1
|
Nashawi H, Foltz CT, Smail MA, Buesing DR, Herman JP, Ulrich-Lai YM. The impact of limited sucrose intake on perineuronal nets of parvalbumin interneurons in the basolateral amygdala: A potential role in stress resilience. Physiol Behav 2025; 290:114774. [PMID: 39631451 DOI: 10.1016/j.physbeh.2024.114774] [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: 08/13/2024] [Revised: 10/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Natural rewards like regular sucrose consumption can buffer physiological and behavioral stress responses, likely mediated, at least in part, by increased plasticity in parvalbumin-positive (PV+) interneurons in the basolateral amygdala (BLA). As PV+ interneuron plasticity is tightly regulated by specialized extracellular matrix structures called perineuronal nets (PNNs), this study investigated the impact of regular sucrose consumption vs. repetitive stress on the PNNs that surround PV+ interneurons in the BLA, as well as the number of glutamatergic (vGLUT1) and GABAergic (vGAT) appositions that PV+ cells receive. Male rats were given an established limited sucrose intake (LSI) feeding paradigm (vs. water-fed controls) and were co-exposed to a brief restraint stress (vs. no stress controls), daily for 14 days. Sucrose consumption increased the proportion of PV+ cells that were surrounded by PNNs, independent of stress exposure. PV+ cells with PNNs had more vGLUT1-positive and fewer vGAT-positive appositions compared to those lacking PNNs. Additionally, sucrose consumption increased the ratio of excitatory/inhibitory appositions onto PV+ cells, suggesting the possibility of elevated PV+ interneuron tone, leading to greater inhibition of the BLA's stress-excitatory output. These findings indicate that sucrose consumption influences PNN formation and structural plasticity on PV+ interneurons in the BLA, which has implications for understanding the neurological mechanisms underlying stress resilience by natural rewards.
Collapse
Affiliation(s)
- Houda Nashawi
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Corey T Foltz
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA
| | - Marissa A Smail
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Dana R Buesing
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA
| | - James P Herman
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA
| | - Yvonne M Ulrich-Lai
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
2
|
Wallsten B, Gligor AH, Gonzalez AE, Ramos JD, Baratta MV, Sorg BA. Response of parvalbumin interneurons and perineuronal nets in rat medial prefrontal cortex and lateral amygdala to stressor controllability. Brain Res 2025; 1848:149351. [PMID: 39592089 DOI: 10.1016/j.brainres.2024.149351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024]
Abstract
Behavioral control over a stressor limits the impact of the stressor being experienced and produces enduring changes that reduce the effects of future stressors. In rats, these stress-buffering effects of control (escapable stress, ES) require activation of the medial prefrontal cortex (mPFC) and prevent the typical amygdala-dependent behavioral outcomes of uncontrollable stress (inescapable stress, IS). Parvalbumin (PV) interneurons regulate output of excitatory neurons, and most mPFC PV neurons are surrounded by perineuronal nets (PNNs), which regulate firing. We exposed male rats to a single session of ES, IS, or no stress and measured c-Fos expression within PV/PNN-containing cells in mPFC subregions (prelimbic, PL; infralimbic, IL) and in the lateral amygdala. We also measured the number and intensity of PNNs. Within PL and IL PV/PNN cells, both ES and IS increased c-Fos intensity in PV/PNN, non-PV, and non-PNN cells. Within the IL, only ES increased the number of c-Fos-expressing PV/PNN-labeled cells. In the lateral amygdala, only ES increased c-Fos intensity within PV cells and PV/PNN cells. Thus, PV neurons in the IL and lateral amygdala may represent an important substrate by which behavioral control buffers against the amygdala-dependent behavioral outcomes typically observed after uncontrollable stress.
Collapse
Affiliation(s)
- Brittani Wallsten
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Abigail H Gligor
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Angela E Gonzalez
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States; Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States; Program in Neuroscience, Washington State University, Vancouver, WA 98686, United States
| | - Jonathan D Ramos
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80301, United States
| | - Barbara A Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, United States; Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States; Program in Neuroscience, Washington State University, Vancouver, WA 98686, United States.
| |
Collapse
|
3
|
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
|
4
|
Towner TT, Coleman HJ, Goyden MA, Vore AS, Papastrat KM, Varlinskaya EI, Werner DF. Prelimbic cortex perineuronal net expression and social behavior: Impact of adolescent intermittent ethanol exposure. Neuropharmacology 2025; 262:110195. [PMID: 39437849 DOI: 10.1016/j.neuropharm.2024.110195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/05/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Adolescent intermittent ethanol (AIE) exposure in rats leads to social deficits. Parvalbumin (PV) expressing fast-spiking interneurons in the prelimbic cortex (PrL) contribute to social behavior, and perineuronal nets (PNNs) within the PrL preferentially encompass and regulate PV interneurons. AIE exposure increases PNNs, but it is unknown if this upregulation contributes to AIE-induced social impairments. The current study was designed to determine the effect of AIE exposure on PNN expression in the PrL and to assess whether PNN dysregulation contributes to social deficits elicited by AIE. cFos-LacZ male and female rats were exposed every other day to tap water or ethanol (4 g/kg, 25% w/v) via intragastric gavage between postnatal day (P) 25-45. We evaluated neuronal activation by β-galactosidase expression and PNN levels either at the end of the exposure regimen on P45 and/or in adulthood on P70. In addition, we used Chondroitinase ABC (ChABC) to deplete PNNs following adolescent exposure (P48) and allowed for PNN restoration before social testing in adulthhod. AIE exposure increased PNN expression in the PrL of adult males, but decreased PNNs immediately following AIE. Vesicular glutamate transporter 2 (vGlut2) and vesicular GABA transporter (vGat) near PNNs were downregulated only in AIE-exposed females. Gene expression of PNN components was largely unaffected by AIE exposure. Removal and reestablishment of PrL PNNs by ChABC led to upregulation of PNNs and social impairments in males, regardless of adolescent exposure. These data suggest that AIE exposure in males upregulates PrL PNNs that likely contribute to social impairments induced by AIE.
Collapse
Affiliation(s)
- Trevor T Towner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Harper J Coleman
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Matthew A Goyden
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Andrew S Vore
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Kimberly M Papastrat
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - Elena I Varlinskaya
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA
| | - David F Werner
- Neurobiology of Adolescent Drinking in Adulthood Consortium, Center for Development and Behavioral Neuroscience, Department of Psychology, Binghamton University, Binghamton, NY, 13902-6000, USA.
| |
Collapse
|
5
|
Jabłońska J, Wiera G, Mozrzymas JW. Extracellular matrix integrity regulates GABAergic plasticity in the hippocampus. Matrix Biol 2024; 134:184-196. [PMID: 39491759 DOI: 10.1016/j.matbio.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/18/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
The brain's extracellular matrix (ECM) is crucial for neural circuit functionality, synaptic plasticity, and learning. While the role of the ECM in excitatory synapses has been extensively studied, its influence on inhibitory synapses, particularly on GABAergic long-term plasticity, remains poorly understood. This study aims to elucidate the effects of ECM components on inhibitory synaptic transmission and plasticity in the hippocampal CA1 region. We focus on the roles of chondroitin sulfate proteoglycans (CSPGs) and hyaluronic acid in modulating inhibitory postsynaptic currents (IPSCs) at two distinct inhibitory synapses formed by somatostatin (SST)-positive and parvalbumin (PV)-positive interneurons onto pyramidal cells (PCs). Using optogenetic stimulation in brain slices, we observed that acute degradation of ECM constituents by hyaluronidase or chondroitinase-ABC did not affect basal inhibitory synaptic transmission. However, short-term plasticity, particularly burst-induced depression, was enhanced at PV→PC synapses following enzymatic treatments. Long-term plasticity experiments demonstrated that CSPGs are essential for NMDA-induced iLTP at SST→PC synapses, whereas the digestion of hyaluronic acid by hyaluronidase impaired iLTP at PV→PC synapses. This indicates a synapse-specific role of CSPGs and hyaluronic acid in regulating GABAergic plasticity. Additionally, we report the presence of cryptic GABAergic plasticity at PV→PC synapses induced by prolonged NMDA application, which became evident after CSPG digestion and was absent under control conditions. Our results underscore the differential impact of ECM degradation on inhibitory synaptic plasticity, highlighting the synapse-specific interplay between ECM components and specific GABAergic synapses. This offers new perspectives in studies on learning and critical period timing.
Collapse
Affiliation(s)
- Jadwiga Jabłońska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland
| | - Grzegorz Wiera
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland.
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland.
| |
Collapse
|
6
|
Umbricht D, Kas MJH, Dawson GR. The role of biomarkers in clinical development of drugs for neuropsychiatric disorders - A pragmatic guide. Eur Neuropsychopharmacol 2024; 88:66-77. [PMID: 39236552 DOI: 10.1016/j.euroneuro.2024.08.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024]
Abstract
The failure rate of drugs being developed for neuropsychiatric indications remains high. Optimizing drug discovery and development requires not only a better neurobiological understanding of disease aetiology and development, but also the means by which we can measure relevant biological and clinical processes related to disease progression, drug target engagement, and sensitivity to treatment. Here we address the role and key considerations for the selection of biomarkers in clinical drug development for neuropsychiatric disorders. We do not provide an exhaustive list of biomarkers; rather we lay out a pragmatic, well-defined biomarker selection strategy that addresses the main goals for each of the phases in the drug development cycle. We discuss the key questions and issues that concern biomarker selection and implementation in each phase of development. For the better development of biomarkers, we emphasize the need to focus on discrete biological dysfunction and/or symptom domains rather than diagnoses. We also advocate the use of biomarker-based patient stratification in phase 2 and 3 to increase sensitivity and power and reduce costs. Our aim is to enhance precision and chances of success for these complex and heterogeneous brain disorders with a high unmet medical need.
Collapse
Affiliation(s)
- Daniel Umbricht
- xperimed GmbH, Basel, Switzerland; University of Zurich, Switzerland.
| | - Martien J H Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | | |
Collapse
|
7
|
Ju J, Li X, Pan Y, Du J, Yang X, Men S, Liu B, Zhang Z, Zhong H, Mai J, Wang Y, Hou ST. Adenosine mediates the amelioration of social novelty deficits during rhythmic light treatment of 16p11.2 deletion female mice. Mol Psychiatry 2024; 29:3381-3394. [PMID: 38740879 PMCID: PMC11541200 DOI: 10.1038/s41380-024-02596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/16/2024]
Abstract
Non-invasive brain stimulation therapy for autism spectrum disorder (ASD) has shown beneficial effects. Recently, we and others demonstrated that visual sensory stimulation using rhythmic 40 Hz light flicker effectively improved cognitive deficits in mouse models of Alzheimer's disease and stroke. However, whether rhythmic visual 40 Hz light flicker stimulation can ameliorate behavioral deficits in ASD remains unknown. Here, we show that 16p11.2 deletion female mice exhibit a strong social novelty deficit, which was ameliorated by treatment with a long-term 40 Hz light stimulation. The elevated power of local-field potential (LFP) in the prefrontal cortex (PFC) of 16p11.2 deletion female mice was also effectively reduced by 40 Hz light treatment. Importantly, the 40 Hz light flicker reversed the excessive excitatory neurotransmission of PFC pyramidal neurons without altering the firing rate and the number of resident PFC neurons. Mechanistically, 40 Hz light flicker evoked adenosine release in the PFC to modulate excessive excitatory neurotransmission of 16p11.2 deletion female mice. Elevated adenosine functioned through its cognate A1 receptor (A1R) to suppress excessive excitatory neurotransmission and to alleviate social novelty deficits. Indeed, either blocking the A1R using a specific antagonist DPCPX or knocking down the A1R in the PFC using a shRNA completely ablated the beneficial effects of 40 Hz light flicker. Thus, this study identified adenosine as a novel neurochemical mediator for ameliorating social novelty deficit by reducing excitatory neurotransmission during 40 Hz light flicker treatment. The 40 Hz light stimulation warrants further development as a non-invasive ASD therapeutics.
