51
|
Mattera A, Cavallo A, Granato G, Baldassarre G, Pagani M. A Biologically Inspired Neural Network Model to Gain Insight Into the Mechanisms of Post-Traumatic Stress Disorder and Eye Movement Desensitization and Reprocessing Therapy. Front Psychol 2022; 13:944838. [PMID: 35911047 PMCID: PMC9326218 DOI: 10.3389/fpsyg.2022.944838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/06/2022] [Indexed: 01/09/2023] Open
Abstract
Eye movement desensitization and reprocessing (EMDR) therapy is a well-established therapeutic method to treat post-traumatic stress disorder (PTSD). However, how EMDR exerts its therapeutic action has been studied in many types of research but still needs to be completely understood. This is in part due to limited knowledge of the neurobiological mechanisms underlying EMDR, and in part to our incomplete understanding of PTSD. In order to model PTSD, we used a biologically inspired computational model based on firing rate units, encompassing the cortex, hippocampus, and amygdala. Through the modulation of its parameters, we fitted real data from patients treated with EMDR or classical exposure therapy. This allowed us to gain insights into PTSD mechanisms and to investigate how EMDR achieves trauma remission.
Collapse
|
52
|
Perrenoud Q, Leclerc C, Geoffroy H, Vitalis T, Richetin K, Rampon C, Gallopin T. Molecular and electrophysiological features of GABAergic neurons in the dentate gyrus reveal limited homology with cortical interneurons. PLoS One 2022; 17:e0270981. [PMID: 35802727 PMCID: PMC9269967 DOI: 10.1371/journal.pone.0270981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
GABAergic interneurons tend to diversify into similar classes across telencephalic regions. However, it remains unclear whether the electrophysiological and molecular properties commonly used to define these classes are discriminant in the hilus of the dentate gyrus. Here, using patch-clamp combined with single cell RT-PCR, we compare the relevance of commonly used electrophysiological and molecular features for the clustering of GABAergic interneurons sampled from the mouse hilus and primary sensory cortex. While unsupervised clustering groups cortical interneurons into well-established classes, it fails to provide a convincing partition of hilar interneurons. Statistical analysis based on resampling indicates that hilar and cortical GABAergic interneurons share limited homology. While our results do not invalidate the use of classical molecular marker in the hilus, they indicate that classes of hilar interneurons defined by the expression of molecular markers do not exhibit strongly discriminating electrophysiological properties.
Collapse
Affiliation(s)
- Quentin Perrenoud
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Clémence Leclerc
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Hélène Geoffroy
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Tania Vitalis
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Kevin Richetin
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, France
| | - Thierry Gallopin
- Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
- * E-mail:
| |
Collapse
|
53
|
Leal Santos S, Chen BK, Pereira GR, Pham V, Denny CA. Propranolol Administration Modulates Neural Activity in the Hippocampal Hilus During Fear Retrieval. Front Behav Neurosci 2022; 16:919831. [PMID: 35874651 PMCID: PMC9301278 DOI: 10.3389/fnbeh.2022.919831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Altered fear learning is a strong behavioral component of anxiety disorders such as post-traumatic stress disorder (PTSD). Recent efforts have attempted to combine exposure therapies with drugs that target fear memory retrieval and memory reconsolidation, in order to improve treatment efficacy. The noradrenergic (NA) signaling system is of particular interest, due to its role in regulating the stress response and its involvement in fear and learning processes. Importantly, propranolol (P), a non-selective β-adrenergic antagonist, has shown the potential in decreasing exaggerated fear in both humans and animal models. In a previous study, we utilized an activity-dependent tagging murine model to determine the neural mechanisms by which propranolol attenuates learned fear. We found that propranolol acutely decreased memory trace reactivation specifically in the dorsal dentate gyrus (dDG), but not in CA3 or CA1. Here, we extended our previous study by investigating whether propranolol additionally altered activity in the hilus, a polymorphic layer that consists of neurons, mossy cells, and GABAergic interneurons. We found that propranolol acutely reduced overall hilar activity in both the dorsal and ventral hilus. Moreover, we report that propranolol significantly altered the activity of parvalbumin (PV)+ cells in the ventral (vDG), but not dorsal DG (dDG). Together, these results suggest that a β-adrenergic blockade may affect the activity of excitatory and inhibitory cell types in the hilar layer of the DG, and that these alterations may contribute to manipulating fear memory traces.
Collapse
Affiliation(s)
- Sofia Leal Santos
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, United States
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Instituto de Investigação em Ciências da Vida e da Saúde (ICVS)/3Bs - PT Government Associate Laboratory, Guimarães, Portugal
| | - Briana K. Chen
- Neurobiology and Behavior (NB&B) Graduate Program, Columbia University, New York, NY, United States
| | - Guilherme R. Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Instituto de Investigação em Ciências da Vida e da Saúde (ICVS)/3Bs - PT Government Associate Laboratory, Guimarães, Portugal
| | - Vananh Pham
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, United States
| | - Christine A. Denny
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
- Division of Systems Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, United States
- *Correspondence: Christine A. Denny,
| |
Collapse
|
54
|
Kriener B, Hu H, Vervaeke K. Parvalbumin interneuron dendrites enhance gamma oscillations. Cell Rep 2022; 39:110948. [PMID: 35705055 DOI: 10.1016/j.celrep.2022.110948] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 02/07/2022] [Accepted: 05/21/2022] [Indexed: 11/24/2022] Open
Abstract
Dendrites are essential determinants of the input-output relationship of single neurons, but their role in network computations is not well understood. Here, we use a combination of dendritic patch-clamp recordings and in silico modeling to determine how dendrites of parvalbumin (PV)-expressing basket cells contribute to network oscillations in the gamma frequency band. Simultaneous soma-dendrite recordings from PV basket cells in the dentate gyrus reveal that the slope, or gain, of the dendritic input-output relationship is exceptionally low, thereby reducing the cell's sensitivity to changes in its input. By simulating gamma oscillations in detailed network models, we demonstrate that the low gain is key to increase spike synchrony in PV basket cell assemblies when cells are driven by spatially and temporally heterogeneous synaptic inputs. These results highlight the role of inhibitory neuron dendrites in synchronized network oscillations.
Collapse
Affiliation(s)
- Birgit Kriener
- Institute of Basic Medical Sciences, Section of Physiology, University of Oslo, Oslo, Norway
| | - Hua Hu
- Institute of Basic Medical Sciences, Section of Physiology, University of Oslo, Oslo, Norway
| | - Koen Vervaeke
- Institute of Basic Medical Sciences, Section of Physiology, University of Oslo, Oslo, Norway.
| |
Collapse
|
55
|
Li H, Zhao J, Lai L, Xia Y, Wan C, Wei S, Liang J, Chen Y, Xu N. Loss of SST and PV Positive Interneurons in the Ventral Hippocampus Results in Anxiety-like Behavior in 5xFAD Mice. Neurobiol Aging 2022; 117:165-178. [DOI: 10.1016/j.neurobiolaging.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/13/2022] [Accepted: 05/28/2022] [Indexed: 10/18/2022]
|
56
|
Kim SY, Lim W. Population and individual firing behaviors in sparsely synchronized rhythms in the hippocampal dentate gyrus. Cogn Neurodyn 2022; 16:643-665. [PMID: 35603046 PMCID: PMC9120338 DOI: 10.1007/s11571-021-09728-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/26/2021] [Accepted: 10/02/2021] [Indexed: 12/16/2022] Open
Abstract
We investigate population and individual firing behaviors in sparsely synchronized rhythms (SSRs) in a spiking neural network of the hippocampal dentate gyrus (DG). The main encoding granule cells (GCs) are grouped into lamellar clusters. In each GC cluster, there is one inhibitory (I) basket cell (BC) along with excitatory (E) GCs, and they form the E-I loop. Winner-take-all competition, leading to sparse activation of the GCs, occurs in each GC cluster. Such sparsity has been thought to enhance pattern separation performed in the DG. During the winner-take-all competition, SSRs are found to appear in each population of the GCs and the BCs through interaction of excitation of the GCs with inhibition of the BCs. Sparsely synchronized spiking stripes appear successively with the population frequencyf p ( = 13.1 Hz) in the raster plots of spikes. We also note that excitatory hilar mossy cells (MCs) control the firing activity of the GC-BC loop by providing excitation to both the GCs and the BCs. SSR also appears in the population of MCs via interaction with the GCs (i.e., GC-MC loop). Population behaviors in the SSRs are quantitatively characterized in terms of the synchronization measures. In addition, we investigate individual firing activity of GCs, BCs, and MCs in the SSRs. Individual GCs exhibit random spike skipping, leading to a multi-peaked inter-spike-interval histogram, which is well characterized in terms of the random phase-locking degree. In this case, population-averaged mean-firing-rate (MFR) < f i ( GC ) > is less than the population frequency f p . On the other hand, both BCs and MCs show "intrastripe" burstings within stripes, together with random spike skipping. Thus, the population-averaged MFR ⟨ f i ( X ) ⟩ ( X = MC and BC) is larger than f p , in contrast to the case of the GCs. MC loss may occur during epileptogenesis. With decreasing the fraction of the MCs, changes in the population and individual firings in the SSRs are also studied. Finally, quantitative association between the population/individual firing behaviors in the SSRs and the winner-take-all competition is discussed.
Collapse
Affiliation(s)
- Sang-Yoon Kim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| | - Woochang Lim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu, 42411 Korea
| |
Collapse
|
57
|
Simonova NA, Volgushev MA, Malyshev AY. Enhanced Non-Associative Long-Term Potentiation in Immature Granule Cells in the Dentate Gyrus of Adult Rats. Front Synaptic Neurosci 2022; 14:889947. [PMID: 35711669 PMCID: PMC9192440 DOI: 10.3389/fnsyn.2022.889947] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/15/2022] [Indexed: 11/20/2022] Open
Abstract
The dentate gyrus is one of the few sites of neurogenesis in the adult brain. Integration of new-generated granule cells into the hippocampal circuitry provides a substrate for structural plasticity, fundamental for normal function of adult hippocampus. However, mechanisms of synaptic plasticity that mediate integration of new-generated granule cells into the existing circuitry remain poorly understood. Especially mechanisms of plasticity at GABA-ergic synapses remain elusive. Here, we show that postsynaptic spiking without presynaptic activation can induce heterosynaptic, non-associative plasticity at GABA-ergic inputs to both immature and mature granule cells. In both immature and mature neurons, plastic changes were bidirectional and individual inputs could express long-term potentiation (LTP) or long-term depression (LTD), or do not change. However, properties of non-associative plasticity dramatically change with maturation of newly generated granule cells: while in immature cells there was a clear predominance of non-associative LTP and net potentiation across the inputs, in mature neurons, potentiation and depression were balanced with no net change on average. We conclude that GABA-ergic inputs to granule cells are plastic, and that the rules for induction of non-associative plasticity change with maturation. We propose that potentiation-biased non-associative plasticity of GABA-ergic transmission might help to counter-balance an increase of excitatory drive that is facilitated by enhanced LTP at glutamatergic synapses in maturating granule cells. Such mechanism might help to build a strong GABA-ergic input to surviving active new cells, necessary for normal function of mature granule cells, which operate under a tight inhibitory control and generate sparse spiking activity.
