1
|
Vasquez LS, Stack S, Taylor WW, Dias BG. Intergenerational Effects of Stress - A Focus on Learning and Memory. Curr Top Behav Neurosci 2025. [PMID: 40119217 DOI: 10.1007/7854_2025_578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Stress is a ubiquitous facet of life. Ranging in form (e.g., psychosocial, physical, nutritional, economic) and longevity (e.g., acute, chronic), stressors affect the biology of those directly in their line of attack. As is becoming increasingly appreciated, the pernicious effects of stress echo across generations (Dias et al. 2015; Yehuda and Lehrner 2018; Jawaid et al. 2021; Dion et al. 2022; Zhou and Ryan 2023; Dias 2024). With a focus on learning and memory, this chapter addresses how stressors derail learning and memory in the generation directly exposed to them andin future generations. To do so, with a specific emphasis on associative fear conditioning in humans and rodents, we touch upon the relevance of extinction training in the aftermath of such conditioning and the recall of such extinction training as windows into normative and disrupted learning. Next, we briefly discuss underlying neuroanatomical substrates mediating these processes. We then draw attention to influences of postnatal, in utero, and pre-conceptional stress on learning and memory across generations. Finally, we briefly outline biological factors that underlie how learning and memory is derailed by these stressors.
Collapse
Affiliation(s)
- L S Vasquez
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - S Stack
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - W W Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - B G Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA.
- Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
2
|
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
|
3
|
Mueller-Buehl C, Pakusch J, Bader V, Winklhofer KF, Mark MD, Faissner A. Combined loss of brevican, neurocan, tenascin-C and tenascin-R leads to impaired fear retrieval due to perineuronal net loss. Sci Rep 2025; 15:5528. [PMID: 39953103 PMCID: PMC11828866 DOI: 10.1038/s41598-025-89580-2] [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: 10/22/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
In conditions such as neurodegenerative diseases, posttraumatic stress disorder (PTSD), addiction and spinal cord injuries, restricted synaptic plasticity hinders the formation of new neuronal connections, preventing the compensation and treatment of adverse behaviors. Perineuronal nets (PNNs) significantly restrict synaptic plasticity by inhibiting synapse formation. The digestion of PNNs has been associated with short-term cognitive improvements and reduced long-term memory, offering potential therapeutic benefits in PTSD. This study investigates the correlation between PNNs and fear memory processes in extracellular matrix (ECM) mutant mice, particularly focusing on the amygdala-medial prefrontal cortex (mPFC) circuit, which is crucial for fear memory generation and maintenance. Fear conditioning was conducted on mice lacking four key ECM-molecules: brevican, neurocan, tenascin-C and tenascin-R (4x KO). These mice exhibited severe impairments in memory consolidation, as evident by their inability to retrieve previously learned fear memories, coupled with reduced PNN density and disturbed synaptic integrity along their PNNs. Additionally, changes in neural activity in the basolateral amygdala (BL) and reductions in VGAT+ synaptic puncta in the amygdala-mPFC circuit were observed. In contrast, tenascin single KOs showed intact fear behavior and memory compared to their control groups. Impaired fear memory consolidation can be advantageous in certain conditions, such as PTSD, making the 4x KO mice an intriguing model for future fear conditioning studies and highlighting brevican, neurocan, Tnc, and Tnr as compelling targets for further investigation. This study underscores the significance of ECM regulation for synaptic organization and the potential of PNN modulation as a therapeutic target for fear memory-related conditions.
Collapse
Affiliation(s)
- Cornelius Mueller-Buehl
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Johanna Pakusch
- Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, D- 44780, Bochum, Germany
| | - Verian Bader
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, D-44780, Bochum, Germany
- Department Biochemistry of Neurodegenerative Diseases, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, D-44780, Bochum, Germany
| | - Konstanze F Winklhofer
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, D-44780, Bochum, Germany
- Cluster of Excellence RESOLV, D-44780, Bochum, Germany
| | - Melanie D Mark
- Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, D- 44780, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, D-44780, Bochum, Germany.
| |
Collapse
|
4
|
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
|
5
|
Zhang Q, Zhang Y, Cong P, Wu Q, Wan H, Huang X, Li X, Li Z, Li J, Wu H, Tian L, Xiong L. Connexin 43 contributes to perioperative neurocognitive disorder by attenuating perineuronal net of hippocampus in aged mice. Cell Mol Life Sci 2025; 82:37. [PMID: 39762568 PMCID: PMC11703800 DOI: 10.1007/s00018-024-05530-7] [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: 08/21/2024] [Revised: 11/12/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Perioperative neurocognitive disorder (PND) is a prevalent form of cognitive impairment in elderly patients following anesthesia and surgery. The underlying mechanisms of PND are closely related to perineuronal nets (PNNs). PNNs, which are complexes of extracellular matrix primarily surrounding neurons in the hippocampus, play a critical role in neurocognitive function. Connexin 43 (Cx43) contributes to cognitive function by modulating the components of PNNs. This study was designed to investigate the specific regulatory mechanisms of Cx43 on PNNs and its pivotal role in the development of PND. METHODS Eighteen-month-old wild-type and Gja1fl/fl C57BL/6 mice were subjected to abdominal surgery under 1.4% isoflurane anesthesia. Cognitive functions, particularly learning and memory, were evaluated via the Y-maze test, Barnes maze (BM) and contextual fear conditioning test (CFT). The mRNA and protein expression levels of Cx43 were assessed by using quantitative reverse transcription polymerase chain reaction (qRT-PCR), fluorescent in situ hybridization (FISH), western blotting and flow cytometry. The quantity of PNNs was measured by Wisteria floribunda agglutinin (WFA) and Aggrecan staining. RESULTS Aged mice subjected to anesthesia and surgery exhibited deficits in hippocampus-dependent cognitive functions, which were accompanied by increased Cx43 mRNA and protein expression. Conditional knockout (cKO) of Cx43 in astrocytes alleviated cognitive deficits and promoted the number of PNNs and dendritic spines in the hippocampus by targeting Dmp1. Knockdown of Dmp1 attenuated the beneficial effects of Cx43 cKO on cognitive deficits induced by anesthesia and surgery. CONCLUSION Our findings indicate that anesthesia and surgery induce an increase in Cx43 expression, which inhibits the formation of PNNs and dendritic spines in hippocampus by suppressing Dmp1 transcription, leading to cognitive deficits in aged mice. These results offer new mechanistic insights into the pathogenesis of PND and identify potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Qian Zhang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Yuxin Zhang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Peilin Cong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Qianqian Wu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Hanxi Wan
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Xinwei Huang
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Xinyang Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Zhouxiang Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Jingxuan Li
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China
| | - Huanghui Wu
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China.
| | - Li Tian
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China.
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Department of Anesthesiology and Perioperative MedicineSchool of Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1239 Sanmen Road, Hongkou District, Shanghai, 200434, China.
| |
Collapse
|
6
|
Banovac I, Prkačin MV, Kirchbaum I, Trnski-Levak S, Bobić-Rasonja M, Sedmak G, Petanjek Z, Jovanov-Milosevic N. Morphological and Molecular Characteristics of Perineuronal Nets in the Human Prefrontal Cortex-A Possible Link to Microcircuitry Specialization. Mol Neurobiol 2025; 62:1094-1111. [PMID: 38958887 PMCID: PMC11711633 DOI: 10.1007/s12035-024-04306-1] [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: 02/15/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Perineuronal nets (PNNs) are a type of extracellular matrix (ECM) that play a significant role in synaptic activity and plasticity of interneurons in health and disease. We researched PNNs' regional and laminar representation and molecular composition using immunohistochemistry and transcriptome analysis of Brodmann areas (BA) 9, 14r, and 24 in 25 human postmortem brains aged 13-82 years. The numbers of VCAN- and NCAN-expressing PNNs, relative to the total number of neurons, were highest in cortical layers I and VI while WFA-binding (WFA+) PNNs were most abundant in layers III-V. The ECM glycosylation pattern was the most pronounced regional difference, shown by a significantly lower proportion of WFA+ PNNs in BA24 (3.27 ± 0.69%) compared to BA9 (6.32 ± 1.73%; P = 0.0449) and BA14 (5.64 ± 0.71%; P = 0.0278). The transcriptome of late developmental and mature stages revealed a relatively stable expression of PNN-related transcripts (log2-transformed expression values: 6.5-8.5 for VCAN and 8.0-9.5 for NCAN). Finally, we propose a classification of PNNs that envelop GABAergic neurons in the human cortex. The significant differences in PNNs' morphology, distribution, and molecular composition strongly suggest an involvement of PNNs in specifying distinct microcircuits in particular cortical regions and layers.
Collapse
Affiliation(s)
- Ivan Banovac
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Matija Vid Prkačin
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Ivona Kirchbaum
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Sara Trnski-Levak
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Mihaela Bobić-Rasonja
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Department of Biology, University of Zagreb School of Medicine, Šalata 3, HR-10000, Zagreb, Croatia
| | - Goran Sedmak
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Natasa Jovanov-Milosevic
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia.
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia.
- Department of Biology, University of Zagreb School of Medicine, Šalata 3, HR-10000, Zagreb, Croatia.
| |
Collapse
|
7
|
Whitten CJ, Kelly JR, Gillespie AL, Brooks HJB, Hooker MK, Temple AR, Hennon LM, Kilgore BJ, Singamaneni YB, Schulz KM, Burghardt GM, Cooper MA. From Play Date to Stress Fate: Juvenile Social Play Rescues Stress-Induced Changes in Adult Social Behavior. Dev Psychobiol 2025; 67:e70020. [PMID: 39878574 DOI: 10.1002/dev.70020] [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: 08/21/2024] [Revised: 12/09/2024] [Accepted: 01/05/2025] [Indexed: 01/31/2025]
Abstract
Long-term effects of social play on neural and behavioral development remain unclear. We investigated whether just 1 h of juvenile social play could rescue the effects of play deprivation on stress-related behavior and markers of neural plasticity. Syrian hamsters were reared from postnatal days 21-43 in three conditions: peer isolation, peer isolation with daily social play sessions (dyadic play), or group-housed with littermates. In adulthood, subjects were exposed to acute social defeat stress, and we examined changes in perineuronal net (PNN) expression surrounding parvalbumin (PV) neurons in the prelimbic (PL), infralimbic (IL), and basolateral amygdala (BLA). Peer deprivation led to exaggerated submissive and defensive behavior in a conditioned defeat test, but 1 h of dyadic play rescued the heightened conditioned defeat response in both males and females. In females, play deprivation reduced PNN/PV coexpression in the PL and IL compared to control groups with opportunities for social play. Males exposed to peer isolation showed elevated agonistic behavior when returned to their littermates compared to males exposed to 1-h play encounters. These findings indicate juvenile social play has long-lasting effects on PNN expression surrounding PV cells in the medial prefrontal cortex, which allows for the development of species' typical agonistic behavior and greater stress resistance in adulthood. The ability of just 1 h of social play to rescue the effects of peer isolation highlights the powerful role of social interactions in neural and behavioral development.