Collapse
Affiliation(s)
- Jun Ju
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Xuanyi Li
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Yifan Pan
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Jun Du
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, 100850, Beijing, China
| | - Xinyi Yang
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Siqi Men
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Bo Liu
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Zhenyu Zhang
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Haolin Zhong
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Jinyuan Mai
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China
| | - Yizheng Wang
- Huashan Hospital, Fudan University, Shanghai, PR China
| | - Sheng-Tao Hou
- Brain Research Centre, Department of Neuroscience, School of Life Sciences, Southern University of Science and Technology, 1088 Xueyuan Blvd, Nanshan District, Shenzhen, 518055, Guangdong, PR China.
| |
Collapse
|
8
|
Klimczak P, Alcaide J, Gramuntell Y, Castillo-Gómez E, Varea E, Perez-Rando M, Nacher J. Long-term effects of a double hit murine model for schizophrenia on parvalbumin expressing cells and plasticity-related molecules in the thalamic reticular nucleus and the habenula. Transl Psychiatry 2024; 14:450. [PMID: 39448557 PMCID: PMC11502763 DOI: 10.1038/s41398-024-03166-6] [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: 03/07/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
The exposure to aversive experiences during early-life affects brain maturation and induces changes in behavior. Additionally, when these experiences coincide with subtle neurodevelopmental alterations, they may contribute to the emergence of psychiatric disorders, such as schizophrenia. Studies in patients and animal models have identified changes in parvalbumin (PV) expressing inhibitory neurons, highlighting their significance in the etiology of this disorder. Most studies have been focused on the cortex, but PV+ neurons also provide inhibitory input to diencephalic regions, particularly to the thalamus (through cells in the thalamic reticular nucleus, TRN) and the habenula. Remarkably, alterations in both nuclei have been described in schizophrenia. Some of these changes in PV+ cells may be mediated by perineuronal nets (PNN), specialized regions of the extracellular matrix that often surround them and regulate their synaptic input and activity. Interestingly, the physiological maturation and integration of PV+ neurons, which involves the assembly of PNN, occurs during early postnatal life. Plasticity molecules associated to inhibitory neurons, such as PSA-NCAM, or NMDA receptors (NMDAR) can also influence the structure and function of these cells. Growing evidence also indicates that glial cells regulate the physiology of PV+ neurons by influencing their maturation and modulating their synaptic connectivity. To explore the impact of early-life aversive experiences and concomitant subtle neurodevelopmental alterations on diencephalic PV+ cells, we analyzed adult male mice subjected to a double-hit model (DHM) of schizophrenia, combining a single injection of an NMDAR antagonist at P7 and post-weaning social isolation. We observed that exploratory behavior, PV+ neurons and their associated PNN, as well as PSA-NCAM and NMDAR expression and glial cells, in the TRN and the habenula were affected by the DHM or one of its factors. To our knowledge, this is the first report on such alterations in these diencephalic structures in an animal model combining neurodevelopmental alterations and early-life stress during adolescence. Our findings complement previous work on PV+ neurons in cortical regions and underscore the importance of studying diencephalic inhibitory networks and their intricate interactions with aversive experiences and neurodevelopmental alterations during early life in the context of schizophrenia.
Collapse
Affiliation(s)
- Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain
- Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| | - Emilio Varea
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Valencia, 46100, Spain.
- Spanish National Network for Research in Mental Health CIBERSAM, Madrid, 28029, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain.
| |
Collapse
|
9
|
Sterin I, Niazi A, Kim J, Park J, Park S. Dynamic Organization of Neuronal Extracellular Matrix Revealed by HaloTag-HAPLN1. J Neurosci 2024; 44:e0666242024. [PMID: 39251350 PMCID: PMC11502233 DOI: 10.1523/jneurosci.0666-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/04/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows a net-like structure around a subset of neurons, a ring-like structure at the nodes of Ranvier, and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to hyaluronan proteoglycan link protein 1, which links hyaluronan and proteoglycans. Expression or application of the probe in primary rat neuronal cultures enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in mouse brains of either sex reveals detailed ECM architectures around excitatory neurons and developmentally regulated dendritic ECM. Our study uncovers extensive structural features of the brain's ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
- Neuroscience Program, University of Utah, Salt Lake City, Utah 84112
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
10
|
Curto Y, Carceller H, Klimczak P, Perez-Rando M, Wang Q, Grewe K, Kawaguchi R, Rizzoli S, Geschwind D, Nave KA, Teruel-Marti V, Singh M, Ehrenreich H, Nácher J. Erythropoietin restrains the inhibitory potential of interneurons in the mouse hippocampus. Mol Psychiatry 2024; 29:2979-2996. [PMID: 38622200 PMCID: PMC11449791 DOI: 10.1038/s41380-024-02528-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 04/17/2024]
Abstract
Severe psychiatric illnesses, for instance schizophrenia, and affective diseases or autism spectrum disorders, have been associated with cognitive impairment and perturbed excitatory-inhibitory balance in the brain. Effects in juvenile mice can elucidate how erythropoietin (EPO) might aid in rectifying hippocampal transcriptional networks and synaptic structures of pyramidal lineages, conceivably explaining mitigation of neuropsychiatric diseases. An imminent conundrum is how EPO restores synapses by involving interneurons. By analyzing ~12,000 single-nuclei transcriptomic data, we generated a comprehensive molecular atlas of hippocampal interneurons, resolved into 15 interneuron subtypes. Next, we studied molecular alterations upon recombinant human (rh)EPO and saw that gene expression changes relate to synaptic structure, trans-synaptic signaling and intracellular catabolic pathways. Putative ligand-receptor interactions between pyramidal and inhibitory neurons, regulating synaptogenesis, are altered upon rhEPO. An array of in/ex vivo experiments confirms that specific interneuronal populations exhibit reduced dendritic complexity, synaptic connectivity, and changes in plasticity-related molecules. Metabolism and inhibitory potential of interneuron subgroups are compromised, leading to greater excitability of pyramidal neurons. To conclude, improvement by rhEPO of neuropsychiatric phenotypes may partly owe to restrictive control over interneurons, facilitating re-connectivity and synapse development.
Collapse
Affiliation(s)
- Yasmina Curto
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
- Neuroplasticity Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Héctor Carceller
- Neuroplasticity Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
- Spanish National Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Patrycja Klimczak
- Neuroplasticity Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
- Spanish National Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Marta Perez-Rando
- Neuroplasticity Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
- Spanish National Network for Research in Mental Health (CIBERSAM), Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Qing Wang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Katharina Grewe
- Department of Neuro- & Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Riki Kawaguchi
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Silvio Rizzoli
- Department of Neuro- & Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Daniel Geschwind
- Institute of Precision Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany
| | - Vicent Teruel-Marti
- Neuronal Circuits Laboratory, Department of Anatomy and Human Embryology, University of Valencia, Valencia, Spain
| | - Manvendra Singh
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany.
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, City Campus, Göttingen, Germany.
- Georg-August-University, Göttingen, Germany.
- Experimental Medicine, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J 5, Mannheim, Germany.
| | - Juan Nácher
- Neuroplasticity Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain.
- Spanish National Network for Research in Mental Health (CIBERSAM), Madrid, Spain.
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain.
| |
Collapse
|
11
|
Alcaide J, Gramuntell Y, Klimczak P, Bueno-Fernandez C, Garcia-Verellen E, Guicciardini C, Sandi C, Castillo-Gómez E, Crespo C, Perez-Rando M, Nacher J. Long term effects of peripubertal stress on the thalamic reticular nucleus of female and male mice. Neurobiol Dis 2024; 200:106642. [PMID: 39173845 DOI: 10.1016/j.nbd.2024.106642] [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: 07/13/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024] Open
Abstract
Adverse experiences during infancy and adolescence have an important and enduring effect on the brain and are predisposing factors for mental disorders, particularly major depression. This impact is particularly notable in regions with protracted development, such as the prefrontal cortex. The inhibitory neurons of this cortical region are altered by peripubertal stress (PPS), particularly in female mice. In this study we have explored whether the inhibitory circuits of the thalamus are impacted by PPS in male and female mice. This diencephalic structure, as the prefrontal cortex, also completes its development during postnatal life and is affected by adverse experiences. The long-term changes induced by PPS were exclusively found in adult female mice. We have found that PPS increases depressive-like behavior and induces changes in parvalbumin-expressing (PV+) cells of the thalamic reticular nucleus (TRN). We observed reductions in the volume of the TRN, together with those of parameters related to structures/molecules that regulate the plasticity and connectivity of PV+ cells: perineuronal nets, matricellular structures surrounding PV+ neurons, and the polysialylated form of the neural cell adhesion molecule (PSA-NCAM). The expression of the GluN1, but not of GluN2C, NMDA receptor subunit was augmented in the TRN after PPS. An increase in the fluorescence intensity of PV+ puncta was also observed in the synaptic output of TRN neurons in the lateral posterior thalamic nucleus. These results demonstrate that the inhibitory circuits of the thalamus, as those of the prefrontal cortex, are vulnerable to the effects of aversive experiences during early life, particularly in females. This vulnerability is probably related to the protracted development of the TRN and might contribute to the development of psychiatric disorders.
Collapse
Affiliation(s)
- Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Clara Bueno-Fernandez
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Erica Garcia-Verellen
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Chiara Guicciardini
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| | - Carlos Crespo
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain.
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain.
| |
Collapse
|
12
|
Wingert JC, Ramos JD, Reynolds SX, Gonzalez AE, Rose RM, Hegarty DM, Aicher SA, Bailey LG, Brown TE, Abbas AI, Sorg BA. Perineuronal Nets in the Rat Medial Prefrontal Cortex Alter Hippocampal-Prefrontal Oscillations and Reshape Cocaine Self-Administration Memories. J Neurosci 2024; 44:e0468242024. [PMID: 38991791 PMCID: PMC11340292 DOI: 10.1523/jneurosci.0468-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
The medial prefrontal cortex (mPFC) is a major contributor to relapse to cocaine in humans and to reinstatement in rodent models of cocaine use disorder. The output from the mPFC is potently modulated by parvalbumin (PV)-containing fast-spiking interneurons, the majority of which are surrounded by perineuronal nets. We previously showed that treatment with chondroitinase ABC (ABC) reduced the consolidation and reconsolidation of a cocaine conditioned place preference memory. However, self-administration memories are more difficult to disrupt. Here we report in male rats that ABC treatment in the mPFC attenuated the consolidation and blocked the reconsolidation of a cocaine self-administration memory. However, reconsolidation was blocked when rats were given a novel, but not familiar, type of retrieval session. Furthermore, ABC treatment prior to, but not after, memory retrieval blocked reconsolidation. This same treatment did not alter a sucrose memory, indicating specificity for cocaine-induced memory. In naive rats, ABC treatment in the mPFC altered levels of PV intensity and cell firing properties. In vivo recordings from the mPFC and dorsal hippocampus (dHIP) during the novel retrieval session revealed that ABC prevented reward-associated increases in high-frequency oscillations and synchrony of these oscillations between the dHIP and mPFC. Together, this is the first study to show that ABC treatment disrupts reconsolidation of the original memory when combined with a novel retrieval session that elicits coupling between the dHIP and mPFC. This coupling after ABC treatment may serve as a fundamental signature for how to disrupt reconsolidation of cocaine memories and reduce relapse.
