1
|
Ueno H, Takahashi Y, Mori S, Kitano E, Murakami S, Wani K, Matsumoto Y, Okamoto M, Ishihara T. Postnatal expression of Cat-315-positive perineuronal nets in the SAMP10 mouse primary somatosensory cortex. IBRO Neurosci Rep 2025; 18:244-256. [PMID: 39935854 PMCID: PMC11810707 DOI: 10.1016/j.ibneur.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
Perineuronal nets (PNNs) form at the end of the critical period of plasticity in the mouse primary somatosensory cortex. PNNs are said to have functions that control neuroplasticity and provide neuroprotection. However, it is not clear which molecules in PNNs have these functions. We have previously reported that Cat-315-positive molecules were not expressed in the PNNs of the senescence-accelerated model (SAM)P10 strain model mice at 12 months of age. To confirm whether the loss of Cat-315-positive molecules occurred early in life in SAMP10 mice, we examined Cat-315-positive PNNs in the primary somatosensory cortex during postnatal development. This research helps to elucidate the function of PNNs and the mechanism of cognitive decline associated with ageing. To confirm whether Cat-315-positive PNNs changed in an age-dependent manner in SAMP10 mice, we examined the primary somatosensory cortex at 21, 28, and 56 days after birth. We compared these results with those of senescence-accelerated mouse-resistant (SAMR) mice. In SAMP10 mice, Cat-315-positive PNNs were expressed in the primary somatosensory cortex early after birth, but their expression was significantly lower than that in SAMR1 mice. Many other molecules that calibrated the PNN were unchanged between SAMP10 and SAMR1 mice. This study revealed that the expression of the Cat-315 epitope was decreased in the primary somatosensory cortex of SAMP10 mice during postnatal development. SAMP10 mice have had histological abnormalities in their brains since early life. Furthermore, using SAMP10 will be useful in elucidating the mechanism of age-related abnormalities in brain function as well as in elucidating the function and structure of PNNs.
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, Okayama 701-0193, Japan
| | - Yu Takahashi
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Sachiko Mori
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Eriko Kitano
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan
| |
Collapse
|
2
|
Deane KE, Binder DK, Razak KA. Cortical layer-specific abnormalities in auditory responses in a mouse model of Fragile X Syndrome. Neurobiol Dis 2025; 212:106963. [PMID: 40393552 DOI: 10.1016/j.nbd.2025.106963] [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: 02/07/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025] Open
Abstract
Fragile X Syndrome (FXS) is a leading genetic cause of autism spectrum disorders (ASD)- associated behaviors, including sensory processing deficits. Sensory sensitivity and temporal processing deficits in the auditory domain will affect development of language and cognitive functions. The mouse model for FXS, Fmr1 KO, has shown remarkably similar auditory processing phenotypes to patients with FXS. In vitro cortical slice recordings show layer-specific differences in Fmr1 KO mouse local circuits, but it is unclear how these differences translate to changes in sensory processing. In this study, we used a depth multielectrode to record in vivo spikes and local field potentials across layers of the auditory cortex in Fmr1 KO and wildtype mice (WT), converting the latter to current source density (CSD) profiles for improved spatial resolution analysis. We observed reduced CSD sink amplitudes and inter-trial phase coherence, and an increase in trial-to-trial variability for temporally modulated stimuli in the KO mice. Results indicated a differential cortical layer pattern of activity in KO mice, with higher baseline gamma power in superficial and deep layers and higher resting delta and theta power in granular layers. Significantly elevated inter-trial variability was observed for CSD and spikes in KO mice. Auditory steady state responses to clicks or gaps at 40 Hz showed considerable trial-to-trial variability in a layer-specific manner in KO mice. Neural generators in the Fmr1 KO mouse auditory cortex failed to detect short gaps in noise, indicating severe temporal processing deficits. Altogether, this study indicates layer-specific cortical mechanisms of sensory hypersensitivity and temporal processing deficits in FXS.
Collapse
Affiliation(s)
- Katrina E Deane
- Psychology Department, University of California, Riverside, 900 University Ave, Riverside, CA 92521, USA
| | - Devin K Binder
- Graduate Neuroscience Program, University of California, Riverside, 900 University Ave, Riverside, CA 92521, USA; Biomedical Sciences, School of Medicine, University of California, Riverside, 900 University Ave, Riverside, CA 92521, USA
| | - Khaleel A Razak
- Psychology Department, University of California, Riverside, 900 University Ave, Riverside, CA 92521, USA; Graduate Neuroscience Program, University of California, Riverside, 900 University Ave, Riverside, CA 92521, USA.
| |
Collapse
|
3
|
Hussain A, Razak KA. Perineuronal net degradation causes a delayed change in resting and sound evoked responses in the mouse auditory cortex. Neuroscience 2025; 577:252-263. [PMID: 40389125 DOI: 10.1016/j.neuroscience.2025.05.026] [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: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Perineuronal nets (PNNs) are extracellular matrix assemblies that preferentially cover parvalbumin-expressing (PV+) interneurons in the neocortex. PV+ cells and PNNs are impaired in a variety of neurodevelopmental disorders including Fragile X Syndrome and schizophrenia. In both of these disorders, electroencephalograph (EEG) recordings show similar phenotypes, including elevated resting gamma band power and reduced temporal fidelity in the 40 Hz auditory steady state response (ASSR). Whether there is a causal link between PNN integrity and EEG abnormalities remains unclear. We tested this link by recording EEG responses in the auditory cortex (AC) in wildtype mice in which PNNs were enzymatically degraded (Chondroitinase ABC or ChABC). EEGs were recorded at two different time points (4- or 14-days post injection, cross-sectional design). In comparison to saline control, ChABC injected mice showed a ∼50 % reduction in PNN density after 4-days. However, there was no difference in resting EEG power spectral density, auditory event-related potential amplitudes or ASSR temporal fidelity between saline and ChABC mice. At the 14-day time point, there was a recovery of PNN density in the AC. Interestingly, EEG responses were abnormal at this time point, with elevated gamma band activity and reduced ASSR temporal fidelity. Thus, the electrophysiological consequences of PNN loss are not seen acutely, but over a delayed time course, suggesting abnormal plasticity after a circuit perturbation. Taken together, these data indicate acute shaping of auditory cortical responses is less dependent on PNNs, but long-term stability of responses following a circuit perturbation depends on the integrity of PNNs.
Collapse
Affiliation(s)
- A Hussain
- Graduate Neuroscience Program, University of California, Riverside, USA
| | - K A Razak
- Graduate Neuroscience Program, University of California, Riverside, USA; Department of Psychology, University of California, Riverside, USA.
| |
Collapse
|
4
|
Mazi AR, Karakoc Y, Demirtas C, Aykin U, Yildirim M. Extracellular Matrix Alterations Due to Early-Life Adversity: Implications for Auditory Learning in Male Sprague-Dawley Rats. Mol Neurobiol 2025; 62:6490-6502. [PMID: 39812993 PMCID: PMC11953085 DOI: 10.1007/s12035-025-04690-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
This study aimed to investigate the impact of early childhood chronic stress on the development of the brain extracellular matrix (ECM) and how alterations in the ECM following early-life adversity (ELA) affect auditory learning and cognitive flexibility. ELA was induced through a combination of maternal separation and neonatal isolation in male Sprague-Dawley rats, and the success of the ELA model was assessed behaviorally and biochemically. A cortex-dependent go/no-go task with two phases was used to determine the impact of ELA on auditory learning and cognitive flexibility. The effects of the ECM on cognition were tested via the enzymatic removal of the ECM. The molecular structure of the adult ECM was examined via immunohistochemistry. ELA impaired initial auditory learning but did not significantly affect cognitive flexibility. Hyase injection into the auditory cortex (ACx) restored initial learning. ELA rats display a reduced perineural net (PNN) and parvalbumin + cell density. Our findings reveal that ELA induces significant alterations in the ECM within the ACx, accompanied by impaired initial auditory learning. Although PNN density is already lower in ELA rats, degrading the ECM facilitates the repair of auditory learning. A reduced PNN number in ELA rats fails to enhance learning unless supplemented with Hyase injection.
Collapse
Affiliation(s)
- Aise Rumeysa Mazi
- Department of Biophysics, Hamidiye Faculty of Medicine, University of Health Sciences, Selimiye Mah. Tibbiye Cad. No:38, 34668, Uskudar, Istanbul, Turkey.
| | - Yunus Karakoc
- Department of Biophysics, Hamidiye Faculty of Medicine, University of Health Sciences, Selimiye Mah. Tibbiye Cad. No:38, 34668, Uskudar, Istanbul, Turkey
| | - Cumaali Demirtas
- Department of Physiology, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Ugur Aykin
- Department of Physiology, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Mehmet Yildirim
- Department of Physiology, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
5
|
Maheu MG, James N, Clark Z, Yang A, Patel R, Beaudette SM, MacPherson REK, Duarte-Guterman P. Running to remember: The effects of exercise on perineuronal nets, microglia, and hippocampal angiogenesis in female and male mice. Behav Brain Res 2025; 484:115478. [PMID: 39956366 DOI: 10.1016/j.bbr.2025.115478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/24/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
Exercise is accepted as a positive health behaviour; however, the mechanisms of exercise on neuroprotection and cognitive health are not completely understood. The purpose of this study was to explore the neurobiological benefits of chronic treadmill exercise in female and male mice through its role in microglial content and morphology, cerebral vascularization, and perineuronal net (PNN) expression. We further examined how these neurobiological changes relate to spatial memory outcomes. Adult mice were assigned to a sedentary or treadmill exercise group for eight weeks. During the final week, all mice were trained on a spatial memory task (Barnes maze) and brains were collected for immunohistochemistry. Exercised mice made fewer errors than sedentary mice during the first two days of training and probe trial. Females, regardless of exercise training, made fewer errors during Barnes maze training and demonstrated a greater frequency of spatial strategy use compared to males. Exercised mice, regardless of sex, had fewer PNNs in the dentate gyrus of the hippocampus compared to sedentary controls. The number of PNNs in the dorsal dentate gyrus was positively correlated with total errors during training. During the probe, greater errors correlated with more PNNs among the exercised group only. Microglia count and cerebral vascularization were not affected by exercise, although proportions of microglia type (ameboid, stout/thick, and thick/thin) were regulated by exercise in the ventral dentate gyrus. We conclude that exercise decreases PNNs in the dentate gyrus in both sexes and this may be related to better spatial learning and memory.
Collapse
Affiliation(s)
- Madeleine G Maheu
- Department of Psychology, Brock University, St. Catharines, ON, Canada; Department of Health Sciences, Brock University, St. Catharines, ON, Canada; Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| | - Noah James
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Zach Clark
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Alex Yang
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Ridhi Patel
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| | - Shawn M Beaudette
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada; Department of Kinesiology, Brock University, St. Catharines, ON, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada; Centre for Neuroscience, Brock University, St. Catharines, ON, Canada.
| | - Paula Duarte-Guterman
- Department of Psychology, Brock University, St. Catharines, ON, Canada; Centre for Neuroscience, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
6
|
Otsuka MY, Essel LB, Sinha A, Nickerson G, Mejia SM, Edge A, Matthews RT, Bouyain S. Aggrecan immobilizes to perineuronal nets through hyaluronan-dependent and hyaluronan-independent binding activities. J Biol Chem 2025; 301:108525. [PMID: 40273987 DOI: 10.1016/j.jbc.2025.108525] [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: 12/10/2024] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Aggrecan (ACAN) is a large, secreted chondroitin sulfate proteoglycan that includes three globular regions named G1, G2, G3, and is decorated with multiple glycosaminoglycan attachments between its G2 and G3 domains. The N-terminal G1 region interacts with the glycosaminoglycan hyaluronan (HA), which is an essential component of the vertebrate extracellular matrix. In the central nervous system, ACAN is found in perineuronal nets (PNNs), honeycomb-like structures that localize to the surface of parvalbumin-positive neurons in specific neural circuits. PNNs regulate the plasticity of the central nervous system, and it is believed that association between ACAN and HA is a foundational event in the assembly of these reticular structures. Here, we report the cocrystal structure of the G1 region of ACAN in the absence and presence of a HA decasaccharide and analyze the importance of the HA-binding activity of ACAN for its integration into PNNs. We demonstrate that the single immunoglobulin domain and the two Link modules that comprise the G1 region form a single structural unit, and that HA is clamped inside a groove that spans the length of the tandem Link domains. Introducing point mutations in the glycosaminoglycan-binding site eliminates HA-binding activity in ACAN, but, surprisingly, only decreases the integration of ACAN into PNNs. Thus, these results suggest that ACAN can be recruited into PNNs independently of its HA-binding activity.
