
| | - Eilif Muller
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Campus Biotech, Geneva, Switzerland
| | - Michael W Reimann
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Campus Biotech, Geneva, Switzerland
| | - Srikanth Ramaswamy
- Blue Brain Project, École Polytechnique Fédérale de Lausanne, Campus Biotech, Geneva, Switzerland
| |
Collapse
|
6
|
Shi A, Petrache AL, Shi J, Ali AB. Preserved Calretinin Interneurons in an App Model of Alzheimer's Disease Disrupt Hippocampal Inhibition via Upregulated P2Y1 Purinoreceptors. Cereb Cortex 2020; 30:1272-1290. [PMID: 31407772 PMCID: PMC7132926 DOI: 10.1093/cercor/bhz165] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/15/2019] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
To understand the pathogenesis of specific neuronal circuit dysfunction in Alzheimer's disease (AD), we investigated the fate of three subclasses of "modulatory interneurons" in hippocampal CA1 using the AppNL-F/NL-F knock-in mouse model of AD. Cholecystokinin- and somatostatin-expressing interneurons were aberrantly hyperactive preceding the presence of the typical AD hallmarks: neuroinflammation and amyloid-β (Aβ) accumulation. These interneurons showed an age-dependent vulnerability to Aβ penetration and a reduction in density and coexpression of the inhibitory neurotransmitter GABA synthesis enzyme, glutamic acid decarboxylase 67 (GAD67), suggesting a loss in their inhibitory function. However, calretinin (CR) interneurons-specialized to govern only inhibition, showed resilience to Aβ accumulation, preservation of structure, and displayed synaptic hyperinhibition, despite the lack of inhibitory control of CA1 excitatory pyramidal cells from midstages of the disease. This aberrant inhibitory homeostasis observed in CA1 CR cells and pyramidal cells was "normalized" by blocking P2Y1 purinoreceptors, which were "upregulated" and strongly expressed in CR cells and astrocytes in AppNL-F/NL-F mice in the later stages of AD. In summary, AD-associated cell-type selective destruction of inhibitory interneurons and disrupted inhibitory homeostasis rectified by modulation of the upregulated purinoreceptor system may serve as a novel therapeutic strategy to normalize selective dysfunctional synaptic homeostasis during pathogenesis of AD.
Collapse
Affiliation(s)
- Anqi Shi
- UCL School of Pharmacy, London, WC1N 1AX, UK
| | | | - Jiachen Shi
- UCL School of Pharmacy, London, WC1N 1AX, UK
| | - Afia B Ali
- UCL School of Pharmacy, London, WC1N 1AX, UK
| |
Collapse
|
7
|
Booker SA, Vida I. Morphological diversity and connectivity of hippocampal interneurons. Cell Tissue Res 2018; 373:619-641. [PMID: 30084021 PMCID: PMC6132631 DOI: 10.1007/s00441-018-2882-2] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022]
Abstract
The mammalian forebrain is constructed from ensembles of neurons that form local microcircuits giving rise to the exquisite cognitive tasks the mammalian brain can perform. Hippocampal neuronal circuits comprise populations of relatively homogenous excitatory neurons, principal cells and exceedingly heterogeneous inhibitory neurons, the interneurons. Interneurons release GABA from their axon terminals and are capable of controlling excitability in every cellular compartment of principal cells and interneurons alike; thus, they provide a brake on excess activity, control the timing of neuronal discharge and provide modulation of synaptic transmission. The dendritic and axonal morphology of interneurons, as well as their afferent and efferent connections within hippocampal circuits, is central to their ability to differentially control excitability, in a cell-type- and compartment-specific manner. This review aims to provide an up-to-date compendium of described hippocampal interneuron subtypes, with respect to their morphology, connectivity, neurochemistry and physiology, a full understanding of which will in time help to explain the rich diversity of neuronal function.
Collapse
Affiliation(s)
- Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK.
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, EH8 9XD, UK.
| | - Imre Vida
- Institute for Integrative Neuroanatomy, Charité - Universitätmedizin Berlin, Berlin, Germany.
| |
Collapse
|
8
|
Khan AA, Shekh-Ahmad T, Khalil A, Walker MC, Ali AB. Cannabidiol exerts antiepileptic effects by restoring hippocampal interneuron functions in a temporal lobe epilepsy model. Br J Pharmacol 2018; 175:2097-2115. [PMID: 29574880 PMCID: PMC5979781 DOI: 10.1111/bph.14202] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/13/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
Background and Purpose A non‐psychoactive phytocannabinoid, cannabidiol (CBD), shows promising results as an effective potential antiepileptic drug in some forms of refractory epilepsy. To elucidate the mechanisms by which CBD exerts its anti‐seizure effects, we investigated its effects at synaptic connections and on the intrinsic membrane properties of hippocampal CA1 pyramidal cells and two major inhibitory interneurons: fast spiking, parvalbumin (PV)‐expressing and adapting, cholecystokinin (CCK)‐expressing interneurons. We also investigated whether in vivo treatment with CBD altered the fate of CCK and PV interneurons using immunohistochemistry. Experimental Approach Electrophysiological intracellular whole‐cell recordings combined with neuroanatomy were performed in acute brain slices of rat temporal lobe epilepsy in in vivo (induced by kainic acid) and in vitro (induced by Mg2+‐free solution) epileptic seizure models. For immunohistochemistry experiments, CBD was administered in vivo (100 mg·kg−1) at zero time and 90 min post status epilepticus, induced with kainic acid. Key Results Bath application of CBD (10 μM) dampened excitability at unitary synapses between pyramidal cells but enhanced inhibitory synaptic potentials elicited by fast spiking and adapting interneurons at postsynaptic pyramidal cells. Furthermore, CBD restored impaired membrane excitability of PV, CCK and pyramidal cells in a cell type‐specific manner. These neuroprotective effects of CBD were corroborated by immunohistochemistry experiments that revealed a significant reduction in atrophy and death of PV‐ and CCK‐expressing interneurons after CBD treatment. Conclusions and Implications Our data suggest that CBD restores excitability and morphological impairments in epileptic models to pre‐epilepsy control levels through multiple mechanisms to reinstate normal network function.
Collapse
Affiliation(s)
| | | | | | | | - Afia B Ali
- UCL School of Pharmacy, London, WC1N 1AX, UK
| |
Collapse
|
9
|
Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ. Hippocampal GABAergic Inhibitory Interneurons. Physiol Rev 2017; 97:1619-1747. [PMID: 28954853 DOI: 10.1152/physrev.00007.2017] [Citation(s) in RCA: 495] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022] Open
Abstract
In the hippocampus GABAergic local circuit inhibitory interneurons represent only ~10-15% of the total neuronal population; however, their remarkable anatomical and physiological diversity allows them to regulate virtually all aspects of cellular and circuit function. Here we provide an overview of the current state of the field of interneuron research, focusing largely on the hippocampus. We discuss recent advances related to the various cell types, including their development and maturation, expression of subtype-specific voltage- and ligand-gated channels, and their roles in network oscillations. We also discuss recent technological advances and approaches that have permitted high-resolution, subtype-specific examination of their roles in numerous neural circuit disorders and the emerging therapeutic strategies to ameliorate such pathophysiological conditions. The ultimate goal of this review is not only to provide a touchstone for the current state of the field, but to help pave the way for future research by highlighting where gaps in our knowledge exist and how a complete appreciation of their roles will aid in future therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ramesh Chittajallu
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Michael T Craig
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ludovic Tricoire
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Jason C Wester
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Chris J McBain
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
10
|
Kohus Z, Káli S, Rovira‐Esteban L, Schlingloff D, Papp O, Freund TF, Hájos N, Gulyás AI. Properties and dynamics of inhibitory synaptic communication within the CA3 microcircuits of pyramidal cells and interneurons expressing parvalbumin or cholecystokinin. J Physiol 2016; 594:3745-74. [PMID: 27038232 PMCID: PMC4929320 DOI: 10.1113/jp272231] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/21/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS To understand how a network operates, its elements must be identified and characterized, and the interactions of the elements need to be studied in detail. In the present study, we describe quantitatively the connectivity of two classes of inhibitory neurons in the hippocampal CA3 area (parvalbumin-positive and cholecystokinin-positive interneurons), a key region for the generation of behaviourally relevant synchronous activity patterns. We describe how interactions among these inhibitory cells and their local excitatory target neurons evolve over the course of physiological and pathological activity patterns. The results of the present study enable the construction of precise neuronal network models that may help us understand how network dynamics is generated and how it can underlie information processing and pathological conditions in the brain. We show how inhibitory dynamics between parvalbumin-positive basket cells and pyramidal cells could contribute to sharp wave-ripple generation. ABSTRACT Different hippocampal activity patterns are determined primarily by the interaction of excitatory cells and different types of interneurons. To understand the mechanisms underlying the generation of different network dynamics, the properties of synaptic transmission need to be uncovered. Perisomatic inhibition is critical for the generation of sharp wave-ripples, gamma oscillations and pathological epileptic activities. Therefore, we aimed to quantitatively and systematically characterize the temporal properties of the synaptic transmission between perisomatic inhibitory neurons and pyramidal cells in the CA3 area of mouse hippocampal slices, using action potential patterns recorded during physiological and pathological network states. Parvalbumin-positive (PV+) and cholecystokinin-positive (CCK+) interneurons showed distinct intrinsic physiological features. Interneurons of the same type formed reciprocally connected subnetworks, whereas the connectivity between interneuron classes was sparse. The characteristics of unitary interactions depended on the identity of both synaptic partners, whereas the short-term plasticity of synaptic transmission depended mainly on the presynaptic cell type. PV+ interneurons showed frequency-dependent depression, whereas more complex dynamics characterized the output of CCK+ interneurons. We quantitatively captured the dynamics of transmission at these different types of connection with simple mathematical models, and describe in detail the response to physiological and pathological discharge patterns. Our data suggest that the temporal propeties of PV+ interneuron transmission may contribute to sharp wave-ripple generation. These findings support the view that intrinsic and synaptic features of PV+ cells make them ideally suited for the generation of physiological network oscillations, whereas CCK+ cells implement a more subtle, graded control in the hippocampus.