Collapse
Affiliation(s)
- Jereme C Wingert
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | - Jonathan D Ramos
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | | | - Angela E Gonzalez
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
- Program in Neuroscience, Washington State University, Vancouver, Washington 98686
| | - R Mae Rose
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
| | - Deborah M Hegarty
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Sue A Aicher
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239
| | - Lydia G Bailey
- Program in Neuroscience, Washington State University, Pullman, Washington 99164
| | - Travis E Brown
- Program in Neuroscience, Washington State University, Pullman, Washington 99164
| | - Atheir I Abbas
- Departments of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
- Psychiatry, Oregon Health & Science University, Portland, Oregon 97239
- Research Division, VA Portland Health Care System, Portland, Oregon 97239
| | - Barbara A Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, Oregon 97232
- Program in Neuroscience, Washington State University, Vancouver, Washington 98686
| |
Collapse
|
13
|
Théberge S, Belliveau C, Xie D, Khalaf R, Perlman K, Rahimian R, Davoli MA, Turecki G, Mechawar N. Parvalbumin interneurons in human ventromedial prefrontal cortex: a comprehensive post-mortem study of myelination and perineuronal nets in neurotypical individuals and depressed suicides with and without a history of child abuse. Cereb Cortex 2024; 34:bhae197. [PMID: 38760318 PMCID: PMC11101286 DOI: 10.1093/cercor/bhae197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
Cortical parvalbumin interneurons (PV+) are major regulators of excitatory/inhibitory information processing, and their maturation is associated with the opening of developmental critical periods (CP). Recent studies reveal that cortical PV+ axons are myelinated, and that myelination along with perineuronal net (PNN) maturation around PV+ cells is associated with the closures of CP. Although PV+ interneurons are susceptible to early-life stress, their relationship between their myelination and PNN coverage remains unexplored. This study compared the fine features of PV+ interneurons in well-characterized human post-mortem ventromedial prefrontal cortex samples (n = 31) from depressed suicides with or without a history of child abuse (CA) and matched controls. In healthy controls, 81% of all sampled PV+ interneurons displayed a myelinated axon, while a subset (66%) of these cells also displayed a PNN, proposing a relationship between both attributes. Intriguingly, a 3-fold increase in the proportion of unmyelinated PV+ interneurons with a PNN was observed in CA victims, along with greater PV-immunofluorescence intensity in myelinated PV+ cells with a PNN. This study, which is the first to provide normative data on myelination and PNNs around PV+ interneurons in human neocortex, sheds further light on the cellular and molecular consequences of early-life adversity on cortical PV+ interneurons.
Collapse
Affiliation(s)
- Stéphanie Théberge
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Av des Pins Ouest, H3A 1A1, Montreal, QC, Canada
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Av des Pins Ouest, H3A 1A1, Montreal, QC, Canada
| | - Dongyue Xie
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
| | - Roy Khalaf
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
| | - Kelly Perlman
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Av des Pins Ouest, H3A 1A1, Montreal, QC, Canada
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
| | - Maria Antonietta Davoli
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Av des Pins Ouest, H3A 1A1, Montreal, QC, Canada
- Department of Psychiatry, McGill University, 1033 Av des Pins Ouest, H3A 1A1, Montréal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, H4H 1R3, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Av des Pins Ouest, H3A 1A1, Montreal, QC, Canada
- Department of Psychiatry, McGill University, 1033 Av des Pins Ouest, H3A 1A1, Montréal, QC, Canada
| |
Collapse
|
14
|
Spoleti E, La Barbera L, Cauzzi E, De Paolis ML, Saba L, Marino R, Sciamanna G, Di Lazzaro V, Keller F, Nobili A, Krashia P, D'Amelio M. Dopamine neuron degeneration in the Ventral Tegmental Area causes hippocampal hyperexcitability in experimental Alzheimer's Disease. Mol Psychiatry 2024; 29:1265-1280. [PMID: 38228889 PMCID: PMC11189820 DOI: 10.1038/s41380-024-02408-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Early and progressive dysfunctions of the dopaminergic system from the Ventral Tegmental Area (VTA) have been described in Alzheimer's Disease (AD). During the long pre-symptomatic phase, alterations in the function of Parvalbumin interneurons (PV-INs) are also observed, resulting in cortical hyperexcitability represented by subclinical epilepsy and aberrant gamma-oscillations. However, it is unknown whether the dopaminergic deficits contribute to brain hyperexcitability in AD. Here, using the Tg2576 mouse model of AD, we prove that reduced hippocampal dopaminergic innervation, due to VTA dopamine neuron degeneration, impairs PV-IN firing and gamma-waves, weakens the inhibition of pyramidal neurons and induces hippocampal hyperexcitability via lower D2-receptor-mediated activation of the CREB-pathway. These alterations coincide with reduced PV-IN numbers and Perineuronal Net density. Importantly, L-DOPA and the selective D2-receptor agonist quinpirole rescue p-CREB levels and improve the PV-IN-mediated inhibition, thus reducing hyperexcitability. Moreover, similarly to quinpirole, sumanirole - another D2-receptor agonist and a known anticonvulsant - not only increases p-CREB levels in PV-INs but also restores gamma-oscillations in Tg2576 mice. Conversely, blocking the dopaminergic transmission with sulpiride (a D2-like receptor antagonist) in WT mice reduces p-CREB levels in PV-INs, mimicking what occurs in Tg2576. Overall, these findings support the hypothesis that the VTA dopaminergic system integrity plays a key role in hippocampal PV-IN function and survival, disclosing a relevant contribution of the reduced dopaminergic tone to aberrant gamma-waves, hippocampal hyperexcitability and epileptiform activity in early AD.
Collapse
Affiliation(s)
- Elena Spoleti
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Emma Cauzzi
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Luana Saba
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Ramona Marino
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Giuseppe Sciamanna
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- UniCamillus International University of Health Sciences, 00131, Rome, Italy
| | - Vincenzo Di Lazzaro
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Operative Research Unit of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Rome, Italy
| | - Flavio Keller
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Paraskevi Krashia
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy.
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.
| |
Collapse
|
15
|
Jakovljević A, Stamenković V, Poleksić J, Hamad MIK, Reiss G, Jakovcevski I, Andjus PR. The Role of Tenascin-C on the Structural Plasticity of Perineuronal Nets and Synaptic Expression in the Hippocampus of Male Mice. Biomolecules 2024; 14:508. [PMID: 38672524 PMCID: PMC11047978 DOI: 10.3390/biom14040508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Neuronal plasticity is a crucial mechanism for an adapting nervous system to change. It is shown to be regulated by perineuronal nets (PNNs), the condensed forms of the extracellular matrix (ECM) around neuronal bodies. By assessing the changes in the number, intensity, and structure of PNNs, the ultrastructure of the PNN mesh, and the expression of inhibitory and excitatory synaptic inputs on these neurons, we aimed to clarify the role of an ECM glycoprotein, tenascin-C (TnC), in the dorsal hippocampus. To enhance neuronal plasticity, TnC-deficient (TnC-/-) and wild-type (TnC+/+) young adult male mice were reared in an enriched environment (EE) for 8 weeks. Deletion of TnC in TnC-/- mice showed an ultrastructural reduction of the PNN mesh and an increased inhibitory input in the dentate gyrus (DG), and an increase in the number of PNNs with a rise in the inhibitory input in the CA2 region. EE induced an increased inhibitory input in the CA2, CA3, and DG regions; in DG, the change was also followed by an increased intensity of PNNs. No changes in PNNs or synaptic expression were found in the CA1 region. We conclude that the DG and CA2 regions emerged as focal points of alterations in PNNs and synaptogenesis with EE as mediated by TnC.
Collapse
Affiliation(s)
- Ana Jakovljević
- Center for Laser Microscopy, Institute for Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vera Stamenković
- Center for Integrative Brain Research, Seattle Children’s Research Institute, 1900 9th Ave, Seattle, WA 98125, USA;
| | - Joko Poleksić
- Institute of Anatomy “Niko Miljanic”, School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Gebhard Reiss
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Pavle R. Andjus
- Center for Laser Microscopy, Institute for Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
16
|
Kumar P, Goettemoeller AM, Espinosa-Garcia C, Tobin BR, Tfaily A, Nelson RS, Natu A, Dammer EB, Santiago JV, Malepati S, Cheng L, Xiao H, Duong DD, Seyfried NT, Wood LB, Rowan MJM, Rangaraju S. Native-state proteomics of Parvalbumin interneurons identifies unique molecular signatures and vulnerabilities to early Alzheimer's pathology. Nat Commun 2024; 15:2823. [PMID: 38561349 PMCID: PMC10985119 DOI: 10.1038/s41467-024-47028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Dysfunction in fast-spiking parvalbumin interneurons (PV-INs) may represent an early pathophysiological perturbation in Alzheimer's Disease (AD). Defining early proteomic alterations in PV-INs can provide key biological and translationally-relevant insights. We used cell-type-specific in-vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state PV-IN proteomes. PV-IN proteomic signatures include high metabolic and translational activity, with over-representation of AD-risk and cognitive resilience-related proteins. In bulk proteomes, PV-IN proteins were associated with cognitive decline in humans, and with progressive neuropathology in humans and the 5xFAD mouse model of Aβ pathology. PV-IN CIBOP in early stages of Aβ pathology revealed signatures of increased mitochondria and metabolism, synaptic and cytoskeletal disruption and decreased mTOR signaling, not apparent in whole-brain proteomes. Furthermore, we demonstrated pre-synaptic defects in PV-to-excitatory neurotransmission, validating our proteomic findings. Overall, in this study we present native-state proteomes of PV-INs, revealing molecular insights into their unique roles in cognitive resiliency and AD pathogenesis.
Collapse
Affiliation(s)
- Prateek Kumar
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Annie M Goettemoeller
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Claudia Espinosa-Garcia
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Brendan R Tobin
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Ali Tfaily
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Ruth S Nelson
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Aditya Natu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric B Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Juliet V Santiago
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Sneha Malepati
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lihong Cheng
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Duc D Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Levi B Wood
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
- School of Chemical and Biological Engineering, GeoInsrgia titute of Technology, Atlanta, GA, 30322, USA
| | - Matthew J M Rowan
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Srikant Rangaraju
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
17
|
Wingert JC, Ramos JD, Reynolds SX, Gonzalez AE, Rose RM, Hegarty DM, Aicher SA, Bailey LG, Brown TE, Abbas AI, Sorg BA. Perineuronal nets in the rat medial prefrontal cortex alter hippocampal-prefrontal oscillations and reshape cocaine self-administration memories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.577568. [PMID: 38370716 PMCID: PMC10871211 DOI: 10.1101/2024.02.05.577568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
The medial prefrontal cortex (mPFC) is a major contributor to relapse to cocaine in humans and to reinstatement behavior in rodent models of cocaine use disorder. Output from the mPFC is modulated by parvalbumin (PV)-containing fast-spiking interneurons, the majority of which are surrounded by perineuronal nets (PNNs). Here we tested whether chondroitinase ABC (ABC)- mediated removal of PNNs prevented the acquisition or reconsolidation of a cocaine self-administration memory. ABC injections into the dorsal mPFC prior to training attenuated the acquisition of cocaine self-administration. Also, ABC given 3 days prior to but not 1 hr after memory reactivation blocked cue-induced reinstatement. However, reduced reinstatement was present only in rats given a novel reactivation contingency, suggesting that PNNs are required for the updating of a familiar memory. In naive rats, ABC injections into mPFC did not alter excitatory or inhibitory puncta on PV cells but reduced PV intensity. Whole-cell recordings revealed a greater inter-spike interval 1 hr after ABC, but not 3 days later. In vivo recordings from the mPFC and dorsal hippocampus (dHIP) during novel memory reactivation revealed that ABC in the mPFC prevented reward-associated increases in beta and gamma activity as well as phase-amplitude coupling between the dHIP and mPFC. Together, our findings show that PNN removal attenuates the acquisition of cocaine self-administration memories and disrupts reconsolidation of the original memory when combined with a novel reactivation session. Further, reduced dHIP/mPFC coupling after PNN removal may serve as a key biomarker for how to disrupt reconsolidation of cocaine memories and reduce relapse.