Collapse
Affiliation(s)
- Matthew Y Otsuka
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Leslie B Essel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Ashis Sinha
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Gabrielle Nickerson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Seth M Mejia
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Ashley Edge
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Russell T Matthews
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA.
| | - Samuel Bouyain
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, USA.
| |
Collapse
|
7
|
Baidoe-Ansah D, Mirzapourdelavar H, Aleshin S, Schott BH, Seidenbecher C, Kaushik R, Dityatev A. Neurocan regulates axon initial segment organization and neuronal activity. Matrix Biol 2025; 136:22-35. [PMID: 39788215 DOI: 10.1016/j.matbio.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The neural extracellular matrix (ECM) accumulates in the form of perineuronal nets (PNNs), particularly around fast-spiking GABAergic interneurons in the cortex and hippocampus, but also around synapses and in association with the axon initial segments (AIS) and nodes of Ranvier. Increasing evidence highlights the role of Neurocan (Ncan), a brain-specific component of ECM, in the pathophysiology of neuropsychiatric disorders like bipolar disorder and schizophrenia. Ncan localizes at PNNs, perisynaptically, and at the nodes of Ranvier and the AIS, highlighting its potential role in regulating axonal excitability. Here, we used knockdown and knockout approaches in mouse primary cortical neurons in combination with immunocytochemistry, Western blotting and electrophysiological techniques to characterize the role of Ncan in the organization of PNNs and AISs and regulation of neuronal activity. We found that reduced Ncan levels led to remodeling of PNNs around neurons via upregulation of aggrecan mRNA and protein levels, increased expression of activity-dependent c-Fos and FosB genes and elevated spontaneous synaptic activity. The latter correlated with increased levels of ankyrin-G in the AIS, particularly in excitatory neurons, and with the elevated expression of Nav1.6 channels. Our results suggest that Ncan regulates the expression of key proteins in PNNs and AISs and provide new insights into its role in fine-tuning neuronal functions.
Collapse
Affiliation(s)
- David Baidoe-Ansah
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Stepan Aleshin
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Björn Hendrik Schott
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Constanze Seidenbecher
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Rahul Kaushik
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
8
|
Kuhl LM, Jeffers MS, Hristozov N, Karthikeyan S, McDonald MW, Hufnagel A, Carter A, Dancause N, Corbett D. Post-Stroke Recovery in Relation to Parvalbumin-Positive Interneurons and Perineuronal Nets. Neurorehabil Neural Repair 2025; 39:286-296. [PMID: 39819253 PMCID: PMC11982584 DOI: 10.1177/15459683241309567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
BACKGROUND There is a critical time window of post-stroke neuroplasticity when spontaneous behavioral recovery occurs. Potential factors responsible for this heightened plasticity are the reduction of parvalbumin-immunoreactive (PV+) interneuron inhibitory signaling and the disappearance of extracellular matrix synaptic stabilizers called perineuronal net(s; PNN/PNNs). OBJECTIVE This study investigated whether behavioral recovery during this critical period following stroke is associated with changes in densities of PV+ interneurons and PNNs.MethodsMale, Sprague-Dawley rats received forelimb motor cortex stroke (n = 43) using endothelin-1, or vehicle injections (n = 44). Cohorts of rats underwent a battery of motor tests and were sacrificed within the post-stroke critical window on day 1, and 1, 2, 4, and 6 weeks. Using immunofluorescent labeling, PNNs (wisteria floribunda agglutinin; WFA+ cells), PV+ interneurons, and cells expressing both PV and PNNs were quantified in contra- and ipsilesional cortices to elucidate their spatial-temporal profiles following stroke.ResultsPV+ interneuron density decreased significantly at 1-day post-stroke in the lateral ipsilesional cortex, while the density of PNNs was significantly lower up to 4 weeks post-stroke in the lateral ipsilesional cortex and at 1 and 2 weeks post-stroke in the medial ipsilesional cortex. Reduction of combined PV+/PNN signaling coincided with spontaneous behavioral recovery.ConclusionsThese results suggest that post-stroke behavioral recovery corresponds to an early reduction in PV+/PNN co-labeled cells in conjunction with an early temporally-dependent reduction in PV+ interneuron signaling and chronic disappearance of PNNs. Interventions targeting PNNs or PV+ interneuron signaling have significant potential for extending the critical window of recovery following stroke.
Collapse
Affiliation(s)
- Lydia M. Kuhl
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, AB, Calgary, Canada
| | - Matthew S. Jeffers
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Nicolay Hristozov
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sudhir Karthikeyan
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Matthew W. McDonald
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Aisha Hufnagel
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Anthony Carter
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Numa Dancause
- Département de Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre interdisciplaire de recherche sur le cerveau et l’apprentissage, Université de Montréal, Montréal, QC, Canada
| | - Dale Corbett
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
Wang X, Li K, Guo L, Liu X, Guo Y, Zhang W. The Influence of Changes in Microglia Development on the Plasticity of the Developing Visual Cortex Circuit in Juvenile Mice. Invest Ophthalmol Vis Sci 2025; 66:45. [PMID: 40244609 PMCID: PMC12013681 DOI: 10.1167/iovs.66.4.45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose To investigate the role of microglial subtypes in mouse visual cortex development, focusing on ocular dominance plasticity and interactions with GABAergic neurons and the extracellular matrix. Methods Immunofluorescence and single-nucleus RNA-sequencing (snRNA-seq) were used to study microglia in the binocular primary visual cortex (V1) from postnatal day (P) 11 to P42. Gene ontology (GO) analysis assessed synapse organization, and the impact of microglial disruption on ocular dominance plasticity was examined. Visual evoked potentials and miniature postsynaptic current recordings are used to monitor functional changes in V1. Results Microglia underwent a marked expansion between P11 and P21 and stabilized after P35, coinciding with notable changes in gene expression that aligned with synaptic remodeling. GO analysis at P14 and P28 revealed significant enrichment in synaptic organization linked to microglia. Single-nucleus RNA sequencing identified six distinct microglial clusters, among which two functionally relevant subpopulations were closely linked to cortical synaptic plasticity. One cluster, enriched in inflammatory responses and endocytosis, peaked at P21, whereas another cluster, associated with synapse organization and signaling, exhibited dynamic changes after eye opening and during the critical period, significantly influencing cortical synaptic plasticity. In parallel, perineuronal nets (PNNs) and PV(+) interneuron populations increased and reached steady levels by P42, suggesting that microglia help coordinate the timing of inhibitory circuit maturation. Disrupting microglial function during the critical period impaired ocular dominance plasticity, but this effect was reversed after treatment cessation. Mechanistically, microglial depletion enhanced PV(+) interneuron numbers, elevated PNN expression, and altered synapse development. Conclusions Our findings highlight specific microglial subtypes as key regulators of cortical synapse development and plasticity through their interactions with PV(+) interneurons and PNNs. These insights advance our understanding of microglial contributions to visual cortex development and provide potential avenues for targeting microglial function to modulate cortical plasticity.
Collapse
Affiliation(s)
- Xuechun Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Kuan Li
- Department of Respiratory Medicine, Haihe Hospital, Tianjin University, Tianjin, China
| | - Lingzhi Guo
- Institute of Ophthalmology, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Xinlong Liu
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
| | - Yatu Guo
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| | - Wei Zhang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| |
Collapse
|
10
|
Okabe N, Wei X, Abumeri F, Batac J, Hovanesyan M, Dai W, Azarapetian S, Campagna J, Pilati N, Marasco A, Alvaro G, Gunthorpe MJ, Varghese J, Cramer SC, Mody I, Carmichael ST. Parvalbumin interneurons regulate rehabilitation-induced functional recovery after stroke and identify a rehabilitation drug. Nat Commun 2025; 16:2556. [PMID: 40089466 PMCID: PMC11910580 DOI: 10.1038/s41467-025-57860-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Motor disability is a critical impairment in stroke patients. Rehabilitation has a limited effect on recovery; but there is no medical therapy for post-stroke recovery. The biological mechanisms of rehabilitation in the brain remain unknown. Here, using a photothrombotic stroke model in male mice, we demonstrate that rehabilitation after stroke selectively enhances synapse formation in presynaptic parvalbumin interneurons and postsynaptic neurons in the rostral forelimb motor area with axonal projections to the caudal forelimb motor area where stroke was induced (stroke-projecting neuron). Rehabilitation improves motor performance and neuronal functional connectivity, while inhibition of stroke-projecting neurons diminishes motor recovery. Stroke-projecting neurons show decreased dendritic spine density, reduced external synaptic inputs, and a lower proportion of parvalbumin synapse in the total GABAergic input. Parvalbumin interneurons regulate neuronal functional connectivity, and their activation during training is necessary for recovery. Furthermore, gamma oscillation, a parvalbumin-regulated rhythm, is increased with rehabilitation-induced recovery in animals after stroke and stroke patients. Pharmacological enhancement of parvalbumin interneuron function improves motor recovery after stroke, reproducing rehabilitation recovery. These findings identify brain circuits that mediate rehabilitation-recovery and the possibility for rational selection of pharmacological agents to deliver the first molecular-rehabilitation therapeutic.
Collapse
Affiliation(s)
- Naohiko Okabe
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
| | - Xiaofei Wei
- Department of Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Farah Abumeri
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Jonathan Batac
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Mary Hovanesyan
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Weiye Dai
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Srbui Azarapetian
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Jesus Campagna
- The Drug Discovery Lab, Mary S. Easton Center for Alzheimer's Disease Research, Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Nadia Pilati
- Autifony Srl, Istituto di Ricerca Pediatrica Citta' della Speranza, Via Corso Stati Uniti, 4f, 35127, Padua, Italy
| | - Agostino Marasco
- Autifony Srl, Istituto di Ricerca Pediatrica Citta' della Speranza, Via Corso Stati Uniti, 4f, 35127, Padua, Italy
| | - Giuseppe Alvaro
- Autifony Srl, Istituto di Ricerca Pediatrica Citta' della Speranza, Via Corso Stati Uniti, 4f, 35127, Padua, Italy
| | - Martin J Gunthorpe
- Autifony Therapeutics Limited, Stevenage Bioscience Catalyst, Stevenage, SG1 2FX, UK
| | - John Varghese
- The Drug Discovery Lab, Mary S. Easton Center for Alzheimer's Disease Research, Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Steven C Cramer
- Department of Neurology, UCLA, California Rehabilitation Institute, Los Angeles, CA, 90095, USA
| | - Istvan Mody
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
11
|
Obray JD, Denton AR, Carroll-Deaton J, Marquardt K, Chandler LJ, Scofield MD. Enhanced fear extinction through infralimbic perineuronal net digestion: The modulatory role of adolescent alcohol exposure. Alcohol 2025; 123:57-67. [PMID: 39710305 DOI: 10.1016/j.alcohol.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Perineuronal nets (PNNs) are specialized components of the extracellular matrix that play a critical role in learning and memory. In a Pavlovian fear conditioning paradigm, degradation of PNNs affects the formation and storage of fear memories. This study examined the impact of adolescent intermittent ethanol (AIE) exposure by vapor inhalation on the expression of PNNs in the adult rat prelimbic (PrL) and infralimbic (IfL) subregions of the medial prefrontal cortex. Results indicated that following AIE, the total number of PNN positive cells in the PrL cortex increased in layer II/III but did not change in layer V. Conversely, in the IfL cortex, the number of PNN positive cells decreased in layer V, with no change in layer II/III. In addition, the intensity of PNN staining was significantly altered by AIE exposure, which narrowed the distribution of signal intensity, reducing the number of high and low intensity PNNs. Given these changes in PNNs, the next experiment assessed the effects of AIE and PNN digestion on extinction of a conditioned fear memory. In Air control rats, digestion of PNNs by bilateral infusion of Chondroitinase ABC (ChABC) into the IfL cortex enhanced fear extinction and reduced contextual fear renewal. In contrast, both fear extinction learning and contextual fear renewal remained unchanged following PNN digestion in AIE exposed rats. These results highlight the sensitivity of prefrontal PNNs to adolescent alcohol exposure and suggest that ChABC-induced plasticity is reduced in the IfL cortex following AIE exposure.