Collapse
Affiliation(s)
- Conner J Whitten
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Jeffrey R Kelly
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Alex L Gillespie
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Heather J B Brooks
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Mackenzie K Hooker
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Anna R Temple
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Leila M Hennon
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Benjamin J Kilgore
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Yaswanth B Singamaneni
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Kalynn M Schulz
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Gordon M Burghardt
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| | - Matthew A Cooper
- Department of Psychology, The University of Tennessee Knoxville, Knoxville, Tennessee, USA
| |
Collapse
|
8
|
Shaughnessy EK, Horne BW, Huhman KL. Perineuronal Nets in Syrian Hamsters: Anatomical Localization, Sex Differences, Diurnal Variation, and Response to Social Stress. Brain Behav 2024; 14:e70189. [PMID: 39711016 DOI: 10.1002/brb3.70189] [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: 12/13/2023] [Revised: 11/01/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
PURPOSE Perineuronal nets (PNNs) are extracellular matrix proteoglycans surrounding neurons and glia. It has been suggested that PNNs are involved in the pathophysiology of multiple CNS illnesses, including stress-related neuropsychiatric disorders like schizophrenia, major depressive disorder, and anxiety disorders. METHOD Before examining the putative role of PNNs in stress-related responses, we described for the first time the anatomical distribution in Syrian hamsters (Mesocricetus auratus), an excellent model organism for studying social stress and circadian rhythms. RESULTS We observed PNNs throughout the hamster cortex and hippocampus but found low to no expression in subcortical regions such as the hypothalamus, thalamus, and striatum, sites where they are observed in rats and mice. We further demonstrated that PNNs are dynamically regulated in a sex-dependent manner in response to acute social stress, specifically in hippocampal area CA1. We did not observe a difference in PNNs between the beginning of the dark versus light phase of the light-dark cycle in hamsters, despite other laboratory rodents showing diurnal variation in PNNs. Finally, we also demonstrated that there are sex differences in PNN expression in the somatosensory cortex and the basolateral amygdala in hamsters, suggesting that sex as a biological variable should be considered in studies of PNN function. CONCLUSION Together, the data from the current study suggest that a comparative approach will be necessary to fully elucidate the functional role of PNNs and, further, that Syrian hamsters are a valuable model in this endeavor.
Collapse
Affiliation(s)
| | - Benjamin W Horne
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Kim L Huhman
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Dargam S, de Olmos S, Marcos Pautassi R, Lorenzo A. Footshock drives remodeling of perineuronal nets in retrosplenial cortex during contextual fear memory formation. Neurobiol Learn Mem 2024; 215:107990. [PMID: 39401564 DOI: 10.1016/j.nlm.2024.107990] [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: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
The retrosplenial cortex (RSC) plays a critical role in complex cognitive functions such as contextual fear memory formation and consolidation. Perineuronal nets (PNNs) are specialized structures of the extracellular matrix that modulate synaptic plasticity by enwrapping the soma, proximal neurites and synapsis mainly on fast spiking inhibitory GABAergic interneurons that express parvalbumin (PV). PNNs change after contextual fear conditioning (CFC) in amygdala or hippocampus, yet it is unknown if similar remodeling takes place at RSC. Here, we used Wisteria floribunda agglutinin (WFA), a ubiquitous marker of PNNs, to study the remodeling of PNNs in RSC during the acquisition or retrieval of contextual fear conditioning (CFC). Adult male mice were exposed to paired presentations of a context and footshock, or to either of these stimuli alone (control groups). The mere exposure of animals to the footshock, either alone or paired with the context, evoked a significant expansion of PNNs, both in the number of WFA positive neurons and in the area occupied by WFA staining, across the entire RSC. This was not associated with c-Fos expression in RSC nor correlated with c-Fos expression in individual PNNs-expressing neurons in RSC, suggesting that PNNs remodeling is triggered by inputs external to the RSC. We also found that PNNs remodeling was independent of the level of PV expression. Notably, PNNs in RSC remained expanded long-after CFC. These results suggest that, in male mice, the threatening experience is the main cause of PNNs remodeling in the RSC.
Collapse
Affiliation(s)
- Salome Dargam
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - Soledad de Olmos
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - Ricardo Marcos Pautassi
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - Alfredo Lorenzo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.
| |
Collapse
|
10
|
Hazlett MF, Hall VL, Patel E, Halvorsen A, Calakos N, West AE. The Perineuronal Net Protein Brevican Acts in Nucleus Accumbens Parvalbumin-Expressing Interneurons of Adult Mice to Regulate Excitatory Synaptic Inputs and Motivated Behaviors. Biol Psychiatry 2024; 96:694-707. [PMID: 38346480 PMCID: PMC11315813 DOI: 10.1016/j.biopsych.2024.02.003] [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: 08/28/2023] [Revised: 01/13/2024] [Accepted: 02/07/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Experience-dependent functional adaptation of nucleus accumbens (NAc) circuitry underlies the development and expression of reward-motivated behaviors. Parvalbumin-expressing GABAergic (gamma-aminobutyric acidergic) interneurons (PVINs) within the NAc are required for this process. Perineuronal nets (PNNs) are extracellular matrix structures enriched around PVINs that arise during development and have been proposed to mediate brain circuit stability. However, their function in the adult NAc is largely unknown. Here, we studied the developmental emergence and adult regulation of PNNs in the NAc of male and female mice and examined the cellular and behavioral consequences of reducing the PNN component brevican in NAc PVINs. METHODS We characterized the expression of PNN components in mouse NAc using immunofluorescence and RNA in situ hybridization. We lowered brevican in NAc PVINs of adult mice using an intersectional viral and genetic method and quantified the effects on synaptic inputs to NAc PVINs and reward-motivated learning. RESULTS PNNs around NAc PVINs were developmentally regulated and appeared during adolescence. In the adult NAc, PVIN PNNs were also dynamically regulated by cocaine. Transcription of the gene that encodes brevican was regulated in a cell type- and isoform-specific manner in the NAc, with the membrane-tethered form of brevican being highly enriched in PVINs. Lowering brevican in NAc PVINs of adult mice decreased their excitatory inputs and enhanced both short-term novel object recognition and cocaine-induced conditioned place preference. CONCLUSIONS Regulation of brevican in NAc PVINs of adult mice modulates their excitatory synaptic drive and sets experience thresholds for the development of motivated behaviors driven by rewarding stimuli.
Collapse
Affiliation(s)
- Mariah F Hazlett
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Victoria L Hall
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Esha Patel
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Aaron Halvorsen
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Nicole Calakos
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina; Department of Neurology, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina; Duke Institute for Brain Sciences, Duke University Medical Center, Durham, North Carolina
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina.
| |
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. 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
|
12
|
Gao JH, Liu YY, Xu HX, Wu K, Zhang LL, Cheng P, Peng XH, Cao JL, Hua R, Zhang YM. Divergent input patterns to the central lateral amygdala play a duet in fear memory formation. iScience 2024; 27:110886. [PMID: 39319272 PMCID: PMC11421289 DOI: 10.1016/j.isci.2024.110886] [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: 01/15/2024] [Revised: 04/18/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
Somatostatin (SOM)-expressing neurons in the central lateral amygdala (CeL) are responsible for fear memory learning, but the circuit and molecular mechanisms underlying this biology remain elusive. Here, we found that glutamatergic neurons in the lateral parabrachial nucleus (LPB) directly dominated the activity of CeLSOM neurons, and that selectively inhibiting the LPBGlu→CeLSOM pathway suppressed fear memory acquisition. By contrast, inhibiting CeL-projecting glutamatergic neurons in the paraventricular thalamic nucleus (PVT) interfered with consolidation-related processes. Notably, CeLSOM-innervating neurons in the LPB were modulated by presynaptic cannabinoid receptor 1 (CB1R), and knock down of CB1Rs in LPB glutamatergic neurons enhanced excitatory transmission to the CeL and partially rescued the impairment in fear memory induced by CB1R activation in the CeL. Overall, our study reveals the mechanisms by which CeLSOM neurons mediate the formation of fear memories during fear conditioning in mice, which may provide a new direction for the clinical research of fear-related disorders.
Collapse
Affiliation(s)
- Jing-Hua Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Department of Anesthesiology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng 224008, Jiangsu, China
| | - Yue-Ying Liu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Hui-Xiang Xu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Ke Wu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Le-le Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Peng Cheng
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Xiao-Han Peng
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Jun-Li Cao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Rong Hua
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yong-Mei Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
13
|
Tziridis K, Maul A, Rasheed J, Krauss P, Schilling A, Schulze H. Tinnitus is associated with increased extracellular matrix density in the auditory cortex of Mongolian gerbils. BMC Neurosci 2024; 25:52. [PMID: 39420272 PMCID: PMC11484117 DOI: 10.1186/s12868-024-00904-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Most scientists agree that subjective tinnitus is the pathological result of an interaction of damage to the peripheral auditory system and central neuroplastic adaptations. Here we investigate such tinnitus related adaptations in the primary auditory cortex (AC) 7 and 13 days after noise trauma induction of tinnitus by quantifying the density of the extracellular matrix (ECM) in the AC of Mongolian gerbils (Meriones unguiculatus). The ECM density has been shown to be relevant for neuroplastic processes and synaptic stability within the cortex. We utilized a mild monaural acoustic noise trauma in overall 22 gerbils to induce tinnitus and a sham exposure in 16 control (C) animals. Tinnitus was assessed by a behavioral response paradigm. Animals were separated for a presence (T) or absence (NT) of a tinnitus percept by a behavioral task. The ECM density 7 and 13 days after trauma was quantified using immunofluorescence luminance of Wisteria floribunda lectin-fluoresceine-5-isothiocyanate (WFA-FITC) on histological slices of the primary AC, relative to the non-auditory brainstem as a reference area. At both timepoints, we found that the WFA-FITC luminance of the AC of NT animals was not significantly different from that of C animals. However, we found a significant increase of luminance in T animals' ACs compared to NT or C animals' cortices. This effect was found exclusively on the AC side contralateral to the trauma ear. These results point to a hemisphere specific process of stabilization of synaptic connections in primary AC, which may be involved in the chronic manifestation of tinnitus.
