1
|
Jabłońska J, Wiera G, Mozrzymas JW. Extracellular matrix integrity regulates GABAergic plasticity in the hippocampus. Matrix Biol 2024:S0945-053X(24)00129-X. [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] [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
|
2
|
Liu K, Gao YZ, Wu XM, Hu XY, Shi CN, He QL, Wu HP, Yao H, Ma DQ, Yang JJ, Ji MH. Microglia phagocytosis of PNNs mediates PV-positive interneuron dysfunction and associated gamma oscillations in neuroinflammation-induced cognitive impairment in mice. Neuropharmacology 2024; 262:110205. [PMID: 39489286 DOI: 10.1016/j.neuropharm.2024.110205] [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: 09/02/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Neuroinflammation, characterized by activation of glial cells, plays a critical role in central nervous system disorders. However, the precise mechanisms of neuroinflammation contributing to cognitive impairment remain elusive. Perineuronal nets (PNNs) are extracellular matrixes that envelop the cell bodies and dendrites of parvalbumin (PV)-positive interneurons and may be mediated by apolipoprotein E (ApoE) gene. To investigate whether disruption of PNNs associated with ApoE is implicated in neuroinflammation-induced cognitive impairment, we established a neuroinflammation model by administering lipopolysaccharides (LPS) at 0.5 mg/kg for 7 consecutive days. Cognitive function was assessed using the open field, Y-maze, and novel object recognition tests, and neural oscillations were also recorded. Furthermore, differentially expressed genes in microglia within the hippocampus were identified through single-cell RNA sequencing analysis. Overexpression of hyaluronan and proteoglycan link protein 1 (Hapln1) and ApoE knockdown were carried out through adeno-associated virus (AAV) injection to C57BL/6J mice and CX3CR1-CreERT2 mice, respectively. It was found that LPS-induced neuroinflammation impaired cognitive function by reducing PNNs and PV-positive interneurons' outputs, as well as disrupting gamma (γ) oscillations in the hippocampal CA1. Overexpression of Hapln1 was able to restore PV-positive interneurons and γ oscillations, ultimately alleviating the cognitive impairment. Mechanistically, LPS-triggered microglial activation leads to the phagocytosis of PNNs, a process influenced by ApoE. Notably, prevention of PNNs engulfment through targeting microglial ApoE in the CA1 improved cognitive impairment. Collectively, our study suggested that microglial phagocytosis of PNNs plays a key role in neuroinflammation-induced cognitive impairment, which is probably mediated by the ApoE.
Collapse
Affiliation(s)
- Kai Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Zhu Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Miao Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Yi Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cui-Na Shi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiu-Li He
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Peng Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Yao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Da-Qing Ma
- Division of Anaesthetics, Pain Medicine & Intensive Care, Department of Surgery & Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK; Perioperative and Systems Medicine Laboratory, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Mu-Huo Ji
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Herzberg MP, Nielsen AN, Luby J, Sylvester CM. Measuring neuroplasticity in human development: the potential to inform the type and timing of mental health interventions. Neuropsychopharmacology 2024; 50:124-136. [PMID: 39103496 PMCID: PMC11525577 DOI: 10.1038/s41386-024-01947-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/07/2024]
Abstract
Neuroplasticity during sensitive periods, the molecular and cellular process of enduring neural change in response to external stimuli during windows of high environmental sensitivity, is crucial for adaptation to expected environments and has implications for psychiatry. Animal research has characterized the developmental sequence and neurobiological mechanisms that govern neuroplasticity, yet gaps in our ability to measure neuroplasticity in humans limit the clinical translation of these principles. Here, we present a roadmap for the development and validation of neuroimaging and electrophysiology measures that index neuroplasticity to begin to address these gaps. We argue that validation of measures to track neuroplasticity in humans will elucidate the etiology of mental illness and inform the type and timing of mental health interventions to optimize effectiveness. We outline criteria for evaluating putative neuroimaging measures of plasticity in humans including links to neurobiological mechanisms shown to govern plasticity in animal models, developmental change that reflects heightened early life plasticity, and prediction of neural and/or behavior change. These criteria are applied to three putative measures of neuroplasticity using electroencephalography (gamma oscillations, aperiodic exponent of power/frequency) or functional magnetic resonance imaging (amplitude of low frequency fluctuations). We discuss the use of these markers in psychiatry, envision future uses for clinical and developmental translation, and suggest steps to address the limitations of the current putative neuroimaging measures of plasticity. With additional work, we expect these markers will significantly impact mental health and be used to characterize mechanisms, devise new interventions, and optimize developmental trajectories to reduce psychopathology risk.
Collapse
Affiliation(s)
- Max P Herzberg
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA.
| | - Ashley N Nielsen
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA.
| | - Joan Luby
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Chad M Sylvester
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
4
|
Dargam S, de Olmos S, Marcos Pautassi R, Lorenzo A. Footshock drives remodeling of perineuronal nets in retrosplenial cortex during contextual fear memory formation. Neurobiol Learn Mem 2024; 215:107990. [PMID: 39401564 DOI: 10.1016/j.nlm.2024.107990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
The retrosplenial cortex (RSC) plays a critical role in complex cognitive functions such as contextual fear memory formation and consolidation. Perineuronal nets (PNNs) are specialized structures of the extracellular matrix that modulate synaptic plasticity by enwrapping the soma, proximal neurites and synapsis mainly on fast spiking inhibitory GABAergic interneurons that express parvalbumin (PV). PNNs change after contextual fear conditioning (CFC) in amygdala or hippocampus, yet it is unknown if similar remodeling takes place at RSC. Here, we used Wisteria floribunda agglutinin (WFA), a ubiquitous marker of PNNs, to study the remodeling of PNNs in RSC during the acquisition or retrieval of contextual fear conditioning (CFC). Adult male mice were exposed to paired presentations of a context and footshock, or to either of these stimuli alone (control groups). The mere exposure of animals to the footshock, either alone or paired with the context, evoked a significant expansion of PNNs, both in the number of WFA positive neurons and in the area occupied by WFA staining, across the entire RSC. This was not associated with c-Fos expression in RSC nor correlated with c-Fos expression in individual PNNs-expressing neurons in RSC, suggesting that PNNs remodeling is triggered by inputs external to the RSC. We also found that PNNs remodeling was independent of the level of PV expression. Notably, PNNs in RSC remained expanded long-after CFC. These results suggest that, in male mice, the threatening experience is the main cause of PNNs remodeling in the RSC.
Collapse
Affiliation(s)
- Salome Dargam
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - Soledad de Olmos
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - Ricardo Marcos Pautassi
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina
| | - Alfredo Lorenzo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Friuli 2434, 5016, Córdoba, Argentina.
| |
Collapse
|
5
|
Hazlett MF, Hall VL, Patel E, Halvorsen A, Calakos N, West AE. The Perineuronal Net Protein Brevican Acts in Nucleus Accumbens Parvalbumin-Expressing Interneurons of Adult Mice to Regulate Excitatory Synaptic Inputs and Motivated Behaviors. Biol Psychiatry 2024; 96:694-707. [PMID: 38346480 PMCID: PMC11315813 DOI: 10.1016/j.biopsych.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/13/2024] [Accepted: 02/07/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Experience-dependent functional adaptation of nucleus accumbens (NAc) circuitry underlies the development and expression of reward-motivated behaviors. Parvalbumin-expressing GABAergic (gamma-aminobutyric acidergic) interneurons (PVINs) within the NAc are required for this process. Perineuronal nets (PNNs) are extracellular matrix structures enriched around PVINs that arise during development and have been proposed to mediate brain circuit stability. However, their function in the adult NAc is largely unknown. Here, we studied the developmental emergence and adult regulation of PNNs in the NAc of male and female mice and examined the cellular and behavioral consequences of reducing the PNN component brevican in NAc PVINs. METHODS We characterized the expression of PNN components in mouse NAc using immunofluorescence and RNA in situ hybridization. We lowered brevican in NAc PVINs of adult mice using an intersectional viral and genetic method and quantified the effects on synaptic inputs to NAc PVINs and reward-motivated learning. RESULTS PNNs around NAc PVINs were developmentally regulated and appeared during adolescence. In the adult NAc, PVIN PNNs were also dynamically regulated by cocaine. Transcription of the gene that encodes brevican was regulated in a cell type- and isoform-specific manner in the NAc, with the membrane-tethered form of brevican being highly enriched in PVINs. Lowering brevican in NAc PVINs of adult mice decreased their excitatory inputs and enhanced both short-term novel object recognition and cocaine-induced conditioned place preference. CONCLUSIONS Regulation of brevican in NAc PVINs of adult mice modulates their excitatory synaptic drive and sets experience thresholds for the development of motivated behaviors driven by rewarding stimuli.
Collapse
Affiliation(s)
- Mariah F Hazlett
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Victoria L Hall
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Esha Patel
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Aaron Halvorsen
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Nicole Calakos
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina; Department of Neurology, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina; Duke Institute for Brain Sciences, Duke University Medical Center, Durham, North Carolina
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
6
|
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
|
7
|
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 2024; 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] [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
|
8
|
Vadisiute A, Meijer E, Therpurakal RN, Mueller M, Szabó F, Messore F, Jursenas A, Bredemeyer O, Krone LB, Mann E, Vyazovskiy V, Hoerder-Suabedissen A, Molnár Z. Glial cells undergo rapid changes following acute chemogenetic manipulation of cortical layer 5 projection neurons. Commun Biol 2024; 7:1286. [PMID: 39384971 PMCID: PMC11464517 DOI: 10.1038/s42003-024-06994-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 09/30/2024] [Indexed: 10/11/2024] Open
Abstract
Bidirectional communication between neurons and glial cells is crucial to establishing and maintaining normal brain function. Some of these interactions are activity-dependent, yet it remains largely unexplored how acute changes in neuronal activity affect glial-to-neuron and neuron-to-glial dynamics. Here, we use excitatory and inhibitory designer receptors exclusively activated by designer drugs (DREADD) to study the effects of acute chemogenetic manipulations of a subpopulation of layer 5 cortical projection and dentate gyrus neurons in adult (Rbp4Cre) mouse brains. We show that acute chemogenetic neuronal activation reduces synaptic density, and increases microglia and astrocyte reactivity, but does not affect parvalbumin (PV+) neurons, only perineuronal nets (PNN). Conversely, acute silencing increases synaptic density and decreases glial reactivity. We show fast glial response upon clozapine-N-oxide (CNO) administration in cortical and subcortical regions. Together, our work provides evidence of fast, activity-dependent, bidirectional interactions between neurons and glial cells.