Collapse
|
18
|
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
|
19
|
Wang AS, Wan X, Storch DS, Li VY, Cornez G, Balthazart J, Cisneros-Franco JM, de Villers-Sidani E, Sakata JT. Cross-species conservation in the regulation of parvalbumin by perineuronal nets. Front Neural Circuits 2023; 17:1297643. [PMID: 38179221 PMCID: PMC10766385 DOI: 10.3389/fncir.2023.1297643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024] Open
Abstract
Parvalbumin (PV) neurons play an integral role in regulating neural dynamics and plasticity. Therefore, understanding the factors that regulate PV expression is important for revealing modulators of brain function. While the contribution of PV neurons to neural processes has been studied in mammals, relatively little is known about PV function in non-mammalian species, and discerning similarities in the regulation of PV across species can provide insight into evolutionary conservation in the role of PV neurons. Here we investigated factors that affect the abundance of PV in PV neurons in sensory and motor circuits of songbirds and rodents. In particular, we examined the degree to which perineuronal nets (PNNs), extracellular matrices that preferentially surround PV neurons, modulate PV abundance as well as how the relationship between PV and PNN expression differs across brain areas and species and changes over development. We generally found that cortical PV neurons that are surrounded by PNNs (PV+PNN neurons) are more enriched with PV than PV neurons without PNNs (PV-PNN neurons) across both rodents and songbirds. Interestingly, the relationship between PV and PNN expression in the vocal portion of the basal ganglia of songbirds (Area X) differed from that in other areas, with PV+PNN neurons having lower PV expression compared to PV-PNN neurons. These relationships remained consistent across development in vocal motor circuits of the songbird brain. Finally, we discovered a causal contribution of PNNs to PV expression in songbirds because degradation of PNNs led to a diminution of PV expression in PV neurons. These findings reveal a conserved relationship between PV and PNN expression in sensory and motor cortices and across songbirds and rodents and suggest that PV neurons could modulate plasticity and neural dynamics in similar ways across songbirds and rodents.
Collapse
Affiliation(s)
- Angela S. Wang
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Xinghaoyun Wan
- Department of Biology, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | | | - Vivian Y. Li
- Department of Biology, McGill University, Montreal, QC, Canada
| | - Gilles Cornez
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | - Jacques Balthazart
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | | | - Etienne de Villers-Sidani
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Jon T. Sakata
- Department of Biology, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, QC, Canada
| |
Collapse
|
20
|
Smail MA, Smith BL, Shukla R, Alganem K, Eby HM, Bollinger JL, Parikh RK, Chambers JB, Reigle JK, Moloney RD, Nawreen N, Wohleb ES, Pantazopoulos H, McCullumsmith RE, Herman JP. Molecular neurobiology of loss: a role for basolateral amygdala extracellular matrix. Mol Psychiatry 2023; 28:4729-4741. [PMID: 37644175 PMCID: PMC10914625 DOI: 10.1038/s41380-023-02231-8] [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: 02/06/2023] [Revised: 08/01/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
Psychological loss is a common experience that erodes well-being and negatively impacts quality of life. The molecular underpinnings of loss are poorly understood. Here, we investigate the mechanisms of loss using an environmental enrichment removal (ER) paradigm in male rats. The basolateral amygdala (BLA) was identified as a region of interest, demonstrating differential Fos responsivity to ER and having an established role in stress processing and adaptation. A comprehensive multi-omics investigation of the BLA, spanning multiple cohorts, platforms, and analyses, revealed alterations in microglia and the extracellular matrix (ECM). Follow-up studies indicated that ER decreased microglia size, complexity, and phagocytosis, suggesting reduced immune surveillance. Loss also substantially increased ECM coverage, specifically targeting perineuronal nets surrounding parvalbumin interneurons, suggesting decreased plasticity and increased inhibition within the BLA following loss. Behavioral analyses suggest that these molecular effects are linked to impaired BLA salience evaluation, leading to a mismatch between stimulus and reaction intensity. These loss-like behaviors could be rescued by depleting BLA ECM during the removal period, helping us understand the mechanisms underlying loss and revealing novel molecular targets to ameliorate its impact.
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA.
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA.
| | - Brittany L Smith
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Rammohan Shukla
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Khaled Alganem
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Hunter M Eby
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Ria K Parikh
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - James B Chambers
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - James K Reigle
- Department of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rachel D Moloney
- School of Pharmacy, University College Cork, Cork, Ireland, UK
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland, UK
- APC Microbiome Ireland, University College Cork, Cork, Ireland, UK
| | - Nawshaba Nawreen
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Harry Pantazopoulos
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, USA
| | - Robert E McCullumsmith
- Department of Neurosciences, University of Toledo, Toledo, OH, USA
- Neurosciences Institute, ProMedica, Toledo, OH, USA
| | - James P Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Veterans Affairs Medical Center, Cincinnati, OH, USA
- Department of Neurology, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
21
|
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: 2] [Impact Index Per Article: 1.0] [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
|
22
|
Wang AS, Wan X, Storch DS, Cornez G, Balthazart J, Cisneros-Franco JM, de Villers-Sidani E, Sakata JT. Cross-species conservation in the regulation of parvalbumin by perineuronal nets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.13.557580. [PMID: 37745532 PMCID: PMC10515890 DOI: 10.1101/2023.09.13.557580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Parvalbumin (PV) neurons play an integral role in regulating neural dynamics and plasticity. Therefore, understanding the factors that regulate PV expression is important for revealing modulators of brain function. While the contribution of PV neurons to neural processes has been studied in mammals, relatively little is known about PV function in non-mammalian species, and discerning similarities in the regulation of PV across species can provide insight into evolutionary conservation in the role of PV neurons. Here we investigated factors that affect the abundance of PV in PV neurons in sensory and motor circuits of songbirds and rodents. In particular, we examined the degree to which perineuronal nets (PNNs), extracellular matrices that preferentially surround PV neurons, modulate PV abundance as well as how the relationship between PV and PNN expression differs across brain areas and species and changes over development. We generally found that cortical PV neurons that are surrounded by PNNs (PV+PNN neurons) are more enriched with PV than PV neurons without PNNs (PV-PNN neurons) across both rodents and songbirds. Interestingly, the relationship between PV and PNN expression in the vocal portion of the basal ganglia of songbirds (Area X) differed from that in other areas, with PV+PNN neurons having lower PV expression compared to PV-PNN neurons. These relationships remained consistent across development in vocal motor circuits of the songbird brain. Finally, we discovered a causal contribution of PNNs to PV expression in songbirds because degradation of PNNs led to a diminution of PV expression in PV neurons. These findings in reveal a conserved relationship between PV and PNN expression in sensory and motor cortices and across songbirds and rodents and suggest that PV neurons could modulate plasticity and neural dynamics in similar ways across songbirds and rodents.
Collapse
Affiliation(s)
- Angela S. Wang
- Department of Biology, McGill University, Montreal, Canada
| | - Xinghaoyun Wan
- Department of Biology, McGill University, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | | | - Gilles Cornez
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | - Jacques Balthazart
- Laboratory of Behavioral Neuroendocrinology, GIGA Neurosciences, University of Liege, Liege, Belgium
| | | | - Etienne de Villers-Sidani
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, Canada
| | - Jon T. Sakata
- Department of Biology, McGill University, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Centre for Research in Brain, Language and Music, McGill University, Montreal, Canada
| |
Collapse
|
23
|
Lupori L, Totaro V, Cornuti S, Ciampi L, Carrara F, Grilli E, Viglione A, Tozzi F, Putignano E, Mazziotti R, Amato G, Gennaro C, Tognini P, Pizzorusso T. A comprehensive atlas of perineuronal net distribution and colocalization with parvalbumin in the adult mouse brain. Cell Rep 2023; 42:112788. [PMID: 37436896 DOI: 10.1016/j.celrep.2023.112788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/03/2023] [Accepted: 06/25/2023] [Indexed: 07/14/2023] Open
Abstract
Perineuronal nets (PNNs) surround specific neurons in the brain and are involved in various forms of plasticity and clinical conditions. However, our understanding of the PNN role in these phenomena is limited by the lack of highly quantitative maps of PNN distribution and association with specific cell types. Here, we present a comprehensive atlas of Wisteria floribunda agglutinin (WFA)-positive PNNs and colocalization with parvalbumin (PV) cells for over 600 regions of the adult mouse brain. Data analysis shows that PV expression is a good predictor of PNN aggregation. In the cortex, PNNs are dramatically enriched in layer 4 of all primary sensory areas in correlation with thalamocortical input density, and their distribution mirrors intracortical connectivity patterns. Gene expression analysis identifies many PNN-correlated genes. Strikingly, PNN-anticorrelated transcripts are enriched in synaptic plasticity genes, generalizing PNNs' role as circuit stability factors.
Collapse
Affiliation(s)
| | | | - Sara Cornuti
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Luca Ciampi
- Institute of Information Science and Technologies (ISTI-CNR), 56124 Pisa, Italy
| | - Fabio Carrara
- Institute of Information Science and Technologies (ISTI-CNR), 56124 Pisa, Italy
| | - Edda Grilli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | | | | | | | | | - Giuseppe Amato
- Institute of Information Science and Technologies (ISTI-CNR), 56124 Pisa, Italy
| | - Claudio Gennaro
- Institute of Information Science and Technologies (ISTI-CNR), 56124 Pisa, Italy
| | - Paola Tognini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Tommaso Pizzorusso
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy; Institute of Neuroscience (IN-CNR), 56124 Pisa, Italy.
| |
Collapse
|
24
|
Engel K, Lee HN, Tewari BP, Lewkowicz AP, Ireland DDC, Manangeeswaran M, Verthelyi D. Neonatal Zika virus infection causes transient perineuronal net degradation. Front Cell Neurosci 2023; 17:1187425. [PMID: 37496706 PMCID: PMC10366369 DOI: 10.3389/fncel.2023.1187425] [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: 03/16/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023] Open
Abstract
Perineuronal nets (PNNs) form a specialized extracellular matrix that predominantly surrounds parvalbumin (PV)-expressing GABAergic inhibitory interneurons and help regulate neuronal activity. Their formation early in the postnatal period is regulated by neuronal signaling and glial activation raising concerns that part of the long-term effects ascribed to perinatal viral infections could be mediated by altered PNN formation. Previously, we developed a model of neonatal Zika virus (ZIKV) infection where mice have lifelong neurological sequelae that includes motor disfunction and reduced anxiety coupled with a persistent low-grade expression in proinflammatory markers despite resolving the acute infection. Here, we demonstrate that ZIKV infection to P1 neonatal mice results in a reduction of PNN formation during the acute disease with significant reduction in Wisteria floribunda agglutinin (WFA) staining at the peak of infection [15 days post infection (dpi)] that persisted after the symptoms resolved (30 dpi). At 60 dpi, when there is residual inflammation in the CNS, the number of WFA+ cells and the level of WFA staining as well as levels of aggrecan and brevican in the brains of convalescent mice were not different from those in uninfected controls, however, there was increased frequency of PNNs with an immature phenotype. Over time the impact of the perinatal infection became less evident and there were no clear differences in PNN morphology between the groups at 1 year post infection. Of note, the reduction in PNNs during acute ZIKV infection was not associated with decreased mRNA levels of aggrecan or brevican, but increased levels of degraded aggrecan and brevican indicating increased PNN degradation. These changes were associated with increased expression of matrix metalloproteinase 12 (MMP12) and MMP19, but not MMP9, a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) or ADAMTS5. Together our findings indicate that infection at the time of PNN development interferes with PNN formation, but the nets can reform once the infection and inflammation subside.