Collapse
Affiliation(s)
- Konstantin Tziridis
- Experimental Otolaryngology, Head and Neck Surgery, University Hospital Erlangen, ENT Hospital, Waldstrasse 1, 91054, Erlangen, Germany.
| | - Antonia Maul
- Experimental Otolaryngology, Head and Neck Surgery, University Hospital Erlangen, ENT Hospital, Waldstrasse 1, 91054, Erlangen, Germany
| | - Jwan Rasheed
- Experimental Otolaryngology, Head and Neck Surgery, University Hospital Erlangen, ENT Hospital, Waldstrasse 1, 91054, Erlangen, Germany
| | - Patrick Krauss
- Experimental Otolaryngology, Head and Neck Surgery, University Hospital Erlangen, ENT Hospital, Waldstrasse 1, 91054, Erlangen, Germany
- Friedrich-Alexander University Erlangen-Nürnberg, CCN group, pattern recognition lab, Immerwahrstrasse 2A, 91058, Erlangen, Germany
| | - Achim Schilling
- Experimental Otolaryngology, Head and Neck Surgery, University Hospital Erlangen, ENT Hospital, Waldstrasse 1, 91054, Erlangen, Germany
- Friedrich-Alexander University Erlangen-Nürnberg, CCN group, pattern recognition lab, Immerwahrstrasse 2A, 91058, Erlangen, Germany
| | - Holger Schulze
- Experimental Otolaryngology, Head and Neck Surgery, University Hospital Erlangen, ENT Hospital, Waldstrasse 1, 91054, Erlangen, Germany
| |
Collapse
|
14
|
Huang Z, Wei X, Tian J, Fu Y, Dong J, Wang Y, Shi J, Lu L, Zhang W. A disinhibitory microcircuit of the orbitofrontal cortex mediates cocaine preference in mice. Mol Psychiatry 2024; 29:3160-3169. [PMID: 38698268 DOI: 10.1038/s41380-024-02579-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/05/2024]
Abstract
Both clinical and animal studies showed that the impaired functions of the orbitofrontal cortex (OFC) underlie the compulsive drug-seeking behavior of drug addiction. However, the functional changes of the microcircuit in the OFC and the underlying molecular mechanisms in drug addiction remain elusive, and little is known for whether microcircuits in the OFC contributed to drug addiction-related behaviors. Utilizing the cocaine-induced conditioned-place preference model, we found that the malfunction of the microcircuit led to disinhibition in the OFC after cocaine withdrawal. We further showed that enhanced Somatostatin-Parvalbumin (SST-PV) inhibitory synapse strength changed microcircuit function, and SST and PV inhibitory neurons showed opposite contributions to the drug addiction-related behavior of mice. Brevican of the perineuronal nets of PV neurons regulated SST-PV synapse strength, and the knockdown of Brevican alleviated cocaine preference. These results reveal a novel molecular mechanism of the regulation of microcircuit function and a novel circuit mechanism of the OFC in gating cocaine preference.
Collapse
Affiliation(s)
- Ziran Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Xiaoyan Wei
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jing Tian
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Yangxue Fu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jihui Dong
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Yihui Wang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital); Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100191, China
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China.
| |
Collapse
|
15
|
Sanchez B, Kraszewski P, Lee S, Cope EC. From molecules to behavior: Implications for perineuronal net remodeling in learning and memory. J Neurochem 2024; 168:1854-1876. [PMID: 38158878 DOI: 10.1111/jnc.16036] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Perineuronal nets (PNNs) are condensed extracellular matrix (ECM) structures found throughout the central nervous system that regulate plasticity. They consist of a heterogeneous mix of ECM components that form lattice-like structures enwrapping the cell body and proximal dendrites of particular neurons. During development, accumulating research has shown that the closure of various critical periods of plasticity is strongly linked to experience-driven PNN formation and maturation. PNNs provide an interface for synaptic contacts within the holes of the structure, generally promoting synaptic stabilization and restricting the formation of new synaptic connections in the adult brain. In this way, they impact both synaptic structure and function, ultimately influencing higher cognitive processes. PNNs are highly plastic structures, changing their composition and distribution throughout life and in response to various experiences and memory disorders, thus serving as a substrate for experience- and disease-dependent cognitive function. In this review, we delve into the proposed mechanisms by which PNNs shape plasticity and memory function, highlighting the potential impact of their structural components, overall architecture, and dynamic remodeling on functional outcomes in health and disease.
Collapse
Affiliation(s)
- Brenda Sanchez
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| | - Piotr Kraszewski
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| | - Sabrina Lee
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| | - Elise C Cope
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| |
Collapse
|
16
|
Chelini G, Mirzapourdelavar H, Durning P, Baidoe-Ansah D, Sethi MK, O'Donovan SM, Klengel T, Balasco L, Berciu C, Boyer-Boiteau A, McCullumsmith R, Ressler KJ, Zaia J, Bozzi Y, Dityatev A, Berretta S. Focal clusters of peri-synaptic matrix contribute to activity-dependent plasticity and memory in mice. Cell Rep 2024; 43:114112. [PMID: 38676925 PMCID: PMC11251421 DOI: 10.1016/j.celrep.2024.114112] [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: 07/10/2023] [Revised: 11/09/2023] [Accepted: 03/28/2024] [Indexed: 04/29/2024] Open
Abstract
Recent findings show that effective integration of novel information in the brain requires coordinated processes of homo- and heterosynaptic plasticity. In this work, we hypothesize that activity-dependent remodeling of the peri-synaptic extracellular matrix (ECM) contributes to these processes. We show that clusters of the peri-synaptic ECM, recognized by CS56 antibody, emerge in response to sensory stimuli, showing temporal and spatial coincidence with dendritic spine plasticity. Using CS56 co-immunoprecipitation of synaptosomal proteins, we identify several molecules involved in Ca2+ signaling, vesicle cycling, and AMPA-receptor exocytosis, thus suggesting a role in long-term potentiation (LTP). Finally, we show that, in the CA1 hippocampal region, the attenuation of CS56 glycoepitopes, through the depletion of versican as one of its main carriers, impairs LTP and object location memory in mice. These findings show that activity-dependent remodeling of the peri-synaptic ECM regulates the induction and consolidation of LTP, contributing to hippocampal-dependent memory.
Collapse
Affiliation(s)
- Gabriele Chelini
- Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; Center for Mind/Brain Sciences, University of Trento, Rovereto 38068 Trento, Italy
| | - Hadi Mirzapourdelavar
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases, Magdeburg 39120 Saxony-Anhalt, Germany
| | - Peter Durning
- Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA 02478, USA
| | - David Baidoe-Ansah
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases, Magdeburg 39120 Saxony-Anhalt, Germany
| | - Manveen K Sethi
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Sinead M O'Donovan
- Cognitive Disorders Research Laboratory, University of Toledo, Toledo, OH 43606, USA
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; Translational Molecular Genomics Laboratory, Mclean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Luigi Balasco
- Center for Mind/Brain Sciences, University of Trento, Rovereto 38068 Trento, Italy
| | - Cristina Berciu
- Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA 02478, USA
| | - Anne Boyer-Boiteau
- Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA 02478, USA
| | - Robert McCullumsmith
- Cognitive Disorders Research Laboratory, University of Toledo, Toledo, OH 43606, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA; Neurobiology of Fear Laboratory, McLean Hospital, Belmont, MA 02478, USA
| | - Joseph Zaia
- Center for Biomedical Mass Spectrometry, Department of Biochemistry and Cell Biology, Boston University School of Medicine, Boston, MA 02118, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Yuri Bozzi
- Center for Mind/Brain Sciences, University of Trento, Rovereto 38068 Trento, Italy; CNR Neuroscience Institute Pisa, 56124 Pisa, Italy
| | - Alexander Dityatev
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases, Magdeburg 39120 Saxony-Anhalt, Germany; Medical Faculty, Otto von Guericke University, Magdeburg 39106 Saxony-Anhalt, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, Magdeburg 39106 Saxony-Anhalt, Germany
| | - Sabina Berretta
- Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
17
|
Schreurs BG, O'Dell DE, Wang D. The Role of Cerebellar Intrinsic Neuronal Excitability, Synaptic Plasticity, and Perineuronal Nets in Eyeblink Conditioning. BIOLOGY 2024; 13:200. [PMID: 38534469 DOI: 10.3390/biology13030200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
Evidence is strong that, in addition to fine motor control, there is an important role for the cerebellum in cognition and emotion. The deep nuclei of the mammalian cerebellum also contain the highest density of perineural nets-mesh-like structures that surround neurons-in the brain, and it appears there may be a connection between these nets and cognitive processes, particularly learning and memory. Here, we review how the cerebellum is involved in eyeblink conditioning-a particularly well-understood form of learning and memory-and focus on the role of perineuronal nets in intrinsic membrane excitability and synaptic plasticity that underlie eyeblink conditioning. We explore the development and role of perineuronal nets and the in vivo and in vitro evidence that manipulations of the perineuronal net in the deep cerebellar nuclei affect eyeblink conditioning. Together, these findings provide evidence of an important role for perineuronal net in learning and memory.
Collapse
Affiliation(s)
- Bernard G Schreurs
- Department of Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| | - Deidre E O'Dell
- Department of Biology, Earth and Environmental Sciences, Pennsylvania Western (PennWest) University, California, PA 15419, USA
| | - Desheng Wang
- Department of Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
18
|
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
|
19
|
Teng Y, Niu J, Liu Y, Wang H, Chen J, Kong Y, Wang L, Lian B, Wang W, Sun H, Yue K. Ketamine alleviates fear memory and spatial cognition deficits in a PTSD rat model via the BDNF signaling pathway of the hippocampus and amygdala. Behav Brain Res 2024; 459:114792. [PMID: 38048914 DOI: 10.1016/j.bbr.2023.114792] [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: 10/07/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is associated with traumatic stress experiences. This condition can be accompanied by learning and cognitive deficits. Studies have demonstrated that ketamine can rapidly and significantly alleviate symptoms in patients with chronic PTSD. Nonetheless, the effects of ketamine on neurocognitive impairment and its mechanism of action in PTSD remain unclear. METHODS In this study, different concentrations of ketamine (5, 10, 15, and 20 mg/kg, i.p.) were evaluated in rat models of single prolonged stress and electrophonic shock (SPS&S). Expression levels of brain-derived neurotrophic factor (BDNF) and post-synaptic density-95 (PSD-95) in the hippocampus (HIP) and amygdala (AMG) were determined by Western blot analysis and immunohistochemistry. RESULTS The data showed that rats subjected to SPS&S exhibited significant PTSD-like cognitive impairment. The effect of ketamine on SPS&S-induced neurocognitive function showed a U-shaped dose effect in rats. A single administration of ketamine at a dosage of 10-15 mg/kg resulted in significant changes in behavioral outcomes. These manifestations of improvement in cognitive function and molecular changes were reversed at high doses (15-20 mg/kg). CONCLUSION Overall, ketamine reversed SPS&S-induced fear and spatial memory impairment and the down-regulation of BDNF and BDNF-related PSD-95 signaling in the HIP and AMG. A dose equal to 15 mg/kg rapidly reversed the behavioral and molecular changes and promoted the amelioration of cognitive dysfunction. The enhanced association of BDNF signaling with PSD-95 effects could be involved in the therapeutic efficiency of ketamine for PTSD.