Collapse
Affiliation(s)
- Z. Kohus
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
- János Szentágothai, PhD Program of Semmelweis UniversityBudapestHungary
| | - S. Káli
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
- Péter Pázmány Catholic UniversityFaculty of Information TechnologyBudapestHungary
| | - L. Rovira‐Esteban
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
| | - D. Schlingloff
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
- János Szentágothai, PhD Program of Semmelweis UniversityBudapestHungary
| | - O. Papp
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
| | - T. F. Freund
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
- Péter Pázmány Catholic UniversityFaculty of Information TechnologyBudapestHungary
| | - N. Hájos
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
| | - A. I. Gulyás
- Institute of Experimental MedicineHungarian Academy of SciencesBudapestHungary
| |
Collapse
|
11
|
Booker SA, Althof D, Gross A, Loreth D, Müller J, Unger A, Fakler B, Varro A, Watanabe M, Gassmann M, Bettler B, Shigemoto R, Vida I, Kulik Á. KCTD12 Auxiliary Proteins Modulate Kinetics of GABABReceptor-Mediated Inhibition in Cholecystokinin-Containing Interneurons. Cereb Cortex 2016; 27:2318-2334. [DOI: 10.1093/cercor/bhw090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
12
|
Time-coded neurotransmitter release at excitatory and inhibitory synapses. Proc Natl Acad Sci U S A 2016; 113:E1108-15. [PMID: 26858411 DOI: 10.1073/pnas.1525591113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Communication between neurons at chemical synapses is regulated by hundreds of different proteins that control the release of neurotransmitter that is packaged in vesicles, transported to an active zone, and released when an input spike occurs. Neurotransmitter can also be released asynchronously, that is, after a delay following the spike, or spontaneously in the absence of a stimulus. The mechanisms underlying asynchronous and spontaneous neurotransmitter release remain elusive. Here, we describe a model of the exocytotic cycle of vesicles at excitatory and inhibitory synapses that accounts for all modes of vesicle release as well as short-term synaptic plasticity (STSP). For asynchronous release, the model predicts a delayed inertial protein unbinding associated with the SNARE complex assembly immediately after vesicle priming. Experiments are proposed to test the model's molecular predictions for differential exocytosis. The simplicity of the model will also facilitate large-scale simulations of neural circuits.
Collapse
|
13
|
Dentate cannabinoid-sensitive interneurons undergo unique and selective strengthening of mutual synaptic inhibition in experimental epilepsy. Neurobiol Dis 2016; 89:23-35. [PMID: 26804027 DOI: 10.1016/j.nbd.2016.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 01/12/2016] [Accepted: 01/19/2016] [Indexed: 11/22/2022] Open
Abstract
Altered inhibition is a salient feature of hippocampal network reorganization in epilepsy. Hippocampal pyramidal cells and dentate granule cells show specific reduction in cannabinoid receptor type 1 (CB1R)-sensitive GABAergic inputs in experimental epilepsy. In the dentate gyrus, CB1Rs regulate synaptic release from accommodating interneurons (AC-INs) with adapting firing characteristics and axonal projections in the molecular layer, but not from fast-spiking basket cells (FS-BCs). However, it is not known whether the intrinsic physiology and synaptic inhibition of AC-INs responsible for CB1R-sensitive inhibition is altered in epilepsy. Using the pilocarpine-induced status epilepticus (SE) model of epilepsy, we find that the basic physiological characteristics of AC-INs in epileptic rats are not different from age-matched controls. In paired interneuronal recordings, the amplitude of unitary inhibitory synaptic currents (uIPSCs) between AC-INs doubled after SE. Non-stationary noise analysis revealed that the post-SE strengthening of synapses between AC-INs resulted from an increase in postsynaptic receptors. Baseline synaptic release and CB1R antagonist enhancement of release at synapses between AC-INs were not different between control and post-SE rats. Additionally, uIPSC amplitude in FS-BCs to AC-INs pairs was unchanged after SE indicating input-specific microcircuit alterations in inhibitory inputs to AC-INs. At the network level, AC-INs showed no reduction in spontaneous and miniature inhibitory synaptic current (sIPSC or mIPSC) frequency or amplitude after SE. However, AC-IN mIPSC amplitude was persistently enhanced in post-SE and epileptic rats. CB1R agonist reduced the amplitude and suppressed a greater proportion of sIPSCs in AC-INs from post-SE and epileptic rats demonstrating a novel, cell-type specific increase in CB1R-sensitive inhibition of AC-INs after SE. This unique post-SE strengthening of inhibition between AC-INs could lead to activity-dependent suppression of AC-IN firing and compromise dentate CB1R-sensitive inhibition in epilepsy.
Collapse
|
14
|
Basu J, Zaremba JD, Cheung SK, Hitti FL, Zemelman BV, Losonczy A, Siegelbaum SA. Gating of hippocampal activity, plasticity, and memory by entorhinal cortex long-range inhibition. Science 2016; 351:aaa5694. [PMID: 26744409 PMCID: PMC4920085 DOI: 10.1126/science.aaa5694] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The cortico-hippocampal circuit is critical for storage of associational memories. Most studies have focused on the role in memory storage of the excitatory projections from entorhinal cortex to hippocampus. However, entorhinal cortex also sends inhibitory projections, whose role in memory storage and cortico-hippocampal activity remains largely unexplored. We found that these long-range inhibitory projections enhance the specificity of contextual and object memory encoding. At the circuit level, these γ-aminobutyric acid (GABA)-releasing projections target hippocampal inhibitory neurons and thus act as a disinhibitory gate that transiently promotes the excitation of hippocampal CA1 pyramidal neurons by suppressing feedforward inhibition. This enhances the ability of CA1 pyramidal neurons to fire synaptically evoked dendritic spikes and to generate a temporally precise form of heterosynaptic plasticity. Long-range inhibition from entorhinal cortex may thus increase the precision of hippocampal-based long-term memory associations by assessing the salience of mnemonormation to the immediate sensory input.
Collapse
Affiliation(s)
- Jayeeta Basu
- Department of Neuroscience, Kavli Brain Institute, Columbia University Medical Center, 1051 Riverside Drive, New York, NY 10032, USA.
| | - Jeffrey D Zaremba
- Department of Neuroscience, Kavli Brain Institute, Columbia University Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| | - Stephanie K Cheung
- Department of Neuroscience, Kavli Brain Institute, Columbia University Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| | - Frederick L Hitti
- Department of Neuroscience, Kavli Brain Institute, Columbia University Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| | | | - Attila Losonczy
- Department of Neuroscience, Kavli Brain Institute, Columbia University Medical Center, 1051 Riverside Drive, New York, NY 10032, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, Kavli Brain Institute, Columbia University Medical Center, 1051 Riverside Drive, New York, NY 10032, USA.
| |
Collapse
|
15
|
Allene C, Lourenço J, Bacci A. The neuronal identity bias behind neocortical GABAergic plasticity. Trends Neurosci 2015; 38:524-34. [PMID: 26318208 DOI: 10.1016/j.tins.2015.07.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/18/2022]
Abstract
In the neocortex, different types of excitatory and inhibitory neurons connect to one another following a detailed blueprint, defining functionally-distinct subnetworks, whose activity and modulation underlie complex cognitive functions. We review the cell-autonomous plasticity of perisomatic inhibition onto principal excitatory neurons. We propose that the tendency of different cortical layers to exhibit depression or potentiation of perisomatic inhibition is dictated by the specific identities of principal neurons (PNs). These are mainly defined by their projection targets and by their preference to be innervated by specific perisomatic-targeting basket cell types. Therefore, principal neurons responsible for relaying information to subcortical nuclei are differentially inhibited and show specific forms of plasticity compared to other PNs that are specialized in more associative functions.