Collapse
Affiliation(s)
- Natalia A. Simonova
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| | - Maxim A. Volgushev
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, United States
- *Correspondence: Maxim A. Volgushev
| | - Alexey Y. Malyshev
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
58
|
Yen TY, Huang X, MacLaren DAA, Schlesiger MI, Monyer H, Lien CC. Inhibitory projections connecting the dentate gyri in the two hemispheres support spatial and contextual memory. Cell Rep 2022; 39:110831. [PMID: 35584671 DOI: 10.1016/j.celrep.2022.110831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/17/2021] [Accepted: 04/27/2022] [Indexed: 12/01/2022] Open
Abstract
The dentate gyrus (DG) receives substantial input from the homologous brain area of the contralateral hemisphere. This input is by and large excitatory. Viral-tracing experiments provided anatomical evidence for the existence of GABAergic connectivity between the two DGs, but the function of these projections has remained elusive. Combining electrophysiological and optogenetic approaches, we demonstrate that somatostatin-expressing contralateral DG (SOM+ cDG)-projecting neurons preferentially engage dendrite-targeting interneurons over principal neurons. Single-unit recordings from freely moving mice reveal that optogenetic stimulation of SOM+ cDG projections modulates the activity of GABAergic neurons and principal neurons over multiple timescales. Importantly, we demonstrate that optogenetic silencing of SOM+ cDG projections during spatial memory encoding, but not during memory retrieval, results in compromised DG-dependent memory. Moreover, optogenetic stimulation of SOM+ cDG projections is sufficient to disrupt contextual memory recall. Collectively, our findings reveal that SOM+ long-range projections mediate inter-DG inhibition and contribute to learning and memory.
Collapse
Affiliation(s)
- Ting-Yun Yen
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Xu Huang
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Duncan Archibald Allan MacLaren
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Magdalene Isabell Schlesiger
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology at the Medical Faculty of Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Cheng-Chang Lien
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| |
Collapse
|
59
|
Potrebić M, Pavković Ž, Puškaš N, Pešić V. The Influence of Social Isolation on Social Orientation, Sociability, Social Novelty Preference, and Hippocampal Parvalbumin-Expressing Interneurons in Peripubertal Rats - Understanding the Importance of Meeting Social Needs in Adolescence. Front Behav Neurosci 2022; 16:872628. [PMID: 35592640 PMCID: PMC9113078 DOI: 10.3389/fnbeh.2022.872628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
The fulfillment of belonging needs underlies a variety of behaviors. In order to understand how social needs unmet during maturation shape everyday life, we examined social motivation and cognition in peripubertal rats, as a rodent model of adolescence, subjected to social isolation (SI) during early and early-to-mid adolescence. The behavioral correlates of social orientation (social space preference), sociability (preference for social over non-social novelty), and social novelty preference (SNP) were examined in group-housed (GH) and single-housed (SH) rats in a 3-chamber test. The response to social odors was examined to gain insights into the developmental role of social odors in motivated social behavior. Differentiation between appetitive (number of visits/approaches) and consummatory (exploratory time) aspects of motivated social behavior was done to determine which facet of social motivation characterizes maturation when social needs are met and which aspect dominates when social needs are unsatisfied. The SI-sensitive parvalbumin-expressing interneurons (PVI) in the hippocampus were examined using immunohistochemistry. The main findings are the following: (1) in GH rats, the preference for social space is not evident regardless of animals' age, while sociability becomes apparent in mid-adolescence strictly through consummatory behavior, along with complete SNP (appetitive, consummatory); (2) SH promotes staying in a social chamber/space regardless of animals' age and produces an appetitive preference for it only in early-adolescent animals; (3) SH promotes sociability (appetitive, consummatory) regardless of the animals' age and prevents the SNP; (4) the preference for a social odor is displayed in all the groups through consummatory behavior, while appetitive behavior is evident only in SH rats; (5) the response to social odors does not commensurate directly to the response to conspecifics; (6) SH does not influence PVI in the hippocampus, except in the case of early-adolescence when a transient decrease in the dentate gyrus is observed. These results accentuate the developmental complexity of social motivation and cognition, and the power of SI in adolescence to infringe social maturation at different functional levels, promoting appetitive behavior toward peers overall but harming the interest for social novelty. The findings emphasize the importance of the fulfillment of basic social needs in the navigation through the social world.
Collapse
Affiliation(s)
- Milica Potrebić
- Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Željko Pavković
- Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nela Puškaš
- Institute of Histology and Embryology “Aleksandar Đ. Kostić”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna Pešić
- Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
60
|
Dohm-Hansen S, Donoso F, Lucassen PJ, Clarke G, Nolan YM. The gut microbiome and adult hippocampal neurogenesis: A new focal point for epilepsy? Neurobiol Dis 2022; 170:105746. [DOI: 10.1016/j.nbd.2022.105746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/13/2022] [Accepted: 04/29/2022] [Indexed: 02/07/2023] Open
|
61
|
Kaneko K, Currin CB, Goff KM, Wengert ER, Somarowthu A, Vogels TP, Goldberg EM. Developmentally regulated impairment of parvalbumin interneuron synaptic transmission in an experimental model of Dravet syndrome. Cell Rep 2022; 38:110580. [PMID: 35354025 PMCID: PMC9003081 DOI: 10.1016/j.celrep.2022.110580] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 01/09/2022] [Accepted: 03/06/2022] [Indexed: 11/25/2022] Open
Abstract
Dravet syndrome is a neurodevelopmental disorder characterized by epilepsy, intellectual disability, and sudden death due to pathogenic variants in SCN1A with loss of function of the sodium channel subunit Nav1.1. Nav1.1-expressing parvalbumin GABAergic interneurons (PV-INs) from young Scn1a+/− mice show impaired action potential generation. An approach assessing PV-IN function in the same mice at two time points shows impaired spike generation in all Scn1a+/− mice at postnatal days (P) 16–21, whether deceased prior or surviving to P35, with normalization by P35 in surviving mice. However, PV-IN synaptic transmission is dysfunctional in young Scn1a+/− mice that did not survive and in Scn1a+/− mice ≥ P35. Modeling confirms that PV-IN axonal propagation is more sensitive to decreased sodium conductance than spike generation. These results demonstrate dynamic dysfunction in Dravet syndrome: combined abnormalities of PV-IN spike generation and propagation drives early disease severity, while ongoing dysfunction of synaptic transmission contributes to chronic pathology. Dravet syndrome is caused by variants in SCN1A with loss of function of Nav1.1 sodium channels. Kaneko et al. use the “mini-slice” to record at two developmental time points. Impaired spike generation of Nav1.1-expressing PV interneurons in Scn1a+/− mice is transient, while abnormalities of PV interneuron synaptic transmission persist.
Collapse
Affiliation(s)
- Keisuke Kaneko
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Christopher B Currin
- The Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, Austria
| | - Kevin M Goff
- Medical Scientist Training Program (MSTP), The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Eric R Wengert
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Ala Somarowthu
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Tim P Vogels
- The Institute of Science and Technology Austria, Am Campus 1, Klosterneuburg, Austria
| | - Ethan M Goldberg
- Division of Neurology, Department of Pediatrics, The Children's Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
62
|
Tomé DF, Sadeh S, Clopath C. Coordinated hippocampal-thalamic-cortical communication crucial for engram dynamics underneath systems consolidation. Nat Commun 2022; 13:840. [PMID: 35149680 PMCID: PMC8837777 DOI: 10.1038/s41467-022-28339-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 01/13/2022] [Indexed: 11/09/2022] Open
Abstract
Systems consolidation refers to the time-dependent reorganization of memory representations or engrams across brain regions. Despite recent advancements in unravelling this process, the exact mechanisms behind engram dynamics and the role of associated pathways remain largely unknown. Here we propose a biologically-plausible computational model to address this knowledge gap. By coordinating synaptic plasticity timescales and incorporating a hippocampus-thalamus-cortex circuit, our model is able to couple engram reactivations across these regions and thereby reproduce key dynamics of cortical and hippocampal engram cells along with their interdependencies. Decoupling hippocampal-thalamic-cortical activity disrupts systems consolidation. Critically, our model yields testable predictions regarding hippocampal and thalamic engram cells, inhibitory engrams, thalamic inhibitory input, and the effect of thalamocortical synaptic coupling on retrograde amnesia induced by hippocampal lesions. Overall, our results suggest that systems consolidation emerges from coupled reactivations of engram cells in distributed brain regions enabled by coordinated synaptic plasticity timescales in multisynaptic subcortical-cortical circuits.
Collapse
Affiliation(s)
| | - Sadra Sadeh
- Department of Bioengineering, Imperial College London, London, UK
| | - Claudia Clopath
- Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
63
|
Knipper M, Singer W, Schwabe K, Hagberg GE, Li Hegner Y, Rüttiger L, Braun C, Land R. Disturbed Balance of Inhibitory Signaling Links Hearing Loss and Cognition. Front Neural Circuits 2022; 15:785603. [PMID: 35069123 PMCID: PMC8770933 DOI: 10.3389/fncir.2021.785603] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Neuronal hyperexcitability in the central auditory pathway linked to reduced inhibitory activity is associated with numerous forms of hearing loss, including noise damage, age-dependent hearing loss, and deafness, as well as tinnitus or auditory processing deficits in autism spectrum disorder (ASD). In most cases, the reduced central inhibitory activity and the accompanying hyperexcitability are interpreted as an active compensatory response to the absence of synaptic activity, linked to increased central neural gain control (increased output activity relative to reduced input). We here suggest that hyperexcitability also could be related to an immaturity or impairment of tonic inhibitory strength that typically develops in an activity-dependent process in the ascending auditory pathway with auditory experience. In these cases, high-SR auditory nerve fibers, which are critical for the shortest latencies and lowest sound thresholds, may have either not matured (possibly in congenital deafness or autism) or are dysfunctional (possibly after sudden, stressful auditory trauma or age-dependent hearing loss linked with cognitive decline). Fast auditory processing deficits can occur despite maintained basal hearing. In that case, tonic inhibitory strength is reduced in ascending auditory nuclei, and fast inhibitory parvalbumin positive interneuron (PV-IN) dendrites are diminished in auditory and frontal brain regions. This leads to deficits in central neural gain control linked to hippocampal LTP/LTD deficiencies, cognitive deficits, and unbalanced extra-hypothalamic stress control. Under these conditions, a diminished inhibitory strength may weaken local neuronal coupling to homeostatic vascular responses required for the metabolic support of auditory adjustment processes. We emphasize the need to distinguish these two states of excitatory/inhibitory imbalance in hearing disorders: (i) Under conditions of preserved fast auditory processing and sustained tonic inhibitory strength, an excitatory/inhibitory imbalance following auditory deprivation can maintain precise hearing through a memory linked, transient disinhibition that leads to enhanced spiking fidelity (central neural gain⇑) (ii) Under conditions of critically diminished fast auditory processing and reduced tonic inhibitory strength, hyperexcitability can be part of an increased synchronization over a broader frequency range, linked to reduced spiking reliability (central neural gain⇓). This latter stage mutually reinforces diminished metabolic support for auditory adjustment processes, increasing the risks for canonical dementia syndromes.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- *Correspondence: Marlies Knipper,
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Kerstin Schwabe
- Experimental Neurosurgery, Department of Neurosurgery, Hannover Medical School, Hanover, Germany
| | - Gisela E. Hagberg
- Department of Biomedical Magnetic Resonance, University Hospital Tübingen (UKT), Tübingen, Germany
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Yiwen Li Hegner
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Christoph Braun
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Rüdiger Land
- Department of Experimental Otology, Institute for Audioneurotechnology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
64
|
Kim SY, Lim W. Dynamical origin for winner-take-all competition in a biological network of the hippocampal dentate gyrus. Phys Rev E 2022; 105:014418. [PMID: 35193268 DOI: 10.1103/physreve.105.014418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
We consider a biological network of the hippocampal dentate gyrus (DG). Computational models suggest that the DG would be a preprocessor for pattern separation (i.e., a process transforming a set of similar input patterns into distinct nonoverlapping output patterns) which could facilitate pattern storage and retrieval in the CA3 area of the hippocampus. The main encoding cells in the DG are the granule cells (GCs) which receive the input from the entorhinal cortex (EC) and send their output to the CA3. We note that the activation degree of GCs is very low (∼5%). This sparsity has been thought to enhance the pattern separation. We investigate the dynamical origin for winner-take-all (WTA) competition which leads to sparse activation of the GCs. The whole GCs are grouped into lamellar clusters. In each cluster, there is one inhibitory (I) basket cell (BC) along with excitatory (E) GCs. There are three kinds of external inputs into the GCs: the direct excitatory EC input; the indirect feedforward inhibitory EC input, mediated by the HIPP (hilar perforant path-associated) cells; and the excitatory input from the hilar mossy cells (MCs). The firing activities of the GCs are determined via competition between the external E and I inputs. The E-I conductance ratio R_{E-I}^{(con)}^{*} (given by the time average of the ratio of the external E to I conductances) may represent well the degree of such external E-I input competition. It is thus found that GCs become active when their R_{E-I}^{(con)}^{*} is larger than a threshold R_{th}^{*}, and then the mean firing rates of the active GCs are strongly correlated with R_{E-I}^{(con)}^{*}. In each cluster, the feedback inhibition from the BC may select the winner GCs. GCs with larger R_{E-I}^{(con)}^{*} than the threshold R_{th}^{*} survive, and they become winners; all the other GCs with smaller R_{E-I}^{(con)}^{*} become silent. In this way, WTA competition occurs via competition between the firing activity of the GCs and the feedback inhibition from the BC in each cluster. Finally, we also study the effects of MC death and adult-born immature GCs on the WTA competition.