Collapse
Affiliation(s)
- Yue Teng
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - JiaYao Niu
- School of Clinical Medicine, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yang Liu
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Han Wang
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - JinHong Chen
- School of Continuing Education, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - YuJia Kong
- School of Public Health, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Ling Wang
- Clinical Competency Training Center, Medical experiment and training center, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Bo Lian
- Department of Bioscience and Technology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - WeiWen Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100864, PR China
| | - HongWei Sun
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China.
| | - KuiTao Yue
- The Medical imaging Center, Affiliated Hospital of Weifang Medical University, 2428# Yuhe Road, Weifang, Shandong 261053, PR China.
| |
Collapse
|
20
|
Hu R, He K, Chen B, Chen Y, Zhang J, Wu X, Shi M, Wu L, Ma R. Electroacupuncture promotes the repair of the damaged spinal cord in mice by mediating neurocan-perineuronal net. CNS Neurosci Ther 2024; 30:e14468. [PMID: 37950551 PMCID: PMC10805400 DOI: 10.1111/cns.14468] [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: 06/28/2023] [Revised: 08/06/2023] [Accepted: 08/29/2023] [Indexed: 11/12/2023] Open
Abstract
AIMS This study aimed to investigate the effect of perineuronal net (PNN) and neurocan (NCAN) on spinal inhibitory parvalbumin interneuron (PV-IN), and the mechanism of electroacupuncture (EA) in promoting spinal cord injury (SCI) repair through neurocan in PNN. METHODS A mouse model of SCI was established. Sham-operated mice or SCI model mice were treated with chondroitin sulfate ABC (ChABC) enzyme or control vehicle for 2 weeks (i.e., sham+veh group, sham+ChABC group, SCI+veh group, and SCI+ChABC group, respectively), and then spinal cord tissues were taken from the T10 lesion epicenter for RNA sequencing (RNA-seq). MSigDB Hallmark and C5 databases for functional analysis, analysis strategies such as differential expression gene analysis (DEG), Kyoto Encyclopedia of Genes and Genomes (KEGG), gene set enrichment analysis (GSEA), and protein-protein interaction (PPI). According to the results of RNA-seq analysis, the expression of NCAN was knocked down or overexpressed by virus intervention, or/and EA intervention. Polymerase chain reaction (PCR), immunofluorescence, western blot, electrophysiological, and behavioral tests were performed. RESULTS After the successful establishment of SCI model, the motor dysfunction of lower limbs, and the expression of PNN core glycan protein at the epicenter of SCI were reduced. RNA-seq and PCR showed that PNN core proteoglycans except NCAN showed the same expression trend in normal and injured spinal cord treated with ChABC. KEGG and GSEA showed that PNN is mainly associated with inhibitory GABA neuronal function in injured spinal cord tissue, and PPI showed that NCAN in PNN can be associated with inhibitory neuronal function through parvalbumin (PV). Calcium imaging showed that local parvalbumin interneuron (PV-IN) activity decreased after PNN destruction, whether due to ChABC treatment or surgical bruising of the spinal cord. Overexpression of neurocan in injured spinal cord can enhance local PV-IN activity. PCR and western blot suggested that overexpression or knockdown of neurocan could up-regulate or down-regulate the expression of GAD. At the same time, the activity of PV-IN in the primary motor cortex (M1) and the primary sensory cortex of lower (S1HL) extremity changed synchronously. In addition, overexpression of neurocan improved the electrical activity of the lower limb and promoted functional repair of the paralyzed hind limb. EA intervention reversed the down-regulation of neurocan, enhanced the expression of PNN in the lesioned area, M1 and S1HL. CONCLUSION Neurocan in PNN can regulate the activity of PV-IN, and EA can promote functional recovery of mice with SCI by upregulating neurocan expression in PNN.
Collapse
Affiliation(s)
- Rong Hu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Kelin He
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
- Department of Acupuncture and MoxibustionThird Affiliated Hospital of Zhejiang Chinese Medical UniversityZhejiangChina
| | - Bowen Chen
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Yi Chen
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Jieqi Zhang
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Xingying Wu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Mengting Shi
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Lei Wu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
- Department of Acupuncture and MoxibustionThird Affiliated Hospital of Zhejiang Chinese Medical UniversityZhejiangChina
| | - Ruijie Ma
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
- Department of Acupuncture and MoxibustionThird Affiliated Hospital of Zhejiang Chinese Medical UniversityZhejiangChina
| |
Collapse
|
21
|
Yuan S, Shi J, Tang X, Deng B, Wu Z, Qiu B, Lin S, Ji C, Wang L, Cui S, Xu N, Yao L. The Role of Perineuronal Nets in the Contralateral Hemisphere in the Electroacupuncture-Mediated Rehabilitation of Poststroke Dysphagia Mice. eNeuro 2023; 10:ENEURO.0234-23.2023. [PMID: 37977825 DOI: 10.1523/eneuro.0234-23.2023] [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: 07/05/2023] [Revised: 10/09/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Acupuncture at Lianquan (CV23) acupoint has been shown to improve swallowing function in poststroke dysphagia (PSD). This improvement is supposed to be associated with the regulation of neuronal activity in the contralateral primary motor cortex (M1), while the underlying mechanism still needs to be elucidated. Perineuronal nets (PNNs) are well-known to be involved in the regulation of neuronal activity. Thus, we here aimed to detect the role of PNNs in the contralateral M1 hemisphere in the electroacupuncture (EA)-mediated effect in male mice. The results were obtained from a combination of methods, including in vitro slice electrophysiological recording, in vivo electrophysiological recording, and immunofluorescent staining in male mice. These results showed a decrease of the excitatory postsynaptic currents (sEPSCs) and no alteration of the inhibitory postsynaptic currents (sIPSCs) in the GABAergic neurons and the tonic inhibition in the excitatory neurons in the contralateral M1 after stroke induction, and EA recovered the impaired sEPSCs in the GABAergic neurons. We further found that the effect of EA-induced increase of c-Fos expression, enhancement of spike firing, potentiation of sEPSCs in the excitatory neurons, and improvement of swallowing function were all blocked by the removal of PNNs in the contralateral M1. In conclusion, the PNNs in the contralateral M1 was suggested to be participated in stroke pathogenesis and might be associated with the EA-mediated swallowing function rehabilitation of PSD in male mice. Our study provides insight into how PNNs might be involved in the mechanism of EA treatment for stroke rehabilitation.
Collapse
Affiliation(s)
- Si Yuan
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
- Department of Rehabilitation of Traditional Chinese Medicine, Hunan University of Chinese Medicine, 410208, Changsha, Hunan Province, China
| | - Jiahui Shi
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Xiaorong Tang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Bing Deng
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Zhennan Wu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Bo Qiu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Shumin Lin
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Chang Ji
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province 510630, China
| | - Lin Wang
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Shuai Cui
- Research Institute of Acupuncture and Meridian, College of Acupuncture and Moxibustion, Anhui University of Chinese Medicine, Hefei, Anhui Province 230012, China
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| | - Lulu Yao
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510006, China
| |
Collapse
|
22
|
Almassri LS, Ohl AP, Iafrate MC, Wade AD, Tokar NJ, Mafi AM, Beebe NL, Young JW, Mellott JG. Age-related upregulation of perineuronal nets on inferior collicular cells that project to the cochlear nucleus. Front Aging Neurosci 2023; 15:1271008. [PMID: 38053844 PMCID: PMC10694216 DOI: 10.3389/fnagi.2023.1271008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
Introduction Disruptions to the balance of excitation and inhibition in the inferior colliculus (IC) occur during aging and underlie various aspects of hearing loss. Specifically, the age-related alteration to GABAergic neurotransmission in the IC likely contributes to the poorer temporal precision characteristic of presbycusis. Perineuronal nets (PNs), a specialized form of the extracellular matrix, maintain excitatory/inhibitory synaptic environments and reduce structural plasticity. We sought to determine whether PNs increasingly surround cell populations in the aged IC that comprise excitatory descending projections to the cochlear nucleus. Method We combined Wisteria floribunda agglutinin (WFA) staining for PNs with retrograde tract-tracing in three age groups of Fischer Brown Norway (FBN) rats. Results The data demonstrate that the percentage of IC-CN cells with a PN doubles from ~10% at young age to ~20% at old age. This was true in both lemniscal and non-lemniscal IC. Discussion Furthermore, the increase of PNs occurred on IC cells that make both ipsilateral and contralateral descending projections to the CN. These results indicate that reduced structural plasticity in the elderly IC-CN pathway, affecting excitatory/inhibitory balance and, potentially, may lead to reduced temporal precision associated with presbycusis.
Collapse
Affiliation(s)
- Laila S. Almassri
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Andrew P. Ohl
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Milena C. Iafrate
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Aidan D. Wade
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Nick J. Tokar
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Amir M. Mafi
- The Ohio State College of Medicine, The Ohio State, Columbus, OH, United States
| | - Nichole L. Beebe
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Jesse W. Young
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Jeffrey G. Mellott
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| |
Collapse
|
23
|
Carceller H, Gramuntell Y, Klimczak P, Nacher J. Perineuronal Nets: Subtle Structures with Large Implications. Neuroscientist 2023; 29:569-590. [PMID: 35872660 DOI: 10.1177/10738584221106346] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Perineuronal nets (PNNs) are specialized structures of the extracellular matrix that surround the soma and proximal dendrites of certain neurons in the central nervous system, particularly parvalbumin-expressing interneurons. Their appearance overlaps the maturation of neuronal circuits and the closure of critical periods in different regions of the brain, setting their connectivity and abruptly reducing their plasticity. As a consequence, the digestion of PNNs, as well as the removal or manipulation of their components, leads to a boost in this plasticity and can play a key role in the functional recovery from different insults and in the etiopathology of certain neurologic and psychiatric disorders. Here we review the structure, composition, and distribution of PNNs and their variation throughout the evolutive scale. We also discuss methodological approaches to study these structures. The function of PNNs during neurodevelopment and adulthood is discussed, as well as the influence of intrinsic and extrinsic factors on these specialized regions of the extracellular matrix. Finally, we review current data on alterations in PNNs described in diseases of the central nervous system (CNS), focusing on psychiatric disorders. Together, all the data available point to the PNNs as a promising target to understand the physiology and pathologic conditions of the CNS.
Collapse
Affiliation(s)
- Héctor Carceller
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
24
|
Graham G, Chimenti MS, Knudtson KL, Grenard DN, Co L, Sumner M, Tchou T, Bieszczad KM. Learning induces unique transcriptional landscapes in the auditory cortex. Hear Res 2023; 438:108878. [PMID: 37659220 PMCID: PMC10529106 DOI: 10.1016/j.heares.2023.108878] [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: 04/03/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023]
Abstract
Learning can induce neurophysiological plasticity in the auditory cortex at multiple timescales. Lasting changes to auditory cortical function that persist over days, weeks, or even a lifetime, require learning to induce de novo gene expression. Indeed, transcription is the molecular determinant for long-term memories to form with a lasting impact on sound-related behavior. However, auditory cortical genes that support auditory learning, memory, and acquired sound-specific behavior are largely unknown. Using an animal model of adult, male Sprague-Dawley rats, this report is the first to identify genome-wide changes in learning-induced gene expression within the auditory cortex that may underlie long-lasting discriminative memory formation of acoustic frequency cues. Auditory cortical samples were collected from animals in the initial learning phase of a two-tone discrimination sound-reward task known to induce sound-specific neurophysiological and behavioral effects. Bioinformatic analyses on gene enrichment profiles from bulk RNA sequencing identified cholinergic synapse (KEGG rno04725), extra-cellular matrix receptor interaction (KEGG rno04512), and neuroactive receptor interaction (KEGG rno04080) among the top biological pathways are likely to be important for auditory discrimination learning. The findings characterize candidate effectors underlying the early stages of changes in cortical and behavioral function to ultimately support the formation of long-term discriminative auditory memory in the adult brain. The molecules and mechanisms identified are potential therapeutic targets to facilitate experiences that induce long-lasting changes to sound-specific auditory function in adulthood and prime for future gene-targeted investigations.