Collapse
Affiliation(s)
- Camille Allene
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris 6), Unité Mixte de Recherche S 1127; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127; Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225; Institut du Cerveau et de la Moelle épinière (ICM), 75013 Paris, France
| | - Joana Lourenço
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris 6), Unité Mixte de Recherche S 1127; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127; Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225; Institut du Cerveau et de la Moelle épinière (ICM), 75013 Paris, France
| | - Alberto Bacci
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC Paris 6), Unité Mixte de Recherche S 1127; Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127; Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225; Institut du Cerveau et de la Moelle épinière (ICM), 75013 Paris, France.
| |
Collapse
|
16
|
Alger BE, Nagode DA, Tang AH. Muscarinic cholinergic receptors modulate inhibitory synaptic rhythms in hippocampus and neocortex. Front Synaptic Neurosci 2014; 6:18. [PMID: 25249974 PMCID: PMC4155787 DOI: 10.3389/fnsyn.2014.00018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 07/29/2014] [Indexed: 01/08/2023] Open
Abstract
Activation of muscarinic acetylcholine (ACh) receptors (mAChRs) powerfully affects many neuronal properties as well as numerous cognitive behaviors. Small neuronal circuits constitute an intermediate level of organization between neurons and behaviors, and mAChRs affect interactions among cells that compose these circuits. Circuit activity is often assessed by extracellular recordings of the local field potentials (LFPs), which are analogous to in vivo EEGs, generated by coordinated neuronal interactions. Coherent forms of physiologically relevant circuit activity manifest themselves as rhythmic oscillations in the LFPs. Frequencies of rhythmic oscillations that are most closely associated with animal behavior are in the range of 4–80 Hz, which is subdivided into theta (4–14 Hz), beta (15–29 Hz) and gamma (30–80 Hz) bands. Activation of mAChRs triggers rhythmic oscillations in these bands in the hippocampus and neocortex. Inhibitory responses mediated by GABAergic interneurons constitute a prominent feature of these oscillations, and indeed, appear to be their major underlying factor in many cases. An important issue is which interneurons are involved in rhythm generation. Besides affecting cellular and network properties directly, mAChRs can cause the mobilization of endogenous cannabinoids (endocannabinoids, eCBs) that, by acting on the principal cannabinoid receptor of the brain, CB1R, regulate the release of certain neurotransmitters, including GABA. CB1Rs are heavily expressed on only a subset of interneurons and, at lower density, on glutamatergic neurons. Exogenous cannabinoids typically disrupt oscillations in the theta (θ) and gamma (γ) ranges, which probably contributes to the behavioral effects of these drugs. It is important to understand how neuronal circuit activity is affected by mAChR-driven eCBs, as this information will provide deeper insight into the actions of ACh itself, as well as into the effects of eCBs and exogenous cannabinoids in animal behavior. After covering some basic aspects of the mAChR system, this review will focus on recent findings concerning the mechanisms and circuitry that generate θ and γ rhythms in hippocampus and neocortex. The ability of optogenetic methods to probe the many roles of ACh in rhythm generation is highlighted.
Collapse
Affiliation(s)
- Bradley E Alger
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA ; Department of Psychiatry, University of Maryland School of Medicine Baltimore, MD, USA ; Program in Neuroscience, Graduate School, University of Maryland Baltimore Baltimore, MD, USA
| | - Daniel A Nagode
- Department of Biology, University of Maryland College Park College Park, MD, USA
| | - Ai-Hui Tang
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA
| |
Collapse
|
17
|
Abstract
Little is known about how neuron firing recorded in vivo retrogradely influences synaptic strength. We injected the firing of a rat hippocampal neurogliaform cell (NGFC), a widely expressed GABAergic neuron type, detected in vivo during theta rhythm, into NGFCs of rat or neuronal nitric oxide synthase (nNOS)-Cre-tdTomato mouse recorded in vitro. We found that the "in vivo firing pattern" produced a transient firing-induced suppression of synaptic inhibition (FSI) evoked by a presynaptic NGFC. Imaging experiments demonstrate that FSI was associated with action potential backpropagation (bAP) and a supralinear increase in dendritic Ca(2+). The application of the L-type Ca(2+) channel antagonist nimodipine blocked FSI. Further pharmacological experiments, such as the application of a nitric oxide-sensitive guanylyl cyclase (NO-sGC) receptor antagonist, a NOS inhibitor, and NO donors, suggested that NO released from postsynaptic cells mediated FSI and likely activated presynaptic receptors to inhibit GABA release. The in vivo firing pattern modulated the size of unitary EPSPs impinging on NGFCs through FSI and not via a direct effect on excitatory synaptic transmission. Our data demonstrate: (1) retrograde signaling initiated by in vivo firing pattern, (2) interneuron bAPs detected with fast temporal resolution, and (3) a novel role for NO expressed by specific interneuron types.
Collapse
|
18
|
Huang CC, Lin YS, Lee CC, Hsu KS. Cell type-specific expression of Eps8 in the mouse hippocampus. BMC Neurosci 2014; 15:26. [PMID: 24533597 PMCID: PMC3937003 DOI: 10.1186/1471-2202-15-26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 02/13/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epidermal growth factor receptor substrate 8 (Eps8) is a multifunctional protein that regulates actin cytoskeleton dynamics and architecture through its barbed-end capping and bundling activities. In cultured hippocampal neurons, Eps8 is enriched at dendritic spine heads and is required for spine morphogenesis; however, the detailed expression pattern of Eps8 in the hippocampus has not yet been explored. RESULTS Here, we demonstrate that endogenous Eps8 protein is restrictively expressed in neurons (NeuN-positive), but not in glial cells (glial fibrillary acidic protein-positive) in area CA1 of the mouse hippocampus. Surprisingly, Eps8 immunoreactivity is rarely found in pyramidal cell somata, but is expressed predominantly in the somata and dendrites of 67 kDa isoform of glutamic acid decarboxylase-expressing GABAergic interneurons in the stratum radiatum and at the border of stratum radiatum and lacunosum-moleculare of area CA1. On the basis of co-localizing markers, we found that Eps8 is not present in perisomatic inhibitory parvalbumin-expressing cells or calretinin-expressing interneurons. However, Eps8 is richly expressed in calbindin-expressing interneurons. Furthermore, Eps8 is also present in cholecystokinin-expressing interneurons, but not in somatostatin-expressing interneurons in area CA1 stratum pyramidale and stratum radiatum. CONCLUSIONS These results reveal a previously unknown cell type-specific expression pattern of endogenous Eps8 protein in the mouse hippocampus and speculate that the role of Eps8 in controlling and orchestrating neuronal morphogenesis and structural plasticity might be more prominent in interneurons than in pyramidal cells of the hippocampus.
Collapse
Affiliation(s)
| | | | | | - Kuei-Sen Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
19
|
Wei XF, Zhang YH, Zhang LL, Xu SJ, Chen XW, Wang C, Si JQ, Ma KT, Wang QW. CCK-8S increases the firing frequency of CCK-positive neurons and facilitates excitatory synaptic transmission in primary rat hippocampal neurons. Neurosci Lett 2013; 549:34-9. [PMID: 23827217 DOI: 10.1016/j.neulet.2013.06.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 06/13/2013] [Accepted: 06/20/2013] [Indexed: 11/16/2022]
Abstract
The neuropeptide cholecystokinin octapeptide (CCK) is involved in a variety of brain functions. In the hippocampus, most CCK is released from CCK-positive (CCK+) neurons, but the effects of CCK on CCK+ neurons are poorly understood. We employed primary hippocampal cultures to explore the modulatory effect of CCK on CCK+ neurons. CCK-8S (0.2 μM) was added to the culture medium from day in vitro 2 (DIV-2) to DIV-11. An adenovirus integrated with the CCK promoter was used to label CCK+ neurons. Whole-cell patch clamp recording was carried on to record the electrophysiology properties. The results show that: (1) CCK-8S significantly decreased membrane capacity but increased the membrane resistance (Rm) of CCK+ neurons, (2) CCK-8S increased action potential (AP) firing frequency of CCK+ neurons but did not affect the firing pattern, (3) CCK-8S facilitated CCK+ neuron excitatory synaptic transmission but attenuated inhibitory synaptic transmission, and (4) the expression of postsynaptic density-95 (PSD-95) in cultured hippocampal neurons was elevated by CCK-8S treatment. Our results demonstrate that CCK-8S significantly alters the membrane electrophysiological characteristics and synaptic activity of cultured hippocampal CCK+ neurons. These findings may enhance our understanding of the modulatory effect of CCK in the brain.
Collapse
Affiliation(s)
- Xiao-Fei Wei
- Zhejiang Provincial Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Ningbo University, School of Medicine, Ningbo 315211, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Synaptic kainate receptors in CA1 interneurons gate the threshold of theta-frequency-induced long-term potentiation. J Neurosci 2013; 32:18215-26. [PMID: 23238735 DOI: 10.1523/jneurosci.2327-12.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Theta oscillations (4-12 Hz) in neuronal networks are known to predispose the synapses involved to plastic changes and may underlie their association with learning behaviors. The lowered threshold for synaptic plasticity during theta oscillations is thought to be due to decreased GABAergic inhibition. Interneuronal kainate receptors (KARs) regulate GABAergic transmission and are implicated in theta activity; however, the physiological significance of this regulation is unknown. In rat hippocampus, we show that during theta activity, there is excitatory postsynaptic drive to CA1 interneurons mediated by KARs. This promotes feedforward inhibition of pyramidal neurons, raising the threshold for induction of theta-burst long-term potentiation. These results identify a novel mechanism whereby the activation of postsynaptic KARs in CA1 interneurons gate changes in synaptic efficacy to a physiologically relevant patterned stimulation.