Collapse
Affiliation(s)
- Kaliroi Engel
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ha-Na Lee
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Bhanu P. Tewari
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Aaron P. Lewkowicz
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Derek D. C. Ireland
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Mohanraj Manangeeswaran
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Daniela Verthelyi
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
25
|
Lavertu-Jolin M, Chattopadhyaya B, Chehrazi P, Carrier D, Wünnemann F, Leclerc S, Dumouchel F, Robertson D, Affia H, Saba K, Gopal V, Patel AB, Andelfinger G, Pineyro G, Di Cristo G. Acan downregulation in parvalbumin GABAergic cells reduces spontaneous recovery of fear memories. Mol Psychiatry 2023; 28:2946-2963. [PMID: 37131076 PMCID: PMC10615765 DOI: 10.1038/s41380-023-02085-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/04/2023]
Abstract
While persistence of fear memories is essential for survival, a failure to inhibit fear in response to harmless stimuli is a feature of anxiety disorders. Extinction training only temporarily suppresses fear memory recovery in adults, but it is highly effective in juvenile rodents. Maturation of GABAergic circuits, in particular of parvalbumin-positive (PV+) cells, restricts plasticity in the adult brain, thus reducing PV+ cell maturation could promote the suppression of fear memories following extinction training in adults. Epigenetic modifications such as histone acetylation control gene accessibility for transcription and help couple synaptic activity to changes in gene expression. Histone deacetylase 2 (Hdac2), in particular, restrains both structural and functional synaptic plasticity. However, whether and how Hdac2 controls the maturation of postnatal PV+ cells is not well understood. Here, we show that PV+- cell specific Hdac2 deletion limits spontaneous fear memory recovery in adult mice, while enhancing PV+ cell bouton remodeling and reducing perineuronal net aggregation around PV+ cells in prefrontal cortex and basolateral amygdala. Prefrontal cortex PV+ cells lacking Hdac2, show reduced expression of Acan, a critical perineuronal net component, which is rescued by Hdac2 re-expression. Pharmacological inhibition of Hdac2 before extinction training is sufficient to reduce both spontaneous fear memory recovery and Acan expression in wild-type adult mice, while these effects are occluded in PV+-cell specific Hdac2 conditional knockout mice. Finally, a brief knock-down of Acan expression mediated by intravenous siRNA delivery before extinction training but after fear memory acquisition is sufficient to reduce spontaneous fear recovery in wild-type mice. Altogether, these data suggest that controlled manipulation of PV+ cells by targeting Hdac2 activity, or the expression of its downstream effector Acan, promotes the long-term efficacy of extinction training in adults.
Collapse
Affiliation(s)
- Marisol Lavertu-Jolin
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | | | - Pegah Chehrazi
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Denise Carrier
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
| | - Florian Wünnemann
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
- Heidelberg University, Faculty of Medicine & Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Séverine Leclerc
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
| | - Félix Dumouchel
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Derek Robertson
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
| | - Hicham Affia
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
| | - Kamal Saba
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vijaya Gopal
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Anant Bahadur Patel
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Gregor Andelfinger
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| | - Graçiela Pineyro
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada
| | - Graziella Di Cristo
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, QC, Canada.
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
26
|
Biro L, Miskolczi C, Szebik H, Bruzsik B, Varga ZK, Szente L, Toth M, Halasz J, Mikics E. Post-weaning social isolation in male mice leads to abnormal aggression and disrupted network organization in the prefrontal cortex: Contribution of parvalbumin interneurons with or without perineuronal nets. Neurobiol Stress 2023; 25:100546. [PMID: 37323648 PMCID: PMC10265620 DOI: 10.1016/j.ynstr.2023.100546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023] Open
Abstract
Adverse social experiences during childhood increase the risk of developing aggression-related psychopathologies. The prefrontal cortex (PFC) is a key regulator of social behavior, where experience-dependent network development is tied to the maturation of parvalbumin-positive (PV+) interneurons. Maltreatment in childhood could impact PFC development and lead to disturbances in social behavior during later life. However, our knowledge regarding the impact of early-life social stress on PFC operation and PV+ cell function is still scarce. Here, we used post-weaning social isolation (PWSI) to model early-life social neglect in mice and to study the associated neuronal changes in the PFC, additionally distinguishing between the two main subpopulations of PV+ interneurons, i.e. those without or those enwrapped by perineuronal nets (PNN). For the first time to such detailed extent in mice, we show that PWSI induced disturbances in social behavior, including abnormal aggression, excessive vigilance and fragmented behavioral organization. PWSI mice showed altered resting-state and fighting-induced co-activation patterns between orbitofrontal and medial PFC (mPFC) subregions, with a particularly highly elevated activity in the mPFC. Surprisingly, aggressive interaction was associated with a higher recruitment of mPFC PV+ neurons that were surrounded by PNN in PWSI mice that seemed to mediate the emergence of social deficits. PWSI did not affect the number of PV+ neurons and PNN density, but enhanced PV and PNN intensity as well as cortical and subcortical glutamatergic drive onto mPFC PV+ neurons. Our results suggest that the increased excitatory input of PV+ cells could emerge as a compensatory mechanism for the PV+ neuron-mediated impaired inhibition of mPFC layer 5 pyramidal neurons, since we found lower numbers of GABAergic PV+ puncta on the perisomatic region of these cells. In conclusion, PWSI leads to altered PV-PNN activity and impaired excitatory/inhibitory balance in the mPFC, which possibly contributes to social behavioral disruptions seen in PWSI mice. Our data advances our understanding on how early-life social stress can impact the maturing PFC and lead to the development of social abnormalities in adulthood.
Collapse
Affiliation(s)
- Laszlo Biro
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
| | - Christina Miskolczi
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
- Janos Szentagothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Ulloi ut 26., Hungary
| | - Huba Szebik
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
- Janos Szentagothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Ulloi ut 26., Hungary
| | - Biborka Bruzsik
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
| | - Zoltan Kristof Varga
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
| | - Laszlo Szente
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
- Janos Szentagothai Doctoral School of Neurosciences, Semmelweis University, 1085 Budapest, Ulloi ut 26., Hungary
| | - Mate Toth
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
| | - Jozsef Halasz
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
| | - Eva Mikics
- Institute of Experimental Medicine, Laboratory of Translational Behavioural Neuroscience, 1083 Budapest, Szigony utca 43., Hungary
| |
Collapse
|
27
|
Kumar P, Goettemoeller AM, Espinosa-Garcia C, Tobin BR, Tfaily A, Nelson RS, Natu A, Dammer EB, Santiago JV, Malepati S, Cheng L, Xiao H, Duong D, Seyfried NT, Wood LB, Rowan MJ, Rangaraju S. Native-state proteomics of Parvalbumin interneurons identifies novel molecular signatures and metabolic vulnerabilities to early Alzheimer's disease pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541038. [PMID: 37292756 PMCID: PMC10245729 DOI: 10.1101/2023.05.17.541038] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One of the earliest pathophysiological perturbations in Alzheimer's Disease (AD) may arise from dysfunction of fast-spiking parvalbumin (PV) interneurons (PV-INs). Defining early protein-level (proteomic) alterations in PV-INs can provide key biological and translationally relevant insights. Here, we use cell-type-specific in vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state proteomes of PV interneurons. PV-INs exhibited proteomic signatures of high metabolic, mitochondrial, and translational activity, with over-representation of causally linked AD genetic risk factors. Analyses of bulk brain proteomes indicated strong correlations between PV-IN proteins with cognitive decline in humans, and with progressive neuropathology in humans and mouse models of Aβ pathology. Furthermore, PV-IN-specific proteomes revealed unique signatures of increased mitochondrial and metabolic proteins, but decreased synaptic and mTOR signaling proteins in response to early Aβ pathology. PV-specific changes were not apparent in whole-brain proteomes. These findings showcase the first native state PV-IN proteomes in mammalian brain, revealing a molecular basis for their unique vulnerabilities in AD.
Collapse
|
28
|
Chehrazi P, Lee KKY, Lavertu-Jolin M, Abbasnejad Z, Carreño-Muñoz MI, Chattopadhyaya B, Di Cristo G. p75 neurotrophin receptor in pre-adolescent prefrontal PV interneurons promotes cognitive flexibility in adult mice. Biol Psychiatry 2023:S0006-3223(23)01238-6. [PMID: 37120061 DOI: 10.1016/j.biopsych.2023.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/31/2023] [Accepted: 04/16/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND Parvalbumin (PV)-positive GABAergic cells provide robust perisomatic inhibition to neighboring pyramidal neurons and regulate brain oscillations. Alterations in PV interneuron connectivity and function in the medial prefrontal cortex (mPFC) have been consistently reported in psychiatric disorders associated with cognitive rigidity, suggesting that PV cell deficits could be a core cellular phenotype in these disorders. p75 neurotrophin receptor (p75NTR) regulates the time course of PV cell maturation in a cell-autonomous fashion. Whether p75NTR expression during postnatal development affects adult prefrontal PV cell connectivity and cognitive function is unknown. METHODS We generated transgenic mice with conditional knockout (cKO) of p75NTR in postnatal PV cells. We analysed PV cell connectivity and recruitment following a tail pinch, by immunolabeling and confocal imaging, in naïve mice or following p75NTR re-expression in pre- or post-adolescent mice using Cre-dependent viral vectors. Cognitive flexibility was evaluated using behavioral tests. RESULTS PV cell-specific p75NTR deletion increased both PV cell synapse density and the proportion of PV cells surrounded by perineuronal nets, a marker of mature PV cells, in adult mPFC but not visual cortex. Both phenotypes were rescued by viral-mediated re-introduction of p75NTR in pre-adolescent but not post-adolescent mPFC. Prefrontal cortical PV cells failed to upregulate c-Fos following a tail-pinch stimulation in adult cKO mice. Finally, cKO mice showed impaired fear memory extinction learning as well as deficits in a attention set-shifting task. CONCLUSION These findings suggest that p75NTR expression in adolescent PV cells contributes to the fine tuning of their connectivity and promotes cognitive flexibility in adulthood.
Collapse
Affiliation(s)
- Pegah Chehrazi
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, Canada; Department of Neurosciences, Université de Montréal, Montréal, Canada
| | - Karen Ka Yan Lee
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, Canada; Department of Neurosciences, Université de Montréal, Montréal, Canada
| | - Marisol Lavertu-Jolin
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, Canada; Department of Neurosciences, Université de Montréal, Montréal, Canada
| | - Zahra Abbasnejad
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, Canada; Department of Neurosciences, Université de Montréal, Montréal, Canada
| | - Maria Isabel Carreño-Muñoz
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, Canada; Department of Neurosciences, Université de Montréal, Montréal, Canada
| | | | - Graziella Di Cristo
- Centre de Recherche, CHU Sainte-Justine (CHUSJ), Montréal, Canada; Department of Neurosciences, Université de Montréal, Montréal, Canada.
| |
Collapse
|
29
|
Reduced inhibitory and excitatory input onto parvalbumin interneurons mediated by perineuronal net might contribute to cognitive impairments in a mouse model of sepsis-associated encephalopathy. Neuropharmacology 2023; 225:109382. [PMID: 36543316 DOI: 10.1016/j.neuropharm.2022.109382] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is commonly defined as diffuse brain dysfunction and can manifest as delirium to coma. Accumulating evidence has suggested that perineuronal net (PNN) plays an important role in the modulation of the synaptic plasticity of central nervous system. We here investigated the role of PNN in SAE induced by lipopolysaccharide (LPS) injection. Behavioral tests were performed by open field, Y-maze, and fear conditioning tests at the indicated time points. The densities of vesicular γ-aminobutyric acid transporter, vesicular glutamate transporter 1, PNN, and parvalbumin (PV) in the hippocampus were evaluated by immunofluorescence. Matrix metalloproteinases-9 (MMP-9) expression and its activity were detected by Western blot and gel zymography, respectively. Local field potential was recorded by in vivo electrophysiology. LPS-treated mice displayed significant cognitive impairments, coincided with activated MMP-9, decreased PNN and PV densities, reduced inhibitory and excitatory input onto PV interneurons enwrapped by PNN, and decreased gamma oscillations in hippocampal CA1. Notably, MMP-9 inhibitor SB-3CT treatment rescued most of these abnormalities. Taken together, our study demonstrates that active MMP-9 mediated PNN remodeling, leading to reduced inhibitory and excitatory input onto PV interneurons and abnormal gamma oscillations in hippocampal CA1, which consequently contributed to cognitive impairments after LPS injection.