Collapse
Affiliation(s)
- G Graham
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ, USA; Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - M S Chimenti
- Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - K L Knudtson
- Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - D N Grenard
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - L Co
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - M Sumner
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - T Tchou
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - K M Bieszczad
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ, USA; Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA; Rutgers Center for Cognitive Science, Rutgers Univ., Piscataway, NJ, USA; Dept. of Otolaryngology-Head and Neck Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
25
|
Graham G, Chimenti MS, Knudtson KL, Grenard DN, Co L, Sumner M, Tchou T, Bieszczad KM. Learning induces unique transcriptional landscapes in the auditory cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.15.536914. [PMID: 37090563 PMCID: PMC10120736 DOI: 10.1101/2023.04.15.536914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Learning can induce neurophysiological plasticity in the auditory cortex at multiple timescales. Lasting changes to auditory cortical function that persist over days, weeks, or even a lifetime, require learning to induce de novo gene expression. Indeed, transcription is the molecular determinant for long-term memories to form with a lasting impact on sound-related behavior. However, auditory cortical genes that support auditory learning, memory, and acquired sound-specific behavior are largely unknown. This report is the first to identify in young adult male rats (Sprague-Dawley) genome-wide changes in learning-induced gene expression within the auditory cortex that may underlie the formation of long-lasting discriminative memory for acoustic frequency cues. Auditory cortical samples were collected from animals in the initial learning phase of a two-tone discrimination sound-reward task known to induce sound-specific neurophysiological and behavioral effects (e.g., Shang et al., 2019). Bioinformatic analyses on gene enrichment profiles from bulk RNA sequencing identified cholinergic synapse (KEGG 04725), extra-cellular matrix receptor interaction (KEGG 04512) , and neuroactive ligand-receptor interaction (KEGG 04080) as top biological pathways for auditory discrimination learning. The findings characterize key candidate effectors underlying changes in cortical function that support the initial formation of long-term discriminative auditory memory in the adult brain. The molecules and mechanisms identified are potential therapeutic targets to facilitate lasting changes to sound-specific auditory function in adulthood and prime for future gene-targeted investigations.
Collapse
Affiliation(s)
- G Graham
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - M S Chimenti
- Iowa Institute of Human Genetics, Univ. of Iowa Carver College of Medicine, Iowa City, IA
| | - K L Knudtson
- Iowa Institute of Human Genetics, Univ. of Iowa Carver College of Medicine, Iowa City, IA
| | - D N Grenard
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - L Co
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - M Sumner
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - T Tchou
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - K M Bieszczad
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
- Rutgers Center for Cognitive Science, Rutgers Univ., Piscataway, NJ
- Dept. of Otolaryngology-Head and Neck Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
26
|
Liu L, Zhang Y, Men S, Li X, Hou ST, Ju J. Elimination of perineuronal nets in CA1 disrupts GABA release and long-term contextual fear memory retention. Hippocampus 2023; 33:862-871. [PMID: 36709413 DOI: 10.1002/hipo.23503] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 12/19/2022] [Accepted: 01/11/2023] [Indexed: 01/30/2023]
Abstract
Perineuronal nets (PNNs) which mostly surround the parvalbumin (PV) neurons, have been shown to play critical roles in neural plasticity. Recently, PNNs have been shown to regulate fear-associated memory, but the molecular mechanism is still unclear. In this study, we found that removal of PNNs in vivo using chondroitinase ABC (ChABC) injection resulted in reduced firing rate of PV neurons and decreased inhibitory synaptic transmission in both PV neurons and excitatory neurons in the CA1 hippocampus. Interestingly, altered synaptic transmission appears to be mediated by presynaptic changes. Furthermore, ChABC treatment disrupts long-term contextual fear memory retention. These results suggest PNNs might alter fear memory by reducing the presynaptic GABA release.
Collapse
Affiliation(s)
- Luping Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yujie Zhang
- The Pediatric Neurology, Shenzhen Children's Hospital, Shenzhen, China
| | - Siqi Men
- Brain Research Centre and Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Xuanyi Li
- Brain Research Centre and Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Sheng-Tao Hou
- Brain Research Centre and Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jun Ju
- Brain Research Centre and Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
27
|
Akitake B, Douglas HM, LaFosse PK, Beiran M, Deveau CE, O'Rawe J, Li AJ, Ryan LN, Duffy SP, Zhou Z, Deng Y, Rajan K, Histed MH. Amplified cortical neural responses as animals learn to use novel activity patterns. Curr Biol 2023; 33:2163-2174.e4. [PMID: 37148876 DOI: 10.1016/j.cub.2023.04.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 05/08/2023]
Abstract
Cerebral cortex supports representations of the world in patterns of neural activity, used by the brain to make decisions and guide behavior. Past work has found diverse, or limited, changes in the primary sensory cortex in response to learning, suggesting that the key computations might occur in downstream regions. Alternatively, sensory cortical changes may be central to learning. We studied cortical learning by using controlled inputs we insert: we trained mice to recognize entirely novel, non-sensory patterns of cortical activity in the primary visual cortex (V1) created by optogenetic stimulation. As animals learned to use these novel patterns, we found that their detection abilities improved by an order of magnitude or more. The behavioral change was accompanied by large increases in V1 neural responses to fixed optogenetic input. Neural response amplification to novel optogenetic inputs had little effect on existing visual sensory responses. A recurrent cortical model shows that this amplification can be achieved by a small mean shift in recurrent network synaptic strength. Amplification would seem to be desirable to improve decision-making in a detection task; therefore, these results suggest that adult recurrent cortical plasticity plays a significant role in improving behavioral performance during learning.
Collapse
Affiliation(s)
- Bradley Akitake
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hannah M Douglas
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul K LaFosse
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Manuel Beiran
- Nash Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Ciana E Deveau
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonathan O'Rawe
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anna J Li
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lauren N Ryan
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Samuel P Duffy
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhishang Zhou
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yanting Deng
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kanaka Rajan
- Nash Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark H Histed
- Unit on Neural Computation and Behavior, National Institute of Mental Health Intramural Program, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Liu P, Zhao Y, Xiong W, Pan Y, Zhu M, Zhu X. Degradation of Perineuronal Nets in the Cerebellar Interpositus Nucleus Ameliorated Social Deficits in Shank3-deficient Mice. Neuroscience 2023; 511:29-38. [PMID: 36587867 DOI: 10.1016/j.neuroscience.2022.12.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/08/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022]
Abstract
Perineuronal nets (PNNs) are structures that contain extracellular matrix chondroitin sulfate proteoglycan and surround the soma and dendrites of various neuronal cell types. They are involved in synaptic plasticity and undertake important physiological functions. Altered expression of PNNs has been demonstrated in the brains of autism-related animal models. However, the underlying mechanism is still unknown. In this study, we demonstrated that the PNNs in the cerebellum are involved in modulating social and repetitive/inflexible behaviors in Shank3B-/- mice, an established animal model of autism spectrum disorder. First, we performed wisteria floribunda agglutinin staining of the whole brain of Shank3B-/- mice, and found wisteria floribunda agglutinin-positive PNNs are significantly increased in the cerebellar interpositus nucleus (IntP) in Shank3B-/- mice compared to control littermates. After degradation of PNNs in the IntP by chondroitinase ABC, the repetitive behaviors of Shank3B-/- mice were decreased, while their social behaviors were ameliorated. These results suggested that PNNs homeostasis is involved in the regulation of social behavior, revealing a potential therapeutic strategy targeting PNNs in the IntP for the treatment of autism spectrum disorder.
Collapse
Affiliation(s)
- Peng Liu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yulu Zhao
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wenchao Xiong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Yida Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minzhen Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xinhong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; School of Psychology, Shenzhen University, Shenzhen 518060, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China.
| |
Collapse
|
29
|
Xu Z, Geron E, Pérez-Cuesta LM, Bai Y, Gan WB. Generalized extinction of fear memory depends on co-allocation of synaptic plasticity in dendrites. Nat Commun 2023; 14:503. [PMID: 36720872 PMCID: PMC9889816 DOI: 10.1038/s41467-023-35805-9] [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: 10/31/2021] [Accepted: 01/03/2023] [Indexed: 02/02/2023] Open
Abstract
Memories can be modified by new experience in a specific or generalized manner. Changes in synaptic connections are crucial for memory storage, but it remains unknown how synaptic changes associated with different memories are distributed within neuronal circuits and how such distributions affect specific or generalized modification by novel experience. Here we show that fear conditioning with two different auditory stimuli (CS) and footshocks (US) induces dendritic spine elimination mainly on different dendritic branches of layer 5 pyramidal neurons in the mouse motor cortex. Subsequent fear extinction causes CS-specific spine formation and extinction of freezing behavior. In contrast, spine elimination induced by fear conditioning with >2 different CS-USs often co-exists on the same dendritic branches. Fear extinction induces CS-nonspecific spine formation and generalized fear extinction. Moreover, activation of somatostatin-expressing interneurons increases the occurrence of spine elimination induced by different CS-USs on the same dendritic branches and facilitates the generalization of fear extinction. These findings suggest that specific or generalized modification of existing memories by new experience depends on whether synaptic changes induced by previous experiences are segregated or co-exist at the level of individual dendritic branches.
Collapse
Affiliation(s)
- Zhiwei Xu
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
- Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Erez Geron
- Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Luis M Pérez-Cuesta
- Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Yang Bai
- Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Wen-Biao Gan
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
- Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
30
|
Chemistry and Function of Glycosaminoglycans in the Nervous System. ADVANCES IN NEUROBIOLOGY 2023; 29:117-162. [DOI: 10.1007/978-3-031-12390-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
31
|
Benbenishty A, Peled-Hajaj S, Krishnaswamy VR, Har-Gil H, Havusha-Laufer S, Ruggiero A, Slutsky I, Blinder P, Sagi I. Longitudinal in vivo imaging of perineuronal nets. NEUROPHOTONICS 2023; 10:015008. [PMID: 36970015 PMCID: PMC10037344 DOI: 10.1117/1.nph.10.1.015008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/23/2023] [Indexed: 06/18/2023]
Abstract
SIGNIFICANCE Perineuronal nets (PNNs) are extracellular matrix structures implicated in learning, memory, information processing, synaptic plasticity, and neuroprotection. However, our understanding of mechanisms governing the evidently important contribution of PNNs to central nervous system function is lacking. A primary cause for this gap of knowledge is the absence of direct experimental tools to study their role in vivo. AIM We introduce a robust approach for quantitative longitudinal imaging of PNNs in brains of awake mice at subcellular resolution. APPROACH We label PNNs in vivo with commercially available compounds and monitor their dynamics with two-photon imaging. RESULTS Using our approach, we show that it is possible to longitudinally follow the same PNNs in vivo while monitoring degradation and reconstitution of PNNs. We demonstrate the compatibility of our method to simultaneously monitor neuronal calcium dynamics in vivo and compare the activity of neurons with and without PNNs. CONCLUSION Our approach is tailored for studying the intricate role of PNNs in vivo, while paving the road for elucidating their role in different neuropathological conditions.