Collapse
|
21
|
Chamberland S, Topolnik L. Inhibitory control of hippocampal inhibitory neurons. Front Neurosci 2012; 6:165. [PMID: 23162426 PMCID: PMC3496901 DOI: 10.3389/fnins.2012.00165] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 10/24/2012] [Indexed: 11/13/2022] Open
Abstract
Information processing within neuronal networks is determined by a dynamic partnership between principal neurons and local circuit inhibitory interneurons. The population of GABAergic interneurons is extremely heterogeneous and comprises, in many brain regions, cells with divergent morphological and physiological properties, distinct molecular expression profiles, and highly specialized functions. GABAergic interneurons have been studied extensively during the past two decades, especially in the hippocampus, which is a relatively simple cortical structure. Different types of hippocampal inhibitory interneurons control spike initiation [e.g., axo-axonic and basket cells (BCs)] and synaptic integration (e.g., bistratified and oriens–lacunosum moleculare interneurons) within pyramidal neurons and synchronize local network activity, providing a means for functional segregation of neuronal ensembles and proper routing of hippocampal information. Thus, it is thought that, at least in the hippocampus, GABAergic inhibitory interneurons represent critical regulating elements at all stages of information processing, from synaptic integration and spike generation to large-scale network activity. However, this raises an important question: if inhibitory interneurons are fundamental for network computations, what are the mechanisms that control the activity of the interneurons themselves? Given the essential role of synaptic inhibition in the regulation of neuronal activity, it would be logical to expect that specific inhibitory mechanisms have evolved to control the operation of interneurons. Here, we review the mechanisms of synaptic inhibition of interneurons and discuss their role in the operation of hippocampal inhibitory circuits.
Collapse
Affiliation(s)
- Simon Chamberland
- Axis of Cellular and Molecular Neuroscience, IUSMQ, Department of Biochemistry, Microbiology and Bio-informatics, Université Laval Québec, QC, Canada
| | | |
Collapse
|
22
|
Involvement of pre- and postsynaptic NMDA receptors at local circuit interneuron connections in rat neocortex. Neuroscience 2012; 228:179-89. [PMID: 23079623 PMCID: PMC3546165 DOI: 10.1016/j.neuroscience.2012.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/27/2012] [Accepted: 10/07/2012] [Indexed: 11/23/2022]
Abstract
To investigate the involvement of N-Methyl-D-aspartate (NMDA) receptors in local neocortical synaptic transmission, dual whole-cell recordings - combined with biocytin labelling - were obtained from bitufted adapting, multipolar adapting or multipolar non-adapting interneurons and pyramidal cells in layers II-V of rat (postnatal days 17-22) sensorimotor cortex. The voltage dependency of the amplitude of Excitatory postsynaptic potentials (EPSPs) received by the three types of interneuron appeared to coincide with the interneuron subclass; upon depolarisation, EPSPs received by multipolar non-adapting interneurons either decreased in amplitude or appeared insensitive, multipolar adapting interneuron EPSP amplitudes increased or appeared insensitive, whereas bitufted interneuron EPSP amplitudes increased or decreased. Connections were challenged with the NMDA receptor antagonist d-(-)-2-amino-5-phosphonopentanoic acid (d-AP5) (50μM) revealing NMDA receptors to contribute to EPSPs received by all cell types, this also abolished the non-conventional voltage dependency. Reciprocal connections were frequent between pyramidal cells and multipolar interneurons, and inhibitory postsynaptic potentials (IPSPs) elicited in pyramidal cells by both multipolar adapting and multipolar non-adapting interneurons were sensitive to a significant reduction in amplitude by d-AP5. The involvement of presynaptic NMDA receptors was indicated by coefficient of variation analysis and an increase in the failures of transmission. Furthermore, by loading MK-801 into the pre- or postsynaptic neurons, we observed that a reduction in inhibition requires presynaptic and not postsynaptic NMDA receptors. These results suggest that NMDA receptors possess pre- and postsynaptic roles at selective neocortical synapses that are probably important in governing spike-timing and information flow.
Collapse
|
23
|
De-May CL, Ali AB. Cell type-specific regulation of inhibition via cannabinoid type 1 receptors in rat neocortex. J Neurophysiol 2012; 109:216-24. [PMID: 23054605 DOI: 10.1152/jn.00272.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Endogenous cannabinoid type 1 (CB1) receptors demonstrate a cell type-specific expression and are potent modulators of synaptic transmission within the central nervous system. We aimed to investigate whether two classes of multipolar interneuron in the neocortex displayed a form of short-term synaptic plasticity, depolarization-induced suppression of inhibition (DSI), and whether the DSI was mediated by a common receptor. Paired whole cell recordings combined with biocytin labeling were performed between pyramidal cells and either multipolar adapting or multipolar nonadapting interneurons in layers II-IV of male Wistar rat (postnatal day 17-22) somatosensory cortex. Inhibitory postsynaptic potentials elicited by multipolar adapting interneurons were sensitive to DSI, which was blocked by the CB1 receptor antagonist AM-251 (8 μM), indicating that the suppression of inhibition was mediated by CB1 receptors. Two subpopulations of multipolar nonadapting interneuron-to-pyramidal cell connections were discovered on the basis of their susceptibility to DSI. Whereas 50% were insensitive to DSI, the remaining half were sensitive to DSI, which could not be prevented by AM-251. DSI at these connections was also insensitive to the group I (mGluRIa) and III metabotropic glutamate receptor antagonists (RS)-1-aminoindan-1,5-dicarboxylic acid (100 μM) and (RS)-α-cyclopropyl-4-phosphonophenylglycine (100 μM) and the group III agonist l-2-amino-4-phosphonobutanoate (50 μM). However, multipolar nonadapting interneuron-to-pyramidal cell connections were sensitive to the endocannabinoid anandamide (9 μM), mimicking the effects of DSI, which also could not be prevented by AM-251, implying a CB1 receptor-independent suppression of inhibition. These results reveal an interneuron type-specific modulation of synaptic transmission via CB receptors in the neocortex.
Collapse
Affiliation(s)
- Claire L De-May
- Department of Pharmacology, University College London School of Pharmacy, London, United Kingdom
| | | |
Collapse
|
24
|
|
25
|
Cellular neurochemical characterization and subcellular localization of phospholipase C β1 in rat brain. Neuroscience 2012; 222:239-68. [PMID: 22735577 DOI: 10.1016/j.neuroscience.2012.06.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 06/15/2012] [Accepted: 06/15/2012] [Indexed: 11/21/2022]
Abstract
The present study describes a complete and detailed neuroanatomical distribution map of the phospholipase C beta1 (PLCβ1) isoform along the adult rat neuraxis, and defines the phenotype of cells expressing PLCβ1, along with its subcellular localization in cortical neurons as assessed by double-immunofluorescence staining and confocal laser scanning. Immunohistochemical labeling revealed a considerable morphological heterogeneity among PLCβ1-positive cells in the cortex, even though there was a marked predominance of pyramidal morphologies. As an exception to the general non-matching distribution of GFAP and PLCβ1, a high degree of co-expression was observed in radial glia-like processes of the spinal cord white matter. In the somatosensory cortex, the proportion of GABAergic neurons co-stained with PLCβ1 was similar (around 2/3) in layers I, II-III, IV and VI, and considerably lower in layer V (around 2/5). Double immunofluorescence against PLCβ1 and nuclear speckle markers SC-35 and NeuN/Fox3 in isolated nuclei from the rat cortex showed a high overlap of both markers with PLCβ1 within the nuclear matrix. In contrast, there was no apparent co-localization with markers of the nuclear envelope and lamina. Finally, to assess whether the subcellular expression pattern of PLCβ1 involved specifically one of the two splice variants of PLCβ1, we carried out Western blot experiments in cortical subcellular fractions. Notably, PLCβ1a/1b ratios were statistically higher in the cytoplasm than in the nuclear and plasma membrane fractions. These results provide a deeper knowledge of the cellular distribution of the PLCβ1 isoform in different cell subtypes of the rat brain, and of its presence in the neuronal nuclear compartment.
Collapse
|
26
|
Mercer A, Botcher NA, Eastlake K, Thomson AM. SP-SR interneurones: a novel class of neurones of the CA2 region of the hippocampus. Hippocampus 2012; 22:1758-69. [PMID: 22431345 DOI: 10.1002/hipo.22010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2012] [Indexed: 11/10/2022]
Abstract
The CA2 region of the hippocampus has distinctive properties and inputs and may be linked with the pathology of specific psychiatric and neurological disorders. It is, therefore, important to understand CA2 circuitry and its involvement in the circuitry of the hippocampus. Properties of CA2 basket cells have been reported. However, other classes of CA2 interneurones with cell bodies located in stratum pyramidale remained to be described. In this study, the unusual axonal arbors of a novel subclass of dendrite-preferring CA2 interneurones whose somata are located in the pyramidal cell layer was revealed following intracellular recordings and biocytin labeling. One to four apical dendrites emerged from the soma, branched in stratum radiatum (SR) forming a tuft, but rarely penetrated stratum lacunosum-moleculare (SLM). One or two basal dendrites branched close to the soma, the branches extended through stratum oriens (SO) and often reached the alveus. Unlike CA2 bistratified cells, the axons of these cells arborized almost exclusively in SR with few, if any, branches extending to stratum pyramidale (SP), SO, or SLM. These interneurones again, unlike bistratified cells, were immunonegative for parvalbumin and cholecystokinin. Electrophysiologically, they were similar to some CA2 basket and bistratified cells in that they presented a "sag" in response to hyperpolarizing current injections and displayed spike frequency adaptation. They targeted the apical dendrites of neighboring CA2 pyramidal cells and received inputs from them.