Collapse
Affiliation(s)
- Sang-Yoon Kim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu 42411, Korea
| | - Woochang Lim
- Institute for Computational Neuroscience and Department of Science Education, Daegu National University of Education, Daegu 42411, Korea
| |
Collapse
|
65
|
Guzman SJ, Schlögl A, Espinoza C, Zhang X, Suter BA, Jonas P. How connectivity rules and synaptic properties shape the efficacy of pattern separation in the entorhinal cortex-dentate gyrus-CA3 network. NATURE COMPUTATIONAL SCIENCE 2021; 1:830-842. [PMID: 38217181 DOI: 10.1038/s43588-021-00157-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/12/2021] [Indexed: 01/15/2024]
Abstract
Pattern separation is a fundamental brain computation that converts small differences in input patterns into large differences in output patterns. Several synaptic mechanisms of pattern separation have been proposed, including code expansion, inhibition and plasticity; however, which of these mechanisms play a role in the entorhinal cortex (EC)-dentate gyrus (DG)-CA3 circuit, a classical pattern separation circuit, remains unclear. Here we show that a biologically realistic, full-scale EC-DG-CA3 circuit model, including granule cells (GCs) and parvalbumin-positive inhibitory interneurons (PV+-INs) in the DG, is an efficient pattern separator. Both external gamma-modulated inhibition and internal lateral inhibition mediated by PV+-INs substantially contributed to pattern separation. Both local connectivity and fast signaling at GC-PV+-IN synapses were important for maximum effectiveness. Similarly, mossy fiber synapses with conditional detonator properties contributed to pattern separation. By contrast, perforant path synapses with Hebbian synaptic plasticity and direct EC-CA3 connection shifted the network towards pattern completion. Our results demonstrate that the specific properties of cells and synapses optimize higher-order computations in biological networks and might be useful to improve the deep learning capabilities of technical networks.
Collapse
Affiliation(s)
- S Jose Guzman
- IST Austria, Klosterneuburg, Austria
- Institute of Molecular Biotechnology, Vienna, Austria
| | | | - Claudia Espinoza
- IST Austria, Klosterneuburg, Austria
- Medical University of Austria, Division of Cognitive Neurobiology, Vienna, Austria
| | - Xiaomin Zhang
- IST Austria, Klosterneuburg, Austria
- Brain Research Institute, University of Zürich, Zurich, Switzerland
| | | | | |
Collapse
|
66
|
Aertsen A. Insights into hippocampal network function. NATURE COMPUTATIONAL SCIENCE 2021; 1:782-783. [PMID: 38217180 DOI: 10.1038/s43588-021-00159-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Affiliation(s)
- Ad Aertsen
- Bernstein Center Freiburg and Faculty of Biology, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
67
|
Reid HMO, Snowden TM, Shkolnikov I, Breit KR, Rodriguez C, Thomas JD, Christie BR. Prenatal alcohol and cannabis exposure can have opposing and region-specific effects on parvalbumin interneuron numbers in the hippocampus. Alcohol Clin Exp Res 2021; 45:2246-2255. [PMID: 34523142 PMCID: PMC8642289 DOI: 10.1111/acer.14708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND We recently showed that alcohol and cannabis can interact prenatally, and in a recent review paper, we identified parvalbumin-positive (PV) interneurons in the hippocampus as a potential point of convergence for these teratogens. METHODS A 2 (Ethanol [EtOH], Air) × 2 (tetrahydrocannabinol [THC], Vehicle) design was used to expose pregnant Sprague-Dawley rats to either EtOH or air, in addition to either THC or the inhalant vehicle solution, during gestational days 5-20. Immunohistochemistry was performed to detect PV interneurons in 1 male and 1 female pup from each litter at postnatal day 70. RESULTS Significant between-group and subregion-specific effects were found in the dorsal cornu ammonis 1 (CA1) subfield and the ventral dentate gyrus (DG). In the dorsal CA1 subfield, there was an increase in the number of PV interneurons in both the EtOH and EtOH +THC groups, but a decrease with THC alone. There were fewer changes in interneuron numbers overall in the DG, though there was a sex difference, with a decrease in the number of PV interneurons in the THC-exposed group in males. There was also a greater cell layer volume in the DG in the EtOH +THC group than the control group, and in the CA1 region in the EtOH group compared to the control and THC groups. CONCLUSIONS Prenatal exposure to alcohol and THC differentially affects parvalbumin-positive interneuron numbers in the hippocampus, indicating that both individual and combined exposure can impact the balance of excitation and inhibition in a structure critically involved in learning and memory processes.
Collapse
Affiliation(s)
- Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Taylor M Snowden
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Irene Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Kristen R Breit
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
- Department of Psychology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | - Cristina Rodriguez
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Jennifer D Thomas
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program, Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, USA
| |
Collapse
|
68
|
Baccini G, Wulff P. Commentary on: Peng, Y et al., Structured recurrent inhibition in the presubiculum could improve information processing. Pflugers Arch 2021; 473:1691-1693. [PMID: 34648053 PMCID: PMC8528764 DOI: 10.1007/s00424-021-02626-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Gilda Baccini
- Institute of Physiology, Christian-Albrechts-University Kiel, 24098, Kiel, Germany.
| | - Peer Wulff
- Institute of Physiology, Christian-Albrechts-University Kiel, 24098, Kiel, Germany.
| |
Collapse
|
69
|
Level of hM4D(Gi) DREADD Expression Determines Inhibitory and Neurotoxic Effects in the Hippocampus. eNeuro 2021; 8:ENEURO.0105-21.2021. [PMID: 34620623 PMCID: PMC8570686 DOI: 10.1523/eneuro.0105-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Selective neuromodulation using designer receptors exclusively activated by designer drugs (DREADDs) has become an increasingly important research tool, as well as an emerging therapeutic approach. However, the safety profile of DREADD expression is unknown. Here, different titers of adeno-associated viral (AAV) vector were administered in an attempt to vary total expression levels of the inhibitory DREADD hM4D(Gi) in excitatory hippocampal neurons. Male Sprague Dawley rats were injected with AAV2/7 encoding DREADD-mCherry, DREADD, or mCherry. Pronounced neuronal loss and neuroinflammatory reactions were observed after transduction with the high titer DREADD AAV, which also resulted in the highest DREADD expression levels. No such effects were observed in the mCherry control group, despite an equally high titer, nor in conditions where lower viral vector titers were injected. In the high titer DREADD conditions, dentate gyrus (DG) evoked potentials were inhibited on clozapine-induced activation of hM4D(Gi), while in low titer conditions DG evoked potentials were enhanced. Recordings of single neuronal activity nevertheless indicated a reduction in spontaneous firing of granule cell layer neurons. Our results indicate that prolonged, high levels of DREADD expression can have neurotoxic effects and that chemogenetic suppression of excitatory hippocampal neurons can paradoxically enhance DG evoked potentials.
Collapse
|
70
|
Pinna A, Colasanti A. The Neurometabolic Basis of Mood Instability: The Parvalbumin Interneuron Link-A Systematic Review and Meta-Analysis. Front Pharmacol 2021; 12:689473. [PMID: 34616292 PMCID: PMC8488267 DOI: 10.3389/fphar.2021.689473] [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: 03/31/2021] [Accepted: 08/18/2021] [Indexed: 12/23/2022] Open
Abstract
The neurobiological bases of mood instability are poorly understood. Neuronal network alterations and neurometabolic abnormalities have been implicated in the pathophysiology of mood and anxiety conditions associated with mood instability and hence are candidate mechanisms underlying its neurobiology. Fast-spiking parvalbumin GABAergic interneurons modulate the activity of principal excitatory neurons through their inhibitory action determining precise neuronal excitation balance. These interneurons are directly involved in generating neuronal networks activities responsible for sustaining higher cerebral functions and are especially vulnerable to metabolic stress associated with deficiency of energy substrates or mitochondrial dysfunction. Parvalbumin interneurons are therefore candidate key players involved in mechanisms underlying the pathogenesis of brain disorders associated with both neuronal networks' dysfunction and brain metabolism dysregulation. To provide empirical support to this hypothesis, we hereby report meta-analytical evidence of parvalbumin interneurons loss or dysfunction in the brain of patients with Bipolar Affective Disorder (BPAD), a condition primarily characterized by mood instability for which the pathophysiological role of mitochondrial dysfunction has recently emerged as critically important. We then present a comprehensive review of evidence from the literature illustrating the bidirectional relationship between deficiency in mitochondrial-dependent energy production and parvalbumin interneuron abnormalities. We propose a mechanistic explanation of how alterations in neuronal excitability, resulting from parvalbumin interneurons loss or dysfunction, might manifest clinically as mood instability, a poorly understood clinical phenotype typical of the most severe forms of affective disorders. The evidence we report provides insights on the broader therapeutic potential of pharmacologically targeting parvalbumin interneurons in psychiatric and neurological conditions characterized by both neurometabolic and neuroexcitability abnormalities.