Collapse
Affiliation(s)
- Amit Benbenishty
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Shany Peled-Hajaj
- Tel Aviv University, Neurobiology, Biochemistry, and Biophysics School, Tel Aviv, Israel
- Tel Aviv University, Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv, Israel
| | | | - Hagai Har-Gil
- Tel Aviv University, Neurobiology, Biochemistry, and Biophysics School, Tel Aviv, Israel
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
| | - Sapir Havusha-Laufer
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| | - Antonella Ruggiero
- Tel Aviv University, Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Inna Slutsky
- Tel Aviv University, Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv, Israel
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
| | - Pablo Blinder
- Tel Aviv University, Neurobiology, Biochemistry, and Biophysics School, Tel Aviv, Israel
- Tel Aviv University, Sagol School of Neuroscience, Tel Aviv, Israel
| | - Irit Sagi
- The Weizmann Institute of Science, Department of Immunology and Regenerative Biology, Rehovot, Israel
| |
Collapse
|
32
|
Singh S, Topolnik L. Inhibitory circuits in fear memory and fear-related disorders. Front Neural Circuits 2023; 17:1122314. [PMID: 37035504 PMCID: PMC10076544 DOI: 10.3389/fncir.2023.1122314] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/17/2023] [Indexed: 04/11/2023] Open
Abstract
Fear learning and memory rely on dynamic interactions between the excitatory and inhibitory neuronal populations that make up the prefrontal cortical, amygdala, and hippocampal circuits. Whereas inhibition of excitatory principal cells (PCs) by GABAergic neurons restrains their excitation, inhibition of GABAergic neurons promotes the excitation of PCs through a process called disinhibition. Specifically, GABAergic interneurons that express parvalbumin (PV+) and somatostatin (SOM+) provide inhibition to different subcellular domains of PCs, whereas those that express the vasoactive intestinal polypeptide (VIP+) facilitate disinhibition of PCs by inhibiting PV+ and SOM+ interneurons. Importantly, although the main connectivity motifs and the underlying network functions of PV+, SOM+, and VIP+ interneurons are replicated across cortical and limbic areas, these inhibitory populations play region-specific roles in fear learning and memory. Here, we provide an overview of the fear processing in the amygdala, hippocampus, and prefrontal cortex based on the evidence obtained in human and animal studies. Moreover, focusing on recent findings obtained using genetically defined imaging and intervention strategies, we discuss the population-specific functions of PV+, SOM+, and VIP+ interneurons in fear circuits. Last, we review current insights that integrate the region-specific inhibitory and disinhibitory network patterns into fear memory acquisition and fear-related disorders.
Collapse
Affiliation(s)
- Sanjay Singh
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Quebec City, QC, Canada
- Neuroscience Axis, CRCHUQ, Laval University, Quebec City, QC, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Quebec City, QC, Canada
- Neuroscience Axis, CRCHUQ, Laval University, Quebec City, QC, Canada
- *Correspondence: Lisa Topolnik
| |
Collapse
|
33
|
Lépine M, Douceau S, Devienne G, Prunotto P, Lenoir S, Regnauld C, Pouettre E, Piquet J, Lebouvier L, Hommet Y, Maubert E, Agin V, Lambolez B, Cauli B, Ali C, Vivien D. Parvalbumin interneuron-derived tissue-type plasminogen activator shapes perineuronal net structure. BMC Biol 2022; 20:218. [PMID: 36199089 PMCID: PMC9535866 DOI: 10.1186/s12915-022-01419-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background Perineuronal nets (PNNs) are specialized extracellular matrix structures mainly found around fast-spiking parvalbumin (FS-PV) interneurons. In the adult, their degradation alters FS-PV-driven functions, such as brain plasticity and memory, and altered PNN structures have been found in neurodevelopmental and central nervous system disorders such as Alzheimer’s disease, leading to interest in identifying targets able to modify or participate in PNN metabolism. The serine protease tissue-type plasminogen activator (tPA) plays multifaceted roles in brain pathophysiology. However, its cellular expression profile in the brain remains unclear and a possible role in matrix plasticity through PNN remodeling has never been investigated. Result By combining a GFP reporter approach, immunohistology, electrophysiology, and single-cell RT-PCR, we discovered that cortical FS-PV interneurons are a source of tPA in vivo. We found that mice specifically lacking tPA in FS-PV interneurons display denser PNNs in the somatosensory cortex, suggesting a role for tPA from FS-PV interneurons in PNN remodeling. In vitro analyses in primary cultures of mouse interneurons also showed that tPA converts plasminogen into active plasmin, which in turn, directly degrades aggrecan, a major structural chondroitin sulfate proteoglycan (CSPG) in PNNs. Conclusions We demonstrate that tPA released from FS-PV interneurons in the central nervous system reduces PNN density through CSPG degradation. The discovery of this tPA-dependent PNN remodeling opens interesting insights into the control of brain plasticity. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01419-8.
Collapse
Affiliation(s)
- Matthieu Lépine
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Sara Douceau
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Gabrielle Devienne
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Paul Prunotto
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Sophie Lenoir
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Caroline Regnauld
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Elsa Pouettre
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Juliette Piquet
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Laurent Lebouvier
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Eric Maubert
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Véronique Agin
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Bertrand Lambolez
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Bruno Cauli
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France.
| | - Denis Vivien
- Department of clinical research, CHU de Caen Normandie, Caen, France
| |
Collapse
|
34
|
Fawcett JW, Fyhn M, Jendelova P, Kwok JCF, Ruzicka J, Sorg BA. The extracellular matrix and perineuronal nets in memory. Mol Psychiatry 2022; 27:3192-3203. [PMID: 35760878 PMCID: PMC9708575 DOI: 10.1038/s41380-022-01634-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023]
Abstract
All components of the CNS are surrounded by a diffuse extracellular matrix (ECM) containing chondroitin sulphate proteoglycans (CSPGs), heparan sulphate proteoglycans (HSPGs), hyaluronan, various glycoproteins including tenascins and thrombospondin, and many other molecules that are secreted into the ECM and bind to ECM components. In addition, some neurons, particularly inhibitory GABAergic parvalbumin-positive (PV) interneurons, are surrounded by a more condensed cartilage-like ECM called perineuronal nets (PNNs). PNNs surround the soma and proximal dendrites as net-like structures that surround the synapses. Attention has focused on the role of PNNs in the control of plasticity, but it is now clear that PNNs also play an important part in the modulation of memory. In this review we summarize the role of the ECM, particularly the PNNs, in the control of various types of memory and their participation in memory pathology. PNNs are now being considered as a target for the treatment of impaired memory. There are many potential treatment targets in PNNs, mainly through modulation of the sulphation, binding, and production of the various CSPGs that they contain or through digestion of their sulphated glycosaminoglycans.
Collapse
Affiliation(s)
- James W Fawcett
- John van Geest Centre for Brain Repair, Department Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK.
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic.
| | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Pavla Jendelova
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Jessica C F Kwok
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jiri Ruzicka
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Barbara A Sorg
- Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| |
Collapse
|
35
|
Ojala KE, Staib M, Gerster S, Ruff CC, Bach DR. Inhibiting Human Aversive Memory by Transcranial Theta-Burst Stimulation to the Primary Sensory Cortex. Biol Psychiatry 2022; 92:149-157. [PMID: 35410762 DOI: 10.1016/j.biopsych.2022.01.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/14/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Predicting adverse events from past experience is fundamental for many biological organisms. However, some individuals suffer from maladaptive memories that impair behavioral control and well-being, e.g., after psychological trauma. Inhibiting the formation and maintenance of such memories would have high clinical relevance. Previous preclinical research has focused on systemically administered pharmacological interventions, which cannot be targeted to specific neural circuits in humans. Here, we investigated the potential of noninvasive neural stimulation on the human sensory cortex in inhibiting aversive memory in a laboratory threat conditioning model. METHODS We build on an emerging nonhuman literature suggesting that primary sensory cortices may be crucially required for threat memory formation and consolidation. Immediately before conditioning innocuous somatosensory stimuli (conditioned stimuli [CS]) to aversive electric stimulation, healthy human participants received continuous theta-burst transcranial magnetic stimulation (cTBS) to individually localized primary somatosensory cortex in either the CS-contralateral (experimental) or CS-ipsilateral (control) hemisphere. We measured fear-potentiated startle to infer threat memory retention on the next day, as well as skin conductance and pupil size during learning. RESULTS After overnight consolidation, threat memory was attenuated in the experimental group compared with the control cTBS group. There was no evidence that this differed between simple and complex CS or that CS identification or initial learning were affected by cTBS. CONCLUSIONS Our results suggest that cTBS to the primary sensory cortex inhibits threat memory, likely by an impact on postlearning consolidation. We propose that noninvasive targeted stimulation of the sensory cortex may provide a new avenue for interfering with aversive memories in humans.
Collapse
Affiliation(s)
- Karita E Ojala
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland; Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland.
| | - Matthias Staib
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland; Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland
| | - Samuel Gerster
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland
| | - Christian C Ruff
- Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland; Zurich Center for Neuroeconomics, Department of Economics, University of Zürich, Zürich, Switzerland
| | - Dominik R Bach
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland; Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland; Wellcome Centre for Human Neuroimaging and Max-Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, London, United Kingdom.
| |
Collapse
|
36
|
Neurocan regulates vulnerability to stress and the anti-depressant effect of ketamine in adolescent rats. Mol Psychiatry 2022; 27:2522-2532. [PMID: 35264728 DOI: 10.1038/s41380-022-01495-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 12/13/2022]
Abstract
Depression is more prevalent among adolescents than adults, but the underlying mechanisms remain largely unknown. Using a subthreshold chronic stress model, here we show that developmentally regulated expressions of the perineuronal nets (PNNs), and one of the components, Neurocan in the prelimbic cortex (PrL) are important for the vulnerability to stress and depressive-like behaviors in both adolescent and adult rats. Reduction of PNNs or Neurocan with pharmacological or viral methods to mimic the expression of PNNs in the PrL during adolescence compromised resilience to stress in adult rats, while virally mediated overexpression of Neurocan reversed vulnerability to stress in adolescent rats. Ketamine, a recent-approved drug for treatment-resistant depression rescued impaired function of Parvalbumin-positive neurons function, increased expression of PNNs in the PrL, and reversed depressive-like behaviors in adolescent rats. Furthermore, we show that Neurocan mediates the anti-depressant effect of ketamine, virally mediated reduction of Neurocan in the PrL abolished the anti-depressant effect of ketamine in adolescent rats. Our findings show an important role of Neurocan in depression in adolescence, and suggest a novel mechanism for the anti-depressant effect of ketamine.