Collapse
Affiliation(s)
- Audrey Mercer
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom.
| | | | | | | |
Collapse
|
27
|
Keimpema E, Straiker A, Mackie K, Harkany T, Hjerling-Leffler J. Sticking out of the crowd: the molecular identity and development of cholecystokinin-containing basket cells. J Physiol 2012; 590:703-14. [PMID: 22219340 DOI: 10.1113/jphysiol.2011.224386] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Certain essential cognitive processes require the precise temporal interplay between glutamatergic (excitatory) pyramidal cells and γ-aminobutyric acid (GABA)-releasing inhibitory interneurons in the hippocampus. Basket cells, the main class of interneurons, target pyramidal cell somata and proximal dendrites and thus are poised to modify network oscillations. Though only present in limited numbers, the impaired development of basket cells can result in changes in the hippocampal circuitry leading to neurological disorders, such as schizophrenia. The diversity of the spatial origins, neurochemical make-up, cytoarchitecture and network contributions amongst basket cells is a provocative example of interneuron heterogeneity in the hippocampus. This review discusses recent data concerned with the developmental trajectories of one subclass, the cholecystokinin-containing basket cell, and emphasizes the significance of the short-range intercellular guidance cues that have recently emerged to impact the formation and function of their inhibitory synapses.
Collapse
Affiliation(s)
- Erik Keimpema
- Division of Molecular Neurobiology, Department of Medical Biochemistry & Biophysics, Scheeles väg 1:A1, Karolinska Institutet, S-17177 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
28
|
Iball J, Ali AB. Endocannabinoid Release Modulates Electrical Coupling between CCK Cells Connected via Chemical and Electrical Synapses in CA1. Front Neural Circuits 2011; 5:17. [PMID: 22125513 PMCID: PMC3222094 DOI: 10.3389/fncir.2011.00017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/07/2011] [Indexed: 11/13/2022] Open
Abstract
Electrical coupling between some subclasses of interneurons is thought to promote coordinated firing that generates rhythmic synchronous activity in cortical regions. Synaptic activity of cholecystokinin (CCK) interneurons which co-express cannabinoid type-1 (CB1) receptors are powerful modulators of network activity via the actions of endocannabinoids. We investigated the modulatory actions of endocannabinoids between chemically and electrically connected synapses of CCK cells using paired whole-cell recordings combined with biocytin and double immunofluorescence labeling in acute slices of rat hippocampus at P18-20 days. CA1 stratum radiatum CCK Schaffer collateral-associated cells were coupled electrically with each other as well as CCK basket cells and CCK cells with axonal projections expanding to dentate gyrus. Approximately 50% of electrically coupled cells received facilitating, asynchronously released inhibitory postsynaptic potential (IPSPs) that curtailed the steady-state coupling coefficient by 57%. Tonic CB1 receptor activity which reduces inhibition enhanced electrical coupling between cells that were connected via chemical and electrical synapses. Blocking CB1 receptors with antagonist, AM-251 (5 μM) resulted in the synchronized release of larger IPSPs and this enhanced inhibition further reduced the steady-state coupling coefficient by 85%. Depolarization induced suppression of inhibition (DSI), maintained the asynchronicity of IPSP latency, but reduced IPSP amplitudes by 95% and enhanced the steady-state coupling coefficient by 104% and IPSP duration by 200%. However, DSI did not did not enhance electrical coupling at purely electrical synapses. These data suggest that different morphological subclasses of CCK interneurons are interconnected via gap junctions. The synergy between the chemical and electrical coupling between CCK cells probably plays a role in activity-dependent endocannabinoid modulation of rhythmic synchronization.
Collapse
Affiliation(s)
- Jonathan Iball
- Department of Pharmacology, The School of Pharmacy, University of London London, UK
| | | |
Collapse
|
29
|
Salesse C, Mueller CL, Chamberland S, Topolnik L. Age-dependent remodelling of inhibitory synapses onto hippocampal CA1 oriens-lacunosum moleculare interneurons. J Physiol 2011; 589:4885-901. [PMID: 21825029 DOI: 10.1113/jphysiol.2011.215244] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Stratum oriens-lacunosum moleculare interneurons (O-LM INs) represent the major element of the hippocampal feedback inhibitory circuit, which provides inhibition to the distal dendritic sites of CA1 pyramidal neurons. Although the intrinsic conductance profile and the properties of glutamatergic transmission to O-LM INs have become a subject of intense investigation, far less is known about the properties of the inhibitory synapses formed onto these cells. Here, we used whole-cell patch-clamp recordings in acute mouse hippocampal slices to study the properties and plasticity of GABAergic inhibitory synapses onto O-LM INs. Surprisingly, we found that the kinetics of inhibitory postsynaptic currents (IPSCs) were slower in mature synapses (P26-40) due to the synaptic incorporation of the α5 subunit of the GABA(A) receptor (a5-GABA(A)R). Moreover, this age-dependent synaptic expression of a5-GABA(A)Rs was directly associated with the emergence of long-term potentiation at IN inhibitory synapses. Finally, the slower time course of IPSCs observed in O-LM INs of mature animals had a profound effect on IN excitability by significantly delaying its spike firing. Our data suggest that GABAergic synapses onto O-LM INs undergo significant modifications during postnatal maturation. The developmental switch in IPSC properties and plasticity is controlled by the synaptic incorporation of the a5-GABA(A)R subunit and may represent a potential mechanism for the age-dependent modifications in the inhibitory control of the hippocampal feedback inhibitory circuit.
Collapse
Affiliation(s)
- Charleen Salesse
- Axis of Cellular and Molecular Neuroscience, CRULRG, Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Québec, PQ, Canada
| | | | | | | |
Collapse
|
30
|
Méndez P, Bacci A. Assortment of GABAergic plasticity in the cortical interneuron melting pot. Neural Plast 2011; 2011:976856. [PMID: 21785736 PMCID: PMC3139185 DOI: 10.1155/2011/976856] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 05/01/2011] [Indexed: 12/30/2022] Open
Abstract
Cortical structures of the adult mammalian brain are characterized by a spectacular diversity of inhibitory interneurons, which use GABA as neurotransmitter. GABAergic neurotransmission is fundamental for integrating and filtering incoming information and dictating postsynaptic neuronal spike timing, therefore providing a tight temporal code used by each neuron, or ensemble of neurons, to perform sophisticated computational operations. However, the heterogeneity of cortical GABAergic cells is associated to equally diverse properties governing intrinsic excitability as well as strength, dynamic range, spatial extent, anatomical localization, and molecular components of inhibitory synaptic connections that they form with pyramidal neurons. Recent studies showed that similarly to their excitatory (glutamatergic) counterparts, also inhibitory synapses can undergo activity-dependent changes in their strength. Here, some aspects related to plasticity and modulation of adult cortical and hippocampal GABAergic synaptic transmission will be reviewed, aiming at providing a fresh perspective towards the elucidation of the role played by specific cellular elements of cortical microcircuits during both physiological and pathological operations.
Collapse
Affiliation(s)
- Pablo Méndez
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Alberto Bacci
- European Brain Research Institute (EBRI), Via del Fosso di Fiorano 64, 00143 Rome, Italy
| |
Collapse
|
31
|
Nakamura NH, Akiyama K, Naito T. Suppression of cAMP-dependent gene expression by cholecystokinin in the hippocampus. Neuroscience 2011; 187:15-23. [PMID: 21540082 DOI: 10.1016/j.neuroscience.2011.04.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 03/18/2011] [Accepted: 04/13/2011] [Indexed: 11/25/2022]
Abstract
Our previous study suggests that "the neuropeptidergic system" might promote a diversity of the mechanisms that regulate signal transmission in the hippocampus. Cholecystokinin (CCK) is the mostly expressed neuropeptide gene in the hippocampus. Here, we investigated whether CCK regulates immediate-early genes (Egr1/zif268 and Fos), critical indicators of cortical neuronal activity. We showed that CCK increased Egr1/zif268 promoter activity in a neuronal cell line, which is transfected with CCK(B) receptor. Unexpectedly, in living hippocampal slices, CCK significantly suppressed cAMP-induced expression of Egr1/zif268 and Fos through CCK(B) receptor activation. This suppression was involved in activating GABA(B) and cannabinoid 1 receptors. In addition to transient CCK modulation of action potentials on hippocampal principal neurons, we suggest that release of endogenous CCK might indirectly produce the suppression of cAMP-dependent gene expression in the hippocampus.
Collapse
Affiliation(s)
- N H Nakamura
- Okinawa Institute of Science and Technology, Okinawa 904-2234, Japan.
| | | | | |
Collapse
|
32
|
Evstratova A, Chamberland S, Topolnik L. Cell type-specific and activity-dependent dynamics of action potential-evoked Ca2+ signals in dendrites of hippocampal inhibitory interneurons. J Physiol 2011; 589:1957-77. [PMID: 21486769 DOI: 10.1113/jphysiol.2010.204255] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In most central neurons, action potentials (APs), generated in the initial axon segment, propagate back into dendrites and trigger considerable Ca(2+) entry via activation of voltage-sensitive calcium channels (VSCCs). Despite the similarity in its underlying mechanisms, however, AP-evoked dendritic Ca(2+) signalling often demonstrates a cell type-specific profile that is determined by the neuron dendritic properties. Using two-photon Ca(2+) imaging in combination with patch-clamp whole-cell recordings,we found that in distinct types of hippocampal inhibitory interneurons Ca(2+) transients evoked by backpropagating APs not only were shaped by the interneuron-specific properties of dendritic Ca(2+) handling but also involved specific Ca(2+) mechanisms that were regulated dynamically by distinct activity patterns. In dendrites of regularly spiking basket cells, AP-evoked Ca(2+) rises were of large amplitude and fast kinetics; however, they decreased with membrane hyperpolarization or following high-frequency firing episodes. In contrast, AP-evoked Ca(2+) elevations in dendrites of Schaffer collateral-associated cells exhibited significantly smaller amplitude and slower kinetics, but increased with membrane hyperpolarization. These cell type-specific properties of AP-evoked dendritic Ca(2+) signalling were determined by distinct endogenous buffer capacities of the interneurons examined and by specific types of VSCCs recruited by APs during different patterns of activity. Furthermore, AP-evoked Ca(2+) transients summated efficiently during theta-like bursting and were associated with the induction of long-term potentiation at inhibitory synapses onto both types of interneurons. Therefore, the cell type-specific profile of AP-evoked dendritic Ca(2+) signalling is shaped in an activity-dependent manner, such that the same pattern of hippocampal activity can be differentially translated into dendritic Ca(2+) signals in different cell types. However, Cell type-specific differences in Ca(2+) signals can be 'smoothed out' by changes in neuronal activity, providing a means for common, cell-type-independent forms of synaptic plasticity.