Collapse
Affiliation(s)
- Antonello Pinna
- School of Life Sciences, University of Sussex, Brighton, United Kingdom.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Alessandro Colasanti
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
71
|
Sparsification of AP firing in adult-born hippocampal granule cells via voltage-dependent α5-GABA A receptors. Cell Rep 2021; 37:109768. [PMID: 34610304 DOI: 10.1016/j.celrep.2021.109768] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/07/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022] Open
Abstract
GABA can depolarize immature neurons close to the action potential (AP) threshold in development and adult neurogenesis. Nevertheless, GABAergic synapses effectively inhibit AP firing in newborn granule cells of the adult hippocampus as early as two weeks post-mitosis. The underlying mechanisms are largely unclear. Here, we analyze GABAergic inputs in newborn hippocampal granule cells mediated by soma-targeting parvalbumin and dendrite-targeting somatostatin interneurons. Surprisingly, both interneuron subtypes activate α5-subunit-containing GABAA receptors (α5-GABAARs) in young neurons, showing a nonlinear voltage dependence with increasing conductance around the AP threshold. By contrast, in mature cells, parvalbumin interneurons mediate linear GABAergic synaptic currents lacking α5-subunits, while somatostatin interneurons continue to target nonlinear α5-GABAARs. Computational modeling shows that the voltage-dependent amplification of α5-GABAAR opening in young neurons is crucial for inhibition of AP firing to generate balanced and sparse firing activity, even with depolarized GABA reversal potential.
Collapse
|
72
|
Giesers NK, Wirths O. Loss of Hippocampal Calretinin and Parvalbumin Interneurons in the 5XFAD Mouse Model of Alzheimer's Disease. ASN Neuro 2021; 12:1759091420925356. [PMID: 32423230 PMCID: PMC7238451 DOI: 10.1177/1759091420925356] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The deposition of amyloid-β peptides in the form of extracellular plaques
and neuronal degeneration belong to the hallmark features of
Alzheimer’s disease (AD). In addition, impaired calcium homeostasis
and altered levels in calcium-binding proteins seem to be associated
with the disease process. In this study, calretinin- (CR) and
parvalbumin- (PV) positive gamma-aminobutyric acid-producing
(GABAergic) interneurons were quantified in different hippocampal
subfields of 12-month-old wild-type mice, as well as in the transgenic
AD mouse models 5XFAD and Tg4-42. While, in comparison with wild-type
mice, CR-positive interneurons were mainly reduced in the CA1 and
CA2/3 regions in plaque-bearing 5XFAD mice, PV-positive interneurons
were reduced in all analyzed subfields including the dentate gyrus. No
reduction in CR- and PV-positive interneuron numbers was detected in
the non-plaque-forming Tg4-42 mouse, although this model has been
previously demonstrated to harbor a massive loss of CA1 pyramidal
neurons. These results provide information about hippocampal
interneuron numbers in two relevant AD mouse models, suggesting that
interneuron loss in this brain region may be related to extracellular
amyloid burden.
Collapse
Affiliation(s)
- Naomi K Giesers
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Oliver Wirths
- Department of Psychiatry and Psychotherapy, Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| |
Collapse
|
73
|
Oulé M, Atucha E, Wells TM, Macharadze T, Sauvage MM, Kreutz MR, Lopez-Rojas J. Dendritic Kv4.2 potassium channels selectively mediate spatial pattern separation in the dentate gyrus. iScience 2021; 24:102876. [PMID: 34386734 PMCID: PMC8346659 DOI: 10.1016/j.isci.2021.102876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/22/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022] Open
Abstract
The capacity to distinguish comparable experiences is fundamental for the recall of similar memories and has been proposed to require pattern separation in the dentate gyrus (DG). However, the cellular mechanisms by which mature granule cells (GCs) of the DG accomplish this function are poorly characterized. Here, we show that Kv4.2 channels selectively modulate the excitability of medial dendrites of dentate GCs. These dendrites are targeted by the medial entorhinal cortex, the main source of spatial inputs to the DG. Accordingly, we found that the spatial pattern separation capability of animals lacking the Kv4.2 channel is significantly impaired. This points to the role of intrinsic excitability in supporting the mnemonic function of the dentate and to the Kv4.2 channel as a candidate substrate promoting spatial pattern separation.
Collapse
Affiliation(s)
- Marie Oulé
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Erika Atucha
- Functional Architecture of Memory Department, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Tenyse M. Wells
- Functional Architecture of Memory Department, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Tamar Macharadze
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
- Department of Anesthesiology and Intensive Care Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Magdalena M. Sauvage
- Functional Architecture of Memory Department, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
- Otto von Guericke University, Medical Faculty, Functional Neuroplasticity Department, 39120, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
| | - Michael R. Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Leibniz Group 'Dendritic Organelles and Synaptic Function', University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), 20251 Hamburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Jeffrey Lopez-Rojas
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA
| |
Collapse
|
74
|
Memel M, Staffaroni AM, Cobigo Y, Casaletto KB, Fonseca C, Bettcher BM, Yassa MA, Elahi FM, Wolf A, Rosen HJ, Kramer JH. APOE moderates the effect of hippocampal blood flow on memory pattern separation in clinically normal older adults. Hippocampus 2021; 31:845-857. [PMID: 33835624 PMCID: PMC8295213 DOI: 10.1002/hipo.23327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/25/2021] [Accepted: 03/07/2021] [Indexed: 11/10/2022]
Abstract
Pattern separation, the ability to differentiate new information from previously experienced similar information, is highly sensitive to hippocampal structure and function and declines with age. Functional MRI studies have demonstrated hippocampal hyperactivation in older adults compared to young, with greater task-related activation associated with worse pattern separation performance. The current study was designed to determine whether pattern separation was sensitive to differences in task-free hippocampal cerebral blood flow (CBF) in 130 functionally intact older adults. Given prior evidence that apolipoprotein E e4 (APOE e4) status moderates the relationship between CBF and episodic memory, we predicted a stronger negative relationship between hippocampal CBF and pattern separation in APOE e4 carriers. An interaction between APOE group and right hippocampal CBF was present, such that greater right hippocampal CBF was related to better lure discrimination in noncarriers, whereas the effect reversed directionality in e4 carriers. These findings suggest that neurovascular changes in the medial temporal lobe may underlie memory deficits in cognitively normal older adults who are APOE e4 carriers.
Collapse
Affiliation(s)
- Molly Memel
- San Francisco VA Medical Center, San Francisco, California
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Adam M. Staffaroni
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Yann Cobigo
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Kaitlin B. Casaletto
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Corrina Fonseca
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Brianne M. Bettcher
- Department of Neurology, University of Colorado Anschutz Medical Campus, CU Alzheimer’s and Cognition Center, Aurora, Colorado
| | - Michael A. Yassa
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, California
| | - Fanny M. Elahi
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Amy Wolf
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Howard J. Rosen
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Joel H. Kramer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| |
Collapse
|
75
|
Johnston S, Parylak SL, Kim S, Mac N, Lim C, Gallina I, Bloyd C, Newberry A, Saavedra CD, Novak O, Gonçalves JT, Gage FH, Shtrahman M. AAV ablates neurogenesis in the adult murine hippocampus. eLife 2021; 10:e59291. [PMID: 34259630 PMCID: PMC8331179 DOI: 10.7554/elife.59291] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) has been widely used as a viral vector across mammalian biology and has been shown to be safe and effective in human gene therapy. We demonstrate that neural progenitor cells (NPCs) and immature dentate granule cells (DGCs) within the adult murine hippocampus are particularly sensitive to rAAV-induced cell death. Cell loss is dose dependent and nearly complete at experimentally relevant viral titers. rAAV-induced cell death is rapid and persistent, with loss of BrdU-labeled cells within 18 hr post-injection and no evidence of recovery of adult neurogenesis at 3 months post-injection. The remaining mature DGCs appear hyperactive 4 weeks post-injection based on immediate early gene expression, consistent with previous studies investigating the effects of attenuating adult neurogenesis. In vitro application of AAV or electroporation of AAV2 inverted terminal repeats (ITRs) is sufficient to induce cell death. Efficient transduction of the dentategyrus (DG)- without ablating adult neurogenesis- can be achieved by injection of rAAV2-retro serotyped virus into CA3. rAAV2-retro results in efficient retrograde labeling of mature DGCs and permits in vivo two-photon calcium imaging of dentate activity while leaving adult neurogenesis intact. These findings expand on recent reports implicating rAAV-linked toxicity in stem cells and other cell types and suggest that future work using rAAV as an experimental tool in the DG and as a gene therapy for diseases of the central nervous system should be carefully evaluated.
Collapse
Affiliation(s)
- Stephen Johnston
- Neurosciences Graduate Program, University of California, San DiegoLa JollaUnited States
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Sarah L Parylak
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Stacy Kim
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
- Department of Neurosciences, University of California, San DiegoLa JollaUnited States
| | - Nolan Mac
- Department of Biology, University of California, San DiegoLa JollaUnited States
| | - Christina Lim
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Iryna Gallina
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Cooper Bloyd
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Alexander Newberry
- Department of Physics, University of California, San DiegoLa JollaUnited States
| | - Christian D Saavedra
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Ondrej Novak
- Laboratory of Experimental Epileptology, Department of Physiology, Second Faculty of Medicine, Charles UniversityPragueUnited Kingdom
| | - J Tiago Gonçalves
- Ruth L. and David S. Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of MedicineBronxUnited States
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of MedicineBronxUnited States
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological StudiesLa JollaUnited States
| | - Matthew Shtrahman
- Department of Neurosciences, University of California, San DiegoLa JollaUnited States
| |
Collapse
|
76
|
Uemura M, Blankvoort S, Tok SSL, Yuan L, Cobar LF, Lit KK, Tashiro A. A neurogenic microenvironment defined by excitatory-inhibitory neuronal circuits in adult dentate gyrus. Cell Rep 2021; 36:109324. [PMID: 34233196 DOI: 10.1016/j.celrep.2021.109324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 05/23/2021] [Accepted: 06/08/2021] [Indexed: 10/20/2022] Open
Abstract
Adult neurogenesis in the dentate gyrus plays a role in adaptive brain functions such as memory formation. Adding new neurons to a specific locus of a neural circuit with functional needs is an efficient way to achieve such an adaptive function. However, it is unknown whether neurogenesis is linked to local functional demands potentially specified by the activity of neuronal circuits. By examining the distribution of neurogenesis and different types of neuronal activity in the dentate gyrus of freely moving adult rats, we find that neurogenesis is positionally associated with active excitatory neurons, some of which show place-cell activity, but is positionally dissociated from a type of interneuron with high-burst tendency. Our finding suggests that the behaviorally relevant activity of excitatory-inhibitory neuronal circuits can define a microenvironment stimulating/inhibiting neurogenesis. Such local regulation of neurogenesis may contribute to strategic recruitment of new neurons to modify functionally relevant neural circuits.
Collapse
Affiliation(s)
- Masato Uemura
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, 7030 Trondheim, Norway
| | - Stefan Blankvoort
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, 7030 Trondheim, Norway
| | - Sean Shui Liang Tok
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Li Yuan
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Luis Fernando Cobar
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Kwok Keung Lit
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Ayumu Tashiro
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology, 7030 Trondheim, Norway; School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore.
| |
Collapse
|
77
|
Peng Y, Barreda Tomas FJ, Pfeiffer P, Drangmeister M, Schreiber S, Vida I, Geiger JRP. Spatially structured inhibition defined by polarized parvalbumin interneuron axons promotes head direction tuning. SCIENCE ADVANCES 2021; 7:7/25/eabg4693. [PMID: 34134979 PMCID: PMC8208710 DOI: 10.1126/sciadv.abg4693] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/04/2021] [Indexed: 05/04/2023]
Abstract
In cortical microcircuits, it is generally assumed that fast-spiking parvalbumin interneurons mediate dense and nonselective inhibition. Some reports indicate sparse and structured inhibitory connectivity, but the computational relevance and the underlying spatial organization remain unresolved. In the rat superficial presubiculum, we find that inhibition by fast-spiking interneurons is organized in the form of a dominant super-reciprocal microcircuit motif where multiple pyramidal cells recurrently inhibit each other via a single interneuron. Multineuron recordings and subsequent 3D reconstructions and analysis further show that this nonrandom connectivity arises from an asymmetric, polarized morphology of fast-spiking interneuron axons, which individually cover different directions in the same volume. Network simulations assuming topographically organized input demonstrate that such polarized inhibition can improve head direction tuning of pyramidal cells in comparison to a "blanket of inhibition." We propose that structured inhibition based on asymmetrical axons is an overarching spatial connectivity principle for tailored computation across brain regions.