Collapse
|
37
|
Kumar A, Biswas A, Bojja SL, Kolathur KK, Volety SM. Emerging therapeutic role of chondroitinase (ChABC) in neurological disorders and cancer. CURRENT DRUG THERAPY 2022. [DOI: 10.2174/1574885517666220331151619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Proteoglycans are essential biomacromolecules that participate in matrix structure and organization, cell proliferation and migration, and cell surface signal transduction. However, their roles in physiology, particularly in CNS remain incompletely deciphered. Numerous studies highlight the elevated levels of chondroitin sulphate proteoglycans (CSPGs) in various diseases like cancers and neurological disorders like spinal cord injury (SCI), traumatic brain damage, neurodegenerative diseases, and are mainly implicated to hinder tissue repair. In such a context, chondroitinase ABC (ChABC), a therapeutic enzyme has shown immense hope to treat these diseases in several preclinical studies, primarily attributed to the digestion of the side chains of the proteoglycan chondroitin sulphate (CS) molecule. Despite extensive research, the progress in evolving the concept of therapeutic targeting of proteoglycans is still in its infancy. This review thus provides fresh insights into the emerging therapeutic applications of ChABC in various diseases apart from SCI and the underlying mechanisms.
Collapse
Affiliation(s)
- Akshara Kumar
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Aishi Biswas
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sree Lalitha Bojja
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Kiran Kumar Kolathur
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Subrahmanyam M Volety
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
38
|
Tanti A, Belliveau C, Nagy C, Maitra M, Denux F, Perlman K, Chen F, Mpai R, Canonne C, Théberge S, McFarquhar A, Davoli MA, Belzung C, Turecki G, Mechawar N. Child abuse associates with increased recruitment of perineuronal nets in the ventromedial prefrontal cortex: a possible implication of oligodendrocyte progenitor cells. Mol Psychiatry 2022; 27:1552-1561. [PMID: 34799691 PMCID: PMC9095471 DOI: 10.1038/s41380-021-01372-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022]
Abstract
Child abuse (CA) is a strong predictor of psychopathologies and suicide, altering normal trajectories of brain development in areas closely linked to emotional responses such as the prefrontal cortex (PFC). Yet, the cellular underpinnings of these enduring effects are unclear. Childhood and adolescence are marked by the protracted formation of perineuronal nets (PNNs), which orchestrate the closure of developmental windows of cortical plasticity by regulating the functional integration of parvalbumin interneurons into neuronal circuits. Using well-characterized post-mortem brain samples, we show that a history of CA is specifically associated with increased densities and morphological complexity of WFL-labeled PNNs in the ventromedial PFC (BA11/12), possibly suggesting increased recruitment and maturation of PNNs. Through single-nucleus sequencing and fluorescent in situ hybridization, we found that the expression of canonical components of PNNs is enriched in oligodendrocyte progenitor cells (OPCs), and that they are upregulated in CA victims. These correlational findings suggest that early-life adversity may lead to persistent patterns of maladaptive behaviors by reducing the neuroplasticity of cortical circuits through the enhancement of developmental OPC-mediated PNN formation.
Collapse
Affiliation(s)
- Arnaud Tanti
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France.
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Malosree Maitra
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Fanny Denux
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Kelly Perlman
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Frank Chen
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Refilwe Mpai
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Candice Canonne
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Stéphanie Théberge
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Ashley McFarquhar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Maria Antonietta Davoli
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | | | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, Canada.
| |
Collapse
|
39
|
Shi Y, Wu X, Zhou J, Cui W, Wang J, Hu Q, Zhang S, Han L, Zhou M, Luo J, Wang Q, Liu H, Feng D, Ge S, Qu Y. Single-Nucleus RNA Sequencing Reveals that Decorin Expression in the Amygdala Regulates Perineuronal Nets Expression and Fear Conditioning Response after Traumatic Brain Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104112. [PMID: 35038242 PMCID: PMC8895134 DOI: 10.1002/advs.202104112] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Indexed: 06/14/2023]
Abstract
Traumatic brain injury (TBI) is a risk factor for posttraumatic stress disorder (PTSD). Augmented fear is a defining characteristic of PTSD, and the amygdala is considered the main brain region to process fear. The mechanism by which the amygdala is involved in fear conditioning after TBI is still unclear. Using single-nucleus RNA sequencing (snRNA-seq), transcriptional changes in cells in the amygdala after TBI are investigated. In total, 72 328 nuclei are obtained from the sham and TBI groups. 7 cell types, and analysis of differentially expressed genes (DEGs) reveals widespread transcriptional changes in each cell type after TBI are identified. In in vivo experiments, it is demonstrated that Decorin (Dcn) expression in the excitatory neurons of the amygdala significantly increased after TBI, and Dcn knockout in the amygdala mitigates TBI-associated fear conditioning. Of note, this effect is caused by a Dcn-mediated decrease in the expression of perineuronal nets (PNNs), which affect the glutamate-γ-aminobutyric acid balance in the amygdala. Finally, the results suggest that Dcn functions by interacting with collagen VI α3 (Col6a3). Consequently, the findings reveal transcriptional changes in different cell types of the amygdala after TBI and provide direct evidence that Dcn relieves fear conditioning by regulating PNNs.
Collapse
Affiliation(s)
- Yingwu Shi
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Xun Wu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Jinpeng Zhou
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Wenxing Cui
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Jin Wang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Qing Hu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Shenghao Zhang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Liying Han
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Meixuan Zhou
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Jianing Luo
- Department of NeurosurgeryWest Theater General HospitalChengduSichuan610083China
| | - Qiang Wang
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Haixiao Liu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Dayun Feng
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Shunnan Ge
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| | - Yan Qu
- Department of NeurosurgeryTangdu HospitalFourth Military Medical UniversityXi'anShaanxi710038China
| |
Collapse
|
40
|
Trnski S, Nikolić B, Ilic K, Drlje M, Bobic-Rasonja M, Darmopil S, Petanjek Z, Hranilovic D, Jovanov-Milosevic N. The Signature of Moderate Perinatal Hypoxia on Cortical Organization and Behavior: Altered PNN-Parvalbumin Interneuron Connectivity of the Cingulate Circuitries. Front Cell Dev Biol 2022; 10:810980. [PMID: 35295859 PMCID: PMC8919082 DOI: 10.3389/fcell.2022.810980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/11/2022] [Indexed: 11/29/2022] Open
Abstract
This study was designed in a rat model to determine the hallmarks of possible permanent behavioral and structural brain alterations after a single moderate hypoxic insult. Eighty-two Wistar Han (RccHan: WIST) rats were randomly subjected to hypoxia (pO2 73 mmHg/2 h) or normoxia at the first postnatal day. The substantially increased blood lactate, a significantly decreased cytochrome-C-oxygenase expression in the brain, and depleted subventricular zone suggested a high vulnerability of subset of cell populations to oxidative stress and consequent tissue response even after a single, moderate, hypoxic event. The results of behavioral tests (open-field, hole-board, social-choice, and T-maze) applied at the 30–45th and 70–85th postnatal days revealed significant hyperactivity and a slower pace of learning in rats subjected to perinatal hypoxia. At 3.5 months after hypoxic insult, the histochemical examination demonstrated a significantly increased number of specific extracellular matrix—perineuronal nets and increased parvalbumin expression in a subpopulation of interneurons in the medial and retrosplenial cingulate cortex of these animals. Conclusively, moderate perinatal hypoxia in rats causes a long-lasting reorganization of the connectivity in the cingulate cortex and consequent alterations of related behavioral and cognitive abilities. This non-invasive hypoxia model in the rat successfully and complementarily models the moderate perinatal hypoxic injury in fetuses and prematurely born human babies and may enhance future research into new diagnostic and therapeutic strategies for perinatal medicine.
Collapse
Affiliation(s)
- Sara Trnski
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Barbara Nikolić
- Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Katarina Ilic
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroimaging, BRAIN Centre, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, United Kingdom
| | - Matea Drlje
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Mihaela Bobic-Rasonja
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Sanja Darmopil
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Zdravko Petanjek
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dubravka Hranilovic
- Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Natasa Jovanov-Milosevic
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
- *Correspondence: Natasa Jovanov-Milosevic,
| |
Collapse
|
41
|
Barahona RA, Morabito S, Swarup V, Green KN. Cortical diurnal rhythms remain intact with microglial depletion. Sci Rep 2022; 12:114. [PMID: 34997092 PMCID: PMC8742049 DOI: 10.1038/s41598-021-04079-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Microglia are subject to change in tandem with the endogenously generated biological oscillations known as our circadian rhythm. Studies have shown microglia harbor an intrinsic molecular clock which regulates diurnal changes in morphology and influences inflammatory responses. In the adult brain, microglia play an important role in the regulation of condensed extracellular matrix structures called perineuronal nets (PNNs), and it has been suggested that PNNs are also regulated in a circadian and diurnal manner. We sought to determine whether microglia mediate the diurnal regulation of PNNs via CSF1R inhibitor dependent microglial depletion in C57BL/6J mice, and how the absence of microglia might affect cortical diurnal gene expression rhythms. While we observe diurnal differences in microglial morphology, where microglia are most ramified at the onset of the dark phase, we do not find diurnal differences in PNN intensity. However, PNN intensity increases across many brain regions in the absence of microglia, supporting a role for microglia in the regulation of PNNs. Here, we also show that cortical diurnal gene expression rhythms are intact, with no cycling gene changes without microglia. These findings demonstrate a role for microglia in the maintenance of PNNs, but not in the maintenance of diurnal rhythms.
Collapse
Affiliation(s)
- Rocio A Barahona
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697, USA
| | - Samuel Morabito
- Mathematical, Computational and Systems Biology (MCSB) Program, University of California, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, 3208 Biological Sciences III, Irvine, CA, 92697, USA.
| |
Collapse
|
42
|
Hodebourg R, Kalivas PW, Kruyer A. Extrasynaptic therapeutic targets in substance use and stress disorders. Trends Pharmacol Sci 2022; 43:56-68. [PMID: 34753604 PMCID: PMC8688303 DOI: 10.1016/j.tips.2021.10.006] [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: 05/27/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
Treatments for substance use and stress disorders are based on ameliorating behavioral symptoms, not on reversing the synaptic pathology that has the potential to cure disorders. This failing arises in part from a research focus on how pre- and postsynaptic physiology is changed even though key neuropathology exists in the perisynaptic neuropil that homeostatically regulates synaptic transmission. We explore recent findings from the substance use and stress disorder literature pointing to a key role for perisynaptic astroglia and signaling in the extracellular matrix (ECM) in regulating synaptic pathology. We conclude that drugs and stress initiate long-lasting changes in brain synapses via enduring neuroadaptations in astroglia and the ECM, and that modulating extrasynaptic regulators may be therapeutically useful.
Collapse
Affiliation(s)
- Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA.
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA
| |
Collapse
|
43
|
Jovasevic V, Zhang H, Petrovic Z, Cicvaric A, Radulovic J. Protocol for assessing the role of hippocampal perineuronal nets in aversive memories. STAR Protoc 2021; 2:100931. [PMID: 34778848 PMCID: PMC8577157 DOI: 10.1016/j.xpro.2021.100931] [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] [Indexed: 11/20/2022] Open
Abstract
Perineuronal nets (PNNs) are emerging as critical regulators of memory-related neuronal processes. However, their exact contribution depends on type of memory, consolidation stage, or brain region, and remains to be fully investigated. We describe here a protocol to evaluate the importance of PNNs in the dorsal hippocampus in different stages of aversive memories using a mouse model. The protocol provides detailed instructions for surgical implantation of hippocampal cannulas, drug infusion, contextual fear conditioning procedures, and immunohistochemistry for PNN visualization. For complete details on the use and execution of this protocol, please refer to Jovasevic et al. (2021).