Collapse
Affiliation(s)
- Auguste Vadisiute
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom.
- St John's College, University of Oxford, St Giles', Oxford, United Kingdom.
| | - Elise Meijer
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
| | - Rajeevan Narayanan Therpurakal
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
- Department of Neurology, Düsseldorf University Hospital, Düsseldorf, Germany
| | - Marissa Mueller
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
| | - Florina Szabó
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
| | - Fernando Messore
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
| | | | - Oliver Bredemeyer
- St John's College, University of Oxford, St Giles', Oxford, United Kingdom
| | - Lukas B Krone
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Centre for Experimental Neurology, University of Bern, Bern, Switzerland
| | - Ed Mann
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
| | - Vladyslav Vyazovskiy
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
- Kavli Institute for Nanoscience Discovery, Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, United Kingdom
- Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, United Kingdom
| | - Anna Hoerder-Suabedissen
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom
- Kavli Institute for Nanoscience Discovery, Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, United Kingdom
- Sleep and Circadian Neuroscience Institute, University of Oxford, Oxford, United Kingdom
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Parks Road, Oxford, OX1 3PT, United Kingdom.
- St John's College, University of Oxford, St Giles', Oxford, United Kingdom.
| |
Collapse
|
9
|
Zhang D, Teng C, Xu Y, Tian L, Cao P, Wang X, Li Z, Guan C, Hu X. Genetic and molecular correlates of cortical thickness alterations in adults with obsessive-compulsive disorder: a transcription-neuroimaging association analysis. Psychol Med 2024; 54:1-10. [PMID: 39363543 PMCID: PMC11496223 DOI: 10.1017/s0033291724001909] [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: 01/25/2024] [Revised: 04/25/2024] [Accepted: 06/11/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Although numerous neuroimaging studies have depicted neural alterations in individuals with obsessive-compulsive disorder (OCD), a psychiatric disorder characterized by intrusive cognitions and repetitive behaviors, the molecular mechanisms connecting brain structural changes and gene expression remain poorly understood. METHODS This study combined the Allen Human Brain Atlas dataset with neuroimaging data from the Meta-Analysis (ENIGMA) consortium and independent cohorts. Later, partial least squares regression and enrichment analysis were performed to probe the correlation between transcription and cortical thickness variation among adults with OCD. RESULTS The cortical map of case-control differences in cortical thickness was spatially correlated with cortical expression of a weighted combination of genes enriched for neurobiologically relevant ontology terms preferentially expressed across different cell types and cortical layers. These genes were specifically expressed in brain tissue, spanning all cortical developmental stages. Protein-protein interaction analysis revealed that these genes coded a network of proteins encompassing various highly interactive hubs. CONCLUSIONS The study findings bridge the gap between neural structure and transcriptome data in OCD, fostering an integrative understanding of the potential biological mechanisms.
Collapse
Affiliation(s)
- Da Zhang
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Changjun Teng
- Department of Medical Psychology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yinhao Xu
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Tian
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ping Cao
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Wang
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zonghong Li
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chengbin Guan
- Department of Medical Psychology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Hu
- Department of Radiology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Faust TE, Devlin BA, Farhy-Tselnicker I, Ferro A, Postolache M, Xin W. Glial Control of Cortical Neuronal Circuit Maturation and Plasticity. J Neurosci 2024; 44:e1208242024. [PMID: 39358028 PMCID: PMC11450532 DOI: 10.1523/jneurosci.1208-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 10/04/2024] Open
Abstract
The brain is a highly adaptable organ that is molded by experience throughout life. Although the field of neuroscience has historically focused on intrinsic neuronal mechanisms of plasticity, there is growing evidence that multiple glial populations regulate the timing and extent of neuronal plasticity, particularly over the course of development. This review highlights recent discoveries on the role of glial cells in the establishment of cortical circuits and the regulation of experience-dependent neuronal plasticity during critical periods of neurodevelopment. These studies provide strong evidence that neuronal circuit maturation and plasticity are non-cell autonomous processes that require both glial-neuronal and glial-glial cross talk to proceed. We conclude by discussing open questions that will continue to guide research in this nascent field.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01605
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina 27708
| | | | - Austin Ferro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724
| | - Maggie Postolache
- Brain Immunology & Glia Center, Washington University School of Medicine, St. Louis, Missouri 63110
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California 94158
| |
Collapse
|
11
|
Zhang L, Wu C, Liu T, Tian Y, Wang D, Wang B, Yin Y. Propofol Protects the Blood-Brain Barrier After Traumatic Brain Injury by Stabilizing the Extracellular Matrix via Prrx1: From Neuroglioma to Neurotrauma. Neurochem Res 2024; 49:2743-2762. [PMID: 38951281 DOI: 10.1007/s11064-024-04202-z] [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: 03/30/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
The purpose of this study is to explore the shared molecular pathogenesis of traumatic brain injury (TBI) and high-grade glioma and investigate the mechanism of propofol (PF) as a potential protective agent. By analyzing the Chinese glioma genome atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases, we compared the transcriptomic data of high-grade glioma and TBI patients to identify common pathological mechanisms. Through bioinformatics analysis, in vitro experiments and in vivo TBI model, we investigated the regulatory effect of PF on extracellular matrix (ECM)-related genes through Prrx1 under oxidative stress. The impact of PF on BBB integrity under oxidative stress was investigated using a dual-layer BBB model, and we explored the protective effect of PF on tight junction proteins and ECM-related genes in mice after TBI. The study found that high-grade glioma and TBI share ECM instability as an important molecular pathological mechanism. PF stabilizes the ECM and protects the BBB by directly binding to Prrx1 or indirectly regulating Prrx1 through miRNAs. In addition, PF reduces intracellular calcium ions and ROS levels under oxidative stress, thereby preserving BBB integrity. In a TBI mouse model, PF protected BBB integrity through up-regulated tight junction proteins and stabilized the expression of ECM-related genes. Our study reveals the shared molecular pathogenesis between TBI and glioblastoma and demonstrate the potential of PF as a protective agent of BBB. This provides new targets and approaches for the development of novel neurotrauma therapeutic drugs.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Chenrui Wu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Tian
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo Wang
- Department of Neurosurgery, Tianjin University Huanhu Hospital, Tianjin, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China.
| | - Yiqing Yin
- Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
| |
Collapse
|
12
|
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
|
13
|
Cadena MA, Sing A, Taylor K, Jin L, Ning L, Amoli MS, Singh Y, Lanjewar SN, Tomov ML, Serpooshan V, Sloan SA. A 3D Bioprinted Cortical Organoid Platform for Modeling Human Brain Development. Adv Healthc Mater 2024; 13:e2401603. [PMID: 38815975 PMCID: PMC11518656 DOI: 10.1002/adhm.202401603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/24/2024] [Indexed: 06/01/2024]
Abstract
The ability to promote three-dimensional (3D) self-organization of induced pluripotent stem cells into complex tissue structures called organoids presents new opportunities for the field of developmental biology. Brain organoids have been used to investigate principles of neurodevelopment and neuropsychiatric disorders and serve as a drug screening and discovery platform. However, brain organoid cultures are currently limited by a lacking ability to precisely control their extracellular environment. Here, this work employs 3D bioprinting to generate a high-throughput, tunable, and reproducible scaffold for controlling organoid development and patterning. Additionally, this approach supports the coculture of organoids and vascular cells in a custom architecture containing interconnected endothelialized channels. Printing fidelity and mechanical assessments confirm that fabricated scaffolds closely match intended design features and exhibit stiffness values reflective of the developing human brain. Using organoid growth, viability, cytoarchitecture, proliferation, and transcriptomic benchmarks, this work finds that organoids cultured within the bioprinted scaffold long-term are healthy and have expected neuroectodermal differentiation. Lastly, this work confirms that the endothelial cells (ECs) in printed channel structures can migrate toward and infiltrate into the embedded organoids. This work demonstrates a tunable 3D culturing platform that can be used to create more complex and accurate models of human brain development and underlying diseases.
Collapse
Affiliation(s)
- Melissa A. Cadena
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anson Sing
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kylie Taylor
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Linqi Jin
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Mechanical Engineering, Cleveland State University, Cleveland, OH, United States
| | - Mehdi Salar Amoli
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Yamini Singh
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - The Brain Organoid Hub
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Samantha N. Lanjewar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Martin L. Tomov
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, US
| | - Steven A. Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
14
|
Zhang N, Song B, Bai P, Du L, Chen L, Xu Y, Zeng T. Perineuronal nets' role in metabolism. Am J Physiol Endocrinol Metab 2024; 327:E411-E421. [PMID: 39140971 DOI: 10.1152/ajpendo.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Perineuronal nets (PNNs), specialized extracellular matrix (ECM) structures that envelop neurons, have recently been recognized as key players in the regulation of metabolism. This review explores the growing body of knowledge concerning PNNs and their role in metabolic control, drawing insights from recent research and relevant studies. The pivotal role of PNNs in the context of energy balance and whole body blood glucose is examined. This review also highlights novel findings, including the effects of astroglia, microglia, sex and gonadal hormones, nutritional regulation, circadian rhythms, and age on PNNs dynamics. These findings illuminate the complex and multifaceted role of PNNs in metabolic health.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Beite Song
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Peng Bai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li Du
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
15
|
Cangalaya C, Sun W, Stoyanov S, Dunay IR, Dityatev A. Integrity of neural extracellular matrix is required for microglia-mediated synaptic remodeling. Glia 2024; 72:1874-1892. [PMID: 38946065 DOI: 10.1002/glia.24588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
Microglia continuously remodel synapses, which are embedded in the extracellular matrix (ECM). However, the mechanisms, which govern this process remain elusive. To investigate the influence of the neural ECM in synaptic remodeling by microglia, we disrupted ECM integrity by injection of chondroitinase ABC (ChABC) into the retrosplenial cortex of healthy adult mice. Using in vivo two-photon microscopy we found that ChABC treatment increased microglial branching complexity and ECM phagocytic capacity and decreased spine elimination rate under basal conditions. Moreover, ECM attenuation largely prevented synaptic remodeling following synaptic stress induced by photodamage of single synaptic elements. These changes were associated with less stable and smaller microglial contacts at the synaptic damage sites, diminished deposition of calreticulin and complement proteins C1q and C3 at synapses and impaired expression of microglial CR3 receptor. Thus, our findings provide novel insights into the function of the neural ECM in deposition of complement proteins and synaptic remodeling by microglia.