Collapse
Affiliation(s)
- Yangfan Peng
- Institute for Neurophysiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Federico J Barreda Tomas
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Philippstr. 13, 10115 Berlin, Germany
| | - Paul Pfeiffer
- Bernstein Center for Computational Neuroscience Berlin, Philippstr. 13, 10115 Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, 10115 Berlin, Germany
| | - Moritz Drangmeister
- Bernstein Center for Computational Neuroscience Berlin, Philippstr. 13, 10115 Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, 10115 Berlin, Germany
| | - Susanne Schreiber
- Bernstein Center for Computational Neuroscience Berlin, Philippstr. 13, 10115 Berlin, Germany
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, 10115 Berlin, Germany
| | - Imre Vida
- Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
| | - Jörg R P Geiger
- Institute for Neurophysiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
| |
Collapse
|
78
|
Vandael D, Okamoto Y, Borges-Merjane C, Vargas-Barroso V, Suter BA, Jonas P. Subcellular patch-clamp techniques for single-bouton stimulation and simultaneous pre- and postsynaptic recording at cortical synapses. Nat Protoc 2021; 16:2947-2967. [PMID: 33990799 DOI: 10.1038/s41596-021-00526-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/01/2021] [Indexed: 02/03/2023]
Abstract
Rigorous investigation of synaptic transmission requires analysis of unitary synaptic events by simultaneous recording from presynaptic terminals and postsynaptic target neurons. However, this has been achieved at only a limited number of model synapses, including the squid giant synapse and the mammalian calyx of Held. Cortical presynaptic terminals have been largely inaccessible to direct presynaptic recording, due to their small size. Here, we describe a protocol for improved subcellular patch-clamp recording in rat and mouse brain slices, with the synapse in a largely intact environment. Slice preparation takes ~2 h, recording ~3 h and post hoc morphological analysis 2 d. Single presynaptic hippocampal mossy fiber terminals are stimulated minimally invasively in the bouton-attached configuration, in which the cytoplasmic content remains unperturbed, or in the whole-bouton configuration, in which the cytoplasmic composition can be precisely controlled. Paired pre-postsynaptic recordings can be integrated with biocytin labeling and morphological analysis, allowing correlative investigation of synapse structure and function. Paired recordings can be obtained from mossy fiber terminals in slices from both rats and mice, implying applicability to genetically modified synapses. Paired recordings can also be performed together with axon tract stimulation or optogenetic activation, allowing comparison of unitary and compound synaptic events in the same target cell. Finally, paired recordings can be combined with spontaneous event analysis, permitting collection of miniature events generated at a single identified synapse. In conclusion, the subcellular patch-clamp techniques detailed here should facilitate analysis of biophysics, plasticity and circuit function of cortical synapses in the mammalian central nervous system.
Collapse
Affiliation(s)
- David Vandael
- IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria
| | - Yuji Okamoto
- IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria
| | | | | | - Benjamin A Suter
- IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria
| | - Peter Jonas
- IST Austria (Institute of Science and Technology Austria), Klosterneuburg, Austria.
| |
Collapse
|
79
|
Comeras LB, Hörmer N, Mohan Bethuraj P, Tasan RO. NPY Released From GABA Neurons of the Dentate Gyrus Specially Reduces Contextual Fear Without Affecting Cued or Trace Fear. Front Synaptic Neurosci 2021; 13:635726. [PMID: 34122036 PMCID: PMC8187774 DOI: 10.3389/fnsyn.2021.635726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/20/2021] [Indexed: 11/15/2022] Open
Abstract
Disproportionate, maladapted, and generalized fear are essential hallmarks of posttraumatic stress disorder (PTSD), which develops upon severe trauma in a subset of exposed individuals. Among the brain areas that are processing fear memories, the hippocampal formation exerts a central role linking emotional-affective with cognitive aspects. In the hippocampus, neuronal excitability is constrained by multiple GABAergic interneurons with highly specialized functions and an extensive repertoire of co-released neuromodulators. Neuropeptide Y (NPY) is one of these co-transmitters that significantly affects hippocampal signaling, with ample evidence supporting its fundamental role in emotional, cognitive, and metabolic circuitries. Here we investigated the role of NPY in relation to GABA, both released from the same interneurons of the dorsal dentate gyrus (DG), in different aspects of fear conditioning. We demonstrated that activation of dentate GABA neurons specifically during fear recall reduced cue-related as well as trace-related freezing behavior, whereas inhibition of the same neurons had no significant effects. Interestingly, concomitant overexpression of NPY in these neurons did not further modify fear recall, neither under baseline conditions nor upon chemogenetic stimulation. However, potentially increased co-release of NPY substantially reduced contextual fear, promoted extinction learning, and long-term suppression of fear in a foreground context–conditioning paradigm. Importantly, NPY in the dorsal DG was not only expressed in somatostatin neurons, but also in parvalbumin-positive basket cells and axoaxonic cells, indicating intense feedback and feedforward modulation of hippocampal signaling and precise curtailing of neuronal engrams. Thus, these findings suggest that co-release of NPY from specific interneuron populations of the dorsal DG modifies dedicated aspects of hippocampal processing by sharpening the activation of neural engrams and the consecutive fear response. Since inappropriate and generalized fear is the major impediment in the treatment of PTSD patients, the dentate NPY system may be a suitable access point to ameliorate PTSD symptoms and improve the inherent disease course.
Collapse
Affiliation(s)
- Lucas B Comeras
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Noa Hörmer
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Ramon O Tasan
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
80
|
Wingert JC, Sorg BA. Impact of Perineuronal Nets on Electrophysiology of Parvalbumin Interneurons, Principal Neurons, and Brain Oscillations: A Review. Front Synaptic Neurosci 2021; 13:673210. [PMID: 34040511 PMCID: PMC8141737 DOI: 10.3389/fnsyn.2021.673210] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/14/2021] [Indexed: 12/11/2022] Open
Abstract
Perineuronal nets (PNNs) are specialized extracellular matrix structures that surround specific neurons in the brain and spinal cord, appear during critical periods of development, and restrict plasticity during adulthood. Removal of PNNs can reinstate juvenile-like plasticity or, in cases of PNN removal during early developmental stages, PNN removal extends the critical plasticity period. PNNs surround mainly parvalbumin (PV)-containing, fast-spiking GABAergic interneurons in several brain regions. These inhibitory interneurons profoundly inhibit the network of surrounding neurons via their elaborate contacts with local pyramidal neurons, and they are key contributors to gamma oscillations generated across several brain regions. Among other functions, these gamma oscillations regulate plasticity associated with learning, decision making, attention, cognitive flexibility, and working memory. The detailed mechanisms by which PNN removal increases plasticity are only beginning to be understood. Here, we review the impact of PNN removal on several electrophysiological features of their underlying PV interneurons and nearby pyramidal neurons, including changes in intrinsic and synaptic membrane properties, brain oscillations, and how these changes may alter the integration of memory-related information. Additionally, we review how PNN removal affects plasticity-associated phenomena such as long-term potentiation (LTP), long-term depression (LTD), and paired-pulse ratio (PPR). The results are discussed in the context of the role of PV interneurons in circuit function and how PNN removal alters this function.
Collapse
Affiliation(s)
- Jereme C Wingert
- Program in Neuroscience, Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Barbara A Sorg
- Program in Neuroscience, Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| |
Collapse
|
81
|
Experience-Dependent Inhibitory Plasticity Is Mediated by CCK+ Basket Cells in the Developing Dentate Gyrus. J Neurosci 2021; 41:4607-4619. [PMID: 33906898 DOI: 10.1523/jneurosci.1207-20.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 11/21/2022] Open
Abstract
Early postnatal experience shapes both inhibitory and excitatory networks in the hippocampus. However, the underlying circuit plasticity is unclear. Using an enriched environment (EE) paradigm during the preweaning period in mice of either sex, we assessed the circuit plasticity of inhibitory cell types in the hippocampus. We found that cholecystokinin (CCK)-expressing basket cells strongly increased somatic inhibition on the excitatory granular cells (GCs) following EE, whereas another pivotal inhibitory cell type, parvalbumin (PV)-expressing cells, did not show changes. Using electrophysiological analysis and the use of cannabinoid receptor 1 (CB1R) agonist WIN 55 212-2, we demonstrate that the change in somatic inhibition from CCK+ neurons increases CB1R-mediated inhibition in the circuit. By inhibiting activity of the entorhinal cortex (EC) using a chemogenetic approach, we further demonstrate that the activity of the projections from the EC mediates the developmental assembly of CCK+ basket cell network. Altogether, our study places the experience-dependent remodeling of CCK+ basket cell innervation as a central process to adjust inhibition in the dentate gyrus and shows that cortical inputs to the hippocampus play an instructional role in controlling the refinement of the synaptic connections during the preweaning period.SIGNIFICANCE STATEMENT Brain plasticity is triggered by experience during postnatal brain development and shapes the maturing neural circuits. In humans, altered experience-dependent plasticity can have long-lasting detrimental effects on circuit function and lead to psychiatric disorders. Yet, the cellular mechanisms governing how early experience fine-tunes the maturing synaptic network is not fully understood. Here, taking advantage of an enrichment-housing paradigm, we unravel a new plasticity mechanism involved in the maintenance of the inhibitory to excitatory balance in the hippocampus. Our findings demonstrate that cortical activity instructs the assembly of the CCK+ basket cell network. Considering the importance of this specific cell type for learning and memory, experience-dependent remodeling of CCK+ cells may be a critical determinant for establishing appropriate neural networks.
Collapse
|
82
|
Trinchero MF, Giacomini D, Schinder AF. Dynamic interplay between GABAergic networks and developing neurons in the adult hippocampus. Curr Opin Neurobiol 2021; 69:124-130. [PMID: 33873060 DOI: 10.1016/j.conb.2021.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/10/2021] [Accepted: 03/14/2021] [Indexed: 01/07/2023]
Abstract
Neurogenesis is a powerful mechanism for structural and functional remodeling that occurs in restricted areas of the adult brain. Although different neurotransmitters regulate various aspects of the progression from neural stem cell quiescence to neuronal maturation, GABA is the main player. The developmental switch from excitation to inhibition combined with a heterogeneous population of GABAergic interneurons that target different subcellular compartments provides multiple points for the regulation of development and function of new neurons. This complexity is enhanced by feedback and feedforward networks that act as sensors and controllers of circuit activity, impinging directly or indirectly onto developing granule cells and, subsequently, on mature neurons. Newly generated granule cells ultimately connect with input and output partners in a manner that is largely sculpted by the activity of local circuits.