Collapse
|
30
|
Tewari BP, Woo AM, Prim CE, Chaunsali L, Kimbrough IF, Engel K, Browning JL, Campbell SL, Sontheimer H. Perineuronal nets support astrocytic ion and glutamate homeostasis at tripartite synapses. RESEARCH SQUARE 2023:rs.3.rs-2501039. [PMID: 36778342 PMCID: PMC9915772 DOI: 10.21203/rs.3.rs-2501039/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Perineuronal nets (PNNs) are dense, negatively charged extracellular matrices that cover the cell body of fast-spiking inhibitory neurons. Synapses can be embedded and stabilized by PNNs believed to prevent synaptic plasticity. We find that in cortical fast-spiking interneurons synaptic terminals localize to perforations in the PNNs, 95% of which contain either excitatory or inhibitory synapses or both. The majority of terminals also colocalize with astrocytic processes expressing Kir4.1 as well as glutamate (Glu) and GABA transporters, hence can be considered tripartite synapses. In the adult brain, degradation of PNNs does not alter axonal terminals but causes expansion of astrocytic coverage of the neuronal somata. However, loss of PNNs impairs astrocytic transmitter and K+ uptake and causes spillage of synaptic Glu into the extrasynaptic space. This data suggests a hitherto unrecognized role of PNNs, to synergize with astrocytes to contain synaptically released signals.
Collapse
Affiliation(s)
- Bhanu P. Tewari
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - AnnaLin M. Woo
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Courtney E. Prim
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lata Chaunsali
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ian F. Kimbrough
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kaliroi Engel
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | | | | | - Harald Sontheimer
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
31
|
Casquero-Veiga M, Lamanna-Rama N, Romero-Miguel D, Rojas-Marquez H, Alcaide J, Beltran M, Nacher J, Desco M, Soto-Montenegro ML. The Poly I:C maternal immune stimulation model shows unique patterns of brain metabolism, morphometry, and plasticity in female rats. Front Behav Neurosci 2023; 16:1022622. [PMID: 36733452 PMCID: PMC9888250 DOI: 10.3389/fnbeh.2022.1022622] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction: Prenatal infections are associated with an increased risk of the onset of schizophrenia. Rodent models of maternal immune stimulation (MIS) have been extensively used in preclinical studies. However, many of these studies only include males, omitting pathophysiological features unique to females. The aim of this study is to characterize the MIS model in female rats using positron emission tomography (PET), structural magnetic resonance imaging (MR), and neuroplasticiy studies. Methods: In gestational day 15, Poly I:C (or Saline) was injected into pregnant Wistar rats to induce the MIS model. Imaging studies: [18F]-fluoro-2-deoxy-D-glucose-PET scans of female-offspring were acquired at post-natal day (PND) 35 and PND100. Furthermore, T2-MR brain images were acquired in adulthood. Differences in FDG uptake and morphometry between groups were assessed with SPM12 and Regions of Interest (ROI) analyses. Ex vivo study: The density of parvalbumin expressing interneurons (PV), perineuronal nets (PNN), and parvalbumin expressing interneurons surrounded by perineuronal nets (PV-PNN) were evaluated in the prelimbic cortex and basolateral amygdala using confocal microscopy. ROIs and neuroplasticity data were analyzed by 2-sample T-test and 2-way-ANOVA analyses, respectively. Results: A significant increase in brain metabolism was found in all animals at adulthood compared to adolescence. MIS hardly modified brain glucose metabolism in females, highlighting a significant hypometabolism in the thalamus at adulthood. In addition, MIS induced gray matter (GM) enlargements in the pituitary, hippocampus, substantia nigra, and cingulate cortex, and GM shrinkages in some thalamic nuclei, cerebelar areas, and brainstem. Moreover, MIS induced white matter shrinkages in the cerebellum, brainstem and corpus callosum, along with cerebrospinal fluid enlargements in the lateral and 4th ventricles. Finally, MIS reduced the density of PV, PNN, and PV-PNN in the basolateral amygdala. Conclusion: Our work showed in vivo the differential pattern of functional and morphometric affectation in the MIS model in females, as well as the deficits caused at the synaptic level according to sex. The differences obtained highlight the relevance of including both sexes in psychiatric research in order to consider their pathophysiological particularities and successfully extend the benefits obtained to the entire patient population.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Cardiovascular Imaging and Population Studies, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Nicolás Lamanna-Rama
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Escuela Técnica Superior de Ingeniería, Universidad Carlos III de Madrid, Madrid, Spain
| | - Diego Romero-Miguel
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Escuela Técnica Superior de Ingeniería, Universidad Carlos III de Madrid, Madrid, Spain
| | - Henar Rojas-Marquez
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain,Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Julia Alcaide
- Neurobiology Unit, Cell Biology Departament, BIOTECMED Institute, Universitat de València, Burjassot, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Marc Beltran
- Neurobiology Unit, Cell Biology Departament, BIOTECMED Institute, Universitat de València, Burjassot, Spain
| | - Juan Nacher
- Neurobiology Unit, Cell Biology Departament, BIOTECMED Institute, Universitat de València, Burjassot, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| | - Manuel Desco
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Advanced Imaging Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Campus de Getafe, Madrid, Spain,*Correspondence: Manuel Desco Maria Luisa Soto-Montenegro
| | - Maria Luisa Soto-Montenegro
- Laboratorio de Imagen Médica, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Alcorcón, Spain,*Correspondence: Manuel Desco Maria Luisa Soto-Montenegro
| |
Collapse
|
32
|
Mueller-Buehl C, Wegrzyn D, Bauch J, Faissner A. Regulation of the E/I-balance by the neural matrisome. Front Mol Neurosci 2023; 16:1102334. [PMID: 37143468 PMCID: PMC10151766 DOI: 10.3389/fnmol.2023.1102334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
In the mammalian cortex a proper excitatory/inhibitory (E/I) balance is fundamental for cognitive functions. Especially γ-aminobutyric acid (GABA)-releasing interneurons regulate the activity of excitatory projection neurons which form the second main class of neurons in the cortex. During development, the maturation of fast-spiking parvalbumin-expressing interneurons goes along with the formation of net-like structures covering their soma and proximal dendrites. These so-called perineuronal nets (PNNs) represent a specialized form of the extracellular matrix (ECM, also designated as matrisome) that stabilize structural synapses but prevent the formation of new connections. Consequently, PNNs are highly involved in the regulation of the synaptic balance. Previous studies revealed that the formation of perineuronal nets is accompanied by an establishment of mature neuronal circuits and by a closure of critical windows of synaptic plasticity. Furthermore, it has been shown that PNNs differentially impinge the integrity of excitatory and inhibitory synapses. In various neurological and neuropsychiatric disorders alterations of PNNs were described and aroused more attention in the last years. The following review gives an update about the role of PNNs for the maturation of parvalbumin-expressing interneurons and summarizes recent findings about the impact of PNNs in different neurological and neuropsychiatric disorders like schizophrenia or epilepsy. A targeted manipulation of PNNs might provide an interesting new possibility to indirectly modulate the synaptic balance and the E/I ratio in pathological conditions.
Collapse
|
33
|
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
|
34
|
Brown TE, Sorg BA. Net gain and loss: influence of natural rewards and drugs of abuse on perineuronal nets. Neuropsychopharmacology 2023; 48:3-20. [PMID: 35568740 PMCID: PMC9700711 DOI: 10.1038/s41386-022-01337-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/26/2022]
Abstract
Overindulgence, excessive consumption, and a pattern of compulsive use of natural rewards, such as certain foods or drugs of abuse, may result in the development of obesity or substance use disorder, respectively. Natural rewards and drugs of abuse can trigger similar changes in the neurobiological substrates that drive food- and drug-seeking behaviors. This review examines the impact natural rewards and drugs of abuse have on perineuronal nets (PNNs). PNNs are specialized extracellular matrix structures that ensheathe certain neurons during development over the critical period to provide synaptic stabilization and a protective microenvironment for the cells they surround. This review also analyzes how natural rewards and drugs of abuse impact the density and maturation of PNNs within reward-associated circuitry of the brain, which may contribute to maladaptive food- and drug-seeking behaviors. Finally, we evaluate the relatively few studies that have degraded PNNs to perturb reward-seeking behaviors. Taken together, this review sheds light on the complex way PNNs are regulated by natural rewards and drugs and highlights a need for future studies to delineate the molecular mechanisms that underlie the modification and maintenance of PNNs following exposure to rewarding stimuli.
Collapse
Affiliation(s)
- Travis E Brown
- Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, 99164, USA.
| | - Barbara A Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR, 97232, USA
| |
Collapse
|
35
|
Spencer KM, Nakhnikian A, Hirano Y, Levin M. The contribution of gamma bursting to spontaneous gamma activity in schizophrenia. Front Hum Neurosci 2023; 17:1130897. [PMID: 37206313 PMCID: PMC10188978 DOI: 10.3389/fnhum.2023.1130897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/07/2023] [Indexed: 05/21/2023] Open
Abstract
Increased spontaneous gamma (30-100 Hz) activity (SGA) has been reported in the auditory cortex in schizophrenia. This phenomenon has been correlated with psychotic symptoms such as auditory hallucinations and could reflect the dysfunction of NMDA receptors on parvalbumin-expressing inhibitory interneurons. Previous findings are from time-averaged spectra, so it is unknown whether increased spontaneous gamma occurs at a constant level, or rather in bursts. To better understand the dynamical nature of spontaneous gamma activity in schizophrenia, here we examined the contribution of gamma bursting and the slope of the EEG spectrum to this phenomenon. The main results from this data set were previously reported. Participants were 24 healthy control participants (HC) and 24 matched participants with schizophrenia (SZ). The data were from EEG recordings during auditory steady-state stimulation, which were localized to bilateral pairs of dipoles in auditory cortex. Time-frequency analysis was performed using Morlet wavelets. Oscillation bursts in the gamma range were defined as periods during which power exceeded 2 standard deviations above the trial-wide average value for at least one cycle. We extracted the burst parameters power, count, and area, as well as non-burst trial power and spectral slope. Gamma burst power and non-burst trial power were greater in SZ than HC, but burst count and area did not differ. Spectral slope was less negative in SZ than HC. Regression modeling found that gamma burst power alone best predicted SGA for both HC and SZ (> = 90% of variance), while spectral slope made a small contribution and non-burst trial power did not influence SGA. Increased SGA in the auditory cortex in schizophrenia is accounted for by increased power within gamma bursts, rather than a tonic increase in gamma-range activity, or a shift in spectral slope. Further research will be necessary to determine if these measures reflect different network mechanisms. We propose that increased gamma burst power is the main component of increased SGA in SZ and could reflect abnormally increased plasticity in cortical circuits due to enhanced plasticity of synapses on parvalbumin-expressing inhibitory interneurons. Thus, increased gamma burst power may be involved in producing psychotic symptoms and cognitive dysfunction.
Collapse
Affiliation(s)
- Kevin M. Spencer
- Research Service, VA Boston Healthcare System, Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- *Correspondence: Kevin M. Spencer,
| | - Alexander Nakhnikian
- Research Service, VA Boston Healthcare System, Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Yoji Hirano
- Research Service, VA Boston Healthcare System, Department of Psychiatry, Harvard Medical School, Boston, MA, United States
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
36
|
Laham BJ, Murthy SS, Hanani M, Clappier M, Boyer S, Vasquez B, Gould E. The estrous cycle modulates early-life adversity effects on mouse avoidance behavior through progesterone signaling. Nat Commun 2022; 13:7537. [PMID: 36476469 PMCID: PMC9729614 DOI: 10.1038/s41467-022-35068-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Early-life adversity (ELA) increases the likelihood of neuropsychiatric diagnoses, which are more prevalent in women than men. Since changes in reproductive hormone levels can also increase the probability of anxiety disorders in women, we examined the effects of ELA on adult female mice across the estrous cycle. We found that during diestrus, when progesterone levels are relatively high, ELA mice exhibit increased avoidance behavior and increased theta oscillation power in the ventral hippocampus (vHIP). We also found that diestrus ELA mice had higher levels of progesterone and lower levels of allopregnanolone, a neurosteroid metabolite of progesterone, in the vHIP compared with control-reared mice. Progesterone receptor antagonism normalized avoidance behavior in ELA mice, while treatment with a negative allosteric modulator of allopregnanolone promoted avoidance behavior in control mice. These results suggest that altered vHIP progesterone and allopregnanolone signaling during diestrus increases avoidance behavior in ELA mice.