Collapse
Affiliation(s)
- Alesya Evstratova
- Axis of Cellular and Molecular Neuroscience, CRULRG, Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Québec, PQ, Canada
| | | | | |
Collapse
|
33
|
Abstract
Plasticity refers to a physiologically measured change that may last for short or long periods of time. Endocannabinoids (ECBs) are prevalent throughout most of the brain, and modulate synaptic transmission in many ways. This chapter will focus on the roles of ECBs in neural plasticity in the mammalian brain. The topics covered can be divided loosely into two themes: how ECBs regulate synaptic plasticity, and how ECBs' actions themselves are regulated by neuronal activity. Because ECBs regulate synaptic plasticity, the modifiability of ECB mobilization constitutes a form of "metaplasticity" (as reported by Abraham and Bear (Trends Neurosci 19:126-130, 1996)), i.e., an upstream process that determines the nature and extent of synaptic plasticity. Many of their basic functions are still being discovered, and while there is consensus on large issues, many points of divergence exist as well. This chapter concentrates on developments in the roles of ECBs in synaptic plasticity that have come to light since the major review by Chevaleyre et al. (Annu Rev Neurosci 29:37-76, 2006).
Collapse
Affiliation(s)
- Bradley E Alger
- Departments of Physiology and Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
34
|
Lee SH, Soltesz I. Requirement for CB1 but not GABAB receptors in the cholecystokinin mediated inhibition of GABA release from cholecystokinin expressing basket cells. J Physiol 2010; 589:891-902. [PMID: 21173082 DOI: 10.1113/jphysiol.2010.198499] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cholecystokinin (CCK) is an abundant neuropeptide involved in normal behaviour and pathophysiological conditions. Recently, CCK was shown to act as a molecular switch for perisomatic inhibition in the hippocampus, by directly depolarizing parvalbumin expressing (PV+) basket cells while indirectly depressing GABA release from CCK expressing (CCK+) basket cells. However, whether these two CCK-mediated effects are causally related is controversial, with one hypothesis proposing that the CCK-induced firing of PV+ basket cells increases the release of GABA, which, in turn, heterosynaptically inhibits GABA release from neighbouring CCK+ basket cell terminals through presynaptic GABAB receptors. Our present data from paired recording experiments from presynaptic basket cells and postsynaptic CA1 pyramidal cells in acute rat brain slices show that the P/Q Ca2+ channel antagonist agatoxin TK (250 nm) abolished GABA release from PV+ basket cells, but it had no effect on the CCK-induced depression of GABA release from CCK+ basket cells. Furthermore, CCK decreased GABA release from CCK+ basket cells even in the presence of the GABAB receptor antagonist CGP55845 (2 μm). In contrast, cannabinoid type-1 (CB1) receptor blockade with AM251 (10 μm) prevented the action of CCK on GABA release both from CCK+ basket cells and dendritically projecting, CCK+ Schaffer collateral-associated interneurons. These results demonstrate that CCK-mediated inhibition of GABA release from CCK+ cells requires no cross-talk between PV+ and CCK+ synapses, but that it critically depends on CB1 receptor-mediated endocannabinoid signalling at both perisomatic and dendritic inputs.
Collapse
Affiliation(s)
- Sang-Hun Lee
- Department of Anatomy and Neurobiology, UC Irvine, Irvine, CA 92697-1280, USA.
| | | |
Collapse
|
35
|
Capogna M. Neurogliaform cells and other interneurons of stratum lacunosum-moleculare gate entorhinal-hippocampal dialogue. J Physiol 2010; 589:1875-83. [PMID: 21135049 DOI: 10.1113/jphysiol.2010.201004] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The stratum lacunosum-moleculare of the hippocampus is an area of integration that receives inputs from extrinsic excitatory fibres including those from the entorhinal cortex, and is under the control of several neuromodulators. A critical aspect is the presence in this hippocampal layer of specific interneurons that are likely to influence the strength and the temporal structure of entorhinal-CA1 hippocampal dynamics. I review here recent data on the physiological role of these interneurons. Special focus is devoted to one interneuron type, the so-called neurogliaform cell, because recent studies have defined its unusual mode of cell-to-cell communication. Neurogliaform cells mediate feedforward inhibition of CA1 pyramidal cells, form a network of cells connected via chemical and electrical synapses, and evoke slow inhibitory synaptic currents mediated by GABA(A) and GABA(B) receptors. The modulation of entorhinal input by neurogliaform cells and their contribution to network theta rhythm are also discussed. I hope that novel information on neurogliaform cells will contribute to the ever-growing appreciation of GABAergic cell type diversity, and will inspire neuroscientists interested not only in synaptic physiology but also in the brain's spatial representation system.
Collapse
Affiliation(s)
- Marco Capogna
- MRC Anatomical Neuropharmacology Unit, Mansfield Road, Oxford OX1 3TH, UK.
| |
Collapse
|
36
|
Ali AB. CB1 modulation of temporally distinct synaptic facilitation among local circuit interneurons mediated by N-type calcium channels in CA1. J Neurophysiol 2010; 105:1051-62. [PMID: 21123660 DOI: 10.1152/jn.00831.2010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
One of the critical factors in determining network behavior of neurons is the influence of local circuit connections among interneurons. The short-term synaptic plasticity and the subtype of presynaptic calcium channels used at local circuit connections of inhibitory interneurons in CA1 were investigated using dual whole-cell recordings combined with biocytin and double immunofluorescence labeling in acute slices of P18- to 21-day-old rat stratum radiatum (SR) and stratum lacunosum molecular (SLM). Two forms of temporally distinct synaptic facilitation were observed among interneuron connections involving presynaptic cholecystokinin (CCK)-positive cells in SR, frequency-dependent facilitation, and a delayed onset of release (45-80 ms) with subsequent facilitation (DORF). Inhibition at both these synapses was under tonic cannabinoid-type 1 (CB1) receptor activity. DORF synapses did not display conventional release-dependent properties; however, blocking CB1 receptors with antagonist AM-251 (10 μM) altered the synaptic transmission to frequency-dependent depression with a fast onset of release (2-4 ms). Presynaptic CCK-negative interneurons in SLM elicited inhibitory postsynaptic potentials (IPSPs) insensitive to CB1 receptor pharmacology displayed frequency-dependent depression. Release of GABA at facilitating synapses was solely mediated via N-type presynaptic calcium channels, whereas depressing synapses utilized P/Q-type channels. These data reveal two distinct models of neurotransmitter release patterns among interneuron circuits that correlate with the subtype of presynaptic calcium channel. These data suggest that endocannabinoids act via CB1 receptors to selectively modulate N-type calcium channels to alter signal transmission.
Collapse
Affiliation(s)
- Afia B Ali
- Department of Pharmacology, University of London, School of Pharmacy, 29/39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
37
|
Chamberland S, Salesse C, Topolnik D, Topolnik L. Synapse-specific inhibitory control of hippocampal feedback inhibitory circuit. Front Cell Neurosci 2010; 4:130. [PMID: 21060720 PMCID: PMC2972748 DOI: 10.3389/fncel.2010.00130] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Accepted: 09/20/2010] [Indexed: 12/12/2022] Open
Abstract
Local circuit and long-range GABAergic projections provide powerful inhibitory control over the operation of hippocampal inhibitory circuits, yet little is known about the input- and target-specific organization of interacting inhibitory networks in relation to their specific functions. Using a combination of two-photon laser scanning photostimulation and whole-cell patch clamp recordings in mice hippocampal slices, we examined the properties of transmission at GABAergic synapses formed onto hippocampal CA1 stratum oriens – lacunosum moleculare (O–LM) interneurons by two major inhibitory inputs: local projection originating from stratum radiatum interneurons and septohippocampal GABAergic terminals. Optical mapping of local inhibitory inputs to O–LM interneurons revealed that vasoactive intestinal polypeptide- and calretinin-positive neurons, with anatomical properties typical of type III interneuron-specific interneurons, provided the major local source of inhibition to O–LM cells. Inhibitory postsynaptic currents evoked by minimal stimulation of this input exhibited small amplitude and significant paired-pulse and multiple-pulse depression during repetitive activity. Moreover, these synapses failed to show any form of long-term synaptic plasticity. In contrast, synapses formed by septohippocampal projection produced higher amplitude and persistent inhibition and exhibited long-term potentiation induced by theta-like activity. These results indicate the input and target-specific segregation in inhibitory control, exerted by two types of GABAergic projections and responsible for distinct dynamics of inhibition in O–LM interneurons. The two inputs are therefore likely to support the differential activity- and brain state-dependent recruitment of hippocampal feedback inhibitory circuits in vivo, crucial for dendritic disinhibition and computations in CA1 pyramidal cells.