Collapse
Affiliation(s)
- Mariela F Trinchero
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA - CONICET), Av. Patricias Argentinas 435, Buenos Aires, C1405BWE, Argentina
| | - Damiana Giacomini
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA - CONICET), Av. Patricias Argentinas 435, Buenos Aires, C1405BWE, Argentina
| | - Alejandro F Schinder
- Laboratory of Neuronal Plasticity, Leloir Institute (IIBBA - CONICET), Av. Patricias Argentinas 435, Buenos Aires, C1405BWE, Argentina.
| |
Collapse
|
83
|
Reid HMO, Chen-Mack N, Snowden T, Christie BR. Understanding Changes in Hippocampal Interneurons Subtypes in the Pathogenesis of Alzheimer's Disease: A Systematic Review. Brain Connect 2021; 11:159-179. [PMID: 33559520 DOI: 10.1089/brain.2020.0879] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: It is becoming increasingly recognized that there is significant interneuron degeneration in Alzheimer's disease. As the hippocampus is integral for learning and memory, we performed a systematic review of primary literature focused on the relationship between Alzheimer's and hippocampal interneurons. In this study, we summarize the experimental work performed to date and identify opportunities for future experiments. Objectives: This PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses)-style systematic review seeks to summarize the findings of all accessible research focused on cholecystokinin (CCK), neuropeptide Y (NPY), parvalbumin (PV), and somatostatin (SOM) interneurons in the hippocampal formation. Results: One thousand five hundred ninety-three articles were pulled from PubMed, PsycInfo, and Web of Science, based on three blocks of search terms. There were 45 articles that met all the predetermined inclusion/exclusion criteria. There is strong evidence that PV interneurons are affected early in the disease by toxic amyloid beta (Aβ) fragments; SOM interneurons are affected indirectly while the SOM neuropeptide may act to slowly worsen toxic Aβ fragment accumulation, whereas NPY- and CCK-positive interneurons are affected later in the progression of the disease. Conclusions: Fewer studies have been performed on NPY and CCK interneurons, and there is room for further investigations regarding the role of PV interneurons in Alzheimer's to help resolve contradictory findings. This review found that PV interneurons are affected early in the disease, but only in Alzheimer's precursor protein but not tau models. NPY and CCK interneurons were found to be affected later in the disease, and SOM interneurons vary greatly. Future studies may consider reporting immunohistochemical studies inclusive of either cell location or morphology-as well as marker to give a more robust picture of the disease.
Collapse
Affiliation(s)
- Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Nathan Chen-Mack
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| | - Taylor Snowden
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| |
Collapse
|
84
|
Song CG, Kang X, Yang F, Du WQ, Zhang JJ, Liu L, Kang JJ, Jia N, Yue H, Fan LY, Wu SX, Jiang W, Gao F. Endocannabinoid system in the neurodevelopment of GABAergic interneurons: implications for neurological and psychiatric disorders. Rev Neurosci 2021; 32:803-831. [PMID: 33781002 DOI: 10.1515/revneuro-2020-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023]
Abstract
In mature mammalian brains, the endocannabinoid system (ECS) plays an important role in the regulation of synaptic plasticity and the functioning of neural networks. Besides, the ECS also contributes to the neurodevelopment of the central nervous system. Due to the increase in the medical and recreational use of cannabis, it is inevitable and essential to elaborate the roles of the ECS on neurodevelopment. GABAergic interneurons represent a group of inhibitory neurons that are vital in controlling neural network activity. However, the role of the ECS in the neurodevelopment of GABAergic interneurons remains to be fully elucidated. In this review, we provide a brief introduction of the ECS and interneuron diversity. We focus on the process of interneuron development and the role of ECS in the modulation of interneuron development, from the expansion of the neural stem/progenitor cells to the migration, specification and maturation of interneurons. We further discuss the potential implications of the ECS and interneurons in the pathogenesis of neurological and psychiatric disorders, including epilepsy, schizophrenia, major depressive disorder and autism spectrum disorder.
Collapse
Affiliation(s)
- Chang-Geng Song
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China.,Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Xin Kang
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Fang Yang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Wan-Qing Du
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Jia-Jia Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Long Liu
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Jun-Jun Kang
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Ning Jia
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Hui Yue
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Lu-Yu Fan
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Sheng-Xi Wu
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| | - Fang Gao
- Department of Neurobiology and Institute of Neurosciences, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an710032, Shaanxi, China
| |
Collapse
|
85
|
Nakajima R, Laskaris N, Rhee JK, Baker BJ, Kosmidis EK. GEVI cell-type specific labelling and a manifold learning approach provide evidence for lateral inhibition at the population level in the mouse hippocampal CA1 area. Eur J Neurosci 2021; 53:3019-3038. [PMID: 33675122 DOI: 10.1111/ejn.15177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/04/2021] [Accepted: 02/22/2021] [Indexed: 01/04/2023]
Abstract
The CA1 area in the mammalian hippocampus is essential for spatial learning. Pyramidal cells are the hippocampus output neurons and their activities are regulated by inhibition exerted by a diversified population of interneurons. Lateral inhibition has been suggested as the mechanism enabling the reconfiguration of pyramidal cell assembly activity observed during spatial learning tasks in rodents. However, lateral inhibition in the CA1 lacks the overwhelming evidence reported in other hippocampal areas such as the CA3 and the dentate gyrus. The use of genetically encoded voltage indicators and fast optical recordings permits the construction of cell-type specific response maps of neuronal activity. Here, we labelled mouse CA1 pyramidal neurons with the genetically encoded voltage indicator ArcLight and optically recorded their response to Schaffer Collaterals stimulation in vitro. By undertaking a manifold learning approach, we report a hyperpolarization-dominated area focused in the perisomatic region of pyramidal cells receiving late excitatory synaptic input. Functional network organization metrics revealed that information transfer was higher in this area. The localized hyperpolarization disappeared when GABAA receptors were pharmacologically blocked. This is the first report where the spatiotemporal pattern of lateral inhibition is visualized in the CA1 by expressing a genetically encoded voltage indicator selectively in principal neurons. Our analysis suggests a fundamental role of lateral inhibition in CA1 information processing.
Collapse
Affiliation(s)
- Ryuichi Nakajima
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Nikolaos Laskaris
- AIIA Lab, Informatics Department, Aristotle University of Thessaloniki, Thessaloniki, Greece.,NeuroInformatics GRoup, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Jun Kyu Rhee
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Bradley J Baker
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Efstratios K Kosmidis
- NeuroInformatics GRoup, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Department of Medicine, Laboratory of Physiology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
86
|
Tuan LH, Tsao CY, Lee LJH, Lee LJ. Voluntary exercise ameliorates synaptic pruning deficits in sleep-deprived adolescent mice. Brain Behav Immun 2021; 93:96-110. [PMID: 33358980 DOI: 10.1016/j.bbi.2020.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/17/2020] [Accepted: 12/16/2020] [Indexed: 11/26/2022] Open
Abstract
Adolescence is a critical period for brain development and adequate sleep during this period is essential for physical function and mental health. Emerging evidence has detailed the neurological impacts of sleep insufficiency on adolescents, as was unveiled by our previous study, microglia, one of the crucial contributors to synaptic pruning, is functionally disrupted by lack of sleep. Here, we provided evidence featuring the protective effect and the underlying mechanisms of voluntary exercise (VE) on microglial functions in an adolescent 72 h sleep deprivation (SD) model. We identified that the aberrant hippocampal neuronal activity and impaired short-term memory performance in sleep-deprived mice were prevented by 11 days of VE. VE significantly normalized the SD-induced dendritic spine increment and maintained the microglial phagocytic ability in sleep-deprived mice. Moreover, we found that the amendment of the noradrenergic signal in the central nervous system may explain the preventative effects of VE on the abnormalities of microglial and neuronal functions caused by SD. These data suggested that VE may confer protection to the microglia-mediated synaptic pruning in the sleep-deprived adolescent brains. Therefore, physical exercise could be a beneficial health practice for the adolescents that copes the adverse influence of inevitable sleep insufficiency.
Collapse
Affiliation(s)
- Li-Heng Tuan
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Yu Tsao
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC
| | - Lukas Jyuhn-Hsiarn Lee
- Division of Environmental Health and Occupational Medicine, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Li-Jen Lee
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University, Taipei, Taiwan, ROC; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan, ROC; Institute of Brain and Mind Sciences, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
87
|
PPM1F in hippocampal dentate gyrus regulates the depression-related behaviors by modulating neuronal excitability. Exp Neurol 2021; 340:113657. [PMID: 33639208 DOI: 10.1016/j.expneurol.2021.113657] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/28/2020] [Accepted: 02/21/2021] [Indexed: 01/21/2023]
Abstract
Major depressive disorder (MDD) is a common, serious, debilitating mental illness. Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F), a serine/threonine phosphatase, has been reported to have multiple biological and cellular functions. However, the effects of PPM1F and its neuronal substrates on depressive behaviors remain largely unknown. Here, we showed that PPM1F is widely distributed in the hippocampus, and chronic unpredictable stress (CUS) can induce increased expression of PPM1F in the hippocampus, which was correlated with depression-associated behaviors. Overexpression of PPM1F mediated by adeno-associated virus (AAV) in the dentate gyrus (DG) produced depression-related behaviors and enhanced susceptibility to subthreshold CUS (SCUS) in both male and female mice, while, knockout of PPM1F in DG produced antidepressant phonotypes under stress conditions. Whole-cell patch-clamp recordings demonstrated that overexpression of PPM1F increased the neuronal excitability of the granule cells in the DG. Consistent with neuronal hyperexcitability, overexpression of PPM1F regulated the expression of certain ion channel genes and induced decreased phosphorylation of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) in hippocampus. These results suggest that PPM1F in the DG regulates depression-related behaviors by modulating neuronal excitability, which might be an important pathological gene for depression or other mental diseases.
Collapse
|
88
|
Lintas A, Sánchez-Campusano R, Villa AEP, Gruart A, Delgado-García JM. Operant conditioning deficits and modified local field potential activities in parvalbumin-deficient mice. Sci Rep 2021; 11:2970. [PMID: 33536607 PMCID: PMC7859233 DOI: 10.1038/s41598-021-82519-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Altered functioning of GABAergic interneurons expressing parvalbumin (PV) in the basal ganglia-thalamo-cortical circuit are likely to be involved in several human psychiatric disorders characterized by deficits in attention and sensory gating with dysfunctional decision-making behavior. However, the contribution of these interneurons in the ability to acquire demanding learning tasks remains unclear. Here, we combine an operant conditioning task with local field potentials simultaneously recorded in several nuclei involved in reward circuits of wild-type (WT) and PV-deficient (PVKO) mice, which are characterized by changes in firing activity of PV-expressing interneurons. In comparison with WT mice, PVKO animals presented significant deficits in the acquisition of the selected learning task. Recordings from prefrontal cortex, nucleus accumbens (NAc) and hippocampus showed significant decreases of the spectral power in beta and gamma bands in PVKO compared with WT mice particularly during the performance of the operant conditioning task. From the first to the last session, at all frequency bands the spectral power in NAc tended to increase in WT and to decrease in PVKO. Results indicate that PV deficiency impairs signaling necessary for instrumental learning and the recognition of natural rewards.