Collapse
Affiliation(s)
- Blake J Laham
- Princeton Neuroscience Institute, Princeton, NJ, 08450, USA
| | | | - Monica Hanani
- Princeton Neuroscience Institute, Princeton, NJ, 08450, USA
| | - Mona Clappier
- Princeton Neuroscience Institute, Princeton, NJ, 08450, USA
| | - Sydney Boyer
- Princeton Neuroscience Institute, Princeton, NJ, 08450, USA
| | - Betsy Vasquez
- Princeton Neuroscience Institute, Princeton, NJ, 08450, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton, NJ, 08450, USA.
| |
Collapse
|
37
|
Jakovljevic A, Agatonovic G, Aleksic D, Aksic M, Reiss G, Förster E, Stamatakis A, Jakovcevski I, Poleksic J. The impact of early life maternal deprivation on the perineuronal nets in the prefrontal cortex and hippocampus of young adult rats. Front Cell Dev Biol 2022; 10. [PMID: 36518543 PMCID: PMC9742529 DOI: 10.3389/fcell.2022.982663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2024] Open
Abstract
Early life stress negatively impacts brain development and affects structure and function of parvalbumin immunopositive (PV+) inhibitory neurons. Main regulators of PV+ interneurons activity and plasticity are perineuronal nets (PNNs), an extracellular matrix formation that enwraps PV+ interneurons mainly in the neocortex and hippocampus. To experimentally address the impact of early life stress on the PNNs and PV+ interneurons in the medial prefrontal cortex and dorsal hippocampus in rats, we employed a 24 h maternal deprivation protocol. We show that maternal deprivation in the medial prefrontal cortex of adult rats caused a decrease in density of overall PNNs and PNNs that enwrap PV+ interneurons in the rostral cingulate cortex. Furthermore, a staining intensity decrease of overall PNNs and PNN+/PV+ cells was found in the prelimbic cortex. Finally, a decrease in both intensity and density of overall PNNs and PNNs surrounding PV+ cells was observed in the infralimbic cortex, together with increase in the intensity of VGAT inhibitory puncta. Surprisingly, maternal deprivation did not cause any changes in the density of PV+ interneurons in the mPFC, neither had it affected PNNs and PV+ interneurons in the hippocampus. Taken together, our findings indicate that PNNs, specifically the ones enwrapping PV+ interneurons in the medial prefrontal cortex, are affected by early life stress.
Collapse
|
38
|
Ali AB, Islam A, Constanti A. The fate of interneurons, GABA A receptor sub-types and perineuronal nets in Alzheimer's disease. Brain Pathol 2022; 33:e13129. [PMID: 36409151 PMCID: PMC9836378 DOI: 10.1111/bpa.13129] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurological disease, which is associated with gradual memory loss and correlated with synaptic hyperactivity and abnormal oscillatory rhythmic brain activity that precedes phenotypic alterations and is partly responsible for the spread of the disease pathology. Synaptic hyperactivity is thought to be because of alteration in the homeostasis of phasic and tonic synaptic inhibition, which is orchestrated by the GABAA inhibitory system, encompassing subclasses of interneurons and GABAA receptors, which play a vital role in cognitive functions, including learning and memory. Furthermore, the extracellular matrix, the perineuronal nets (PNNs) which often go unnoticed in considerations of AD pathology, encapsulate the inhibitory cells and neurites in critical brain regions and have recently come under the light for their crucial role in synaptic stabilisation and excitatory-inhibitory balance and when disrupted, serve as a potential trigger for AD-associated synaptic imbalance. Therefore, in this review, we summarise the current understanding of the selective vulnerability of distinct interneuron subtypes, their synaptic and extrasynaptic GABAA R subtypes as well as the changes in PNNs in AD, detailing their contribution to the mechanisms of disease development. We aim to highlight how seemingly unique malfunction in each component of the interneuronal GABA inhibitory system can be tied together to result in critical circuit dysfunction, leading to the irreversible symptomatic damage observed in AD.
Collapse
|
39
|
John U, Patro N, Patro I. Perineuronal nets: Cruise from a honeycomb to the safety nets. Brain Res Bull 2022; 190:179-194. [DOI: 10.1016/j.brainresbull.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/17/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
|
40
|
Tewari BP, Chaunsali L, Prim CE, Sontheimer H. A glial perspective on the extracellular matrix and perineuronal net remodeling in the central nervous system. Front Cell Neurosci 2022; 16:1022754. [PMID: 36339816 PMCID: PMC9630365 DOI: 10.3389/fncel.2022.1022754] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
A structural scaffold embedding brain cells and vasculature is known as extracellular matrix (ECM). The physical appearance of ECM in the central nervous system (CNS) ranges from a diffused, homogeneous, amorphous, and nearly omnipresent matrix to highly organized distinct morphologies such as basement membranes and perineuronal nets (PNNs). ECM changes its composition and organization during development, adulthood, aging, and in several CNS pathologies. This spatiotemporal dynamic nature of the ECM and PNNs brings a unique versatility to their functions spanning from neurogenesis, cell migration and differentiation, axonal growth, and pathfinding cues, etc., in the developing brain, to stabilizing synapses, neuromodulation, and being an active partner of tetrapartite synapses in the adult brain. The malleability of ECM and PNNs is governed by both intrinsic and extrinsic factors. Glial cells are among the major extrinsic factors that facilitate the remodeling of ECM and PNN, thereby acting as key regulators of diverse functions of ECM and PNN in health and diseases. In this review, we discuss recent advances in our understanding of PNNs and how glial cells are central to ECM and PNN remodeling in normal and pathological states of the CNS.
Collapse
|
41
|
Gonzalez AE, Jorgensen ET, Ramos JD, Harkness JH, Aadland JA, Brown TE, Sorg BA. Impact of Perineuronal Net Removal in the Rat Medial Prefrontal Cortex on Parvalbumin Interneurons After Reinstatement of Cocaine Conditioned Place Preference. Front Cell Neurosci 2022; 16:932391. [PMID: 35966203 PMCID: PMC9366391 DOI: 10.3389/fncel.2022.932391] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
Parvalbumin (PV)-positive cells are GABAergic fast-spiking interneurons that modulate the activity of pyramidal neurons in the medial prefrontal cortex (mPFC) and their output to brain areas associated with learning and memory. The majority of PV cells within the mPFC are surrounded by a specialized extracellular matrix structure called the perineuronal net (PNN). We have shown that removal of PNNs with the enzyme chondroitinase-ABC (Ch-ABC) in the mPFC prevents the consolidation and reconsolidation of cocaine-associated conditioned place preference (CPP) memories. Here we examined the extent to which retrieval of a CPP memory during cocaine-primed reinstatement altered the levels and function of PV neurons and their surrounding PNNs during the reconsolidation period. We further determined the extent to which PNN removal prior to reinstatement altered PV intensity levels and PV cell function. Male Sprague-Dawley rats were trained for cocaine-induced conditioned place preference (CPP) followed by extinction training, microinjection of Ch-ABC in the prelimbic PFC, and cocaine-induced reinstatement. Rats were sacrificed immediately prior to reinstatement or at 2 h, 6 h, or 48 h after reinstatement for immunohistochemistry or 2 h later for electrophysiology. Our findings indicate that PNN removal only partially diminished reinstatement. Cocaine-primed reinstatement produced only minor changes in PNN or PV intensity in vehicle controls. However, after PNN removal, the intensity of remaining PNN-surrounded PV cells was decreased at all times except at 2 h post-reinstatement, at which time cocaine increased PV intensity. Consistent with this, in vehicle controls, PV neurons naturally devoid of PNNs showed a similar pattern to Ch-ABC-treated rats prior to and after cocaine reinstatement, suggesting a protective effect of PNNs on cocaine-induced changes in PV intensity. Using whole-cell patch-clamp, cocaine-primed reinstatement in Ch-ABC-treated rats decreased the number of elicited action potentials but increased excitatory synaptic transmission, which may have been compensatory. These findings suggest that without PNNs, cocaine-induced reinstatement produces rapid changes in PV intensity and PV cell excitability, which may in turn regulate output of the mPFC post-memory retrieval and diminish the maintenance of cocaine memory during reconsolidation.
Collapse
Affiliation(s)
- Angela E. Gonzalez
- Program in Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, United States
- Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Emily T. Jorgensen
- Neuroscience Graduate Program and School of Pharmacy, University of Wyoming, Laramie, WY, United States
| | - Jonathan D. Ramos
- Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| | | | - Jake A. Aadland
- Neuroscience Graduate Program and School of Pharmacy, University of Wyoming, Laramie, WY, United States
| | - Travis E. Brown
- Neuroscience Graduate Program and School of Pharmacy, University of Wyoming, Laramie, WY, United States
| | - Barbara A. Sorg
- Program in Integrative Physiology and Neuroscience, Washington State University, Vancouver, WA, United States
- Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
- *Correspondence: Barbara A. Sorg
| |
Collapse
|
42
|
Gilfarb RA, Leuner B. GABA System Modifications During Periods of Hormonal Flux Across the Female Lifespan. Front Behav Neurosci 2022; 16:802530. [PMID: 35783228 PMCID: PMC9245048 DOI: 10.3389/fnbeh.2022.802530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/21/2022] [Indexed: 01/10/2023] Open
Abstract
The female lifespan is marked by periods of dramatic hormonal fluctuation. Changes in the ovarian hormones estradiol and progesterone, in addition to the progesterone metabolite allopregnanolone, are among the most significant and have been shown to have widespread effects on the brain. This review summarizes current understanding of alterations that occur within the GABA system during the major hormonal transition periods of puberty, the ovarian cycle, pregnancy and the postpartum period, as well as reproductive aging. The functional impacts of altered inhibitory activity during these times are also discussed. Lastly, avenues for future research are identified, which, if pursued, can broaden understanding of the GABA system in the female brain and potentially lead to better treatments for women experiencing changes in brain function at each of these hormonal transition periods.
Collapse
Affiliation(s)
- Rachel A. Gilfarb
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Benedetta Leuner
- Department of Psychology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
- *Correspondence: Benedetta Leuner,
| |
Collapse
|
43
|
Gibel-Russo R, Benacom D, Di Nardo AA. Non-Cell-Autonomous Factors Implicated in Parvalbumin Interneuron Maturation and Critical Periods. Front Neural Circuits 2022; 16:875873. [PMID: 35601531 PMCID: PMC9115720 DOI: 10.3389/fncir.2022.875873] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
From birth to adolescence, the brain adapts to its environmental stimuli through structural and functional remodeling of neural circuits during critical periods of heightened plasticity. They occur across modalities for proper sensory, motor, linguistic, and cognitive development. If they are disrupted by early-life adverse experiences or genetic deficiencies, lasting consequences include behavioral changes, physiological and cognitive deficits, or psychiatric illness. Critical period timing is orchestrated not only by appropriate neural activity but also by a multitude of signals that participate in the maturation of fast-spiking parvalbumin interneurons and the consolidation of neural circuits. In this review, we describe the various signaling factors that initiate critical period onset, such as BDNF, SPARCL1, or OTX2, which originate either from local neurons or glial cells or from extracortical sources such as the choroid plexus. Critical period closure is established by signals that modulate extracellular matrix and myelination, while timing and plasticity can also be influenced by circadian rhythms and by hormones and corticosteroids that affect brain oxidative stress levels or immune response. Molecular outcomes include lasting epigenetic changes which themselves can be considered signals that shape downstream cross-modal critical periods. Comprehensive knowledge of how these signals and signaling factors interplay to influence neural mechanisms will help provide an inclusive perspective on the effects of early adversity and developmental defects that permanently change perception and behavior.