Collapse
Affiliation(s)
- J Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Adam R Denton
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Psychology, Tusculum University, Tusculum, TN 37745, USA
| | - Jayda Carroll-Deaton
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kristin Marquardt
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
12
|
Wallsten B, Gligor AH, Gonzalez AE, Ramos JD, Baratta MV, Sorg BA. Response of parvalbumin interneurons and perineuronal nets in rat medial prefrontal cortex and lateral amygdala to stressor controllability. Brain Res 2025; 1848:149351. [PMID: 39592089 DOI: 10.1016/j.brainres.2024.149351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024]
Abstract
Behavioral control over a stressor limits the impact of the stressor being experienced and produces enduring changes that reduce the effects of future stressors. In rats, these stress-buffering effects of control (escapable stress, ES) require activation of the medial prefrontal cortex (mPFC) and prevent the typical amygdala-dependent behavioral outcomes of uncontrollable stress (inescapable stress, IS). Parvalbumin (PV) interneurons regulate output of excitatory neurons, and most mPFC PV neurons are surrounded by perineuronal nets (PNNs), which regulate firing. We exposed male rats to a single session of ES, IS, or no stress and measured c-Fos expression within PV/PNN-containing cells in mPFC subregions (prelimbic, PL; infralimbic, IL) and in the lateral amygdala. We also measured the number and intensity of PNNs. Within PL and IL PV/PNN cells, both ES and IS increased c-Fos intensity in PV/PNN, non-PV, and non-PNN cells. Within the IL, only ES increased the number of c-Fos-expressing PV/PNN-labeled cells. In the lateral amygdala, only ES increased c-Fos intensity within PV cells and PV/PNN cells. Thus, PV neurons in the IL and lateral amygdala may represent an important substrate by which behavioral control buffers against the amygdala-dependent behavioral outcomes typically observed after uncontrollable stress.
Collapse
Affiliation(s)
- Brittani Wallsten
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Abigail H Gligor
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Angela E Gonzalez
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States; Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States; Program in Neuroscience, Washington State University, Vancouver, WA 98686, United States
| | - Jonathan D Ramos
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, United States
| | - Barbara A Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States; Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States; Program in Neuroscience, Washington State University, Vancouver, WA 98686, United States.
| |
Collapse
|
13
|
Grødem S, Thompson EH, Røe MB, Vatne GH, Nymoen Nystuen I, Buccino A, Otterstad T, Hafting T, Fyhn M, Lensjø KK. Differential impacts of germline and adult aggrecan knockout in PV+ neurons on perineuronal nets and PV+ neuronal function. Mol Psychiatry 2025:10.1038/s41380-025-02894-5. [PMID: 39837996 DOI: 10.1038/s41380-025-02894-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 12/06/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Perineuronal nets (PNNs) are a condensed form of extracellular matrix primarily found around parvalbumin-expressing (PV+) interneurons. The postnatal maturation of PV+ neurons is accompanied with the formation of PNNs and reduced plasticity. Alterations in PNN and PV+ neuron function have been described for mental disorders such as schizophrenia and autism. The formation of PNNs is highly dependent on aggrecan, a proteoglycan encoded by the ACAN gene, but it remains unknown if it is produced by the PV+ neurons themselves. Thus, we established a knockout (KO) mouse model (ACANflx/PVcre) and an adeno-associated virus to specifically eliminate aggrecan production from PV+ neurons, in the germline or adult animals, respectively. The germline KO (ACANflx/PVcre) eliminated the expression of PNNs labeled by Wisteria floribunda agglutinin (WFA), the most commonly used PNN marker. Surprisingly, electrophysiological properties of PV+ interneurons and ocular dominance plasticity of adult ACANflx/PVcre mice were similar to controls. In contrast, AAV-mediated ACAN knockout in adult mice increased ocular dominance plasticity. Moreover, in vivo Chondroitinase ABC treatment of KO mice resulted in reduced firing rate of PV+ cells and increased frequency of spontaneous excitatory postsynaptic currents (sEPSC), a phenotype associated with chABC treatment of WT animals. These findings suggest that compensatory mechanisms may be activated during development in response to the germline loss of aggrecan. Indeed, qPCR of bulk tissue indicates that other PNN components, including neurocan and tenascin-R, are expressed at higher levels in the KO animals. Finally, behavioral testing revealed that ACANflx/PVcre mice had similar long-term memory as controls in the Morris water maze. However, they employed bolder search strategies during spatial learning and showed lower level of anxiety-related behavior in an open field and zero maze.
Collapse
Affiliation(s)
- Sverre Grødem
- Department of Bioscience, University of Oslo, Oslo, Norway
| | | | | | | | | | - Alessio Buccino
- Department of Bioscience, University of Oslo, Oslo, Norway
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | | | - Torkel Hafting
- Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Marianne Fyhn
- Department of Bioscience, University of Oslo, Oslo, Norway
| | - Kristian Kinden Lensjø
- Department of Bioscience, University of Oslo, Oslo, Norway.
- Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
14
|
Wan X, Wang AS, Storch DS, Li VY, Sakata JT. Perineuronal nets in motor circuitry regulate the performance of learned vocalizations in songbirds. Commun Biol 2025; 8:86. [PMID: 39827274 PMCID: PMC11743155 DOI: 10.1038/s42003-025-07520-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025] Open
Abstract
The accurate and reliable performance of learned vocalizations (e.g., speech and birdsong) modulates the efficacy of communication in humans and songbirds. Consequently, it is critical to understand the factors that regulate the performance of learned vocalizations. Across taxa, neural circuits underlying motor learning and control are replete with perineuronal nets (PNNs), and we analyzed how PNNs in vocal motor circuitry regulate the performance of learned song in zebra finches. We report that developmental increases in PNN expression in vocal circuitry are associated with developmental increases in song stereotypy. We also document that enzymatically degrading PNNs in the motor nucleus HVC acutely altered song structure (changes in syllable sequencing and production). Collectively, our data reveal a causal contribution of PNNs to the performance of learned behaviors and, given the parallels in the regulation of birdsong and speech, suggest that PNNs in motor circuitry could modulate speech performance.
Collapse
Affiliation(s)
- Xinghaoyun Wan
- Department of Biology, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Angela S Wang
- Department of Biology, McGill University, Montreal, QC, Canada
| | | | - Vivian Y Li
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Jon T Sakata
- Department of Biology, McGill University, Montreal, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Centre for Research in Brain, Language and Music, McGill University, Montreal, QC, Canada.
| |
Collapse
|
15
|
Sokolov R, Krut' V, Belousov V, Rozov A, Mukhina IV. Hyaluronidase-induced matrix remodeling contributes to long-term synaptic changes. Front Neural Circuits 2025; 18:1441280. [PMID: 39897766 PMCID: PMC11782146 DOI: 10.3389/fncir.2024.1441280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 02/04/2025] Open
Abstract
Extracellular brain space contains water, dissolved ions, and multiple other signaling molecules. The neural extracellular matrix (ECM) is also a significant component of the extracellular space. The ECM is synthesized by neurons, astrocytes, and other types of cells. Hyaluronan, a hyaluronic acid polymer, is a key component of the ECM. The functions of hyaluronan include barrier functions and signaling. In this article, we investigate physiological processes during the acute phase of enzymatic ECM removal. We found that hyaluronidase, an ECM removal agent, triggers simultaneous membrane depolarization and sharp calcium influx into neurons. Spontaneous action potential firing frequency increased rapidly after ECM destruction in interneurons, but not pyramidal neurons. Hyaluronidase-dependent calcium entry can be blocked by a selective antagonist of N-methyl-D-aspartate (NMDA) receptors, revealing these receptors as the main player in the observed phenomenon. Additionally, we demonstrate increased NMDA-dependent long-term potentiation at CA3-to-CA1 synapses during the acute phase of ECM removal. These findings suggest that hyaluronan is a significant synaptic player.
Collapse
Affiliation(s)
- Rostislav Sokolov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Viktoriya Krut'
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Vsevolod Belousov
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Andrey Rozov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Irina V. Mukhina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| |
Collapse
|
16
|
Hsiao Y, Fonseca MA, Tiemroth AS, Vasquez EJ, Gomez AM. Persistent large-scale changes in alternative splicing in prefrontal cortical neuron types following psychedelic exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633439. [PMID: 39868117 PMCID: PMC11761703 DOI: 10.1101/2025.01.16.633439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Psychedelics engage the serotonergic system as potent neuromodulators, increasing neuroplasticity in humans and rodents. Persistent changes in cognitive flexibility, emotional regulation, and social cognition are thought to underlie the therapeutic effects of psychedelics. However, the underlying molecular and cellular basis of psychedelic-induced plasticity remains unclear. Here, we identify persistent, cell type-specific alternative splicing changes in the mouse medial prefrontal cortex (mPFC) induced by a single dose of psychedelics. Combining deep RiboTag sequencing and bioinformatics, we find that a single dose of psychedelics modestly alters gene expression while dramatically shifting patterns of alternative splicing lasting at least a month. We connect our functional enrichment and alternative splicing analysis with changes in the extracellular matrix, synaptic physiology, and intrinsic physiology in parvalbumin interneurons days to a week after psychedelic treatment. Our dataset is an essential resource for understanding the persistent, cell type-specific effects of psychedelics on cortical cell types and functions.
Collapse
|
17
|
Towner TT, Coleman HJ, Goyden MA, Vore AS, Papastrat KM, Varlinskaya EI, Werner DF. Prelimbic cortex perineuronal net expression and social behavior: Impact of adolescent intermittent ethanol exposure. Neuropharmacology 2025; 262:110195. [PMID: 39437849 DOI: 10.1016/j.neuropharm.2024.110195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/05/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Adolescent intermittent ethanol (AIE) exposure in rats leads to social deficits. Parvalbumin (PV) expressing fast-spiking interneurons in the prelimbic cortex (PrL) contribute to social behavior, and perineuronal nets (PNNs) within the PrL preferentially encompass and regulate PV interneurons. AIE exposure increases PNNs, but it is unknown if this upregulation contributes to AIE-induced social impairments. The current study was designed to determine the effect of AIE exposure on PNN expression in the PrL and to assess whether PNN dysregulation contributes to social deficits elicited by AIE. cFos-LacZ male and female rats were exposed every other day to tap water or ethanol (4 g/kg, 25% w/v) via intragastric gavage between postnatal day (P) 25-45. We evaluated neuronal activation by β-galactosidase expression and PNN levels either at the end of the exposure regimen on P45 and/or in adulthood on P70. In addition, we used Chondroitinase ABC (ChABC) to deplete PNNs following adolescent exposure (P48) and allowed for PNN restoration before social testing in adulthhod. AIE exposure increased PNN expression in the PrL of adult males, but decreased PNNs immediately following AIE. Vesicular glutamate transporter 2 (vGlut2) and vesicular GABA transporter (vGat) near PNNs were downregulated only in AIE-exposed females. Gene expression of PNN components was largely unaffected by AIE exposure. Removal and reestablishment of PrL PNNs by ChABC led to upregulation of PNNs and social impairments in males, regardless of adolescent exposure. These data suggest that AIE exposure in males upregulates PrL PNNs that likely contribute to social impairments induced by AIE.