Collapse
Affiliation(s)
- Carla Cangalaya
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Weilun Sun
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Stoyan Stoyanov
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
16
|
Huang Z, Wei X, Tian J, Fu Y, Dong J, Wang Y, Shi J, Lu L, Zhang W. A disinhibitory microcircuit of the orbitofrontal cortex mediates cocaine preference in mice. Mol Psychiatry 2024; 29:3160-3169. [PMID: 38698268 DOI: 10.1038/s41380-024-02579-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/05/2024]
Abstract
Both clinical and animal studies showed that the impaired functions of the orbitofrontal cortex (OFC) underlie the compulsive drug-seeking behavior of drug addiction. However, the functional changes of the microcircuit in the OFC and the underlying molecular mechanisms in drug addiction remain elusive, and little is known for whether microcircuits in the OFC contributed to drug addiction-related behaviors. Utilizing the cocaine-induced conditioned-place preference model, we found that the malfunction of the microcircuit led to disinhibition in the OFC after cocaine withdrawal. We further showed that enhanced Somatostatin-Parvalbumin (SST-PV) inhibitory synapse strength changed microcircuit function, and SST and PV inhibitory neurons showed opposite contributions to the drug addiction-related behavior of mice. Brevican of the perineuronal nets of PV neurons regulated SST-PV synapse strength, and the knockdown of Brevican alleviated cocaine preference. These results reveal a novel molecular mechanism of the regulation of microcircuit function and a novel circuit mechanism of the OFC in gating cocaine preference.
Collapse
Affiliation(s)
- Ziran Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Xiaoyan Wei
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jing Tian
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Yangxue Fu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jihui Dong
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Yihui Wang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China
| | - Lin Lu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital); Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100191, China
| | - Wen Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, 100191, China.
| |
Collapse
|
17
|
Almalki WH, Almujri SS. Therapeutic approaches to microglial dysfunction in Alzheimer's disease: Enhancing phagocytosis and metabolic regulation. Pathol Res Pract 2024; 263:155614. [PMID: 39342887 DOI: 10.1016/j.prp.2024.155614] [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: 08/05/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Microglia are essential in neurogenesis, synaptic pruning, and homeostasis. Nevertheless, aging, and cellular senescence may modify their role, causing them to shift from being shields to being players of neurodegeneration. In the aging brain, the population of microglia increases, followed by enhanced activity of genes related to neuroinflammation. This change increases their ability to cause inflammation, resulting in a long-lasting state of inflammation in the brain that harms the condition of neurons. In Alzheimer's Disease (AD), microglia are located inside amyloid plaques and exhibit an inflammatory phenotype characterized by a diminished ability to engulf and remove waste material, worsening the illness's advancement. Genetic polymorphisms in TREM2, APOE, and CD33 highlight the significant impact of microglial dysfunction in AD. This review examines therapeutic approaches that aim to address microglial dysfunction, such as enhancing the microglial capability to engulf and remove amyloid-β clumps and regulating microglial metabolism and mitochondrial activity. Microglial transplanting and reprogramming advancements show the potential to restore their ability to reduce inflammation. Although there has been notable advancement, there are still voids in our knowledge of microglial biology, including their relationships with other brain cells. Further studies should prioritize the improvement of human AD models, establish standardized methods for characterizing microglia, and explore how various factors influence microglial responses. It is essential to tackle these problems to create effective treatment plans that focus on reducing inflammation in the brain and protecting against damage in age-related neurodegenerative illnesses.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Aseer 61421, Saudi Arabia
| |
Collapse
|
18
|
Brunello CA, Cannarozzo C, Castrén E. Rethinking the role of TRKB in the action of antidepressants and psychedelics. Trends Neurosci 2024:S0166-2236(24)00154-1. [PMID: 39304417 DOI: 10.1016/j.tins.2024.08.011] [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: 03/13/2024] [Revised: 06/20/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
Antidepressant drugs promote neuronal plasticity, and activation of brain-derived neurotrophic factor (BDNF) signaling through its receptor neuronal receptor tyrosine kinase 2 (NTRK2 or TRKB) is among the critical steps in this process. These mechanisms are shared by typical slow-acting antidepressants, fast-acting ketamine, and psychedelic compounds, although the cellular targets of each drug differ. In this opinion, we propose that some of these antidepressants may directly bind to TRKB and allosterically potentiate BDNF signaling, among other possible effects. TRKB activation in parvalbumin-containing interneurons disinhibits cortical networks and reactivates a juvenile-like plasticity window. Subsequent rewiring of aberrant networks, coupled with environmental stimuli, may underlie its clinical antidepressant effects. The end-to-end hypothesis proposed may stimulate the search for new treatment strategies.
Collapse
Affiliation(s)
| | | | - Eero Castrén
- Neuroscience Center - HILIFE, University of Helsinki, Finland.
| |
Collapse
|
19
|
Prieto-López L, Pereiro X, Vecino E. The mechanics of the retina: Müller glia role on retinal extracellular matrix and modelling. Front Med (Lausanne) 2024; 11:1393057. [PMID: 39296899 PMCID: PMC11410058 DOI: 10.3389/fmed.2024.1393057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
The retina is a highly heterogeneous tissue, both cell-wise but also regarding its extracellular matrix (ECM). The stiffness of the ECM is pivotal in retinal development and maturation and has also been associated with the onset and/or progression of numerous retinal pathologies, such as glaucoma, proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), epiretinal membrane (ERM) formation or uveitis. Nonetheless, much remains unknown about the biomechanical milieu of the retina, and specifically the role that Müller glia play as principal mechanosensors and major producers of ECM constituents. So far, new approaches need to be developed to further the knowledge in the field of retinal mechanobiology for ECM-target applications to arise. In this review, we focus on the involvement of Müller glia in shaping and altering the retinal ECM under both physiological and pathological conditions and look into various biomaterial options to more accurately replicate the impact of matrix stiffness in vitro.
Collapse
Affiliation(s)
- Laura Prieto-López
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| |
Collapse
|
20
|
Colodete DA, Grace AA, Guimarães FS, Gomes FV. Degradation of Perineuronal Nets in the Ventral Hippocampus of Adult Rats Recreates an Adolescent-Like Phenotype of Stress Susceptibility. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100338. [PMID: 39099729 PMCID: PMC11295568 DOI: 10.1016/j.bpsgos.2024.100338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 08/06/2024] Open
Abstract
Background Psychiatric disorders often emerge during late adolescence/early adulthood, a period with increased susceptibility to socioenvironmental factors that coincides with incomplete parvalbumin interneuron (PVI) development. Stress during this period causes functional loss of PVIs in the ventral hippocampus (vHip), which has been associated with dopamine system overdrive. This vulnerability persists until the appearance of perineuronal nets (PNNs) around PVIs. We assessed the long-lasting effects of adolescent or adult stress on behavior, ventral tegmental area dopamine neuron activity, and the number of PVIs and their associated PNNs in the vHip. Additionally, we tested whether PNN removal in the vHip of adult rats, proposed to reset PVIs to a juvenile-like state, would recreate an adolescent-like phenotype of stress susceptibility. Methods Male rats underwent a 10-day stress protocol during adolescence or adulthood. Three to 4 weeks poststress, we evaluated behaviors related to anxiety, sociability, and cognition, ventral tegmental area dopamine neuron activity, and the number of PV+ and PNN+ cells in the vHip. Furthermore, adult animals received intra-vHip infusion of ChABC (chondroitinase ABC) to degrade PNNs before undergoing stress. Results Unlike adult stress, adolescent stress induced anxiety responses, reduced sociability, cognitive deficits, ventral tegmental area dopamine system overdrive, and decreased PV+ and PNN+ cells in the vHip. However, intra-vHip ChABC infusion caused the adult stress to produce changes similar to the ones observed after adolescent stress. Conclusions Our findings underscore adolescence as a period of heightened vulnerability to the long-lasting impact of stress and highlight the protective role of PNNs against stress-induced damage in PVIs.
Collapse
Affiliation(s)
- Débora A.E. Colodete
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry, and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Francisco S. Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Sanchez B, Kraszewski P, Lee S, Cope EC. From molecules to behavior: Implications for perineuronal net remodeling in learning and memory. J Neurochem 2024; 168:1854-1876. [PMID: 38158878 DOI: 10.1111/jnc.16036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/30/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Perineuronal nets (PNNs) are condensed extracellular matrix (ECM) structures found throughout the central nervous system that regulate plasticity. They consist of a heterogeneous mix of ECM components that form lattice-like structures enwrapping the cell body and proximal dendrites of particular neurons. During development, accumulating research has shown that the closure of various critical periods of plasticity is strongly linked to experience-driven PNN formation and maturation. PNNs provide an interface for synaptic contacts within the holes of the structure, generally promoting synaptic stabilization and restricting the formation of new synaptic connections in the adult brain. In this way, they impact both synaptic structure and function, ultimately influencing higher cognitive processes. PNNs are highly plastic structures, changing their composition and distribution throughout life and in response to various experiences and memory disorders, thus serving as a substrate for experience- and disease-dependent cognitive function. In this review, we delve into the proposed mechanisms by which PNNs shape plasticity and memory function, highlighting the potential impact of their structural components, overall architecture, and dynamic remodeling on functional outcomes in health and disease.
Collapse
Affiliation(s)
- Brenda Sanchez
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| | - Piotr Kraszewski
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| | - Sabrina Lee
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| | - Elise C Cope
- Department of Neuroscience, University of Virginia School of Medicine, Virginia, USA
| |
Collapse
|
22
|
Beddows CA, Shi F, Horton AL, Dalal S, Zhang P, Ling CC, Yong VW, Loh K, Cho E, Karagiannis C, Rose AJ, Montgomery MK, Gregorevic P, Watt MJ, Packer NH, Parker BL, Brown RM, Moh ESX, Dodd GT. Pathogenic hypothalamic extracellular matrix promotes metabolic disease. Nature 2024; 633:914-922. [PMID: 39294371 PMCID: PMC11424483 DOI: 10.1038/s41586-024-07922-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/07/2024] [Indexed: 09/20/2024]
Abstract
Metabolic diseases such as obesity and type 2 diabetes are marked by insulin resistance1,2. Cells within the arcuate nucleus of the hypothalamus (ARC), which are crucial for regulating metabolism, become insulin resistant during the progression of metabolic disease3-8, but these mechanisms are not fully understood. Here we investigated the role of a specialized chondroitin sulfate proteoglycan extracellular matrix, termed a perineuronal net, which surrounds ARC neurons. In metabolic disease, the perineuronal net of the ARC becomes augmented and remodelled, driving insulin resistance and metabolic dysfunction. Disruption of the perineuronal net in obese mice, either enzymatically or with small molecules, improves insulin access to the brain, reversing neuronal insulin resistance and enhancing metabolic health. Our findings identify ARC extracellular matrix remodelling as a fundamental mechanism driving metabolic diseases.