Collapse
Affiliation(s)
- Simon Chamberland
- Axis of Cellular and Molecular Neuroscience, CRULRG Québec, PQ, Canada
| | | | | | | |
Collapse
|
38
|
Distinct endocannabinoid control of GABA release at perisomatic and dendritic synapses in the hippocampus. J Neurosci 2010; 30:7993-8000. [PMID: 20534847 DOI: 10.1523/jneurosci.6238-09.2010] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Endocannabinoid-mediated retrograde synaptic signaling is a key regulator of GABA release at synapses formed on the perisomatic region of pyramidal cells by basket cells that coexpress the cannabinoid type 1 receptor (CB(1)R) and cholecystokinin (CCK). However, CB(1)R and CCK-positive GABAergic terminals are present on pyramidal cell dendrites as well, but the principles of endocannabinoid control of GABA release in dendrites are not understood. We performed paired recordings from CCK-positive perisomatically (basket cells) or dendritically projecting (Schaffer collateral-associated cells) interneurons and postsynaptic CA1 pyramidal cells to determine the properties of endocannabinoid signaling at GABAergic synapses along the somato-dendritic axis. Although several key elements of the currently known molecular machinery for endocannabinoid synthesis are thought be primarily localized in dendrites, our results revealed that the depolarization-induced suppression of inhibition, the endocannabinoid-mediated tonic inhibition of GABA release, and the metabotropic glutamate receptor activation-induced, CB(1)R-mediated depression of GABA release were all significantly less effective at dendritic compared with perisomatic synapses. In addition, low concentration of exogenous CB(1) receptor agonist inhibited GABA release to a lesser extent at dendritic compared with perisomatic synapses, indicating that presynaptic differences are partly responsible for the differential control of GABA release by endocannabinoids in dendrites. Together, these data demonstrate a novel domain-specific regulation of GABA release by endocannabinoid signaling in the hippocampus.
Collapse
|
39
|
Cell type-specific long-term plasticity at glutamatergic synapses onto hippocampal interneurons expressing either parvalbumin or CB1 cannabinoid receptor. J Neurosci 2010; 30:1337-47. [PMID: 20107060 DOI: 10.1523/jneurosci.3481-09.2010] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Different GABAergic interneuron types have specific roles in hippocampal function, and anatomical as well as physiological features vary greatly between interneuron classes. Long-term plasticity of interneurons has mostly been studied in unidentified GABAergic cells and is known to be very heterogeneous. Here we tested whether cell type-specific plasticity properties in distinct GABAergic interneuron types might underlie this heterogeneity. We show that long-term potentiation (LTP) and depression (LTD), two common forms of synaptic plasticity, are expressed in a highly cell type-specific manner at glutamatergic synapses onto hippocampal GABAergic neurons. Both LTP and LTD are generated in interneurons expressing parvalbumin (PV+), whereas interneurons with similar axon distributions but expressing cannabinoid receptor-1 show no lasting plasticity in response to the same protocol. In addition, LTP or LTD occurs in PV+ interneurons with different efferent target domains. Perisomatic-targeting PV+ basket and axo-axonic interneurons express LTP, whereas glutamatergic synapses onto PV+ bistratified cells display LTD. Both LTP and LTD are pathway specific, independent of NMDA receptors, and occur at synapses with calcium-permeable (CP) AMPA receptors. Plasticity in interneurons with CP-AMPA receptors strongly modulates disynaptic GABAergic transmission onto CA1 pyramidal cells. We propose that long-term plasticity adjusts the synaptic strength between pyramidal cells and interneurons in a cell type-specific manner and, in the defined CA1 interneurons, shifts the spatial pattern of inhibitory weight from pyramidal cell dendrites to the perisomatic region.
Collapse
|
40
|
Ali AB, Todorova M. Asynchronous release of GABA via tonic cannabinoid receptor activation at identified interneuron synapses in rat CA1. Eur J Neurosci 2010; 31:1196-207. [PMID: 20345910 DOI: 10.1111/j.1460-9568.2010.07165.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The influence of local circuit interneurons is thought to play an important role in adjusting synaptic strength via endogenous cannabinoid type 1 (CB1) receptors. Using paired whole-cell recordings, combined with double immunofluorescence and biocytin labelling in acute slices of rat CA1 at postnatal day 18-23, we investigated the properties of Cholecystokinin (CCK)-positive stratum radiatum local circuit interneuron connections that utilised CB1 receptors. Three types of synaptic connections were studied, lacunosum-moleculare-radiatum perforant path-associated (LM-R PPA) to Shaffer collateral-associated (SCA) interneurons, SCA-SCA interneurons and SCA-pyramidal cells. These three synapses were differentially under tonic reduction of inhibition that was blocked by the CB1 receptor inverse agonist AM-251 (10 microM), which enhanced IPSPs. The strength of tonic reduction of inhibition was correlated with asynchronous release which was apparent at connections among interneurons. AM-251 increased the ratio of synchronous to asynchronous release (synchronicity ratio), while the CB receptor agonist anandamide (14 microM) decreased the synchronicity ratio. Fast and slow calcium chelators (BAPTA-AM and EGTA-AM) also increased the synchronicity ratio, accelerated inhibitory time courses and reduced IPSP amplitudes. These data suggest that CB1 receptors at connections among interneuron synapses play a role in tonic suppression of inhibition and govern the asynchronous release of GABA, modulating the time windows of inhibition. Effects of calcium chelators suggest that asynchronous release is a result of a long-lasting presynaptic calcium transients and/or a large distance between calcium source and sensor of exocytosis. These properties of specialised inhibitory neurons may have important modulatory roles in controlling spike timing among local circuit interneurons.
Collapse
Affiliation(s)
- A B Ali
- Department of Pharmacology, School of Pharmacy, University of London, 29/39 Brunswick Square, London WC1N 1AX, UK.
| | | |
Collapse
|
41
|
Ali AB. Presynaptic cell dependent modulation of inhibition in cortical regions. Curr Neuropharmacol 2010; 7:125-31. [PMID: 19949571 PMCID: PMC2730004 DOI: 10.2174/157015909788848875] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 01/15/2009] [Accepted: 02/24/2009] [Indexed: 01/20/2023] Open
Abstract
Several lines of evidence suggest that the modulation of presynaptic GABA release is mediated by a variety of receptors including; presynaptic AMPA, cannabinoid, GABAB, kainate, metabotropic glutamate, NMDA, and opioid receptors. The evidence supporting presynaptic modulation of inhibition is predominantly obtained from studying stimulus elicited, spontaneous or miniature synaptic events, where the information regarding the identity of the presynaptic cell is lost. This article summarises these findings then focuses on another approach to study the presynaptic modulation of GABA release by comparing the modulation of GABA release at unitary synapses identified morphologically, immunocytochemically and electrophysiologically. To date, evidence for cell-type specific regulation of presynaptic inhibition at identified synapses involving most of the above presynaptic receptors does not exist. Therefore, the key presynaptic modulators that will be focused on here are kainate and cannabinoid receptors and their intracellular signalling cascades that orchestrate GABA release. There will be some discussion on presynaptic modulation via opioid receptors at identified synapses. This review provides evidence to suggest a cell-type specific modulation of presynaptic inhibition in cortical regions.
Collapse
Affiliation(s)
- Afia B Ali
- University of London, School of Pharmacy, Department of Pharmacology 29/39 Brunswick Square, London WC1N 1AX, UK.
| |
Collapse
|
42
|
Synaptic activation of kainate receptors gates presynaptic CB(1) signaling at GABAergic synapses. Nat Neurosci 2010; 13:197-204. [PMID: 20081851 DOI: 10.1038/nn.2481] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 12/08/2009] [Indexed: 12/11/2022]
Abstract
Glutamate can control inhibitory synaptic transmission through activation of presynaptic kainate receptors. We found that glutamate released by train stimulation of Schaffer collaterals could lead to either short-term depression or short-term facilitation of inhibitory synaptic transmission in mouse CA1 pyramidal neurons, depending on the presence of cannabinoid type 1 (CB(1)) receptors on GABAergic afferents. The train-induced depression of inhibition (t-Di) required the mobilization of 2-arachidonoylglycerol through postsynaptic activation of metabotropic glutamate receptors and [Ca(2+)] rise. GluK1 (GluR5)-dependent depolarization of GABAergic terminals enabled t-Di by facilitating presynaptic CB(1) signaling. Thus, concerted activation of presynaptic CB(1) receptors and kainate receptors mediates short-term depression of inhibitory synaptic transmission. In contrast, in inhibitory connections expressing GluK1, but not CB(1), receptors, train stimulation of Schaffer collaterals led to short-term facilitation. Thus, activation of kainate receptors by synaptically released glutamate gates presynaptic CB(1) signaling, which in turn controls the direction of short-term heterosynaptic plasticity.
Collapse
|
43
|
Endogenous cannabinoids trigger the depolarization-induced suppression of excitation in the lateral amygdala. Learn Mem 2009; 17:43-9. [PMID: 20042481 DOI: 10.1101/lm.1663410] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The amygdala is a key area of the brain where the emotional memories are stored throughout the lifespan. It is well established that synapses in the lateral nucleus of amygdala (LA) can undergo long-term potentiation, a putative cellular correlate of learning and memory. However, a type of short-term synaptic plasticity, known as depolarization-induced suppression of excitation (DSE), has not been studied previously in the amygdala in general and in the LA in particular. In this study we aimed to prove either the absence or the presence of this phenomenon in the LA. Our data demonstrate for the first time that DSE is present in the LA and that it modulates the cortical excitatory synaptic input into this region. The existence of this type of retrograde neurotransmission in glutamatergic pyramidal neurons of the LA suggests that the axonal terminals of cortical inputs do possess functional type 1 cannabinoid receptors, and provides a novel insight regarding inputs into the LA. Further experiments indeed revealed endocannabinoids as the messenger for this retrograde signaling in the LA. In conclusion, the DSE may play a functional role in synaptic plasticity and related emotional memory processing in the LA.