Collapse
Affiliation(s)
- Trevor T Towner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Harper J Coleman
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Matthew A Goyden
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Andrew S Vore
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Kimberly M Papastrat
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Elena I Varlinskaya
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - David F Werner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA.
| |
Collapse
|
18
|
Li X, Li N, Zhao P, Ren D, Luo B, Zhou T. Perineuronal Nets: From Structure to Neurological Disorders. Curr Med Chem 2025; 32:1685-1701. [PMID: 37946343 DOI: 10.2174/0109298673258290231009111633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/05/2023] [Accepted: 09/01/2023] [Indexed: 11/12/2023]
Abstract
Perineuronal nets (PNN) is condensed extracellular matrix (ECM) in the central nervous system (CNS), which surrounds cell soma, axon initial segments, and synapses. In the brain, most neurons surrounded by PNN are interneurons, especially the parvalbumin-expressing interneurons (PVI). The formation of PNN is involved in the PVI maturation as well as the onset and closure of critical periods for developmental plasticity end. Dysfunction of PVI can lead to some neurological disorders, such as schizophrenia, bipolar depression, and Alzheimer's disease. Similarly, PNN assembling abnormalities are often observed in human patients and animal disease models. PNN is thought to have a neuroprotective effect and interact with signaling molecules to regulate synaptic plasticity and neuronal activity. In this review, we provide an overview of the composition, structure, and functions of PNN. In addition, we highlight abnormal changes in PNN components in pathological conditions. Understanding the roles of different components of PNN will bring us a new perspective on brain plasticity and neurological disorders.
Collapse
Affiliation(s)
- Xianghe Li
- Queen Mary School of Nanchang University, Nanchang 330031, China
| | - Nuojin Li
- Queen Mary School of Nanchang University, Nanchang 330031, China
| | - Pingping Zhao
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Dongyan Ren
- Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Bin Luo
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Tian Zhou
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| |
Collapse
|
19
|
Jabłońska J, Wiera G, Mozrzymas JW. Extracellular matrix integrity regulates GABAergic plasticity in the hippocampus. Matrix Biol 2024; 134:184-196. [PMID: 39491759 DOI: 10.1016/j.matbio.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/18/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
The brain's extracellular matrix (ECM) is crucial for neural circuit functionality, synaptic plasticity, and learning. While the role of the ECM in excitatory synapses has been extensively studied, its influence on inhibitory synapses, particularly on GABAergic long-term plasticity, remains poorly understood. This study aims to elucidate the effects of ECM components on inhibitory synaptic transmission and plasticity in the hippocampal CA1 region. We focus on the roles of chondroitin sulfate proteoglycans (CSPGs) and hyaluronic acid in modulating inhibitory postsynaptic currents (IPSCs) at two distinct inhibitory synapses formed by somatostatin (SST)-positive and parvalbumin (PV)-positive interneurons onto pyramidal cells (PCs). Using optogenetic stimulation in brain slices, we observed that acute degradation of ECM constituents by hyaluronidase or chondroitinase-ABC did not affect basal inhibitory synaptic transmission. However, short-term plasticity, particularly burst-induced depression, was enhanced at PV→PC synapses following enzymatic treatments. Long-term plasticity experiments demonstrated that CSPGs are essential for NMDA-induced iLTP at SST→PC synapses, whereas the digestion of hyaluronic acid by hyaluronidase impaired iLTP at PV→PC synapses. This indicates a synapse-specific role of CSPGs and hyaluronic acid in regulating GABAergic plasticity. Additionally, we report the presence of cryptic GABAergic plasticity at PV→PC synapses induced by prolonged NMDA application, which became evident after CSPG digestion and was absent under control conditions. Our results underscore the differential impact of ECM degradation on inhibitory synaptic plasticity, highlighting the synapse-specific interplay between ECM components and specific GABAergic synapses. This offers new perspectives in studies on learning and critical period timing.
Collapse
Affiliation(s)
- Jadwiga Jabłońska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland
| | - Grzegorz Wiera
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland.
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland.
| |
Collapse
|
20
|
Tahmasian N, Feng MY, Arbabi K, Rusu B, Cao W, Kukreja B, Lubotzky A, Wainberg M, Tripathy SJ, Kalish BT. Neonatal Brain Injury Triggers Niche-Specific Changes to Cellular Biogeography. eNeuro 2024; 11:ENEURO.0224-24.2024. [PMID: 39681473 DOI: 10.1523/eneuro.0224-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Preterm infants are at risk for brain injury and neurodevelopmental impairment due, in part, to white matter injury following chronic hypoxia exposure. However, the precise molecular mechanisms by which neonatal hypoxia disrupts early neurodevelopment are poorly understood. Here, we constructed a brain-wide map of the regenerative response to newborn brain injury using high-resolution imaging-based spatial transcriptomics to analyze over 800,000 cells in a mouse model of chronic neonatal hypoxia. Additionally, we developed a new method for inferring condition-associated differences in cell type spatial proximity, enabling the identification of niche-specific changes in cellular architecture. We observed hypoxia-associated changes in region-specific cell states, cell type composition, and spatial organization. Importantly, our analysis revealed mechanisms underlying reparative neurogenesis and gliogenesis, while also nominating pathways that may impede circuit rewiring following neonatal hypoxia. Altogether, our work provides a comprehensive description of the molecular response to newborn brain injury.
Collapse
Affiliation(s)
- Nareh Tahmasian
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Min Yi Feng
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Keon Arbabi
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Bianca Rusu
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Wuxinhao Cao
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Bharti Kukreja
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Asael Lubotzky
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - Michael Wainberg
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario M5G 1X5, Canada
| | - Shreejoy J Tripathy
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Brian T Kalish
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
21
|
Otsuka MY, Essel LB, Sinha A, Nickerson G, Mejia SM, Matthews RT, Bouyain S. The hyaluronan-binding activity of aggrecan is important, but not essential, for its specific insertion into perineuronal nets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625086. [PMID: 39651196 PMCID: PMC11623537 DOI: 10.1101/2024.11.25.625086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Aggrecan (ACAN) is a large, secreted chondroitin sulfate proteoglycan that includes three globular regions named G1, G2, G3, and is decorated with multiple glycosaminoglycan attachments between its G2 and G3 domains. The N-terminal G1 region interacts with the glycosaminoglycan hyaluronan (HA), which is an essential component of the vertebrate extracellular matrix. In the central nervous system, ACAN is found in perineuronal nets (PNNs), honeycomb-like structures that are enriched on parvalbumin-positive neurons in specific neural circuits. PNNs regulate the plasticity of the central nervous system, and it is believed that association between ACAN and HA is a foundational event in the assembly of these reticular structures. Here, we report the co-crystal structure of the G1 region of ACAN in the absence and presence of an HA decasaccharide and analyze the importance of the HA-binding activity of ACAN for its integration into PNNs. We demonstrate that the single immunoglobulin domain and the two Link modules that comprise the G1 region form a single structural unit, and that HA is clamped inside a groove that spans the length of the tandem Link domains. Introduction of point mutations in the glycosaminoglycan-binding site eliminates HA-binding activity in ACAN, but, surprisingly, only decreases the integration of ACAN into PNNs. Thus, these results suggest that the HA-binding activity of ACAN is important for its recruitment to PNNs, but it does not appear to be essential.
Collapse
|
22
|
Gilbert KF, Amontree M, Deasy S, Ma J, Conant K. Pramipexole, a D3 receptor agonist, increases cortical gamma power and biochemical correlates of cortical excitation; implications for mood disorders. Eur J Neurosci 2024; 60:6490-6508. [PMID: 39410873 DOI: 10.1111/ejn.16570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024]
Abstract
Major depressive disorder (MDD) has been associated with deficits in working memory as well as underlying gamma oscillation power. Consistent with this, overall reductions in cortical excitation have also been described with MDD. In previous work, we have demonstrated that the monoamine reuptake inhibitor venlafaxine increases gamma oscillation power in ex vivo hippocampal slices and that this is associated with concomitant increases in pyramidal arbour and reduced levels of plasticity-restricting perineuronal nets (PNNs). In the present study, we have examined the effects of chronic treatment with pramipexole (PPX), a D3 dopamine receptor agonist, for its effects on gamma oscillation power as measured by in vivo electroencephalography (EEG) recordings in female BALB/c and C57Bl6 mice. We observe a modest but significant increase in 20-50 Hz gamma power with PPX in both strains. Additionally, biochemical analysis of prefrontal cortex lysates from PPX-treated BALB/c mice shows a number of changes that could contribute to, or follow from, increased pyramidal excitability and/or gamma power. PPX-associated changes include reduced levels of specific PNN components as well as tissue inhibitor of matrix metalloproteases-1 (TIMP-1), which inhibits long-term potentiation of synaptic transmission. Consistent with its effects on gamma power, PNN proteins and TIMP-1, chronic PPX treatment also improves working memory and reduces anhedonia. Together these results add to an emerging literature linking extracellular matrix and/or gamma oscillation power to both mood and cognition.
Collapse
Affiliation(s)
- Karli F Gilbert
- Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine (GUMC), Washington, D.C., USA
| | - Matthew Amontree
- Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine (GUMC), Washington, D.C., USA
| | | | - Junfeng Ma
- Department of Oncology, GUMC, Washington, D.C., USA
| | - Katherine Conant
- Interdisciplinary Program in Neuroscience, Georgetown University School of Medicine (GUMC), Washington, D.C., USA
- Department of Neuroscience, GUMC, Washington, D.C., USA
| |
Collapse
|
23
|
Herzberg MP, Nielsen AN, Luby J, Sylvester CM. Measuring neuroplasticity in human development: the potential to inform the type and timing of mental health interventions. Neuropsychopharmacology 2024; 50:124-136. [PMID: 39103496 PMCID: PMC11525577 DOI: 10.1038/s41386-024-01947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024]
Abstract
Neuroplasticity during sensitive periods, the molecular and cellular process of enduring neural change in response to external stimuli during windows of high environmental sensitivity, is crucial for adaptation to expected environments and has implications for psychiatry. Animal research has characterized the developmental sequence and neurobiological mechanisms that govern neuroplasticity, yet gaps in our ability to measure neuroplasticity in humans limit the clinical translation of these principles. Here, we present a roadmap for the development and validation of neuroimaging and electrophysiology measures that index neuroplasticity to begin to address these gaps. We argue that validation of measures to track neuroplasticity in humans will elucidate the etiology of mental illness and inform the type and timing of mental health interventions to optimize effectiveness. We outline criteria for evaluating putative neuroimaging measures of plasticity in humans including links to neurobiological mechanisms shown to govern plasticity in animal models, developmental change that reflects heightened early life plasticity, and prediction of neural and/or behavior change. These criteria are applied to three putative measures of neuroplasticity using electroencephalography (gamma oscillations, aperiodic exponent of power/frequency) or functional magnetic resonance imaging (amplitude of low frequency fluctuations). We discuss the use of these markers in psychiatry, envision future uses for clinical and developmental translation, and suggest steps to address the limitations of the current putative neuroimaging measures of plasticity. With additional work, we expect these markers will significantly impact mental health and be used to characterize mechanisms, devise new interventions, and optimize developmental trajectories to reduce psychopathology risk.
Collapse
Affiliation(s)
- Max P Herzberg
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| | - Ashley N Nielsen
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA.
| | - Joan Luby
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Chad M Sylvester
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
24
|
Aguilar JS, Lasek AW. Modulation of stress-, pain-, and alcohol-related behaviors by perineuronal nets. Neurobiol Stress 2024; 33:100692. [PMID: 39691634 PMCID: PMC11650882 DOI: 10.1016/j.ynstr.2024.100692] [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: 06/18/2024] [Revised: 10/10/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024] Open
Abstract
Perineuronal nets (PNNs) are a special form of central nervous system extracellular matrix enriched in hyaluronan, chondroitin sulfate proteoglycans, tenascins, and link proteins that regulate synaptic plasticity. Most PNNs in the brain surround parvalbumin-expressing inhibitory interneurons, which tightly regulate excitatory/inhibitory balance and brain activity associated with optimal cognitive functioning. Alterations in PNNs have been observed in neurological diseases and psychiatric disorders, suggesting that they may be key contributors to the neuropathological progression and behavioral changes in these diseases. Alcohol use disorder (AUD), major depressive disorder (MDD), and chronic pain are highly comorbid conditions, and changes in PNNs have been observed in animal models of these disorders, as well as postmortem tissue from individuals diagnosed with AUD and MDD. This review focuses on the literature describing stress-, alcohol-, and pain-induced adaptations in PNNs, potential cellular contributors to altered PNNs, and the role of PNNs in behaviors related to these disorders. Medicines that can restore PNNs to a non-pathological state may be a novel therapeutic approach to treating chronic pain, AUD, and MDD.