Collapse
Affiliation(s)
- Cait A Beddows
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Feiyue Shi
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Anna L Horton
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Sagar Dalal
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Ping Zhang
- Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
| | - Chang-Chun Ling
- Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kim Loh
- St Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Victoria, Australia
| | - Chris Karagiannis
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Adam J Rose
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Magdalene K Montgomery
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Neurology, The University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Nicolle H Packer
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Benjamin L Parker
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Robyn M Brown
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Edward S X Moh
- ARC Centre of Excellence in Synthetic Biology, School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Garron T Dodd
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
23
|
Rahimian R, Belliveau C, Simard S, Turecki G, Mechawar N. Perineuronal Net Alterations Following Early-Life Stress: Are Microglia Pulling Some Strings? Biomolecules 2024; 14:1087. [PMID: 39334854 PMCID: PMC11430691 DOI: 10.3390/biom14091087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The extracellular matrix plays a key role in synapse formation and in the modulation of synaptic function in the central nervous system. Recent investigations have revealed that microglia, the resident immune cells of the brain, are involved in extracellular matrix remodeling under both physiological and pathological conditions. Moreover, the dysregulation of both innate immune responses and the extracellular matrix has been documented in stress-related psychopathologies as well as in relation to early-life stress. However, the dynamics of microglial regulation of the ECM and how it can be impacted by early-life adversity have been understudied. This brief review provides an overview of the recent literature on this topic, drawing from both animal model and human post mortem studies. Direct and indirect mechanisms through which microglia may regulate the extracellular matrix-including perineuronal nets-are presented and discussed in light of the interactions with other cell types.
Collapse
Affiliation(s)
- Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Sophie Simard
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, QC H4H 1R3, Canada; (R.R.); (C.B.); (S.S.); (G.T.)
- Department of Psychiatry, McGill University, Montreal, QC H3A 0G4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
24
|
Méndez P, de la Vega-Ruiz R, Montes-Mellado A. Estrogenic regulation of hippocampal inhibitory system across lifespan. J Neuroendocrinol 2024:e13441. [PMID: 39143852 DOI: 10.1111/jne.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Estrogens produced in peripheral tissues and locally in the brain are potent neuromodulators. The function of the hippocampus, a brain region essential for episodic memory and spatial navigation, relies on the activity of ensembles of excitatory neurons whose activity is temporally and spatially coordinated by a wide diversity of inhibitory neurons (INs) types. Over the last years, we have accumulated evidence that indicates that estrogens regulate the function of hippocampal INs through different mechanisms, including transcriptional regulation and rapid nongenomic signaling. Here, we argue that the well-documented influence of estrogens on episodic memory may be related to the actions of local and peripheral estrogens on the heterogenous populations of hippocampal INs. We discuss how physiological changes in peripheral sex hormone levels throughout lifespan may interact with local brain sources to regulate IN function at different stages of life, from early hippocampal development to the aging brain. We conclude that considering INs as mediators of sex hormone actions in the hippocampus across the healthy life span will benefit our understanding of sex-biased neurodevelopmental disorders and physiological aging.
Collapse
|
25
|
Seiler S, Rudolf F, Gomes FR, Pavlovic A, Nebel J, Seidenbecher CI, Foo LC. Astrocyte-derived factors regulate CNS myelination. Glia 2024. [PMID: 39092473 DOI: 10.1002/glia.24596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
The role that astrocytes play in central nervous system (CNS) myelination is poorly understood. We investigated the contribution of astrocyte-derived factors to myelination and revealed a substantial overlap in the secretomes of human and rat astrocytes. Using in vitro myelinating co-cultures of primary retinal ganglion cells and cortical oligodendrocyte precursor cells, we discovered that factors secreted by resting astrocytes, but not reactive astrocytes, facilitated myelination. Soluble brevican emerged as a new enhancer of developmental myelination in vivo, CNS and its absence was linked to remyelination deficits following an immune-mediated damage in an EAE mouse model. The observed reduction of brevican expression in reactive astrocytes and human MS lesions suggested a potential link to the compromised remyelination characteristic of neurodegenerative diseases. Our findings suggested brevican's role in myelination may be mediated through interactions with binding partners such as contactin-1 and tenascin-R. Proteomic analysis of resting versus reactive astrocytes highlighted a shift in protein expression profiles, pinpointing candidates that either facilitate or impede CNS repair, suggesting that depending on their reactivity state, astrocytes play a dual role during myelination.
Collapse
Affiliation(s)
- Sybille Seiler
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Franziska Rudolf
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Filipa Ramilo Gomes
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Anto Pavlovic
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| | - Jana Nebel
- Department Neurochemistry & Molecular Biology, Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Constanze I Seidenbecher
- Department Neurochemistry & Molecular Biology, Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Lynette C Foo
- F. Hoffmann-La Roche, pRED, Neuroscience, Discovery & Translational Area (NRD), Basel, Switzerland
| |
Collapse
|
26
|
Tewari BP, Woo AM, Prim CE, Chaunsali L, Patel DC, Kimbrough IF, Engel K, Browning JL, Campbell SL, Sontheimer H. Astrocytes require perineuronal nets to maintain synaptic homeostasis in mice. Nat Neurosci 2024; 27:1475-1488. [PMID: 39020018 PMCID: PMC11303255 DOI: 10.1038/s41593-024-01714-3] [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: 01/20/2023] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Perineuronal nets (PNNs) are densely packed extracellular matrices that cover the cell body of fast-spiking inhibitory neurons. PNNs stabilize synapses inhibiting synaptic plasticity. Here we show that synaptic terminals of fast-spiking interneurons localize to holes in the PNNs in the adult mouse somatosensory cortex. Approximately 95% of holes in the PNNs contain synapses and astrocytic processes expressing Kir4.1, glutamate and GABA transporters. Hence, holes in the PNNs contain tripartite synapses. In the adult mouse brain, PNN degradation causes an expanded astrocytic coverage of the neuronal somata without altering the axon terminals. The loss of PNNs impairs astrocytic transmitter and potassium uptake, resulting in the spillage of glutamate into the extrasynaptic space. Our data show that PNNs and astrocytes cooperate to contain synaptically released signals in physiological conditions. Their combined action is altered in mouse models of Alzheimer's disease and epilepsy where PNNs are disrupted.
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
| | - Dipan C Patel
- 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
| | | | - Susan L Campbell
- Department of Animal Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
27
|
Lehner A, Hoffmann L, Rampp S, Coras R, Paulsen F, Frischknecht R, Hamer H, Walther K, Brandner S, Hofer W, Pieper T, Reisch L, Bien CG, Blumcke I. Age-dependent increase of perineuronal nets in the human hippocampus and precocious aging in epilepsy. Epilepsia Open 2024; 9:1372-1381. [PMID: 38845524 PMCID: PMC11296138 DOI: 10.1002/epi4.12963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 08/03/2024] Open
Abstract
OBJECTIVE Perineuronal nets (PNN) are specialized extracellular matrix (ECM) components of the central nervous system, frequently accumulating at the surface of inhibitory GABAergic interneurons. While an altered distribution of PNN has been observed in neurological disorders including Alzheimer's disease, schizophrenia and epilepsy, their anatomical distribution also changes during physiological brain maturation and aging. Such an age-dependent shift was experimentally associated also with hippocampal engram formation during brain maturation. Our aim was to histopathologically assess PNN in the hippocampus of adult and pediatric patients with temporal lobe epilepsy (TLE) compared to age-matched post-mortem control subjects and to compare PNN-related changes with memory impairment observed in our patient cohort. METHODS Sixty-six formalin-fixed and paraffin-embedded tissue specimens of the human hippocampus were retrieved from the European Epilepsy Brain Bank. Twenty-nine patients had histopathologically confirmed hippocampal sclerosis (HS), and eleven patients suffered from TLE without HS. PNN were immunohistochemically visualized using an antibody directed against aggrecan and manually counted from hippocampus subfields and the subiculum. RESULTS PNN density increased with age in both human controls and TLE patients. However, their density was significantly higher in all HS patients compared to age-matched controls. Intriguingly, TLE patients presented presurgically with better memory when their hippocampal PNN density was higher (p < 0.05). SIGNIFICANCE Our results were compatible with age-dependent ECM specialization in the human hippocampus and its precocious aging in the epileptic condition. These observations confirm recent experimental animal models and also support the notion that PNN play a role in memory formation in the human brain. PLAIN LANGUAGE SUMMARY "Perineuronal nets" (PNN) are a specialized compartment of the extracellular matrix (ECM), especially surrounding highly active neurons of the mammalian brain. There is evidence that PNN play a role in memory formation, brain maturation, and in some pathologies like Alzheimer's disease, schizophrenia or epilepsy. In this study, we investigated the role of PNN in patients suffering from drug-resistant focal epilepsy compared to controls. We found that with increasing age, more neurons are surrounded by PNN. Similarly, all epilepsy patients but especially patients with better memory performance also had more PNN. This study raises further interest in studying ECM molecules in the human brain under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Annika Lehner
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| | - Lucas Hoffmann
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| | - Stefan Rampp
- Department of NeuroradiologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Department of NeurosurgeryUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
| | - Roland Coras
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, FAU Erlangen‐NürnbergErlangenGermany
| | - Renato Frischknecht
- Department of Biology, Animal PhysiologyFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Hajo Hamer
- Epilepsy Center, Department of NeurologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
| | - Katrin Walther
- Epilepsy Center, Department of NeurologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
| | - Sebastian Brandner
- Department of NeurosurgeryUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Department of NeurosurgeryKlinikum FürthGermany
| | - Wiebke Hofer
- Department of Psychology/Neuropsychology, Center for Pediatric Neurology, Neurorehabilitation, and EpileptologySchön Klinik VogtareuthGermany
| | - Tom Pieper
- Center for Pediatric Neurology, Neurorehabilitation, and EpileptologySchön Klinik VogtareuthGermany
| | - Lea‐Marie Reisch
- Department of Epileptology (Krankenhaus Mara), Medical SchoolBielefeld UniversityBielefeldGermany
| | - Christian G. Bien
- Department of Epileptology (Krankenhaus Mara), Medical SchoolBielefeld UniversityBielefeldGermany
| | - Ingmar Blumcke
- Department of NeuropathologyUniversitätsklinikum Erlangen and FAU Erlangen‐NürnbergErlangenGermany
- Partner of the European Reference Network (ERN) EpiCAREBarcelonaSpain
| |
Collapse
|
28
|
Hernández-del Caño C, Varela-Andrés N, Cebrián-León A, Deogracias R. Neurotrophins and Their Receptors: BDNF's Role in GABAergic Neurodevelopment and Disease. Int J Mol Sci 2024; 25:8312. [PMID: 39125882 PMCID: PMC11311851 DOI: 10.3390/ijms25158312] [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: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Neurotrophins and their receptors are distinctly expressed during brain development and play crucial roles in the formation, survival, and function of neurons in the nervous system. Among these molecules, brain-derived neurotrophic factor (BDNF) has garnered significant attention due to its involvement in regulating GABAergic system development and function. In this review, we summarize and compare the expression patterns and roles of neurotrophins and their receptors in both the developing and adult brains of rodents, macaques, and humans. Then, we focus on the implications of BDNF in the development and function of GABAergic neurons from the cortex and the striatum, as both the presence of BDNF single nucleotide polymorphisms and disruptions in BDNF levels alter the excitatory/inhibitory balance in the brain. This imbalance has different implications in the pathogenesis of neurodevelopmental diseases like autism spectrum disorder (ASD), Rett syndrome (RTT), and schizophrenia (SCZ). Altogether, evidence shows that neurotrophins, especially BDNF, are essential for the development, maintenance, and function of the brain, and disruptions in their expression or signaling are common mechanisms in the pathophysiology of brain diseases.