Collapse
|
44
|
Synaptic cross talk between perisomatic-targeting interneuron classes expressing cholecystokinin and parvalbumin in hippocampus. J Neurosci 2009; 29:4140-54. [PMID: 19339609 DOI: 10.1523/jneurosci.5264-08.2009] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cholescystokinin (CCK)- or parvalbumin (PV)-containing interneurons are the major perisomatic-targeting interneurons in the cerebral cortex, including hippocampus, and are thought to form mutually exclusive networks. We used several techniques to test the alternative hypothesis that CCK and PV cells are coupled by chemical synapses. Triple immunofluorescence confocal microscopy revealed numerous axosomatic, axodendritic, and axoaxonic contacts stained for CCK, PV, and the presynaptic marker synaptophysin. The existence of mutual CCK and PV synapses was supported by dual EM immunolabeling. Paired whole-cell recordings detected unitary GABA(A)ergic synaptic transmission between identified CCK and PV cells, and single CCK cells could transiently inhibit action potential firing of synaptically coupled PV cells. We conclude that the major hippocampal perisomatic-targeting interneurons communicate synaptically. This communication should affect neuronal network activity, including neuronal oscillations, in which the CCK and PV cells have well established roles. The prevalence of CCK and PV networks in other brain regions suggests that internetwork interactions could be generally important.
Collapse
|
45
|
Kano M, Ohno-Shosaku T, Hashimotodani Y, Uchigashima M, Watanabe M. Endocannabinoid-mediated control of synaptic transmission. Physiol Rev 2009; 89:309-80. [PMID: 19126760 DOI: 10.1152/physrev.00019.2008] [Citation(s) in RCA: 1078] [Impact Index Per Article: 71.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The discovery of cannabinoid receptors and subsequent identification of their endogenous ligands (endocannabinoids) in early 1990s have greatly accelerated research on cannabinoid actions in the brain. Then, the discovery in 2001 that endocannabinoids mediate retrograde synaptic signaling has opened up a new era for cannabinoid research and also established a new concept how diffusible messengers modulate synaptic efficacy and neural activity. The last 7 years have witnessed remarkable advances in our understanding of the endocannabinoid system. It is now well accepted that endocannabinoids are released from postsynaptic neurons, activate presynaptic cannabinoid CB(1) receptors, and cause transient and long-lasting reduction of neurotransmitter release. In this review, we aim to integrate our current understanding of functions of the endocannabinoid system, especially focusing on the control of synaptic transmission in the brain. We summarize recent electrophysiological studies carried out on synapses of various brain regions and discuss how synaptic transmission is regulated by endocannabinoid signaling. Then we refer to recent anatomical studies on subcellular distribution of the molecules involved in endocannabinoid signaling and discuss how these signaling molecules are arranged around synapses. In addition, we make a brief overview of studies on cannabinoid receptors and their intracellular signaling, biochemical studies on endocannabinoid metabolism, and behavioral studies on the roles of the endocannabinoid system in various aspects of neural functions.
Collapse
Affiliation(s)
- Masanobu Kano
- Department of Neurophysiology, The University of Tokyo, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
46
|
Antonelli T, Tomasini MC, Mazza R, Fuxe K, Gaetani S, Cuomo V, Tanganelli S, Ferraro L. Cannabinoid CB1 and Cholecystokinin CCK2 Receptors Modulate, in an Opposing Way, Electrically Evoked [3H]GABA Efflux from Rat Cerebral Cortex Cell Cultures: Possible Relevance for Cortical GABA Transmission and Anxiety. J Pharmacol Exp Ther 2009; 329:708-17. [DOI: 10.1124/jpet.109.150649] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
47
|
Modulation of network oscillatory activity and GABAergic synaptic transmission by CB1 cannabinoid receptors in the rat medial entorhinal cortex. Neural Plast 2008; 2008:808564. [PMID: 19079598 PMCID: PMC2593022 DOI: 10.1155/2008/808564] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Accepted: 06/19/2008] [Indexed: 11/25/2022] Open
Abstract
Cannabinoids modulate inhibitory GABAergic neurotransmission in many brain regions. Within the temporal lobe, cannabinoid receptors are highly expressed, and are located presynaptically at inhibitory terminals. Here, we have explored the role of type-1 cannabinoid receptors (CB1Rs) at the level of inhibitory synaptic currents and field-recorded network oscillations. We report that arachidonylcyclopropylamide (ACPA; 10 μM), an agonist at CB1R, inhibits GABAergic synaptic transmission onto both superficial and deep medial entorhinal (mEC) neurones, but this has little effect on network oscillations in beta/gamma frequency bands. By contrast, the CB1R antagonist/inverse agonist LY320135 (500 nM), increased GABAergic synaptic activity and beta/gamma oscillatory activity in superficial mEC, was suppressed, whilst that in deep mEC was enhanced. These data indicate that cannabinoid-mediated effects on inhibitory synaptic activity may be constitutively active in vitro, and that modulation of CB1R activation using inverse agonists unmasks complex effects of CBR function on network activity.
Collapse
|
48
|
Galarreta M, Erdélyi F, Szabó G, Hestrin S. Cannabinoid sensitivity and synaptic properties of 2 GABAergic networks in the neocortex. Cereb Cortex 2008; 18:2296-305. [PMID: 18203691 DOI: 10.1093/cercor/bhm253] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Distinct networks of gamma-aminobutyric acidergic interneurons connected by electrical synapses can promote different patterns of activity in the neocortex. Cannabinoids affect memory and cognition by powerfully modulating a subset of inhibitory synapses; however, very little is known about the synaptic properties of the cannabinoid receptor-expressing neurons (CB(1)-positive irregular spiking [CB(1)-IS]) in the neocortex. Using paired recordings in neocortical slices, we 1st report here that synapses of CB(1)-IS cells, but not synapses of fast-spiking (FS) cells, are suppressed by release of endocannabinoids from pyramidal neurons. CB(1)-IS synapses were characterized by a very high failure rate that contrasted with the high reliability of FS synapses. Furthermore, CB(1)-IS cells received excitatory inputs less frequently compared with FS cells and made significantly less frequent inhibitory contacts onto local pyramids. These distinct synaptic properties together with their characteristic irregular firing suggest that CB(1)-IS cells play different role in neocortical function than that of FS cells. Thus, whereas the synaptic properties of FS cells can allow them to impose high-frequency rhythmic oscillatory activity, those of CB(1)-IS cells may lead to disruption of fast rhythmic oscillations. Our results suggest that activity-dependent release of cannabinoids, by blocking CB(1)-IS synapses, may alter the role of inhibition in neocortical circuits.
Collapse
Affiliation(s)
- Mario Galarreta
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
49
|
Le Vasseur M, Ran I, Lacaille JC. Selective induction of metabotropic glutamate receptor 1– and metabotropic glutamate receptor 5–dependent chemical long-term potentiation at oriens/alveus interneuron synapses of mouse hippocampus. Neuroscience 2008; 151:28-42. [DOI: 10.1016/j.neuroscience.2007.09.071] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 08/29/2007] [Accepted: 09/26/2007] [Indexed: 10/22/2022]
|
50
|
Lemtiri-Chlieh F, Levine ES. Lack of depolarization-induced suppression of inhibition (DSI) in layer 2/3 interneurons that receive cannabinoid-sensitive inhibitory inputs. J Neurophysiol 2007; 98:2517-24. [PMID: 17881480 DOI: 10.1152/jn.00817.2007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In layer 2/3 of neocortex, brief trains of action potentials in pyramidal neurons (PNs) induce the mobilization of endogenous cannabinoids (eCBs), resulting in a depression of GABA release from the terminals of inhibitory interneurons (INs). This depolarization-induced suppression of inhibition (DSI) is mediated by activation of the type 1 cannabinoid receptor (CB1) on presynaptic terminals of a subset of INs. However, it is not clear whether CB1 receptors are also expressed at synapses between INs, and whether INs can release eCBs in response to depolarization. In the present studies, brain slices containing somatosensory cortex were prepared from 14- to 21-day-old CD-1 mice. Whole cell recordings were obtained from layer 2/3 PNs and from INs classified as regular spiking nonpyramidal, irregular spiking, or fast spiking. For all three classes of INs, the cannabinoid agonist WIN55,212-2 suppressed inhibitory synaptic activity, similar to the effect seen in PNs. In addition, trains of action potentials in PNs resulted in significant DSI. In INs, however, DSI was not seen in any cell type, even with prolonged high-frequency spike trains that produced calcium increases comparable to that seen with DSI induction in PNs. In addition, blocking eCB reuptake with AM404, which enhanced DSI in PNs, failed to unmask any DSI in INs. Thus the lack of DSI in INs does not appear to be due to an insufficient increase in intracellular calcium or enhanced reuptake. These results suggest that layer 2/3 INs receive CB1-expressing inhibitory inputs, but that eCBs are not released by these INs.
Collapse
Affiliation(s)
- Fouad Lemtiri-Chlieh
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | |
Collapse
|