Collapse
Affiliation(s)
- Jhoan S. Aguilar
- Center for Clinical Pharmacology, Department of Anesthesiology, Washington University, St Louis, St. Louis, MO, 63110, USA
| | - Amy W. Lasek
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| |
Collapse
|
25
|
Sterin I, Niazi A, Kim J, Park J, Park S. Dynamic Organization of Neuronal Extracellular Matrix Revealed by HaloTag-HAPLN1. J Neurosci 2024; 44:e0666242024. [PMID: 39251350 PMCID: PMC11502233 DOI: 10.1523/jneurosci.0666-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/04/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows a net-like structure around a subset of neurons, a ring-like structure at the nodes of Ranvier, and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to hyaluronan proteoglycan link protein 1, which links hyaluronan and proteoglycans. Expression or application of the probe in primary rat neuronal cultures enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in mouse brains of either sex reveals detailed ECM architectures around excitatory neurons and developmentally regulated dendritic ECM. Our study uncovers extensive structural features of the brain's ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
- Neuroscience Program, University of Utah, Salt Lake City, Utah 84112
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
26
|
Obray JD, Denton AR, Carroll-Deaton J, Marquardt K, Chandler LJ, Scofield MD. Enhanced Fear Extinction Through Infralimbic Perineuronal Net Digestion: The Modulatory Role of Adolescent Alcohol Exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619810. [PMID: 39484370 PMCID: PMC11526981 DOI: 10.1101/2024.10.23.619810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Perineuronal nets (PNNs) are specialized components of the extracellular matrix that play a critical role in learning and memory. In a Pavlovian fear conditioning paradigm, degradation of PNNs affects the formation and storage of fear memories. This study examined the impact of adolescent intermittent ethanol (AIE) exposure by vapor inhalation on the expression of PNNs in the adult rat prelimbic (PrL) and infralimbic (IfL) subregions of the medial prefrontal cortex. Results indicated that following AIE, the total number of PNN positive cells in the PrL cortex increased in layer II/III but did not change in layer V. Conversely, in the IfL cortex, the number of PNN positive cells decreased in layer V, with no change in layer II/III. In addition, the intensity of PNN staining was significantly altered by AIE exposure, which narrowed the distribution of signal intensity, reducing the number of high and low intensity PNNs. Given these changes in PNNs, the next experiment assessed the effects of AIE and PNN digestion on extinction of a conditioned fear memory. In Air control rats, digestion of PNNs by bilateral infusion of Chondroitinase ABC (ChABC) into the IfL cortex enhanced fear extinction and reduced contextual fear renewal. In contrast, both fear extinction learning and contextual fear renewal remained unchanged following PNN digestion in AIE exposed rats. These results highlight the sensitivity of prefrontal PNNs to adolescent alcohol exposure and suggest that ChABC-induced plasticity is reduced in the IfL cortex following AIE exposure.
Collapse
Affiliation(s)
- J. Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Adam R. Denton
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425
- Department of Psychology, Tusculum University, Tusculum, TN 37745
| | - Jayda Carroll-Deaton
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Kristin Marquardt
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - L. Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
| | - Michael D. Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
27
|
Faust TE, Devlin BA, Farhy-Tselnicker I, Ferro A, Postolache M, Xin W. Glial Control of Cortical Neuronal Circuit Maturation and Plasticity. J Neurosci 2024; 44:e1208242024. [PMID: 39358028 PMCID: PMC11450532 DOI: 10.1523/jneurosci.1208-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 10/04/2024] Open
Abstract
The brain is a highly adaptable organ that is molded by experience throughout life. Although the field of neuroscience has historically focused on intrinsic neuronal mechanisms of plasticity, there is growing evidence that multiple glial populations regulate the timing and extent of neuronal plasticity, particularly over the course of development. This review highlights recent discoveries on the role of glial cells in the establishment of cortical circuits and the regulation of experience-dependent neuronal plasticity during critical periods of neurodevelopment. These studies provide strong evidence that neuronal circuit maturation and plasticity are non-cell autonomous processes that require both glial-neuronal and glial-glial cross talk to proceed. We conclude by discussing open questions that will continue to guide research in this nascent field.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina 27708
| | | | - Austin Ferro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | - Maggie Postolache
- Brain Immunology & Glia Center, Washington University School of Medicine, St. Louis, Missouri 63110
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California 94158
| |
Collapse
|
28
|
Lev-Ram V, Lemieux SP, Deerinck TJ, Bushong EA, Perez AJ, Pritchard DR, Toyama BH, Park SKR, McClatchy DB, Savas JN, Whitney M, Adams SR, Ellisman MH, Yates J, Tsien RY. Do Perineuronal Nets Stabilize the Engram of a Synaptic Circuit? Cells 2024; 13:1627. [PMID: 39404392 PMCID: PMC11476018 DOI: 10.3390/cells13191627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024] Open
Abstract
Perineuronal nets (PNNs), a specialized form of extra cellular matrix (ECM), surround numerous neurons in the CNS and allow synaptic connectivity through holes in its structure. We hypothesize that PNNs serve as gatekeepers that guard and protect synaptic territory and thus may stabilize an engram circuit. We present high-resolution and 3D EM images of PNN-engulfed neurons in mice brains, showing that synapses occupy the PNN holes and that invasion of other cellular components is rare. PNN constituents in mice brains are long-lived and can be eroded faster in an enriched environment, while synaptic proteins have a high turnover rate. Preventing PNN erosion by using pharmacological inhibition of PNN-modifying proteases or matrix metalloproteases 9 (MMP9) knockout mice allowed normal fear memory acquisition but diminished long-term memory stabilization, supporting the above hypothesis.
Collapse
Affiliation(s)
- Varda Lev-Ram
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Sakina Palida Lemieux
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Thomas J. Deerinck
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
| | - Eric A. Bushong
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
| | - Alex J. Perez
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
| | - Denise R. Pritchard
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Brandon H. Toyama
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sung Kyu R. Park
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
| | - Daniel B. McClatchy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
| | - Jeffrey N. Savas
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60208, USA
| | - Michael Whitney
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Stephen R. Adams
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
- Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - John Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Roger Y. Tsien
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
- Department of Chemistry & Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
29
|
Birch EE, Duffy KR. Leveraging neural plasticity for the treatment of amblyopia. Surv Ophthalmol 2024; 69:818-832. [PMID: 38763223 PMCID: PMC11380599 DOI: 10.1016/j.survophthal.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024]
Abstract
Amblyopia is a form of visual cortical impairment that arises from abnormal visual experience early in life. Most often, amblyopia is a unilateral visual impairment that can develop as a result of strabismus, anisometropia, or a combination of these conditions that result in discordant binocular experience. Characterized by reduced visual acuity and impaired binocular function, amblyopia places a substantial burden on the developing child. Although frontline treatment with glasses and patching can improve visual acuity, residual amblyopia remains for most children. Newer binocular-based therapies can elicit rapid recovery of visual acuity and may also improve stereoacuity in some children. Nevertheless, for both treatment modalities full recovery is elusive, recurrence of amblyopia is common, and improvements are negligible when treatment is administered at older ages. Insights derived from animal models about the factors that govern neural plasticity have been leveraged to develop innovative treatments for amblyopia. These novel therapies exhibit efficacy to promote recovery, and some are effective even at ages when conventional treatments fail to yield benefit. Approaches for enhancing visual system plasticity and promoting recovery from amblyopia include altering the balance between excitatory and inhibitory mechanisms, reversing the accumulation of proteins that inhibit plasticity, and harnessing the principles of metaplasticity. Although these therapies have exhibited promising results in animal models, their safety and ability to remediate amblyopia need to be evaluated in humans.
Collapse
Affiliation(s)
- Eileen E Birch
- Crystal Charity Ball Pediatric Vision Laboratory, Retina Foundation, Dallas, TX, USA; University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Kevin R Duffy
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
30
|
Rahimian R, Belliveau C, Simard S, Turecki G, Mechawar N. Perineuronal Net Alterations Following Early-Life Stress: Are Microglia Pulling Some Strings? Biomolecules 2024; 14:1087. [PMID: 39334854 PMCID: PMC11430691 DOI: 10.3390/biom14091087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The extracellular matrix plays a key role in synapse formation and in the modulation of synaptic function in the central nervous system. Recent investigations have revealed that microglia, the resident immune cells of the brain, are involved in extracellular matrix remodeling under both physiological and pathological conditions. Moreover, the dysregulation of both innate immune responses and the extracellular matrix has been documented in stress-related psychopathologies as well as in relation to early-life stress. However, the dynamics of microglial regulation of the ECM and how it can be impacted by early-life adversity have been understudied. This brief review provides an overview of the recent literature on this topic, drawing from both animal model and human post mortem studies. Direct and indirect mechanisms through which microglia may regulate the extracellular matrix-including perineuronal nets-are presented and discussed in light of the interactions with other cell types.
Collapse
Affiliation(s)
- Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Sophie Simard
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
31
|
Wingert JC, Ramos JD, Reynolds SX, Gonzalez AE, Rose RM, Hegarty DM, Aicher SA, Bailey LG, Brown TE, Abbas AI, Sorg BA. Perineuronal Nets in the Rat Medial Prefrontal Cortex Alter Hippocampal-Prefrontal Oscillations and Reshape Cocaine Self-Administration Memories. J Neurosci 2024; 44:e0468242024. [PMID: 38991791 PMCID: PMC11340292 DOI: 10.1523/jneurosci.0468-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
The medial prefrontal cortex (mPFC) is a major contributor to relapse to cocaine in humans and to reinstatement in rodent models of cocaine use disorder. The output from the mPFC is potently modulated by parvalbumin (PV)-containing fast-spiking interneurons, the majority of which are surrounded by perineuronal nets. We previously showed that treatment with chondroitinase ABC (ABC) reduced the consolidation and reconsolidation of a cocaine conditioned place preference memory. However, self-administration memories are more difficult to disrupt. Here we report in male rats that ABC treatment in the mPFC attenuated the consolidation and blocked the reconsolidation of a cocaine self-administration memory. However, reconsolidation was blocked when rats were given a novel, but not familiar, type of retrieval session. Furthermore, ABC treatment prior to, but not after, memory retrieval blocked reconsolidation. This same treatment did not alter a sucrose memory, indicating specificity for cocaine-induced memory. In naive rats, ABC treatment in the mPFC altered levels of PV intensity and cell firing properties. In vivo recordings from the mPFC and dorsal hippocampus (dHIP) during the novel retrieval session revealed that ABC prevented reward-associated increases in high-frequency oscillations and synchrony of these oscillations between the dHIP and mPFC. Together, this is the first study to show that ABC treatment disrupts reconsolidation of the original memory when combined with a novel retrieval session that elicits coupling between the dHIP and mPFC. This coupling after ABC treatment may serve as a fundamental signature for how to disrupt reconsolidation of cocaine memories and reduce relapse.
Collapse
Affiliation(s)
- Jereme C Wingert
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | - Jonathan D Ramos
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | | | - Angela E Gonzalez
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
- Program in Neuroscience, Washington State University, Vancouver, Washington 98686
| | - R Mae Rose
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | - Deborah M Hegarty
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Sue A Aicher
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Lydia G Bailey
- Program in Neuroscience, Washington State University, Pullman, Washington 99164
| | - Travis E Brown
- Program in Neuroscience, Washington State University, Pullman, Washington 99164
| | - Atheir I Abbas
- Departments of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
- Psychiatry, Oregon Health & Science University, Portland, Oregon 97239
- Research Division, VA Portland Health Care System, Portland, Oregon 97239
| | - Barbara A Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
- Program in Neuroscience, Washington State University, Vancouver, Washington 98686
| |
Collapse
|
32
|
Carey SD, Conant K, Maguire-Zeiss KA. Short-term exposure to HIV Tat induces glial activation and changes in perineuronal nets. Eur J Neurosci 2024; 60:4303-4316. [PMID: 38844747 DOI: 10.1111/ejn.16427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 08/07/2024]
Abstract
Despite widespread use of combination antiretroviral therapy (cART), there remains a subset of individuals who display cognitive impairment broadly known as HIV-associated neurocognitive disorder (HAND). Interestingly, HIV-infected cells continuously release the HIV-1 protein Tat even in the presence of cART. Persistent exposure to Tat is proposed to increase both neuroinflammation and neurotoxicity. In vitro evidence shows that matrix metalloproteinases (MMPs) are among the neuroinflammatory molecules induced by Tat, which are known to disrupt specialized neuronal extracellular matrix structures called perineuronal nets (PNNs). PNNs predominantly surround parvalbumin interneurons and help to buffer these cells from oxidant stress and to independently increase their excitability. In order to better understand the link between short-term exposure to Tat, neuroinflammation, and PNNs, we explored the direct effects of Tat on glial cells and neurons. Herein, we report that in mixed glial cultures, Tat directly increases the expression of proinflammatory molecules, including MMP-9. Moreover, direct injection of Tat protein into mouse hippocampus increases the expression of astrocyte and microglia markers as well as MMP-9. The number of PNNs is decreased following Tat exposure, followed later by decreased numbers of hippocampal parvalbumin-expressing neurons. In older mice, Tat induced significant increases in the gene expression of proinflammatory molecules including markers of gliosis, MMPs and complement system proteins. Taken together, these data support a direct effect of Tat on glial-derived MMP expression subsequently affecting PNNs and neuronal health, with older mice more susceptible to Tat-induced inflammation.