Collapse
Affiliation(s)
- Carlos Hernández-del Caño
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.H.-d.C.); (N.V.-A.); (A.C.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Natalia Varela-Andrés
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.H.-d.C.); (N.V.-A.); (A.C.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Alejandro Cebrián-León
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.H.-d.C.); (N.V.-A.); (A.C.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Rubén Deogracias
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain; (C.H.-d.C.); (N.V.-A.); (A.C.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
29
|
Banovac I, Prkačin MV, Kirchbaum I, Trnski-Levak S, Bobić-Rasonja M, Sedmak G, Petanjek Z, Jovanov-Milosevic N. Morphological and Molecular Characteristics of Perineuronal Nets in the Human Prefrontal Cortex-A Possible Link to Microcircuitry Specialization. Mol Neurobiol 2024:10.1007/s12035-024-04306-1. [PMID: 38958887 DOI: 10.1007/s12035-024-04306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Perineuronal nets (PNNs) are a type of extracellular matrix (ECM) that play a significant role in synaptic activity and plasticity of interneurons in health and disease. We researched PNNs' regional and laminar representation and molecular composition using immunohistochemistry and transcriptome analysis of Brodmann areas (BA) 9, 14r, and 24 in 25 human postmortem brains aged 13-82 years. The numbers of VCAN- and NCAN-expressing PNNs, relative to the total number of neurons, were highest in cortical layers I and VI while WFA-binding (WFA+) PNNs were most abundant in layers III-V. The ECM glycosylation pattern was the most pronounced regional difference, shown by a significantly lower proportion of WFA+ PNNs in BA24 (3.27 ± 0.69%) compared to BA9 (6.32 ± 1.73%; P = 0.0449) and BA14 (5.64 ± 0.71%; P = 0.0278). The transcriptome of late developmental and mature stages revealed a relatively stable expression of PNN-related transcripts (log2-transformed expression values: 6.5-8.5 for VCAN and 8.0-9.5 for NCAN). Finally, we propose a classification of PNNs that envelop GABAergic neurons in the human cortex. The significant differences in PNNs' morphology, distribution, and molecular composition strongly suggest an involvement of PNNs in specifying distinct microcircuits in particular cortical regions and layers.
Collapse
Affiliation(s)
- Ivan Banovac
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Matija Vid Prkačin
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Ivona Kirchbaum
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Sara Trnski-Levak
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Mihaela Bobić-Rasonja
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Department of Biology, University of Zagreb School of Medicine, Šalata 3, HR-10000, Zagreb, Croatia
| | - Goran Sedmak
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Natasa Jovanov-Milosevic
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia.
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia.
- Department of Biology, University of Zagreb School of Medicine, Šalata 3, HR-10000, Zagreb, Croatia.
| |
Collapse
|
30
|
Oliver D, Chesney E, Cullen AE, Davies C, Englund A, Gifford G, Kerins S, Lalousis PA, Logeswaran Y, Merritt K, Zahid U, Crossley NA, McCutcheon RA, McGuire P, Fusar-Poli P. Exploring causal mechanisms of psychosis risk. Neurosci Biobehav Rev 2024; 162:105699. [PMID: 38710421 PMCID: PMC11250118 DOI: 10.1016/j.neubiorev.2024.105699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/17/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Robust epidemiological evidence of risk and protective factors for psychosis is essential to inform preventive interventions. Previous evidence syntheses have classified these risk and protective factors according to their strength of association with psychosis. In this critical review we appraise the distinct and overlapping mechanisms of 25 key environmental risk factors for psychosis, and link these to mechanistic pathways that may contribute to neurochemical alterations hypothesised to underlie psychotic symptoms. We then discuss the implications of our findings for future research, specifically considering interactions between factors, exploring universal and subgroup-specific factors, improving understanding of temporality and risk dynamics, standardising operationalisation and measurement of risk and protective factors, and developing preventive interventions targeting risk and protective factors.
Collapse
Affiliation(s)
- Dominic Oliver
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK; Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Edward Chesney
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - Alexis E Cullen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, Karolinska Institutet, Sweden
| | - Cathy Davies
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Amir Englund
- Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - George Gifford
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Sarah Kerins
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Yanakan Logeswaran
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Biostatistics & Health Informatics, King's College London, London, UK
| | - Kate Merritt
- Division of Psychiatry, Institute of Mental Health, UCL, London, UK
| | - Uzma Zahid
- Department of Psychology, King's College London, London, UK
| | - Nicolas A Crossley
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Chile
| | - Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Philip McGuire
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; OASIS Service, South London and Maudsley NHS Foundation Trust, London SE11 5DL, UK
| |
Collapse
|
31
|
Talwalkar A, Haden G, Duncan KA. Chondroitin sulfate proteoglycans mRNA expression and degradation in the zebra finch following traumatic brain injury. J Chem Neuroanat 2024; 138:102418. [PMID: 38621597 DOI: 10.1016/j.jchemneu.2024.102418] [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: 01/04/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of fatality and disability worldwide. From minutes to months following damage, injury can result in a complex pathophysiology that can lead to temporary or permanent deficits including an array of neurodegenerative symptoms. These changes can include behavioral dysregulation, memory dysfunctions, and mood changes including depression. The nature and severity of impairments resulting from TBIs vary widely given the range of injury type, location, and extent of brain tissue involved. In response to the injury, the brain induces structural and functional changes to promote repair and minimize injury size. Despite its high prevalence, effective treatment strategies for TBI are limited. PNNs are part of the neuronal extracellular matrix (ECM) that mediate synaptic stabilization in the adult brain and thus neuroplasticity. They are associated mostly with inhibitory GABAergic interneurons and are thought to be responsible for maintaining the excitatory/inhibitory balance of the brain. The major structural components of PNNs include multiple chondroitin sulfate proteoglycans (CSPGs) as well as other structural proteins. Here we examine the effects of injury on CSPG expression, specifically around the changes in the side change moieties. To investigate CSPG expression following injury, adult male and female zebra finches received either a bilateral penetrating, or no injury and qPCR analysis and immunohistochemistry for components of the CSPGs were examined at 1- or 7-days post-injury. Next, to determine if CSPGs and thus PNNs should be a target for therapeutic intervention, CSPG side chains were degraded at the time of injury with chondroitinase ABC (ChABC) CSPGs moieties were examined. Additionally, GABA receptor mRNA and aromatase mRNA expression was quantified following CSPG degradation as they have been implicated in neuronal survival and neurogenesis. Our data indicate the CSPG moieties change following injury, potentially allowing for a brief period of synaptic reorganization, and that treatments that target CSPG side chains are successful in further targeting this brief critical period by decreasing GABA mRNA receptor expression, but also decreasing aromatase expression.
Collapse
Affiliation(s)
- Adam Talwalkar
- Program in Biochemistry, Vassar College, Poughkeepsie, NY 12604, USA
| | - Gage Haden
- Department of Biology, Vassar College, Poughkeepsie, NY 12604, USA
| | - Kelli A Duncan
- Department of Biology, Vassar College, Poughkeepsie, NY 12604, USA; Program in Neuroscience and Behavior, Vassar College, Poughkeepsie, NY 12604, USA.
| |
Collapse
|
32
|
Honeycutt S, Mukherjee A, Paladino M, Gilles-Thomas E, Loney G. Adolescent nicotine exposure promotes adulthood opioid consumption that persists despite adverse consequences and increases the density of insular perineuronal nets. ADDICTION NEUROSCIENCE 2024; 11:100150. [PMID: 38911872 PMCID: PMC11192509 DOI: 10.1016/j.addicn.2024.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Adolescence marks a sensitive period for neurodevelopment wherein exposure to drugs of abuse may disrupt maturation and induce persistent changes in neurophysiology which may exacerbate the risk for developing substance use disorders in adulthood. Adolescent nicotine exposure (ANE) enhances motivation to obtain drugs of abuse, particularly opioids, and increases vulnerability for the development of opioid use disorder (OUD). Here, we characterized ANE effects on learning about the adverse consequences of opioid consumption in adulthood in the absence of further nicotine administration. First, we show that ANE engenders punishment resistant fentanyl self-administration in a heterogenous seeking-taking chain schedule of reinforcement at least at the tested dose of fentanyl (0.75 μg/kg). We found that ANE rats consumed significantly more fentanyl and contingent foot shock punishment was less efficacious in limiting fentanyl seeking in ANE rats, relative to nicotine-naïve controls. Next, we demonstrated that ANE limits learning about the deleterious consequences of acute opioid intoxication in adulthood. In a combined conditioned taste avoidance and place preference paradigm we found that ANE resulted in significant reductions in the strength of morphine-induced CTA, and a simultaneous enhancement of CPP at a higher dose that was less capable of driving reinforcement in naïve controls. Finally, we examined the expression of perineuronal nets (PNNs) within insular cortex (IC) and found ANE rats to have increased density of PNNs across the anterior IC and significantly more parvalbumin-labeled IC cells relative to naïve controls. Together, these data lay the framework for a mechanistic explanation of the extreme comorbidity between nicotine use and development of OUDs.