Collapse
Affiliation(s)
- Alessandra Lintas
- Neuroheuristic Research Group & LABEX, HEC Lausanne, University of Lausanne, Quartier UNIL-Chamberonne, 1015, Lausanne, Switzerland.
| | - Raudel Sánchez-Campusano
- Division of Neurosciences, Pablo de Olavide University, Ctra. de Utrera, km. 1, 41013, Sevilla, Spain
| | - Alessandro E P Villa
- Neuroheuristic Research Group & LABEX, HEC Lausanne, University of Lausanne, Quartier UNIL-Chamberonne, 1015, Lausanne, Switzerland
| | - Agnès Gruart
- Division of Neurosciences, Pablo de Olavide University, Ctra. de Utrera, km. 1, 41013, Sevilla, Spain
| | - José M Delgado-García
- Division of Neurosciences, Pablo de Olavide University, Ctra. de Utrera, km. 1, 41013, Sevilla, Spain
| |
Collapse
|
89
|
Pléau C, Peret A, Pearlstein E, Scalfati T, Vigier A, Marti G, Michel FJ, Marissal T, Crépel V. Dentate Granule Cells Recruited in the Home Environment Display Distinctive Properties. Front Cell Neurosci 2021; 14:609123. [PMID: 33519383 PMCID: PMC7843370 DOI: 10.3389/fncel.2020.609123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/23/2020] [Indexed: 11/13/2022] Open
Abstract
The dentate granule cells (DGCs) play a crucial role in learning and memory. Many studies have described the role and physiological properties of these sparsely active neurons using different behavioral contexts. However, the morpho-functional features of DGCs recruited in mice maintained in their home cage (without training), considered as a baseline condition, have not yet been established. Using fosGFP transgenic mice, we observed ex vivo that DGCs recruited in animals maintained in the home cage condition are mature neurons that display a longer dendritic tree and lower excitability compared with non-activated cells. The higher GABAA receptor-mediated shunting inhibition contributes to the lower excitability of DGCs activated in the home environment by shifting the input resistance towards lower values. Remarkably, that shunting inhibition is neither observed in non-activated DGCs nor in DGCs activated during training in virtual reality. In short, our results suggest that strong shunting inhibition and reduced excitability could constitute a distinctive neural signature of mature DGCs recruited in the context of the home environment.
Collapse
Affiliation(s)
- Claire Pléau
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Angélique Peret
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | | | - Thomas Scalfati
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Alexandre Vigier
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | | | | | - Thomas Marissal
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Valérie Crépel
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| |
Collapse
|
90
|
Fredes F, Silva MA, Koppensteiner P, Kobayashi K, Joesch M, Shigemoto R. Ventro-dorsal Hippocampal Pathway Gates Novelty-Induced Contextual Memory Formation. Curr Biol 2021; 31:25-38.e5. [PMID: 33065009 PMCID: PMC7808756 DOI: 10.1016/j.cub.2020.09.074] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 01/07/2023]
Abstract
Novelty facilitates memory formation and is detected by both the dorsal and ventral hippocampus. Although dentate granule cells (GCs) in the dorsal hippocampus are known to mediate the formation of novelty-induced contextual memories, the required pathways and mechanisms remain unclear. Here we demonstrate that a powerful excitatory pathway from mossy cells (MCs) in the ventral hippocampus to dorsal GCs is necessary and sufficient for driving dorsal GC activation in novel environment exploration. In vivo Ca2+ imaging in freely moving mice indicated that this pathway relays environmental novelty. Furthermore, manipulation of ventral MC activity bidirectionally regulates novelty-induced contextual memory acquisition. Our results show that ventral MC activity gates contextual memory formation through an intra-hippocampal interaction activated by environmental novelty.
Collapse
Affiliation(s)
- Felipe Fredes
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria; Department of Biomedicine, Aarhus University, Ole Worms Alle 6, Building 1182, 8000 Aarhus C, Denmark.
| | - Maria Alejandra Silva
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Peter Koppensteiner
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Myodaiji, Okazaki, Japan
| | - Maximilian Joesch
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
91
|
Groisman AI, Yang SM, Schinder AF. Differential Coupling of Adult-Born Granule Cells to Parvalbumin and Somatostatin Interneurons. Cell Rep 2021; 30:202-214.e4. [PMID: 31914387 PMCID: PMC7011182 DOI: 10.1016/j.celrep.2019.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/16/2019] [Accepted: 11/27/2019] [Indexed: 12/29/2022] Open
Abstract
A strong GABAergic tone imposes sparse levels of activity in the dentate gyrus of the hippocampus. This balance is challenged by the addition of new granule cells (GCs) with high excitability. How developing GCs integrate within local inhibitory networks remains unknown. We used optogenetics to study synaptogenesis between new GCs and GABAergic interneurons expressing parvalbumin (PV-INs) and somatostatin (SST-INs). PV-INs target the soma, and synapses become mature after 6 weeks. This transition is accelerated by exposure to an enriched environment. PV-INs exert efficient control of GC spiking and participate in both feedforward and feedback loops, a mechanism that would favor lateral inhibition and sparse coding. SST-INs target the dendrites, and synapses mature after 8 weeks. Outputs from GCs onto PV-INs develop faster than those onto SST-INs. Our results reveal a long-lasting transition wherein adult-born neurons remain poorly coupled to inhibition, which might enhance activity-dependent plasticity of input and output synapses. Groisman et al. examine the integration of adult-born granule cells (GCs) to inhibitory networks of the adult hippocampus. Synapse maturation is remarkably slow for parvalbumin and somatostatin interneurons, both for connections toward and from GCs. Inhibition controls the activity of new GCs late in development.
Collapse
Affiliation(s)
- Ayelén I Groisman
- Laboratorio de Plasticidad Neuronal, Fundación Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Sung M Yang
- Laboratorio de Plasticidad Neuronal, Fundación Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina
| | - Alejandro F Schinder
- Laboratorio de Plasticidad Neuronal, Fundación Instituto Leloir, Av. Patricias Argentinas 435, C1405BWE Buenos Aires, Argentina.
| |
Collapse
|
92
|
Morales C, Morici JF, Espinosa N, Sacson A, Lara-Vasquez A, García-Pérez MA, Bekinschtein P, Weisstaub NV, Fuentealba P. Dentate Gyrus Somatostatin Cells are Required for Contextual Discrimination During Episodic Memory Encoding. Cereb Cortex 2021; 31:1046-1059. [PMID: 33026440 DOI: 10.1093/cercor/bhaa273] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 11/12/2022] Open
Abstract
Memory systems ought to store and discriminate representations of similar experiences in order to efficiently guide future decisions. This problem is solved by pattern separation, implemented in the dentate gyrus (DG) by granule cells to support episodic memory formation. Pattern separation is enabled by tonic inhibitory bombardment generated by multiple GABAergic cell populations that strictly maintain low activity levels in granule cells. Somatostatin-expressing cells are one of those interneuron populations, selectively targeting the distal dendrites of granule cells, where cortical multimodal information reaches the DG. Nonetheless, somatostatin cells have very low connection probability and synaptic efficacy with both granule cells and other interneuron types. Hence, the role of somatostatin cells in DG circuitry, particularly in the context of pattern separation, remains uncertain. Here, by using optogenetic stimulation and behavioral tasks in mice, we demonstrate that somatostatin cells are required for the acquisition of both contextual and spatial overlapping memories.
Collapse
Affiliation(s)
- Cristian Morales
- Departamento de Psiquiatria, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Catolica de Chile, Santiago 7820436, Chile
| | - Juan Facundo Morici
- Instituto de Neurociencia Cognitiva y Traslacional, Instituto de Neurologia Cognitiva, Consejo Nacional de Investigaciones Cientificas y Tecnicas Fundacion INECO, Universidad Favaloro, 1078 Buenos Aires, Argentina
| | - Nelson Espinosa
- Departamento de Psiquiatria, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Catolica de Chile, Santiago 7820436, Chile
| | - Agostina Sacson
- Instituto de Neurociencia Cognitiva y Traslacional, Instituto de Neurologia Cognitiva, Consejo Nacional de Investigaciones Cientificas y Tecnicas Fundacion INECO, Universidad Favaloro, 1078 Buenos Aires, Argentina
| | - Ariel Lara-Vasquez
- Departamento de Psiquiatria, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Catolica de Chile, Santiago 7820436, Chile
| | - M A García-Pérez
- Departamento de Psiquiatria, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Catolica de Chile, Santiago 7820436, Chile
| | - Pedro Bekinschtein
- Instituto de Neurociencia Cognitiva y Traslacional, Instituto de Neurologia Cognitiva, Consejo Nacional de Investigaciones Cientificas y Tecnicas Fundacion INECO, Universidad Favaloro, 1078 Buenos Aires, Argentina
| | - Noelia V Weisstaub
- Instituto de Neurociencia Cognitiva y Traslacional, Instituto de Neurologia Cognitiva, Consejo Nacional de Investigaciones Cientificas y Tecnicas Fundacion INECO, Universidad Favaloro, 1078 Buenos Aires, Argentina
| | - Pablo Fuentealba
- Departamento de Psiquiatria, Centro Interdisciplinario de Neurociencia, Pontificia Universidad Catolica de Chile, Santiago 7820436, Chile.,Centro de Investigacion en Nanotecnologia y Materiales Avanzados, Pontificia Universidad Catolica de Chile, Santiago 7820436, Chile
| |
Collapse
|
93
|
Nahar L, Delacroix BM, Nam HW. The Role of Parvalbumin Interneurons in Neurotransmitter Balance and Neurological Disease. Front Psychiatry 2021; 12:679960. [PMID: 34220586 PMCID: PMC8249927 DOI: 10.3389/fpsyt.2021.679960] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022] Open
Abstract
While great progress has been made in the understanding of neurological illnesses, the pathologies, and etiologies that give rise to these diseases still remain an enigma, thus, also making treatments for them more challenging. For effective and individualized treatment, it is beneficial to identify the underlying mechanisms that govern the associated cognitive and behavioral processes that go awry in neurological disorders. Parvalbumin fast-spiking interneurons (Pv-FSI) are GABAergic cells that are only a small fraction of the brain's neuronal network, but manifest unique cellular and molecular properties that drastically influence the downstream effects on signaling and ultimately change cognitive behaviors. Proper brain functioning relies heavily on neuronal communication which Pv-FSI regulates, excitatory-inhibitory balances and GABAergic disinhibition between circuitries. This review highlights the depth of Pv-FSI involvement in the cortex, hippocampus, and striatum, as it pertains to expression, neurotransmission, role in neurological disorders, and dysfunction, as well as cognitive behavior and reward-seeking. Recent research has indicated that Pv-FSI play pivotal roles in the molecular pathophysiology and cognitive-behavioral deficits that are core features of many psychiatric disorders, such as schizophrenia, autism spectrum disorders, Alzheimer's disease, and drug addiction. This suggests that Pv-FSI could be viable targets for treatment of these disorders and thus calls for further examination of the undeniable impact Pv-FSI have on the brain and cognitive behavior.
Collapse
Affiliation(s)
- Lailun Nahar
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Blake M Delacroix
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
94
|
Allegra M, Posani L, Gómez-Ocádiz R, Schmidt-Hieber C. Differential Relation between Neuronal and Behavioral Discrimination during Hippocampal Memory Encoding. Neuron 2020; 108:1103-1112.e6. [PMID: 33068531 PMCID: PMC7772055 DOI: 10.1016/j.neuron.2020.09.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/14/2020] [Accepted: 09/24/2020] [Indexed: 12/23/2022]
Abstract
How are distinct memories formed and used for behavior? To relate neuronal and behavioral discrimination during memory formation, we use in vivo 2-photon Ca2+ imaging and whole-cell recordings from hippocampal subregions in head-fixed mice performing a spatial virtual reality task. We find that subthreshold activity as well as population codes of dentate gyrus neurons robustly discriminate across different spatial environments, whereas neuronal remapping in CA1 depends on the degree of difference between visual cues. Moreover, neuronal discrimination in CA1, but not in the dentate gyrus, reflects behavioral performance. Our results suggest that CA1 weights the decorrelated information from the dentate gyrus according to its relevance, producing a map of memory representations that can be used by downstream circuits to guide learning and behavior.