Collapse
Affiliation(s)
- Sean D Carey
- Department of Biology, Georgetown University, Washington DC, United States
| | - Katherine Conant
- Department of Neuroscience, Georgetown University School of Medicine, Washington DC, United States
| | - Kathleen A Maguire-Zeiss
- Department of Neuroscience, Georgetown University School of Medicine, Washington DC, United States
| |
Collapse
|
33
|
Sullivan ED, Dannenhoffer CA, Sutherland EB, Vidrascu EM, Gómez-A A, Boettiger CA, Robinson DL. Effects of adolescent intermittent ethanol exposure on cortical perineuronal net and parvalbumin expression in adulthood mediate behavioral inflexibility. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1507-1518. [PMID: 39073296 PMCID: PMC11305908 DOI: 10.1111/acer.15395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Alcohol is commonly consumed by adolescents in a binge-like pattern, which can lead to long-lasting cognitive deficits, including reduced behavioral flexibility. We and others have determined that adolescent intermittent ethanol (AIE) exposure leads to increased number of perineuronal net (PNN) numbers in brain regions that are important for behavioral flexibility. However, whether altered neurochemistry stemming from AIE exposure plays a significant role in reduced behavioral flexibility is unknown. METHODS We measured the number and size of parvalbumin expressing (PV+) interneurons and associated PNNs within the orbitofrontal cortex (OFC), prelimbic cortex (PrL), infralimbic cortex (IL), and anterior insular cortex (AIC) of female and male rats following AIE or control exposure and subsequent training on an attentional set-shift task (ASST). We then ran analyses to determine whether AIE-induced changes in PV and PNN measures statistically mediated the AIE-induced behavioral deficit in reversal learning. RESULTS We demonstrate that AIE exposure impaired behavioral flexibility on reversal two of the ASST (i.e., recalling the initial learned associations), and led to smaller PV+ cells and increased PNN numbers in the AIC. Interestingly, PNN size and number were not altered in the PrL or IL following AIE exposure, in contrast to prior reports. Mediation analyses suggest that AIE alters behavioral flexibility, at least in part through changes in PV and PNN fluorescent measures in the AIC. CONCLUSIONS This study reveals a significant link between AIE exposure, neural alterations, and diminished behavioral flexibility in rats, and highlights a potential novel mechanism comprising changes in PV and PNN measures within the AIC. Future studies should explore the impact of PNN degradation within the AIC on behavioral flexibility.
Collapse
Affiliation(s)
- Emily D.K. Sullivan
- Bowles Center for Alcohol Studies at University of North Carolina at Chapel Hill, Dept. of Psychiatry, Chapel Hill, NC, 27278, USA
| | - Carol A. Dannenhoffer
- Bowles Center for Alcohol Studies at University of North Carolina at Chapel Hill, Dept. of Psychiatry, Chapel Hill, NC, 27278, USA
| | - Elizabeth B. Sutherland
- Bowles Center for Alcohol Studies at University of North Carolina at Chapel Hill, Dept. of Psychology & Neuroscience, Chapel Hill, NC, 27278, USA
| | - Elena M. Vidrascu
- Bowles Center for Alcohol Studies at University of North Carolina at Chapel Hill, Dept. of Psychology & Neuroscience, Chapel Hill, NC, 27278, USA
| | - Alexander Gómez-A
- Bowles Center for Alcohol Studies at University of North Carolina at Chapel Hill, Dept. of Psychiatry, Chapel Hill, NC, 27278, USA
| | - Charlotte A. Boettiger
- Bowles Center for Alcohol Studies at University of North Carolina at Chapel Hill, Dept. of Psychology & Neuroscience, Chapel Hill, NC, 27278, USA
| | - Donita L. Robinson
- Bowles Center for Alcohol Studies at University of North Carolina at Chapel Hill, Dept. of Psychiatry, Chapel Hill, NC, 27278, USA
| |
Collapse
|
34
|
Honeycutt S, Mukherjee A, Paladino M, Gilles-Thomas E, Loney G. Adolescent nicotine exposure promotes adulthood opioid consumption that persists despite adverse consequences and increases the density of insular perineuronal nets. ADDICTION NEUROSCIENCE 2024; 11:100150. [PMID: 38911872 PMCID: PMC11192509 DOI: 10.1016/j.addicn.2024.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Adolescence marks a sensitive period for neurodevelopment wherein exposure to drugs of abuse may disrupt maturation and induce persistent changes in neurophysiology which may exacerbate the risk for developing substance use disorders in adulthood. Adolescent nicotine exposure (ANE) enhances motivation to obtain drugs of abuse, particularly opioids, and increases vulnerability for the development of opioid use disorder (OUD). Here, we characterized ANE effects on learning about the adverse consequences of opioid consumption in adulthood in the absence of further nicotine administration. First, we show that ANE engenders punishment resistant fentanyl self-administration in a heterogenous seeking-taking chain schedule of reinforcement at least at the tested dose of fentanyl (0.75 μg/kg). We found that ANE rats consumed significantly more fentanyl and contingent foot shock punishment was less efficacious in limiting fentanyl seeking in ANE rats, relative to nicotine-naïve controls. Next, we demonstrated that ANE limits learning about the deleterious consequences of acute opioid intoxication in adulthood. In a combined conditioned taste avoidance and place preference paradigm we found that ANE resulted in significant reductions in the strength of morphine-induced CTA, and a simultaneous enhancement of CPP at a higher dose that was less capable of driving reinforcement in naïve controls. Finally, we examined the expression of perineuronal nets (PNNs) within insular cortex (IC) and found ANE rats to have increased density of PNNs across the anterior IC and significantly more parvalbumin-labeled IC cells relative to naïve controls. Together, these data lay the framework for a mechanistic explanation of the extreme comorbidity between nicotine use and development of OUDs.
Collapse
|
35
|
Iwakura Y, Kobayashi Y, Namba H, Nawa H, Takei N. Epidermal Growth Factor Suppresses the Development of GABAergic Neurons Via the Modulation of Perineuronal Net Formation in the Neocortex of Developing Rodent Brains. Neurochem Res 2024; 49:1347-1358. [PMID: 38353896 DOI: 10.1007/s11064-024-04122-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 04/04/2024]
Abstract
Previously, we reported that epidermal growth factor (EGF) suppresses GABAergic neuronal development in the rodent cortex. Parvalbumin-positive GABAergic neurons (PV neurons) have a unique extracellular structure, perineuronal nets (PNNs). PNNs are formed during the development of PV neurons and are mainly formed from chondroitin sulfate (CS) proteoglycans (CSPGs). We examined the effect of EGF on CSPG production and PNN formation as a potential molecular mechanism for the inhibition of inhibiting GABAergic neuronal development by EGF. In EGF-overexpressing transgenic (EGF-Tg) mice, the number of PNN-positive PV neurons was decreased in the cortex compared with that in wild-type mice, as in our previous report. The amount of CS and neurocan was also lower in the cortex of EGF-Tg mice, with a similar decrease observed in EGF-treated cultured cortical neurons. PD153035, an EGF receptor (ErbB1) kinase inhibitor, prevented those mentioned above excess EGF-induced reduction in PNN. We explored the molecular mechanism underlying the effect of EGF on PNNs using fluorescent substrates for matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases (ADAMs). EGF increased the enzyme activity of MMPs and ADAMs in cultured neurons. These enzyme activities were also increased in the EGF-Tg mice cortex. GM6001, a broad inhibitor of MMPs and ADAMs, also blocked EGF-induced PNN reductions. Therefore, EGF/EGF receptor signals may regulate PNN formation in the developing cortex.
Collapse
Affiliation(s)
- Yuriko Iwakura
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan.
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan.
| | - Yutaro Kobayashi
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Biochemistry, Graduate School of Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hisaaki Namba
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, 640-8156, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, 640-8156, Japan
| | - Nobuyuki Takei
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8122, Japan
| |
Collapse
|
36
|
Lazarov O, Gupta M, Kumar P, Morrissey Z, Phan T. Memory circuits in dementia: The engram, hippocampal neurogenesis and Alzheimer's disease. Prog Neurobiol 2024; 236:102601. [PMID: 38570083 PMCID: PMC11221328 DOI: 10.1016/j.pneurobio.2024.102601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Here, we provide an in-depth consideration of our current understanding of engrams, spanning from molecular to network levels, and hippocampal neurogenesis, in health and Alzheimer's disease (AD). This review highlights novel findings in these emerging research fields and future research directions for novel therapeutic avenues for memory failure in dementia. Engrams, memory in AD, and hippocampal neurogenesis have each been extensively studied. The integration of these topics, however, has been relatively less deliberated, and is the focus of this review. We primarily focus on the dentate gyrus (DG) of the hippocampus, which is a key area of episodic memory formation. Episodic memory is significantly impaired in AD, and is also the site of adult hippocampal neurogenesis. Advancements in technology, especially opto- and chemogenetics, have made sophisticated manipulations of engram cells possible. Furthermore, innovative methods have emerged for monitoring neurons, even specific neuronal populations, in vivo while animals engage in tasks, such as calcium imaging. In vivo calcium imaging contributes to a more comprehensive understanding of engram cells. Critically, studies of the engram in the DG using these technologies have shown the important contribution of hippocampal neurogenesis for memory in both health and AD. Together, the discussion of these topics provides a holistic perspective that motivates questions for future research.
Collapse
Affiliation(s)
- Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Pavan Kumar
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zachery Morrissey
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Trongha Phan
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
37
|
Palmisano A, Pandit S, Smeralda CL, Demchenko I, Rossi S, Battelli L, Rivolta D, Bhat V, Santarnecchi E. The Pathophysiological Underpinnings of Gamma-Band Alterations in Psychiatric Disorders. Life (Basel) 2024; 14:578. [PMID: 38792599 PMCID: PMC11122172 DOI: 10.3390/life14050578] [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: 02/05/2024] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 05/26/2024] Open
Abstract
Investigating the biophysiological substrates of psychiatric illnesses is of great interest to our understanding of disorders' etiology, the identification of reliable biomarkers, and potential new therapeutic avenues. Schizophrenia represents a consolidated model of γ alterations arising from the aberrant activity of parvalbumin-positive GABAergic interneurons, whose dysfunction is associated with perineuronal net impairment and neuroinflammation. This model of pathogenesis is supported by molecular, cellular, and functional evidence. Proof for alterations of γ oscillations and their underlying mechanisms has also been reported in bipolar disorder and represents an emerging topic for major depressive disorder. Although evidence from animal models needs to be further elucidated in humans, the pathophysiology of γ-band alteration represents a common denominator for different neuropsychiatric disorders. The purpose of this narrative review is to outline a framework of converging results in psychiatric conditions characterized by γ abnormality, from neurochemical dysfunction to alterations in brain rhythms.