Collapse
|
33
|
Chang K, Lin L, Cui T, Zhao H, Li J, Liu C, Gao D, Lu S. Zinc-a2-Glycoprotein Acts as a Component of PNN to Protect Hippocampal Neurons from Apoptosis. Mol Neurobiol 2024; 61:3607-3618. [PMID: 38001359 DOI: 10.1007/s12035-023-03771-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023]
Abstract
In the adult mouse brain, perineuronal net (PNN), a highly structured extracellular matrix, surrounds subsets of neurons. The AZGP1 gene encodes zinc-2-glycoprotein (ZAG) is a lipid-mobilizing factor. However, its expression and distribution in the adult brain have been controversial. Here, for the first time, we demonstrate that the secreted ZAG is localized to Wisteria floribunda agglutinin (WFA)-positive PNNs around parvalbumin (PV)-expressing interneurons in the hippocampus, cortex, and a number of other PNN-bearing neurons and co-localizes with aggrecan, one of the components of PNNs. Few ZAG-positive nets were seen in the area without WFA staining by chondroitinase ABC (ChABC) which degrades glycosaminoglycans (GAGs) from the chondroitin sulfate proteoglycans (CSPGs) in the PNN. Reanalysis of single-cell sequencing data revealed that ZAG mRNA was mainly expressed in oligodendrocyte lineages, specifically in olfactory sheathing cells. The ZAG receptor β3 adrenergic receptor (β3AR) is also selectively co-localized with PV interneurons and CA2 pyramidal neurons in the hippocampus. In addition, molecular docking provides valuable new insights on how GAGs interfere with ZAG and ZAG/β3AR complex. Finally, our results indicated that human recombinant ZAG could significantly inhibit serum derivation-induced cell apoptosis in HT22 cells. Our combined experimental and theoretical approach raises a unique hypothesis namely that ZAG may be a crucial functional attribute of PNNs in the brain to protect neuronal cell from apoptosis.
Collapse
Affiliation(s)
- Kewei Chang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Liyan Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Tingting Cui
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Hao Zhao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Jiaxin Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Chang Liu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Dan Gao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| | - Shemin Lu
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
- Department of Biochemistry and Molecular Biology, Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
34
|
Hu J, Zhang S, Wu H, Wang L, Zhang Y, Gao H, Li M, Ren H, Xiao H, Guo K, Li W, Liu Q. 1-Methyltryptophan treatment ameliorates high-fat diet-induced depression in mice through reversing changes in perineuronal nets. Transl Psychiatry 2024; 14:228. [PMID: 38816357 PMCID: PMC11139877 DOI: 10.1038/s41398-024-02938-4] [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: 07/22/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024] Open
Abstract
Depression and obesity are prevalent disorders with significant public health implications. In this study, we used a high-fat diet (HFD)-induced obese mouse model to investigate the mechanism underlying HFD-induced depression-like behaviors. HFD-induced obese mice exhibited depression-like behaviors and a reduction in hippocampus volume, which were reversed by treatment with an indoleamine 2,3-dioxygenase (IDO) inhibitor 1-methyltryptophan (1-MT). Interestingly, no changes in IDO levels were observed post-1-MT treatment, suggesting that other mechanisms may be involved in the anti-depressive effect of 1-MT. We further conducted RNA sequencing analysis to clarify the potential underlying mechanism of the anti-depressive effect of 1-MT in HFD-induced depressive mice and found a significant enrichment of shared differential genes in the extracellular matrix (ECM) organization pathway between the 1-MT-treated and untreated HFD-induced depressive mice. Therefore, we hypothesized that changes in ECM play a crucial role in the anti-depressive effect of 1-MT. To this end, we investigated perineuronal nets (PNNs), which are ECM assemblies that preferentially ensheath parvalbumin (PV)-positive interneurons and are involved in many abnormalities. We found that HFD is associated with excessive accumulation of PV-positive neurons and upregulation of PNNs, affecting synaptic transmission in PV-positive neurons and leading to glutamate-gamma-aminobutyric acid imbalances in the hippocampus. The 1-MT effectively reversed these changes, highlighting a PNN-related mechanism by which 1-MT exerts its anti-depressive effect.
Collapse
Affiliation(s)
- Juntao Hu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shanshan Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Haoran Wu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Leilei Wang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Hongyang Gao
- Electron Microscopy Core Laboratory, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Meihui Li
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hong Ren
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Honglei Xiao
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kun Guo
- Key Laboratory of Carcinogenesis and Cancer Invasion, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Research Center, Institute of Biomedical Science, Fudan University, Shanghai, China.
| | - Wensheng Li
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Qiong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China.
| |
Collapse
|
35
|
Chapman MA, Sorg BA. A Systematic Review of Extracellular Matrix-Related Alterations in Parkinson's Disease. Brain Sci 2024; 14:522. [PMID: 38928523 PMCID: PMC11201521 DOI: 10.3390/brainsci14060522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
The role of the extracellular matrix (ECM) in Parkinson's disease (PD) is not well understood, even though it is critical for neuronal structure and signaling. This systematic review identified the top deregulated ECM-related pathways in studies that used gene set enrichment analyses (GSEA) to document transcriptomic, proteomic, or genomic alterations in PD. PubMed and Google scholar were searched for transcriptomics, proteomics, or genomics studies that employed GSEA on data from PD tissues or cells and reported ECM-related pathways among the top-10 most enriched versus controls. Twenty-seven studies were included, two of which used multiple omics analyses. Transcriptomics and proteomics studies were conducted on a variety of tissue and cell types. Of the 17 transcriptomics studies (16 data sets), 13 identified one or more adhesion pathways in the top-10 deregulated gene sets or pathways, primarily related to cell adhesion and focal adhesion. Among the 8 proteomics studies, 5 identified altered overarching ECM gene sets or pathways among the top 10. Among the 4 genomics studies, 3 identified focal adhesion pathways among the top 10. The findings summarized here suggest that ECM organization/structure and cell adhesion (particularly focal adhesion) are altered in PD and should be the focus of future studies.
Collapse
Affiliation(s)
| | - Barbara A. Sorg
- R.S. Dow Neurobiology, Legacy Research Institute, Portland, OR 97232, USA;
| |
Collapse
|
36
|
Sun C, Wang Y, Jin X, Ni B, Xu B, Hou JJ, Zhong C, Liu J, Wu Y, Song L, Hou L, Yi M, Liu X, Xiong J. Observing perineuronal nets like structures via coaxial scattering quantitative interference imaging at multiple wavelengths. OPTICS EXPRESS 2024; 32:18150-18160. [PMID: 38858978 DOI: 10.1364/oe.521510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/02/2024] [Indexed: 06/12/2024]
Abstract
Perineuronal nets (PNNs) are important functional structures on the surface of nerve cells. Observation of PNNs usually requires dyeing or fluorescent labeling. As a network structure with a micron grid and sub-wavelength thickness but no special optical properties, quantitative phase imaging (QPI) is the only purely optical method for high-resolution imaging of PNNs. We proposed a Scattering Quantitative Interference Imaging (SQII) method which measures the geometric rather than transmission or reflection phase during the scattering process to visualize PNNs. Different from QIP methods, SQII method is sensitive to scattering and not affected by wavelength changes. Via geometric phase shifting method, we simplify the phase shift operation. The SQII method not only focuses on interference phase, but also on the interference contrast. The singularity points and phase lines of the scattering geometric phase depict the edges of the network structure and can be found at the valley area of the interference contrast parameter SINDR under different wavelengths. Our SQII method has its unique imaging properties, is very simple and easy to implement and has more worth for promotion.
Collapse
|
37
|
Patel DC, Swift N, Tewari BP, Browning JL, Prim C, Chaunsali L, Kimbrough IF, Olsen ML, Sontheimer H. Increased expression of chondroitin sulfate proteoglycans in dentate gyrus and amygdala causes postinfectious seizures. Brain 2024; 147:1856-1870. [PMID: 38146224 PMCID: PMC11068111 DOI: 10.1093/brain/awad430] [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: 06/27/2023] [Revised: 10/05/2023] [Accepted: 12/08/2023] [Indexed: 12/27/2023] Open
Abstract
Alterations in the extracellular matrix are common in patients with epilepsy and animal models of epilepsy, yet whether they are the cause or consequence of seizures and epilepsy development is unknown. Using Theiler's murine encephalomyelitis virus (TMEV) infection-induced model of acquired epilepsy, we found de novo expression of chondroitin sulfate proteoglycans (CSPGs), a major extracellular matrix component, in dentate gyrus (DG) and amygdala exclusively in mice with acute seizures. Preventing the synthesis of CSPGs specifically in DG and amygdala by deletion of the major CSPG aggrecan reduced seizure burden. Patch-clamp recordings from dentate granule cells revealed enhanced intrinsic and synaptic excitability in seizing mice that was significantly ameliorated by aggrecan deletion. In situ experiments suggested that dentate granule cell hyperexcitability results from negatively charged CSPGs increasing stationary cations on the membrane, thereby depolarizing neurons, increasing their intrinsic and synaptic excitability. These results show increased expression of CSPGs in the DG and amygdala as one of the causal factors for TMEV-induced acute seizures. We also show identical changes in CSPGs in pilocarpine-induced epilepsy, suggesting that enhanced CSPGs in the DG and amygdala may be a common ictogenic factor and potential therapeutic target.