Collapse
Affiliation(s)
- Vladimir Jovasevic
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL 60611, USA
| | - Hui Zhang
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Zorica Petrovic
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ana Cicvaric
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jelena Radulovic
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
44
|
Crapser JD, Arreola MA, Tsourmas KI, Green KN. Microglia as hackers of the matrix: sculpting synapses and the extracellular space. Cell Mol Immunol 2021; 18:2472-2488. [PMID: 34413489 PMCID: PMC8546068 DOI: 10.1038/s41423-021-00751-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia shape the synaptic environment in health and disease, but synapses do not exist in a vacuum. Instead, pre- and postsynaptic terminals are surrounded by extracellular matrix (ECM), which together with glia comprise the four elements of the contemporary tetrapartite synapse model. While research in this area is still just beginning, accumulating evidence points toward a novel role for microglia in regulating the ECM during normal brain homeostasis, and such processes may, in turn, become dysfunctional in disease. As it relates to synapses, microglia are reported to modify the perisynaptic matrix, which is the diffuse matrix that surrounds dendritic and axonal terminals, as well as perineuronal nets (PNNs), specialized reticular formations of compact ECM that enwrap neuronal subsets and stabilize proximal synapses. The interconnected relationship between synapses and the ECM in which they are embedded suggests that alterations in one structure necessarily affect the dynamics of the other, and microglia may need to sculpt the matrix to modify the synapses within. Here, we provide an overview of the microglial regulation of synapses, perisynaptic matrix, and PNNs, propose candidate mechanisms by which these structures may be modified, and present the implications of such modifications in normal brain homeostasis and in disease.
Collapse
Affiliation(s)
- Joshua D. Crapser
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Miguel A. Arreola
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kate I. Tsourmas
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kim N. Green
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| |
Collapse
|
45
|
Ibrahim P, Almeida D, Nagy C, Turecki G. Molecular impacts of childhood abuse on the human brain. Neurobiol Stress 2021; 15:100343. [PMID: 34141833 PMCID: PMC8187840 DOI: 10.1016/j.ynstr.2021.100343] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/24/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Childhood abuse (CA) is a prevalent global health concern, increasing the risk of negative mental health outcomes later in life. In the literature, CA is commonly defined as physical, sexual, and emotional abuse, as well as neglect. Several mental disorders have been associated with CA, including depression, bipolar disorder, schizophrenia, and post-traumatic stress disorder, along with an increased risk of suicide. It is thought that traumatic life events occurring during childhood and adolescence may have a significant impact on essential brain functions, which may persist throughout adulthood. The interaction between the brain and the external environment can be mediated by epigenetic alterations in gene expression, and there is a growing body of evidence to show that such changes occur as a function of CA. Disruptions in the HPA axis, myelination, plasticity, and signaling have been identified in individuals with a history of CA. Understanding the molecular impact of CA on the brain is essential for the development of treatment and prevention measures. In this review, we will summarize studies that highlight the molecular changes associated with CA in the human brain, along with supporting evidence from peripheral studies and animal models. We will also discuss some of the limitations surrounding the study of CA and propose extracellular vesicles as a promising future approach in the field.
Collapse
Affiliation(s)
- Pascal Ibrahim
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Daniel Almeida
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
46
|
Postnatal Sox6 Regulates Synaptic Function of Cortical Parvalbumin-Expressing Neurons. J Neurosci 2021; 41:8876-8886. [PMID: 34503995 PMCID: PMC8549537 DOI: 10.1523/jneurosci.0021-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 11/29/2022] Open
Abstract
Cortical parvalbumin-expressing (Pvalb+) neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. This class of inhibitory neurons undergoes extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. While several transcription factors, such as Nkx2-1, Lhx6, and Sox6, are known to be necessary for the differentiation of progenitors into Pvalb+ neurons, which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons' innervation and synaptic function remains largely unknown. Because Sox6 is continuously expressed in Pvalb+ neurons until adulthood, we used conditional knock-out strategies to investigate its putative role in the postnatal maturation and synaptic function of cortical Pvalb+ neurons in mice of both sexes. We found that early postnatal loss of Sox6 in Pvalb+ neurons leads to failure of synaptic bouton growth, whereas later removal in mature Pvalb+ neurons in the adult causes shrinkage of already established synaptic boutons. Paired recordings between Pvalb+ neurons and pyramidal neurons revealed reduced release probability and increased failure rate of Pvalb+ neurons' synaptic output. Furthermore, Pvalb+ neurons lacking Sox6 display reduced expression of full-length tropomyosin-receptor kinase B (TrkB), a key modulator of GABAergic transmission. Once re-expressed in neurons lacking Sox6, TrkB was sufficient to rescue the morphologic synaptic phenotype. Finally, we showed that Sox6 mRNA levels were increased by motor training. Our data thus suggest a constitutive role for Sox6 in the maintenance of synaptic output from Pvalb+ neurons into adulthood. SIGNIFICANCE STATEMENT Cortical parvalbumin-expressing (Pvalb+) inhibitory neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. These inhibitory neurons undergo extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. However, it remains largely unknown which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons. Here, we show that the transcription factor Sox6 cell-autonomously regulates the synaptic maintenance and output of Pvalb+ neurons until adulthood, leaving unaffected other maturational features of this neuronal population.
Collapse
|
47
|
Learning-induced plasticity in the barrel cortex is disrupted by inhibition of layer 4 somatostatin-containing interneurons. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119146. [PMID: 34599984 DOI: 10.1016/j.bbamcr.2021.119146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 09/13/2021] [Indexed: 12/26/2022]
Abstract
Gaba-ergic neurons are a diverse cell class with extensive influence over cortical processing, but their role in experience-dependent plasticity is not completely understood. Here we addressed the role of cortical somatostatin- (SOM-INs) and vasoactive intestinal polypeptide- (VIP-INs) containing interneurons in a Pavlovian conditioning where stimulation of the vibrissae is used as a conditioned stimulus and tail shock as unconditioned one. This procedure induces a plastic change observed as an enlargement of the cortical functional representation of vibrissae activated during conditioning. Using layer-targeted, cell-selective DREADD transductions, we examined the involvement of SOM-INs and VIP-INs activity in learning-related plastic changes. Under optical recordings, we injected DREADD-expressing vectors into layer IV (L4) barrels or layer II/III (L2/3) areas corresponding to the activated vibrissae. The activity of the interneurons was modulated during all conditioning sessions, and functional 2-deoxyglucose (2DG) maps were obtained 24 h after the last session. In mice with L4 but not L2/3 SOM-INs suppressed during conditioning, the plastic change of whisker representation was absent. The behavioral effect of conditioning was disturbed. Both L4 SOM-INs excitation and L2/3 VIP-INs inhibition during conditioning did not affect the plasticity or the conditioned response. We found the activity of L4 SOM-INs is indispensable in the formation of learning-induced plastic change. We propose that L4 SOM-INs may provide disinhibition by blocking L4 parvalbumin interneurons, allowing a flow of information into upper cortical layers during learning.
Collapse
|
48
|
Wen Z, Chen ZS, Milad MR. Fear extinction learning modulates large-scale brain connectivity. Neuroimage 2021; 238:118261. [PMID: 34126211 PMCID: PMC8436785 DOI: 10.1016/j.neuroimage.2021.118261] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022] Open
Abstract
Exploring the neural circuits of the extinction of conditioned fear is critical to advance our understanding of fear- and anxiety-related disorders. The field has focused on examining the role of various regions of the medial prefrontal cortex, insular cortex, hippocampus, and amygdala in conditioned fear and its extinction. The contribution of this 'fear network' to the conscious awareness of fear has recently been questioned. And as such, there is a need to examine higher/multiple cortical systems that might contribute to the conscious feeling of fear and anxiety. Herein, we studied functional connectivity patterns across the entire brain to examine the contribution of multiple networks to the acquisition of fear extinction learning and its retrieval. We conducted trial-by-trial analyses on data from 137 healthy participants who underwent a two-day fear conditioning and extinction paradigm in a functional magnetic resonance imaging (fMRI) scanner. We found that functional connectivity across a broad range of brain regions, many of which are part of the default mode, frontoparietal, and ventral attention networks, increased from early to late extinction learning only to a conditioned cue. The increased connectivity during extinction learning predicted the magnitude of extinction memory tested 24 h later. Together, these findings provide evidence supporting recent studies implicating distributed brain regions in learning, consolidation and expression of fear extinction memory in the human brain.
Collapse
Affiliation(s)
- Zhenfu Wen
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Zhe Sage Chen
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, United States; The Neuroscience Institute, New York University School of Medicine, New York, NY, United States
| | - Mohammed R Milad
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States; The Neuroscience Institute, New York University School of Medicine, New York, NY, United States; Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States.
| |
Collapse
|
49
|
Mafi AM, Russ MG, Hofer LN, Pham VQ, Young JW, Mellott JG. Inferior collicular cells that project to the auditory thalamus are increasingly surrounded by perineuronal nets with age. Neurobiol Aging 2021; 105:1-15. [PMID: 34004491 PMCID: PMC8338758 DOI: 10.1016/j.neurobiolaging.2021.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/30/2021] [Accepted: 04/03/2021] [Indexed: 12/20/2022]
Abstract
The age-related loss of GABA in the inferior colliculus (IC) likely plays a role in the development of age-related hearing loss. Perineuronal nets (PNs), specialized aggregates of extracellular matrix, increase with age in the IC. PNs, associated with GABAergic neurotransmission, can stabilize synapses and inhibit structural plasticity. We sought to determine whether PN expression increased on GABAergic and non-GABAergic IC cells that project to the medial geniculate body (MG). We used retrograde tract-tracing in combination with immunohistochemistry for glutamic acid decarboxylase and Wisteria floribunda agglutinin across three age groups of Fischer Brown Norway rats. Results demonstrate that PNs increase with age on lemniscal and non-lemniscal IC-MG cells, however two key differences exist. First, PNs increased on non-lemniscal IC-MG cells during middle-age, but not until old age on lemniscal IC-MG cells. Second, increases of PNs on lemniscal IC-MG cells occurred on non-GABAergic cells rather than on GABAergic cells. These results suggest that synaptic stabilization and reduced plasticity likely occur at different ages on a subset of the IC-MG pathway.
Collapse
Affiliation(s)
- Amir M Mafi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Matthew G Russ
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Lindsay N Hofer
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Vincent Q Pham
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Jesse W Young
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Jeffrey G Mellott
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA.
| |
Collapse
|
50
|
Miyata S. Structural and Functional Remodeling of the Extracellular Matrix during Brain Development and Aging. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2003.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Shinji Miyata
- Faculty of Agriculture, Tokyo University of Agriculture and Technology
| |
Collapse
|