Collapse
Affiliation(s)
- Annalisa Palmisano
- Chair of Lifespan Developmental Neuroscience, Faculty of Psychology, TUD Dresden University of Technology, 01069 Dresden, Germany
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Department of Education, Psychology, and Communication, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Siddhartha Pandit
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
| | - Carmelo L. Smeralda
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Siena Brain Investigation and Neuromodulation (SI-BIN) Laboratory, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, 53100 Siena, Italy;
| | - Ilya Demchenko
- Interventional Psychiatry Program, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (I.D.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Simone Rossi
- Siena Brain Investigation and Neuromodulation (SI-BIN) Laboratory, Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, 53100 Siena, Italy;
| | - Lorella Battelli
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Center for Neuroscience and Cognitive Systems@UniTn, Istituto Italiano di Tecnologia, 38068 Rovereto, Italy
| | - Davide Rivolta
- Department of Education, Psychology, and Communication, University of Bari Aldo Moro, 70121 Bari, Italy;
| | - Venkat Bhat
- Interventional Psychiatry Program, St. Michael’s Hospital—Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (I.D.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Emiliano Santarnecchi
- Precision Neuroscience and Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA (E.S.)
- Department of Neurology and Radiology, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
38
|
Jakovljević A, Stamenković V, Poleksić J, Hamad MIK, Reiss G, Jakovcevski I, Andjus PR. The Role of Tenascin-C on the Structural Plasticity of Perineuronal Nets and Synaptic Expression in the Hippocampus of Male Mice. Biomolecules 2024; 14:508. [PMID: 38672524 PMCID: PMC11047978 DOI: 10.3390/biom14040508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Neuronal plasticity is a crucial mechanism for an adapting nervous system to change. It is shown to be regulated by perineuronal nets (PNNs), the condensed forms of the extracellular matrix (ECM) around neuronal bodies. By assessing the changes in the number, intensity, and structure of PNNs, the ultrastructure of the PNN mesh, and the expression of inhibitory and excitatory synaptic inputs on these neurons, we aimed to clarify the role of an ECM glycoprotein, tenascin-C (TnC), in the dorsal hippocampus. To enhance neuronal plasticity, TnC-deficient (TnC-/-) and wild-type (TnC+/+) young adult male mice were reared in an enriched environment (EE) for 8 weeks. Deletion of TnC in TnC-/- mice showed an ultrastructural reduction of the PNN mesh and an increased inhibitory input in the dentate gyrus (DG), and an increase in the number of PNNs with a rise in the inhibitory input in the CA2 region. EE induced an increased inhibitory input in the CA2, CA3, and DG regions; in DG, the change was also followed by an increased intensity of PNNs. No changes in PNNs or synaptic expression were found in the CA1 region. We conclude that the DG and CA2 regions emerged as focal points of alterations in PNNs and synaptogenesis with EE as mediated by TnC.
Collapse
Affiliation(s)
- Ana Jakovljević
- Center for Laser Microscopy, Institute for Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vera Stamenković
- Center for Integrative Brain Research, Seattle Children’s Research Institute, 1900 9th Ave, Seattle, WA 98125, USA;
| | - Joko Poleksić
- Institute of Anatomy “Niko Miljanic”, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Gebhard Reiss
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Pavle R. Andjus
- Center for Laser Microscopy, Institute for Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
39
|
Paveliev M, Egorchev AA, Musin F, Lipachev N, Melnikova A, Gimadutdinov RM, Kashipov AR, Molotkov D, Chickrin DE, Aganov AV. Perineuronal Net Microscopy: From Brain Pathology to Artificial Intelligence. Int J Mol Sci 2024; 25:4227. [PMID: 38673819 PMCID: PMC11049984 DOI: 10.3390/ijms25084227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Perineuronal nets (PNN) are a special highly structured type of extracellular matrix encapsulating synapses on large populations of CNS neurons. PNN undergo structural changes in schizophrenia, epilepsy, Alzheimer's disease, stroke, post-traumatic conditions, and some other brain disorders. The functional role of the PNN microstructure in brain pathologies has remained largely unstudied until recently. Here, we review recent research implicating PNN microstructural changes in schizophrenia and other disorders. We further concentrate on high-resolution studies of the PNN mesh units surrounding synaptic boutons to elucidate fine structural details behind the mutual functional regulation between the ECM and the synaptic terminal. We also review some updates regarding PNN as a potential pharmacological target. Artificial intelligence (AI)-based methods are now arriving as a new tool that may have the potential to grasp the brain's complexity through a wide range of organization levels-from synaptic molecular events to large scale tissue rearrangements and the whole-brain connectome function. This scope matches exactly the complex role of PNN in brain physiology and pathology processes, and the first AI-assisted PNN microscopy studies have been reported. To that end, we report here on a machine learning-assisted tool for PNN mesh contour tracing.
Collapse
Affiliation(s)
- Mikhail Paveliev
- Neuroscience Center, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Anton A. Egorchev
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Foat Musin
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Nikita Lipachev
- Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, Kazan 420008, Tatarstan, Russia; (N.L.); (A.V.A.)
| | - Anastasiia Melnikova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, Kazan 420015, Tatarstan, Russia;
| | - Rustem M. Gimadutdinov
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Aidar R. Kashipov
- Institute of Artificial Intelligence, Robotics and Systems Engineering, Kazan Federal University, Kremlyovskaya 18, Kazan 420008, Tatarstan, Russia; (A.R.K.); (D.E.C.)
| | - Dmitry Molotkov
- Biomedicum Imaging Unit, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland;
| | - Dmitry E. Chickrin
- Institute of Artificial Intelligence, Robotics and Systems Engineering, Kazan Federal University, Kremlyovskaya 18, Kazan 420008, Tatarstan, Russia; (A.R.K.); (D.E.C.)
| | - Albert V. Aganov
- Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, Kazan 420008, Tatarstan, Russia; (N.L.); (A.V.A.)
| |
Collapse
|
40
|
Sterin I, Niazi A, Kim J, Park J, Park S. Novel extracellular matrix architecture on excitatory neurons revealed by HaloTag-HAPLN1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587384. [PMID: 38585814 PMCID: PMC10996768 DOI: 10.1101/2024.03.29.587384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows an aggregated pattern in a net-like structure around a subset of neurons and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to HAPLN1, which links hyaluronan and proteoglycans. Expression or application of the probe enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in vivo reveals novel forms of ECM architecture around excitatory neurons and developmentally regulated dendritic ECM. Overall, our study uncovers extensive structural features of the brain' ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
- Neuroscience Program, University of Utah, Salt Lake City, Utah, USA
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
41
|
Li X, Wu X, Lu T, Kuang C, Si Y, Zheng W, Li Z, Xue Y. Perineuronal Nets in the CNS: Architects of Memory and Potential Therapeutic Target in Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:3412. [PMID: 38542386 PMCID: PMC10970535 DOI: 10.3390/ijms25063412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
The extracellular matrix (ECM) within the brain possesses a distinctive composition and functionality, influencing a spectrum of physiological and pathological states. Among its constituents, perineuronal nets (PNNs) are unique ECM structures that wrap around the cell body of many neurons and extend along their dendrites within the central nervous system (CNS). PNNs are pivotal regulators of plasticity in CNS, both during development and adulthood stages. Characterized by their condensed glycosaminoglycan-rich structures and heterogeneous molecular composition, PNNs not only offer neuroprotection but also participate in signal transduction, orchestrating neuronal activity and plasticity. Interfering with the PNNs in adult animals induces the reactivation of critical period plasticity, permitting modifications in neuronal connections and promoting the recovery of neuroplasticity following spinal cord damage. Interestingly, in the adult brain, PNN expression is dynamic, potentially modulating plasticity-associated states. Given their multifaceted roles, PNNs have emerged as regulators in the domains of learning, memory, addiction behaviors, and other neuropsychiatric disorders. In this review, we aimed to address how PNNs contribute to the memory processes in physiological and pathological conditions.
Collapse
Affiliation(s)
- Xue Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xianwen Wu
- Department of Laboratory Animal Sciences, Peking University Health Sciences Center, Beijing 100191, China;
| | - Tangsheng Lu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chenyan Kuang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China;
| | - Yue Si
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei Zheng
- Peking-Tsinghua Centre for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China;
| | - Zhonghao Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yanxue Xue
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
| |
Collapse
|
42
|
Morphett JC, Whittaker AL, Reichelt AC, Hutchinson MR. Perineuronal net structure as a non-cellular mechanism contributing to affective state: A scoping review. Neurosci Biobehav Rev 2024; 158:105568. [PMID: 38309496 DOI: 10.1016/j.neubiorev.2024.105568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Affective state encompasses emotional responses to our physiology and influences how we perceive and respond within our environment. In affective disorders such as depression, cognitive adaptability is challenged, and structural and functional brain changes have been identified. However, an incomplete understanding persists of the molecular and cellular mechanisms at play in affective state. An exciting area of newly appreciated importance is perineuronal nets (PNNs); a specialised component of extracellular matrix playing a critical role in neuroprotection and synaptic plasticity. A scoping review found 24 studies demonstrating that PNNs are still a developing field of research with a promising general trend for stress in adulthood to increase the intensity of PNNs, whereas stress in adolescence reduced (potentially developmentally delayed) PNN numbers and intensity, while antidepressants correlated with reduced PNN numbers. Despite promising trends, limited research underscores the need for further exploration, emphasizing behavioral outcomes for validating affective states. Understanding PNNs' role may offer therapeutic insights for depression and inform biomarker development, advancing precision medicine and enhancing well-being.
Collapse
Affiliation(s)
- J C Morphett
- School of Biomedicine, Adelaide Medical School, University of Adelaide, Adelaide, Kaurna Country, Australia.
| | - A L Whittaker
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA, Australia
| | - A C Reichelt
- School of Biomedicine, Adelaide Medical School, University of Adelaide, Adelaide, Kaurna Country, Australia
| | - M R Hutchinson
- School of Biomedicine, Adelaide Medical School, University of Adelaide, Adelaide, Kaurna Country, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, University of Adelaide, Roseworthy, SA, Australia
| |
Collapse
|
43
|
Wingert JC, Ramos JD, Reynolds SX, Gonzalez AE, Rose RM, Hegarty DM, Aicher SA, Bailey LG, Brown TE, Abbas AI, Sorg BA. Perineuronal nets in the rat medial prefrontal cortex alter hippocampal-prefrontal oscillations and reshape cocaine self-administration memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.577568. [PMID: 38370716 PMCID: PMC10871211 DOI: 10.1101/2024.02.05.577568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The medial prefrontal cortex (mPFC) is a major contributor to relapse to cocaine in humans and to reinstatement behavior in rodent models of cocaine use disorder. Output from the mPFC is modulated by parvalbumin (PV)-containing fast-spiking interneurons, the majority of which are surrounded by perineuronal nets (PNNs). Here we tested whether chondroitinase ABC (ABC)- mediated removal of PNNs prevented the acquisition or reconsolidation of a cocaine self-administration memory. ABC injections into the dorsal mPFC prior to training attenuated the acquisition of cocaine self-administration. Also, ABC given 3 days prior to but not 1 hr after memory reactivation blocked cue-induced reinstatement. However, reduced reinstatement was present only in rats given a novel reactivation contingency, suggesting that PNNs are required for the updating of a familiar memory. In naive rats, ABC injections into mPFC did not alter excitatory or inhibitory puncta on PV cells but reduced PV intensity. Whole-cell recordings revealed a greater inter-spike interval 1 hr after ABC, but not 3 days later. In vivo recordings from the mPFC and dorsal hippocampus (dHIP) during novel memory reactivation revealed that ABC in the mPFC prevented reward-associated increases in beta and gamma activity as well as phase-amplitude coupling between the dHIP and mPFC. Together, our findings show that PNN removal attenuates the acquisition of cocaine self-administration memories and disrupts reconsolidation of the original memory when combined with a novel reactivation session. Further, reduced dHIP/mPFC coupling after PNN removal may serve as a key biomarker for how to disrupt reconsolidation of cocaine memories and reduce relapse.