Collapse
Affiliation(s)
- Dipan C Patel
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Nathaniel Swift
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Bhanu P Tewari
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Jack L Browning
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Courtney Prim
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Lata Chaunsali
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Ian F Kimbrough
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| | - Michelle L Olsen
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Harald Sontheimer
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
38
|
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
|
39
|
Ortega JA, Soares de Aguiar GP, Chandravanshi P, Levy N, Engel E, Álvarez Z. Exploring the properties and potential of the neural extracellular matrix for next-generation regenerative therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1962. [PMID: 38723788 DOI: 10.1002/wnan.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024]
Abstract
The extracellular matrix (ECM) is a dynamic and complex network of proteins and molecules that surrounds cells and tissues in the nervous system and orchestrates a myriad of biological functions. This review carefully examines the diverse interactions between cells and the ECM, as well as the transformative chemical and physical changes that the ECM undergoes during neural development, aging, and disease. These transformations play a pivotal role in shaping tissue morphogenesis and neural activity, thereby influencing the functionality of the central nervous system (CNS). In our comprehensive review, we describe the diverse behaviors of the CNS ECM in different physiological and pathological scenarios and explore the unique properties that make ECM-based strategies attractive for CNS repair and regeneration. Addressing the challenges of scalability, variability, and integration with host tissues, we review how advanced natural, synthetic, and combinatorial matrix approaches enhance biocompatibility, mechanical properties, and functional recovery. Overall, this review highlights the potential of decellularized ECM as a powerful tool for CNS modeling and regenerative purposes and sets the stage for future research in this exciting field. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Gisele P Soares de Aguiar
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natacha Levy
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
40
|
Marín O. Parvalbumin interneuron deficits in schizophrenia. Eur Neuropsychopharmacol 2024; 82:44-52. [PMID: 38490084 DOI: 10.1016/j.euroneuro.2024.02.010] [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: 12/21/2023] [Accepted: 02/16/2024] [Indexed: 03/17/2024]
Abstract
Parvalbumin-expressing (PV+) interneurons represent one of the most abundant subclasses of cortical interneurons. Owing to their specific electrophysiological and synaptic properties, PV+ interneurons are essential for gating and pacing the activity of excitatory neurons. In particular, PV+ interneurons are critically involved in generating and maintaining cortical rhythms in the gamma frequency, which are essential for complex cognitive functions. Deficits in PV+ interneurons have been frequently reported in postmortem studies of schizophrenia patients, and alterations in gamma oscillations are a prominent electrophysiological feature of the disease. Here, I summarise the main features of PV+ interneurons and review clinical and preclinical studies linking the developmental dysfunction of cortical PV+ interneurons with the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom; Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
41
|
Parise EM, Gyles TM, Godino A, Sial OK, Browne CJ, Parise LF, Torres-Berrío A, Salery M, Durand-de Cuttoli R, Rivera MT, Cardona-Acosta AM, Holt L, Markovic T, van der Zee YY, Lorsch ZS, Cathomas F, Garon JB, Teague C, Issler O, Hamilton PJ, Bolaños-Guzmán CA, Russo SJ, Nestler EJ. Sex-Specific Regulation of Stress Susceptibility by the Astrocytic Gene Htra1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.588724. [PMID: 38659771 PMCID: PMC11042238 DOI: 10.1101/2024.04.12.588724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Major depressive disorder (MDD) is linked to impaired structural and synaptic plasticity in limbic brain regions. Astrocytes, which regulate synapses and are influenced by chronic stress, likely contribute to these changes. We analyzed astrocyte gene profiles in the nucleus accumbens (NAc) of humans with MDD and mice exposed to chronic stress. Htra1 , which encodes an astrocyte-secreted protease targeting the extracellular matrix (ECM), was significantly downregulated in the NAc of males but upregulated in females in both species. Manipulating Htra1 in mouse NAc astrocytes bidirectionally controlled stress susceptibility in a sex-specific manner. Such Htra1 manipulations also altered neuronal signaling and ECM structural integrity in NAc. These findings highlight astroglia and the brain's ECM as key mediators of sex-specific stress vulnerability, offering new approaches for MDD therapies.
Collapse
|
42
|
Sánchez-Ventura J, Lago N, Penas C, Navarro X, Udina E. Link Protein 1 Is Involved in the Activity-Dependent Modulation of Perineuronal Nets in the Spinal Cord. Int J Mol Sci 2024; 25:4267. [PMID: 38673852 PMCID: PMC11050079 DOI: 10.3390/ijms25084267] [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: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
One of the challenges of the mature nervous system is to maintain the stability of neural networks while providing a degree of plasticity to generate experience-dependent modifications. This plasticity-stability dynamism is regulated by perineuronal nets (PNNs) and is crucial for the proper functioning of the system. Previously, we found a relation between spinal PNNs reduction and maladaptive plasticity after spinal cord injury (SCI), which was attenuated by maintaining PNNs with activity-dependent therapies. Moreover, transgenic mice lacking the cartilage link protein 1 (Crtl1 KO mice) showed aberrant spinal PNNs and increased spinal plasticity. Therefore, the aim of this study is to evaluate the role of link protein 1 in the activity-dependent modulation of spinal PNNs surrounding motoneurons and its impact on the maladaptive plasticity observed following SCI. We first studied the activity-dependent modulation of spinal PNNs using a voluntary wheel-running protocol. This training protocol increased spinal PNNs in WT mice but did not modify PNN components in Crtl1 KO mice, suggesting that link protein 1 mediates the activity-dependent modulation of PNNs. Secondly, a thoracic SCI was performed, and functional outcomes were evaluated for 35 days. Interestingly, hyperreflexia and hyperalgesia found at the end of the experiment in WT-injured mice were already present at basal levels in Crtl1 KO mice and remained unchanged after the injury. These findings demonstrated that link protein 1 plays a dual role in the correct formation and in activity-dependent modulation of PNNs, turning it into an essential element for the proper function of PNN in spinal circuits.
Collapse
Affiliation(s)
| | | | | | - Xavier Navarro
- Department Cell Biology, Physiology and Immunology, Institute of Neuroscience, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain (N.L.); (C.P.)
| | - Esther Udina
- Department Cell Biology, Physiology and Immunology, Institute of Neuroscience, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain (N.L.); (C.P.)
| |
Collapse
|
43
|
Paveliev M, Egorchev AA, Musin F, Lipachev N, Melnikova A, Gimadutdinov RM, Kashipov AR, Molotkov D, Chickrin DE, Aganov AV. Perineuronal Net Microscopy: From Brain Pathology to Artificial Intelligence. Int J Mol Sci 2024; 25:4227. [PMID: 38673819 PMCID: PMC11049984 DOI: 10.3390/ijms25084227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Perineuronal nets (PNN) are a special highly structured type of extracellular matrix encapsulating synapses on large populations of CNS neurons. PNN undergo structural changes in schizophrenia, epilepsy, Alzheimer's disease, stroke, post-traumatic conditions, and some other brain disorders. The functional role of the PNN microstructure in brain pathologies has remained largely unstudied until recently. Here, we review recent research implicating PNN microstructural changes in schizophrenia and other disorders. We further concentrate on high-resolution studies of the PNN mesh units surrounding synaptic boutons to elucidate fine structural details behind the mutual functional regulation between the ECM and the synaptic terminal. We also review some updates regarding PNN as a potential pharmacological target. Artificial intelligence (AI)-based methods are now arriving as a new tool that may have the potential to grasp the brain's complexity through a wide range of organization levels-from synaptic molecular events to large scale tissue rearrangements and the whole-brain connectome function. This scope matches exactly the complex role of PNN in brain physiology and pathology processes, and the first AI-assisted PNN microscopy studies have been reported. To that end, we report here on a machine learning-assisted tool for PNN mesh contour tracing.
Collapse
Affiliation(s)
- Mikhail Paveliev
- Neuroscience Center, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Anton A. Egorchev
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Foat Musin
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Nikita Lipachev
- Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, Kazan 420008, Tatarstan, Russia; (N.L.); (A.V.A.)
| | - Anastasiia Melnikova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, Kazan 420015, Tatarstan, Russia;
| | - Rustem M. Gimadutdinov
- Institute of Computational Mathematics and Information Technologies, Kazan Federal University, Kremlyovskaya 35, Kazan 420008, Tatarstan, Russia; (A.A.E.); (F.M.); (R.M.G.)
| | - Aidar R. Kashipov
- Institute of Artificial Intelligence, Robotics and Systems Engineering, Kazan Federal University, Kremlyovskaya 18, Kazan 420008, Tatarstan, Russia; (A.R.K.); (D.E.C.)
| | - Dmitry Molotkov
- Biomedicum Imaging Unit, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland;
| | - Dmitry E. Chickrin
- Institute of Artificial Intelligence, Robotics and Systems Engineering, Kazan Federal University, Kremlyovskaya 18, Kazan 420008, Tatarstan, Russia; (A.R.K.); (D.E.C.)
| | - Albert V. Aganov
- Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, Kazan 420008, Tatarstan, Russia; (N.L.); (A.V.A.)
| |
Collapse
|
44
|
Tewari BP, Conant K. Editorial: Emerging roles of extracellular matrix in the physiology and pathophysiology of the central nervous system. Front Cell Neurosci 2024; 18:1400652. [PMID: 38638301 PMCID: PMC11024420 DOI: 10.3389/fncel.2024.1400652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024] Open
Affiliation(s)
- Bhanu P. Tewari
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
45
|
Jovasevic V, Wood EM, Cicvaric A, Zhang H, Petrovic Z, Carboncino A, Parker KK, Bassett TE, Moltesen M, Yamawaki N, Login H, Kalucka J, Sananbenesi F, Zhang X, Fischer A, Radulovic J. Formation of memory assemblies through the DNA-sensing TLR9 pathway. Nature 2024; 628:145-153. [PMID: 38538785 PMCID: PMC10990941 DOI: 10.1038/s41586-024-07220-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
As hippocampal neurons respond to diverse types of information1, a subset assembles into microcircuits representing a memory2. Those neurons typically undergo energy-intensive molecular adaptations, occasionally resulting in transient DNA damage3-5. Here we found discrete clusters of excitatory hippocampal CA1 neurons with persistent double-stranded DNA (dsDNA) breaks, nuclear envelope ruptures and perinuclear release of histone and dsDNA fragments hours after learning. Following these early events, some neurons acquired an inflammatory phenotype involving activation of TLR9 signalling and accumulation of centrosomal DNA damage repair complexes6. Neuron-specific knockdown of Tlr9 impaired memory while blunting contextual fear conditioning-induced changes of gene expression in specific clusters of excitatory CA1 neurons. Notably, TLR9 had an essential role in centrosome function, including DNA damage repair, ciliogenesis and build-up of perineuronal nets. We demonstrate a novel cascade of learning-induced molecular events in discrete neuronal clusters undergoing dsDNA damage and TLR9-mediated repair, resulting in their recruitment to memory circuits. With compromised TLR9 function, this fundamental memory mechanism becomes a gateway to genomic instability and cognitive impairments implicated in accelerated senescence, psychiatric disorders and neurodegenerative disorders. Maintaining the integrity of TLR9 inflammatory signalling thus emerges as a promising preventive strategy for neurocognitive deficits.