Collapse
|
44
|
Pesarico AP, Carceller H, Guirado R, Coviello S, Nacher J. Long term effects of 24-h-restraint stress on the connectivity and structure of interneurons in the basolateral amygdala. Prog Neuropsychopharmacol Biol Psychiatry 2022; 115:110512. [PMID: 35066055 DOI: 10.1016/j.pnpbp.2022.110512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/15/2022]
Abstract
The effects of intense stressors can last a long time and may lead to the development of psychiatric disorders, including posttraumatic stress disorder. The basolateral amygdala (BLA) plays a critical role in these diseases and is extremely sensitive to stress. Here, we explored in male and female mice the long-term (35 days) impact of a 24-h restraint stress on BLA circuitry. We used Thy1-YFP mice to discriminate 2 subpopulations of excitatory neurons, which participate in "Fear-On" (Thy1-) and "Fear-Off" (Thy1+) circuits. The stress decreased the density of parvalbumin (PV) + inhibitory neurons in both sexes but did not alter their dendritic complexity. We also analyzed the perisomatic input of basket interneurons on Thy1+ and Thy1- neurons, finding sex dependent effects. In males, we did not find alterations in the density of PV+ puncta or in that of cannabinoid receptor 1 (CB1R) + puncta from cholecystokinin+ basket cells. By contrast, in females we found increased the density of PV+ puncta on Thy1+ neurons and reduced on the Thy1- neurons. This adverse experience also reduced in the long term the density of CB1R+ puncta both on Thy1+ and Thy1- cells in females. The expression of the activity marker FosB was not altered in PV+ interneurons and in Thy1+ neurons of stressed animals. The density of perineuronal nets, a specialized region of the extracellular matrix, which covers particularly PV+ interneurons and regulates their connectivity, was increased by stress in male mice. Our findings indicate that a single stressful event can produce long-term alterations in the inhibitory circuits of the BLA, especially on PV+ neurons and their plasticity, and that there is a differential impact depending on the sex and the fear-related circuits involved.
Collapse
Affiliation(s)
- Ana Paula Pesarico
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Hector Carceller
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
| | - Ramón Guirado
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Simona Coviello
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain.
| |
Collapse
|
45
|
Brandenburg C, Blatt GJ. Region-Specific Alterations of Perineuronal Net Expression in Postmortem Autism Brain Tissue. Front Mol Neurosci 2022; 15:838918. [PMID: 35493330 PMCID: PMC9043328 DOI: 10.3389/fnmol.2022.838918] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic variance in autism spectrum disorder (ASD) is often associated with mechanisms that broadly fall into the category of neuroplasticity. Parvalbumin positive neurons and their surrounding perineuronal nets (PNNs) are important factors in critical period plasticity and have both been implicated in ASD. PNNs are found in high density within output structures of the cerebellum and basal ganglia, two regions that are densely connected to many other brain areas and have the potential to participate in the diverse array of symptoms present in an ASD diagnosis. The dentate nucleus (DN) and globus pallidus (GP) were therefore assessed for differences in PNN expression in human postmortem ASD brain tissue. While Purkinje cell loss is a consistent neuropathological finding in ASD, in this cohort, the Purkinje cell targets within the DN did not show differences in number of cells with or without a PNN. However, the density of parvalbumin positive neurons with a PNN were significantly reduced in the GP internus and externus of ASD cases, which was not dependent on seizure status. It is unclear whether these alterations manifest during development or are a consequence of activity-dependent mechanisms that lead to altered network dynamics later in life.
Collapse
Affiliation(s)
- Cheryl Brandenburg
- Hussman Institute for Autism, Baltimore, MD, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: Cheryl Brandenburg,
| | - Gene J. Blatt
- Hussman Institute for Autism, Baltimore, MD, United States
| |
Collapse
|
46
|
Towner TT, Applegate DT, Varlinskaya EI, Werner DF. Impact of Adolescent Intermittent Ethanol Exposure on Social Investigation, Social Preference, and Neuronal Activation in cFos-LacZ Male and Female Rats. Front Pharmacol 2022; 13:841657. [PMID: 35401161 PMCID: PMC8984146 DOI: 10.3389/fphar.2022.841657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/28/2022] [Indexed: 12/03/2022] Open
Abstract
Adolescence is a sensitive developmental period during which alcohol use is often initiated and consumed in high quantities, often at binge or even high-intensity drinking levels. Our lab has repeatedly found that adolescent intermittent ethanol (AIE) exposure in rats results in long-lasting social impairments, specifically in males, however our knowledge of the neuronal underpinnings to this sex-specific effect of AIE is limited. The present study was designed to test whether social anxiety-like alterations in AIE-exposed males would be accompanied by alterations of neuronal activation across brain regions associated with social behavior, with AIE females demonstrating no social impairments and alterations in neuronal activation. Adolescent male and female cFos-LacZ transgenic rats on a Sprague-Dawley background were exposed to ethanol (4 g/kg, 25% v/v) or water via intragastric gavage every other day during postnatal days (P) 25–45 for a total of 11 exposures (n = 13 per group). Social behavior of adult rats was assessed on P70 using a modified social interaction test, and neuronal activation in brain regions implicated in social responding was assessed via β-galactosidase (β-gal) expression. We found that AIE exposure in males resulted in a significantly lower social preference coefficient relative to water-exposed controls, with no effect evident in females. Exposure-specific relationships between social behavior and neuronal activation were identified, with AIE eliminating correlations found in water controls related to social interaction, and eliciting negative correlations mainly in limbic regions in a sex-specific manner. AIE exposure in the absence of social testing was also found to differentially affect neural activity in the orbitofrontal cortex and central amygdala in males and females. These data suggest that AIE produces sex-specific social impairments that are potentially driven by differential neuronal activation states in regions important for social behavior, including the medial prefrontal and orbitofrontal cortices, nucleus accumbens, lateral septum, and central amygdala. Future studies should be focused on identification of specific neuronal phenotypes activated by interaction with a social partner in AIE-exposed subjects and their control counterparts.
Collapse
|
47
|
Herrmann T, Gerth M, Dittmann R, Pensold D, Ungelenk M, Liebmann L, Hübner CA. Disruption of KCC2 in Parvalbumin-Positive Interneurons Is Associated With a Decreased Seizure Threshold and a Progressive Loss of Parvalbumin-Positive Interneurons. Front Mol Neurosci 2022; 14:807090. [PMID: 35185464 PMCID: PMC8850922 DOI: 10.3389/fnmol.2021.807090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023] Open
Abstract
GABAA receptors are ligand-gated ion channels, which are predominantly permeable for chloride. The neuronal K-Cl cotransporter KCC2 lowers the intraneuronal chloride concentration and thus plays an important role for GABA signaling. KCC2 loss-of-function is associated with seizures and epilepsy. Here, we show that KCC2 is expressed in the majority of parvalbumin-positive interneurons (PV-INs) of the mouse brain. PV-INs receive excitatory input from principle cells and in turn control principle cell activity by perisomatic inhibition and inhibitory input from other interneurons. Upon Cre-mediated disruption of KCC2 in mice, the polarity of the GABA response of PV-INs changed from hyperpolarization to depolarization for the majority of PV-INs. Reduced excitatory postsynaptic potential-spike (E-S) coupling and increased spontaneous inhibitory postsynaptic current (sIPSC) frequencies further suggest that PV-INs are disinhibited upon disruption of KCC2. In vivo, PV-IN-specific KCC2 knockout mice display a reduced seizure threshold and develop spontaneous sometimes fatal seizures. We further found a time dependent loss of PV-INs, which was preceded by an up-regulation of pro-apoptotic genes upon disruption of KCC2.
Collapse
|
48
|
Gómez de San José N, Massa F, Halbgebauer S, Oeckl P, Steinacker P, Otto M. Neuronal pentraxins as biomarkers of synaptic activity: from physiological functions to pathological changes in neurodegeneration. J Neural Transm (Vienna) 2022; 129:207-230. [PMID: 34460014 PMCID: PMC8866268 DOI: 10.1007/s00702-021-02411-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/17/2021] [Indexed: 12/22/2022]
Abstract
The diagnosis of neurodegenerative disorders is often challenging due to the lack of diagnostic tools, comorbidities and shared pathological manifestations. Synaptic dysfunction is an early pathological event in many neurodegenerative disorders, but the underpinning mechanisms are still poorly characterised. Reliable quantification of synaptic damage is crucial to understand the pathophysiology of neurodegeneration, to track disease status and to obtain prognostic information. Neuronal pentraxins (NPTXs) are extracellular scaffolding proteins emerging as potential biomarkers of synaptic dysfunction in neurodegeneration. They are a family of proteins involved in homeostatic synaptic plasticity by recruiting post-synaptic receptors into synapses. Recent research investigates the dynamic changes of NPTXs in the cerebrospinal fluid (CSF) as an expression of synaptic damage, possibly related to cognitive impairment. In this review, we summarise the available data on NPTXs structure and expression patterns as well as on their contribution in synaptic function and plasticity and other less well-characterised roles. Moreover, we propose a mechanism for their involvement in synaptic damage and neurodegeneration and assess their potential as CSF biomarkers for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Federico Massa
- Department of Neurology, University of Ulm, Ulm, Germany
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | | | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE E.V.), Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120, Halle (Saale), Germany.
| |
Collapse
|
49
|
Laham BJ, Gould E. How Stress Influences the Dynamic Plasticity of the Brain’s Extracellular Matrix. Front Cell Neurosci 2022; 15:814287. [PMID: 35145379 PMCID: PMC8821883 DOI: 10.3389/fncel.2021.814287] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse and structured extracellular matrix (ECM) comprise ∼20% of the brain’s volume and play important roles in development and adult plasticity. Perineuronal nets (PNNs), specialized ECM structures that surround certain types of neurons in the brain, emerge during the postnatal period, making their development and maintenance potentially sensitive to experience. Recent studies have shown that stress affects diffuse ECM as well as PNNs, and that such effects are dependent on life stage and brain region. Given that the ECM participates in synaptic plasticity, the generation of neuronal oscillations, and synchronous firing across brain regions, all of which have been linked to cognition and emotional regulation, ECM components may be candidate therapeutic targets for stress-induced neuropsychiatric disease. This review considers the influence of stress over diffuse and structured ECM during postnatal life with a focus on functional outcomes and the potential for translational relevance.
Collapse
|
50
|
Warm D, Schroer J, Sinning A. Gabaergic Interneurons in Early Brain Development: Conducting and Orchestrated by Cortical Network Activity. Front Mol Neurosci 2022; 14:807969. [PMID: 35046773 PMCID: PMC8763242 DOI: 10.3389/fnmol.2021.807969] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/06/2021] [Indexed: 01/22/2023] Open
Abstract
Throughout early phases of brain development, the two main neural signaling mechanisms—excitation and inhibition—are dynamically sculpted in the neocortex to establish primary functions. Despite its relatively late formation and persistent developmental changes, the GABAergic system promotes the ordered shaping of neuronal circuits at the structural and functional levels. Within this frame, interneurons participate first in spontaneous and later in sensory-evoked activity patterns that precede cortical functions of the mature brain. Upon their subcortical generation, interneurons in the embryonic brain must first orderly migrate to and settle in respective target layers before they can actively engage in cortical network activity. During this process, changes at the molecular and synaptic level of interneurons allow not only their coordinated formation but also the pruning of connections as well as excitatory and inhibitory synapses. At the postsynaptic site, the shift of GABAergic signaling from an excitatory towards an inhibitory response is required to enable synchronization within cortical networks. Concomitantly, the progressive specification of different interneuron subtypes endows the neocortex with distinct local cortical circuits and region-specific modulation of neuronal firing. Finally, the apoptotic process further refines neuronal populations by constantly maintaining a controlled ratio of inhibitory and excitatory neurons. Interestingly, many of these fundamental and complex processes are influenced—if not directly controlled—by electrical activity. Interneurons on the subcellular, cellular, and network level are affected by high frequency patterns, such as spindle burst and gamma oscillations in rodents and delta brushes in humans. Conversely, the maturation of interneuron structure and function on each of these scales feeds back and contributes to the generation of cortical activity patterns that are essential for the proper peri- and postnatal development. Overall, a more precise description of the conducting role of interneurons in terms of how they contribute to specific activity patterns—as well as how specific activity patterns impinge on their maturation as orchestra members—will lead to a better understanding of the physiological and pathophysiological development and function of the nervous system.
Collapse
|