Collapse
|
44
|
Wang AS, Wan X, Storch DS, Li VY, Cornez G, Balthazart J, Cisneros-Franco JM, de Villers-Sidani E, Sakata JT. Cross-species conservation in the regulation of parvalbumin by perineuronal nets. Front Neural Circuits 2023; 17:1297643. [PMID: 38179221 PMCID: PMC10766385 DOI: 10.3389/fncir.2023.1297643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024] Open
Abstract
Parvalbumin (PV) neurons play an integral role in regulating neural dynamics and plasticity. Therefore, understanding the factors that regulate PV expression is important for revealing modulators of brain function. While the contribution of PV neurons to neural processes has been studied in mammals, relatively little is known about PV function in non-mammalian species, and discerning similarities in the regulation of PV across species can provide insight into evolutionary conservation in the role of PV neurons. Here we investigated factors that affect the abundance of PV in PV neurons in sensory and motor circuits of songbirds and rodents. In particular, we examined the degree to which perineuronal nets (PNNs), extracellular matrices that preferentially surround PV neurons, modulate PV abundance as well as how the relationship between PV and PNN expression differs across brain areas and species and changes over development. We generally found that cortical PV neurons that are surrounded by PNNs (PV+PNN neurons) are more enriched with PV than PV neurons without PNNs (PV-PNN neurons) across both rodents and songbirds. Interestingly, the relationship between PV and PNN expression in the vocal portion of the basal ganglia of songbirds (Area X) differed from that in other areas, with PV+PNN neurons having lower PV expression compared to PV-PNN neurons. These relationships remained consistent across development in vocal motor circuits of the songbird brain. Finally, we discovered a causal contribution of PNNs to PV expression in songbirds because degradation of PNNs led to a diminution of PV expression in PV neurons. These findings reveal a conserved relationship between PV and PNN expression in sensory and motor cortices and across songbirds and rodents and suggest that PV neurons could modulate plasticity and neural dynamics in similar ways across songbirds and rodents.
Collapse
Affiliation(s)
- Angela S. Wang
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Xinghaoyun Wan
- Department of Biology, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | | | - Vivian Y. Li
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Gilles Cornez
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | - Jacques Balthazart
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | | | - Etienne de Villers-Sidani
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Jon T. Sakata
- Department of Biology, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, QC, Canada
| |
Collapse
|
45
|
Dong Y, Zhao K, Qin X, Du G, Gao L. The mechanisms of perineuronal net abnormalities in contributing aging and neurological diseases. Ageing Res Rev 2023; 92:102092. [PMID: 37839757 DOI: 10.1016/j.arr.2023.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
The perineuronal net (PNN) is a highly latticed extracellular matrix in the central nervous system, which is composed of hyaluronic acid, proteoglycan, hyaluronan and proteoglycan link protein (Hapln), and tenascin. PNN is predominantly distributed in GABAergic interneurons expressing Parvalbumin (PV) and plays a critical role in synaptic function, learning and memory, oxidative stress, and inflammation. In addition, PNN's structure and function are also modulated by a variety of factors, including protein tyrosine phosphatase σ (PTPσ), orthodenticle homeo-box 2 (Otx2), and erb-b2 receptor tyrosine kinase 4 (ErbB4). Glycosaminoglycan (GAG), a component of proteoglycan, also influences PNN through its sulfate mode. PNN undergoes abnormal changes during aging and in various neurological diseases, such as Alzheimer's disease, Parkinson's disease, schizophrenia, autism spectrum disorder, and multiple sclerosis. Nevertheless, there is limited report on the relationship between PNN and aging or age-related neurological diseases. This review elaborates on the mechanisms governing PNN regulation and summarizes how PNN abnormalities contribute to aging and neurological diseases, offering insights for potential treatments.
Collapse
Affiliation(s)
- Yixiao Dong
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Kunkun Zhao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China
| | - Guanhua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan, China.
| |
Collapse
|
46
|
Gray DT, Zempare M, Carey N, Khattab S, Sinakevitch I, De Biase LM, Barnes CA. Extracellular matrix proteoglycans support aged hippocampus networks: a potential cellular-level mechanism of brain reserve. Neurobiol Aging 2023; 131:52-58. [PMID: 37572527 PMCID: PMC10529564 DOI: 10.1016/j.neurobiolaging.2023.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
One hallmark of normative brain aging is vast heterogeneity in whether older people succumb to or resist cognitive decline. Resilience describes a brain's capacity to maintain cognition in the face of aging and disease. One factor influencing resilience is brain reserve-the status of neurobiological resources available to support neuronal circuits as dysfunction accumulates. This study uses a cohort of behaviorally characterized adult, middle-aged, and aged rats to test whether neurobiological factors that protect inhibitory neurotransmission and synapse function represent key components of brain reserve. Histochemical analysis of extracellular matrix proteoglycans, which play critical roles in stabilizing synapses and modulating inhibitory neuron excitability, was conducted alongside analyses of lipofuscin-associated autofluorescence. The findings indicate that aging results in lower proteoglycan density and more lipofuscin in CA3. Aged rats with higher proteoglycan density exhibited better performance on the Morris watermaze, whereas lipofuscin abundance was not related to spatial memory. These data suggest that the local environment around neurons may protect against synapse dysfunction or hyperexcitability and could contribute to brain reserve mechanisms.
Collapse
Affiliation(s)
- Daniel T Gray
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Marc Zempare
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Natalie Carey
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Salma Khattab
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Irina Sinakevitch
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Lindsay M De Biase
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Carol A Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA; Departments of Psychology, Neurology and Neuroscience, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
47
|
Perica MI, Luna B. Impact of stress on excitatory and inhibitory markers of adolescent cognitive critical period plasticity. Neurosci Biobehav Rev 2023; 153:105378. [PMID: 37643681 PMCID: PMC10591935 DOI: 10.1016/j.neubiorev.2023.105378] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Adolescence is a time of significant neurocognitive development. Prolonged maturation of prefrontal cortex (PFC) through adolescence has been found to support improvements in executive function. Changes in excitatory and inhibitory mechanisms of critical period plasticity have been found to be present in the PFC through adolescence, suggesting that environment may have a greater effect on development during this time. Stress is one factor known to affect neurodevelopment increasing risk for psychopathology. However, less is known about how stress experienced during adolescence could affect adolescent-specific critical period plasticity mechanisms and cognitive outcomes. In this review, we synthesize findings from human and animal literatures looking at the experience of stress during adolescence on cognition and frontal excitatory and inhibitory neural activity. Studies indicate enhancing effects of acute stress on cognition and excitation within specific contexts, while chronic stress generally dampens excitatory and inhibitory processes and impairs cognition. We propose a model of how stress could affect frontal critical period plasticity, thus potentially altering neurodevelopmental trajectories that could lead to risk for psychopathology.
Collapse
Affiliation(s)
- Maria I Perica
- Department of Psychology, University of Pittsburgh, PA, USA.
| | - Beatriz Luna
- Department of Psychology, University of Pittsburgh, PA, USA
| |
Collapse
|
48
|
Zhang Y, Guo Z, Yang L, Cheng C, Gai C, Gao Y, Zhang Y, Sun H, Hu D. Possible Involvement of Perineuronal Nets in Anti-Depressant Effects of Electroacupuncture in Chronic-Stress-Induced Depression in Rats. Neurochem Res 2023; 48:3146-3159. [PMID: 37347359 DOI: 10.1007/s11064-023-03970-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 05/29/2023] [Accepted: 06/12/2023] [Indexed: 06/23/2023]
Abstract
Acupuncture can alleviate depression-like behaviors. However, the neural mechanisms behind the anti-depressive effect remain unknown. Perineuronal net (PNN) abnormalities have been reported in multiple psychiatric disorders. This study investigated the modulation and neural mechanism of PNNs in the anti-depressant process of electroacupuncture (EA) at Baihui (GV20) and Yintang (GV29) points. A rat depression model was induced by chronic unpredicted mild stress (CUMS). The results revealed that CUMS, applied for four weeks, specifically reduces PNNs around parvalbumin (PV). In addition, EA and fluoxetine treatments reverse the decrease in PNNs+ cell density and the ratio of PV and PNN double-positive cells to PV+ neurons in the medial prefrontal cortex (mPFC) after CUMS. Furthermore, EA treatment can reverse the decrease in the protein expression of PNN components (aggrecan and brevican) in the mPFC caused by stress. After EA treatment, the decreased expression of GAD67, GLuA1, and PSD95 in the mPFC induced by CUMS for four weeks was also reversed. PNN degradation in mPFC brain areas potentially interferes with the anti-depressant benefits of EA in rats with depression induced by CUMS. EA treatment did not increase PNNs+ cell density and the ratio of PV and PNN double-positive cells to PV+ neurons after PNNs degradation in the mPFC brain region of rats. This finding indicated that the mechanism of acupuncture's anti-depressant effect may be based on reversing the CUMS-induced decline in PNN expression, the functional impairment of γ-aminobutyric acid (GABA) neurons, and the regulation of excitatory synaptic proteins expression.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyu Guo
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Luping Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Cuicui Cheng
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong Gai
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yushan Gao
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Hongmei Sun
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Die Hu
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
49
|
Wang AS, Wan X, Storch DS, Cornez G, Balthazart J, Cisneros-Franco JM, de Villers-Sidani E, Sakata JT. Cross-species conservation in the regulation of parvalbumin by perineuronal nets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.13.557580. [PMID: 37745532 PMCID: PMC10515890 DOI: 10.1101/2023.09.13.557580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Parvalbumin (PV) neurons play an integral role in regulating neural dynamics and plasticity. Therefore, understanding the factors that regulate PV expression is important for revealing modulators of brain function. While the contribution of PV neurons to neural processes has been studied in mammals, relatively little is known about PV function in non-mammalian species, and discerning similarities in the regulation of PV across species can provide insight into evolutionary conservation in the role of PV neurons. Here we investigated factors that affect the abundance of PV in PV neurons in sensory and motor circuits of songbirds and rodents. In particular, we examined the degree to which perineuronal nets (PNNs), extracellular matrices that preferentially surround PV neurons, modulate PV abundance as well as how the relationship between PV and PNN expression differs across brain areas and species and changes over development. We generally found that cortical PV neurons that are surrounded by PNNs (PV+PNN neurons) are more enriched with PV than PV neurons without PNNs (PV-PNN neurons) across both rodents and songbirds. Interestingly, the relationship between PV and PNN expression in the vocal portion of the basal ganglia of songbirds (Area X) differed from that in other areas, with PV+PNN neurons having lower PV expression compared to PV-PNN neurons. These relationships remained consistent across development in vocal motor circuits of the songbird brain. Finally, we discovered a causal contribution of PNNs to PV expression in songbirds because degradation of PNNs led to a diminution of PV expression in PV neurons. These findings in reveal a conserved relationship between PV and PNN expression in sensory and motor cortices and across songbirds and rodents and suggest that PV neurons could modulate plasticity and neural dynamics in similar ways across songbirds and rodents.
Collapse
Affiliation(s)
- Angela S. Wang
- Department of Biology, McGill University, Montreal, Canada
| | - Xinghaoyun Wan
- Department of Biology, McGill University, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | | | - Gilles Cornez
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | - Jacques Balthazart
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | | | - Etienne de Villers-Sidani
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, Canada
| | - Jon T. Sakata
- Department of Biology, McGill University, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, Canada
| |
Collapse
|
50
|
Druga R, Salaj M, Al-Redouan A. Parvalbumin - Positive Neurons in the Neocortex: A Review. Physiol Res 2023; 72:S173-S191. [PMID: 37565421 PMCID: PMC10660579 DOI: 10.33549/physiolres.935005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/02/2023] [Indexed: 12/01/2023] Open
Abstract
The calcium binding protein parvalbumin (PV) in the mammalian neocortex is expressed in a subpopulation of cortical GABAergic inhibitory interneurons. PV - producing interneurons represent the largest subpopulation of neocortical inhibitory cells, exhibit mutual chemical and electrical synaptic contacts and are well known to generate gamma oscillation. This review summarizes basic data of the distribution, afferent and efferent connections and physiological properties of parvalbumin expressing neurons in the neocortex. Basic data about participation of PV-positive neurons in cortical microcircuits are presented. Autaptic connections, metabolism and perineuronal nets (PNN) of PV positive neurons are also discussed.
Collapse
Affiliation(s)
- R Druga
- Department of Anatomy, 2nd Medical Faculty, Charles University Prague, Czech Republic.
| | | | | |
Collapse
|