Collapse
Affiliation(s)
- Vladimir Jovasevic
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M Wood
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana Cicvaric
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hui Zhang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Zorica Petrovic
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anna Carboncino
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kendra K Parker
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Thomas E Bassett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria Moltesen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Naoki Yamawaki
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Hande Login
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- PROMEMO, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Farahnaz Sananbenesi
- Department for Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medical Center, Göttingen, Germany
- Cluster of Excellence MBExC, University of Göttingen, Göttingen, Germany
| | - Xusheng Zhang
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andre Fischer
- Department for Psychiatry and Psychotherapy, German Center for Neurodegenerative Diseases, University Medical Center, Göttingen, Germany
- Cluster of Excellence MBExC, University of Göttingen, Göttingen, Germany
| | - Jelena Radulovic
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- PROMEMO, Aarhus University, Aarhus, Denmark.
- DANDRITE, Aarhus University, Aarhus, Denmark.
- Department of Psychiatry and Behavioral Sciences, Psychiatry Research Institute Montefiore Einstein (PRIME), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
46
|
Miller B, Crider A, Aravamuthan B, Galindo R. Human chorionic gonadotropin decreases cerebral cystic encephalomalacia and parvalbumin interneuron degeneration in a pro-inflammatory model of mouse neonatal hypoxia-ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587006. [PMID: 38585735 PMCID: PMC10996598 DOI: 10.1101/2024.03.27.587006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The pregnancy hormone, human chorionic gonadotropin (hCG) is an immunoregulatory and neurotrophic glycoprotein of potential clinical utility in the neonate at risk for cerebral injury. Despite its well-known role in its ability to modulate the innate immune response during pregnancy, hCG has not been demonstrated to affect the pro-degenerative actions of inflammation in neonatal hypoxia-ischemia (HI). Here we utilize a neonatal mouse model of mild HI combined with intraperitoneal administration of lipopolysaccharide (LPS) to evaluate the neuroprotective actions of hCG in the setting of endotoxin-mediated systemic inflammation. Intraperitoneal treatment of hCG shortly prior to LPS injection significantly decreased tissue loss and cystic degeneration in the hippocampal and cerebral cortex in the term-equivalent neonatal mouse exposed to mild HI. Noting that parvalbumin immunoreactive interneurons have been broadly implicated in neurodevelopmental disorders, it is notable that hCG significantly improved the injury-mediated reduction of these neurons in the cerebral cortex, striatum and hippocampus. The above findings were associated with a decrease in the amount of Iba1 immunoreactive microglia in most of these brain regions. These observations implicate hCG as an agent capable of improving the neurological morbidity associated with peripheral inflammation in the neonate affected by HI. Future preclinical studies should aim at demonstrating added neuroprotective benefit by hCG in the context of therapeutic hypothermia and further exploring the mechanisms responsible for this effect. This research is likely to advance the therapeutic role of gonadotropins as a treatment for neonates with neonatal brain injury.
Collapse
Affiliation(s)
- Ben Miller
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, MO, USA 63110
| | - Alexander Crider
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, MO, USA 63110
| | - Bhooma Aravamuthan
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, MO, USA 63110
| | - Rafael Galindo
- Department of Neurology, Division of Pediatric & Developmental Neurology, Washington University School of Medicine, St. Louis, MO, USA 63110
| |
Collapse
|
47
|
Sterin I, Niazi A, Kim J, Park J, Park S. Novel extracellular matrix architecture on excitatory neurons revealed by HaloTag-HAPLN1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587384. [PMID: 38585814 PMCID: PMC10996768 DOI: 10.1101/2024.03.29.587384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The brain's extracellular matrix (ECM) regulates neuronal plasticity and animal behavior. ECM staining shows an aggregated pattern in a net-like structure around a subset of neurons and diffuse staining in the interstitial matrix. However, understanding the structural features of ECM deposition across various neuronal types and subcellular compartments remains limited. To visualize the organization pattern and assembly process of the hyaluronan-scaffolded ECM in the brain, we fused a HaloTag to HAPLN1, which links hyaluronan and proteoglycans. Expression or application of the probe enables us to identify spatial and temporal regulation of ECM deposition and heterogeneity in ECM aggregation among neuronal populations. Dual-color birthdating shows the ECM assembly process in culture and in vivo. Sparse expression in vivo reveals novel forms of ECM architecture around excitatory neurons and developmentally regulated dendritic ECM. Overall, our study uncovers extensive structural features of the brain' ECM, suggesting diverse roles in regulating neuronal plasticity.
Collapse
Affiliation(s)
- Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
- Neuroscience Program, University of Utah, Salt Lake City, Utah, USA
| | - Jennifer Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
48
|
Martinez CA, Pantazopoulos H, Gisabella B, Stephens ET, Garteiser J, Del Arco A. Choice impulsivity after repeated social stress is associated with increased perineuronal nets in the medial prefrontal cortex. Sci Rep 2024; 14:7093. [PMID: 38528075 DOI: 10.1038/s41598-024-57599-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024] Open
Abstract
Repeated stress can predispose to substance abuse. However, behavioral and neurobiological adaptations that link stress to substance abuse remain unclear. This study investigates whether intermittent social defeat (ISD), a stress protocol that promotes drug-seeking behavior, alters intertemporal decision-making and cortical inhibitory function in the medial prefrontal cortex (mPFC). Male long evans rats were trained in a delay discounting task (DDT) where rats make a choice between a fast (1 s) small reward (1 sugar pellet) and a large reward (3 sugar pellets) that comes with a time delay (10 s or 20 s). A decreased preference for delayed rewards was used as an index of choice impulsivity. Rats were exposed to ISD and tested in the DDT 24 h after each stress episode, and one- and two-weeks after the last stress episode. Immunohistochemistry was performed in rat's brains to evaluate perineuronal nets (PNNs) and parvalbumin GABA interneurons (PV) labeling as markers of inhibitory function in mPFC. ISD significantly decreased the preference for delayed large rewards in low impulsive, but not high impulsive, animals. ISD also increased the density of PNNs in the mPFC. These results suggest that increased choice impulsivity and cortical inhibition predispose animals to seek out rewards after stress.
Collapse
Affiliation(s)
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Barbara Gisabella
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Emily T Stephens
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jacob Garteiser
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alberto Del Arco
- HESRM, School of Applied Sciences, University of Mississippi, Oxford, MS, USA.
- Department of Psychiatry and Human Behavior, Medical School, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
49
|
Santos-Silva T, Lopes CFB, Hazar Ülgen D, Guimarães DA, Guimarães FS, Alberici LC, Sandi C, Gomes FV. Adolescent Stress-Induced Ventral Hippocampus Redox Dysregulation Underlies Behavioral Deficits and Excitatory/Inhibitory Imbalance Related to Schizophrenia. Schizophr Bull 2024:sbae033. [PMID: 38525594 DOI: 10.1093/schbul/sbae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
BACKGROUND AND HYPOTHESIS Redox dysregulation has been proposed as a convergent point of childhood trauma and the emergence of psychiatric disorders, such as schizophrenia (SCZ). A critical region particularly vulnerable to environmental insults during adolescence is the ventral hippocampus (vHip). However, the impact of severe stress on vHip redox states and their functional consequences, including behavioral and electrophysiological changes related to SCZ, are not entirely understood. STUDY DESIGN After exposing adolescent animals to physical stress (postnatal day, PND31-40), we explored social and cognitive behaviors (PND47-49), the basal activity of pyramidal glutamate neurons, the number of parvalbumin (PV) interneurons, and the transcriptomic signature of the vHip (PND51). We also evaluated the impact of stress on the redox system, including mitochondrial respiratory function, reactive oxygen species (ROS) production, and glutathione (GSH) levels in the vHip and serum. STUDY RESULTS Adolescent-stressed animals exhibited loss of sociability, cognitive impairment, and vHip excitatory/inhibitory (E/I) imbalance. Genome-wide transcriptional profiling unveiled the impact of stress on redox system- and synaptic-related genes. Stress impacted mitochondrial respiratory function and changes in ROS levels in the vHip. GSH and glutathione disulfide (GSSG) levels were elevated in the serum of stressed animals, while GSSG was also increased in the vHip and negatively correlated with sociability. Additionally, PV interneuron deficits in the vHip caused by adolescent stress were associated with oxidative stress. CONCLUSIONS Our results highlight the negative impact of adolescent stress on vHip redox regulation and mitochondrial function, which are partially associated with E/I imbalance and behavioral abnormalities related to SCZ.
Collapse
Affiliation(s)
- Thamyris Santos-Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Caio Fábio Baeta Lopes
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Doğukan Hazar Ülgen
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Danielle A Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luciane Carla Alberici
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carmen Sandi
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
50
|
Schreurs BG, O'Dell DE, Wang D. The Role of Cerebellar Intrinsic Neuronal Excitability, Synaptic Plasticity, and Perineuronal Nets in Eyeblink Conditioning. BIOLOGY 2024; 13:200. [PMID: 38534469 DOI: 10.3390/biology13030200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/29/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
Evidence is strong that, in addition to fine motor control, there is an important role for the cerebellum in cognition and emotion. The deep nuclei of the mammalian cerebellum also contain the highest density of perineural nets-mesh-like structures that surround neurons-in the brain, and it appears there may be a connection between these nets and cognitive processes, particularly learning and memory. Here, we review how the cerebellum is involved in eyeblink conditioning-a particularly well-understood form of learning and memory-and focus on the role of perineuronal nets in intrinsic membrane excitability and synaptic plasticity that underlie eyeblink conditioning. We explore the development and role of perineuronal nets and the in vivo and in vitro evidence that manipulations of the perineuronal net in the deep cerebellar nuclei affect eyeblink conditioning. Together, these findings provide evidence of an important role for perineuronal net in learning and memory.
Collapse
Affiliation(s)
- Bernard G Schreurs
- Department of Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| | - Deidre E O'Dell
- Department of Biology, Earth and Environmental Sciences, Pennsylvania Western (PennWest) University, California, PA 15419, USA
| | - Desheng Wang
- Department of Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|