Collapse
Affiliation(s)
- Manuela Allegra
- Department of Neuroscience, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France
| | - Lorenzo Posani
- Department of Neuroscience, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France
| | - Ruy Gómez-Ocádiz
- Department of Neuroscience, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | | |
Collapse
|
95
|
Medeiros DDC, Cota VR, Oliveira ACP, Moreira FA, Moraes MFD. The Endocannabinoid System Activation as a Neural Network Desynchronizing Mediator for Seizure Suppression. Front Behav Neurosci 2020; 14:603245. [PMID: 33281577 PMCID: PMC7691588 DOI: 10.3389/fnbeh.2020.603245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 10/20/2020] [Indexed: 01/08/2023] Open
Abstract
The understanding that hyper-excitability and hyper-synchronism in epilepsy are indissociably bound by a cause-consequence relation has only recently been challenged. Thus, therapeutic strategies for seizure suppression have often aimed at inhibiting excitatory circuits and/or activating inhibitory ones. However, new approaches that aim to desynchronize networks or compromise abnormal coupling between adjacent neural circuitry have been proven effective, even at the cost of enhancing local neuronal activation. Although most of these novel perspectives targeting circuitry desynchronization and network coupling have been implemented by non-pharmacological devices, we argue that there may be endogenous neurochemical systems that act primarily in the desynchronization component of network behavior rather than dampening excitability of individual neurons. This review explores the endocannabinoid system as one such possible pharmacological landmark for mimicking a form of "on-demand" desynchronization analogous to those proposed by deep brain stimulation in the treatment of epilepsy. This essay discusses the evidence supporting the role of the endocannabinoid system in modulating the synchronization and/or coupling of distinct local neural circuitry; which presents obvious implications on the physiological setting of proper sensory-motor integration. Accordingly, the process of ictogenesis involves pathological circuit coupling that could be avoided, or at least have its spread throughout the containment of other areas, if such endogenous mechanisms of control could be activated or potentiated by pharmacological intervention. In addition, we will discuss evidence that supports not only a weaker role played on neuronal excitability but the potential of the endocannabinoid system strengthening its modulatory effect, only when circuitry coupling surpasses a level of activation.
Collapse
Affiliation(s)
- Daniel de Castro Medeiros
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vinícius Rosa Cota
- Laboratório Interdisciplinar de Neuroengenharia e Neurociências, Departamento de Engenharia Elétrica, Universidade Federal de São João Del-Rei, São João Del-Rei, Brazil
| | - Antonio Carlos P Oliveira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabricio A Moreira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Márcio Flávio Dutra Moraes
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Centro de Tecnologia e Pesquisa em Magneto Ressonância, Programa de Pós-Graduação em Engenharia Elétrica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
96
|
Richetin K, Steullet P, Pachoud M, Perbet R, Parietti E, Maheswaran M, Eddarkaoui S, Bégard S, Pythoud C, Rey M, Caillierez R, Q Do K, Halliez S, Bezzi P, Buée L, Leuba G, Colin M, Toni N, Déglon N. Tau accumulation in astrocytes of the dentate gyrus induces neuronal dysfunction and memory deficits in Alzheimer's disease. Nat Neurosci 2020; 23:1567-1579. [PMID: 33169029 DOI: 10.1038/s41593-020-00728-x] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/24/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of the tau protein in neurons, neurodegeneration and memory loss. However, the role of non-neuronal cells in this chain of events remains unclear. In the present study, we found accumulation of tau in hilar astrocytes of the dentate gyrus of individuals with AD. In mice, the overexpression of 3R tau specifically in hilar astrocytes of the dentate gyrus altered mitochondrial dynamics and function. In turn, these changes led to a reduction of adult neurogenesis, parvalbumin-expressing neurons, inhibitory synapses and hilar gamma oscillations, which were accompanied by impaired spatial memory performances. Together, these results indicate that the loss of tau homeostasis in hilar astrocytes of the dentate gyrus is sufficient to induce AD-like symptoms, through the impairment of the neuronal network. These results are important for our understanding of disease mechanisms and underline the crucial role of astrocytes in hippocampal function.
Collapse
Affiliation(s)
- Kevin Richetin
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland. .,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland. .,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Pascal Steullet
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Mathieu Pachoud
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Romain Perbet
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Enea Parietti
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Mathischan Maheswaran
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Séverine Bégard
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Catherine Pythoud
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Maria Rey
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Raphaëlle Caillierez
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Kim Q Do
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Sophie Halliez
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Paola Bezzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Geneviève Leuba
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Morvane Colin
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Nicolas Toni
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| | - Nicole Déglon
- Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.,Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
97
|
Xia Y, Zhang Z, Lin W, Yan J, Zhu C, Yin D, He S, Su Y, Xu N, Caldwell RW, Yao L, Chen Y. Modulating microglia activation prevents maternal immune activation induced schizophrenia-relevant behavior phenotypes via arginase 1 in the dentate gyrus. Neuropsychopharmacology 2020; 45:1896-1908. [PMID: 32599605 PMCID: PMC7608378 DOI: 10.1038/s41386-020-0743-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 12/31/2022]
Abstract
Prenatal infection during pregnancy increases the risk for developing neuropsychiatric disorders such as schizophrenia. This is linked to an inflammatory microglial phenotype in the offspring induced by maternal immune activation (MIA). Microglia are crucial for brain development and maintenance of neuronal niches, however, whether and how their activation is involved in the regulation of neurodevelopment remains unclear. Here, we used a MIA rodent model in which polyinosinic: polycytidylic acid (poly (I:C)) was injected into pregnant mice. We found fewer parvalbumin positive (PV+) cells and impaired GABAergic transmission in the dentate gyrus (DG), accompanied by schizophrenia-like behavior in the adult offspring. Minocycline, a potent inhibitor of microglia activation, successfully prevented the above-mentioned deficits in the offspring. Furthermore, by using microglia-specific arginase 1 (Arg1) ablation as well as overexpression in DG, we identified a critical role of Arg1 in microglia activation to protect against poly (I:C) imparted neuropathology and altered behavior in offspring. Taken together, our results highlight that Arg1-mediated alternative activation of microglia are potential therapeutic targets for psychiatric disorders induced by MIA.
Collapse
Affiliation(s)
- Yucen Xia
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Zhiqing Zhang
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Weipeng Lin
- grid.22069.3f0000 0004 0369 6365Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062 China
| | - Jinglan Yan
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Chuan’an Zhu
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Dongmin Yin
- grid.22069.3f0000 0004 0369 6365Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life Science, East China Normal University, Shanghai, 200062 China
| | - Su He
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Yang Su
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Nenggui Xu
- grid.411866.c0000 0000 8848 7685South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Robert William Caldwell
- grid.410427.40000 0001 2284 9329Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912 USA
| | - Lin Yao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China. .,School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China. .,Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, 510515, China. .,Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
98
|
Metabolic tuning of inhibition regulates hippocampal neurogenesis in the adult brain. Proc Natl Acad Sci U S A 2020; 117:25818-25829. [PMID: 32973092 DOI: 10.1073/pnas.2006138117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hippocampus-engaged behaviors stimulate neurogenesis in the adult dentate gyrus by largely unknown means. To explore the underlying mechanisms, we used tetrode recording to analyze neuronal activity in the dentate gyrus of freely moving adult mice during hippocampus-engaged contextual exploration. We found that exploration induced an overall sustained increase in inhibitory neuron activity that was concomitant with decreased excitatory neuron activity. A mathematical model based on energy homeostasis in the dentate gyrus showed that enhanced inhibition and decreased excitation resulted in a similar increase in neurogenesis to that observed experimentally. To mechanistically investigate this sustained inhibitory regulation, we performed metabolomic and lipidomic profiling of the hippocampus during exploration. We found sustainably increased signaling of sphingosine-1-phosphate, a bioactive metabolite, during exploration. Furthermore, we found that sphingosine-1-phosphate signaling through its receptor 2 increased interneuron activity and thus mediated exploration-induced neurogenesis. Taken together, our findings point to a behavior-metabolism circuit pathway through which experience regulates adult hippocampal neurogenesis.
Collapse
|
99
|
Neuropeptides Modulate Local Astrocytes to Regulate Adult Hippocampal Neural Stem Cells. Neuron 2020; 108:349-366.e6. [PMID: 32877641 DOI: 10.1016/j.neuron.2020.07.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 06/12/2020] [Accepted: 07/29/2020] [Indexed: 12/20/2022]
Abstract
Neural stem cells (NSCs) in the dentate gyrus (DG) reside in a specialized local niche that supports their neurogenic proliferation to produce adult-born neurons throughout life. How local niche cells interact at the circuit level to ensure continuous neurogenesis from NSCs remains unknown. Here we report the role of endogenous neuropeptide cholecystokinin (CCK), released from dentate CCK interneurons, in regulating neurogenic niche cells and NSCs. Specifically, stimulating CCK release supports neurogenic proliferation of NSCs through a dominant astrocyte-mediated glutamatergic signaling cascade. In contrast, reducing dentate CCK induces reactive astrocytes, which correlates with decreased neurogenic proliferation of NSCs and upregulation of genes involved in immune processes. Our findings provide novel circuit-based information on how CCK acts on local astrocytes to regulate the key behavior of adult NSCs.
Collapse
|
100
|
Vaden RJ, Gonzalez JC, Tsai MC, Niver AJ, Fusilier AR, Griffith CM, Kramer RH, Wadiche JI, Overstreet-Wadiche L. Parvalbumin interneurons provide spillover to newborn and mature dentate granule cells. eLife 2020; 9:54125. [PMID: 32602839 PMCID: PMC7326496 DOI: 10.7554/elife.54125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/19/2020] [Indexed: 01/09/2023] Open
Abstract
Parvalbumin-expressing interneurons (PVs) in the dentate gyrus provide activity-dependent regulation of adult neurogenesis as well as maintain inhibitory control of mature neurons. In mature neurons, PVs evoke GABAA postsynaptic currents (GPSCs) with fast rise and decay phases that allow precise control of spike timing, yet synaptic currents with fast kinetics do not appear in adult-born neurons until several weeks after cell birth. Here we used mouse hippocampal slices to address how PVs signal to newborn neurons prior to the appearance of fast GPSCs. Whereas PV-evoked currents in mature neurons exhibit hallmark fast rise and decay phases, newborn neurons display slow GPSCs with characteristics of spillover signaling. We also unmasked slow spillover currents in mature neurons in the absence of fast GPSCs. Our results suggest that PVs mediate slow spillover signaling in addition to conventional fast synaptic signaling, and that spillover transmission mediates activity-dependent regulation of early events in adult neurogenesis.
Collapse
Affiliation(s)
- Ryan J Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Jose Carlos Gonzalez
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Ming-Chi Tsai
- Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, United States
| | - Anastasia J Niver
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Allison R Fusilier
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Chelsea M Griffith
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | - Richard H Kramer
- Department of Molecular and Cellular Biology, University of California Berkeley, Berkeley, United States
| | - Jacques I Wadiche
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, United States
| | | |
Collapse
|