1
|
Mazi AR, Karakoc Y, Demirtas C, Aykin U, Yildirim M. Extracellular Matrix Alterations Due to Early-Life Adversity: Implications for Auditory Learning in Male Sprague-Dawley Rats. Mol Neurobiol 2025; 62:6490-6502. [PMID: 39812993 PMCID: PMC11953085 DOI: 10.1007/s12035-025-04690-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
This study aimed to investigate the impact of early childhood chronic stress on the development of the brain extracellular matrix (ECM) and how alterations in the ECM following early-life adversity (ELA) affect auditory learning and cognitive flexibility. ELA was induced through a combination of maternal separation and neonatal isolation in male Sprague-Dawley rats, and the success of the ELA model was assessed behaviorally and biochemically. A cortex-dependent go/no-go task with two phases was used to determine the impact of ELA on auditory learning and cognitive flexibility. The effects of the ECM on cognition were tested via the enzymatic removal of the ECM. The molecular structure of the adult ECM was examined via immunohistochemistry. ELA impaired initial auditory learning but did not significantly affect cognitive flexibility. Hyase injection into the auditory cortex (ACx) restored initial learning. ELA rats display a reduced perineural net (PNN) and parvalbumin + cell density. Our findings reveal that ELA induces significant alterations in the ECM within the ACx, accompanied by impaired initial auditory learning. Although PNN density is already lower in ELA rats, degrading the ECM facilitates the repair of auditory learning. A reduced PNN number in ELA rats fails to enhance learning unless supplemented with Hyase injection.
Collapse
Affiliation(s)
- Aise Rumeysa Mazi
- Department of Biophysics, Hamidiye Faculty of Medicine, University of Health Sciences, Selimiye Mah. Tibbiye Cad. No:38, 34668, Uskudar, Istanbul, Turkey.
| | - Yunus Karakoc
- Department of Biophysics, Hamidiye Faculty of Medicine, University of Health Sciences, Selimiye Mah. Tibbiye Cad. No:38, 34668, Uskudar, Istanbul, Turkey
| | - Cumaali Demirtas
- Department of Physiology, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Ugur Aykin
- Department of Physiology, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Mehmet Yildirim
- Department of Physiology, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
2
|
Sokolov R, Krut' V, Belousov V, Rozov A, Mukhina IV. Hyaluronidase-induced matrix remodeling contributes to long-term synaptic changes. Front Neural Circuits 2025; 18:1441280. [PMID: 39897766 PMCID: PMC11782146 DOI: 10.3389/fncir.2024.1441280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 02/04/2025] Open
Abstract
Extracellular brain space contains water, dissolved ions, and multiple other signaling molecules. The neural extracellular matrix (ECM) is also a significant component of the extracellular space. The ECM is synthesized by neurons, astrocytes, and other types of cells. Hyaluronan, a hyaluronic acid polymer, is a key component of the ECM. The functions of hyaluronan include barrier functions and signaling. In this article, we investigate physiological processes during the acute phase of enzymatic ECM removal. We found that hyaluronidase, an ECM removal agent, triggers simultaneous membrane depolarization and sharp calcium influx into neurons. Spontaneous action potential firing frequency increased rapidly after ECM destruction in interneurons, but not pyramidal neurons. Hyaluronidase-dependent calcium entry can be blocked by a selective antagonist of N-methyl-D-aspartate (NMDA) receptors, revealing these receptors as the main player in the observed phenomenon. Additionally, we demonstrate increased NMDA-dependent long-term potentiation at CA3-to-CA1 synapses during the acute phase of ECM removal. These findings suggest that hyaluronan is a significant synaptic player.
Collapse
Affiliation(s)
- Rostislav Sokolov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Viktoriya Krut'
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Vsevolod Belousov
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Andrey Rozov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Irina V. Mukhina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| |
Collapse
|
3
|
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 2025; 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] [MESH Headings] [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
|
4
|
Li X, Li N, Zhao P, Ren D, Luo B, Zhou T. Perineuronal Nets: From Structure to Neurological Disorders. Curr Med Chem 2025; 32:1685-1701. [PMID: 37946343 DOI: 10.2174/0109298673258290231009111633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/05/2023] [Accepted: 09/01/2023] [Indexed: 11/12/2023]
Abstract
Perineuronal nets (PNN) is condensed extracellular matrix (ECM) in the central nervous system (CNS), which surrounds cell soma, axon initial segments, and synapses. In the brain, most neurons surrounded by PNN are interneurons, especially the parvalbumin-expressing interneurons (PVI). The formation of PNN is involved in the PVI maturation as well as the onset and closure of critical periods for developmental plasticity end. Dysfunction of PVI can lead to some neurological disorders, such as schizophrenia, bipolar depression, and Alzheimer's disease. Similarly, PNN assembling abnormalities are often observed in human patients and animal disease models. PNN is thought to have a neuroprotective effect and interact with signaling molecules to regulate synaptic plasticity and neuronal activity. In this review, we provide an overview of the composition, structure, and functions of PNN. In addition, we highlight abnormal changes in PNN components in pathological conditions. Understanding the roles of different components of PNN will bring us a new perspective on brain plasticity and neurological disorders.
Collapse
Affiliation(s)
- Xianghe Li
- Queen Mary School of Nanchang University, Nanchang 330031, China
| | - Nuojin Li
- Queen Mary School of Nanchang University, Nanchang 330031, China
| | - Pingping Zhao
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Dongyan Ren
- Institute of Life Science, Nanchang University, Nanchang 330031, China
| | - Bin Luo
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Tian Zhou
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| |
Collapse
|
5
|
Jabłońska J, Wiera G, Mozrzymas JW. Extracellular matrix integrity regulates GABAergic plasticity in the hippocampus. Matrix Biol 2024; 134:184-196. [PMID: 39491759 DOI: 10.1016/j.matbio.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/18/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
The brain's extracellular matrix (ECM) is crucial for neural circuit functionality, synaptic plasticity, and learning. While the role of the ECM in excitatory synapses has been extensively studied, its influence on inhibitory synapses, particularly on GABAergic long-term plasticity, remains poorly understood. This study aims to elucidate the effects of ECM components on inhibitory synaptic transmission and plasticity in the hippocampal CA1 region. We focus on the roles of chondroitin sulfate proteoglycans (CSPGs) and hyaluronic acid in modulating inhibitory postsynaptic currents (IPSCs) at two distinct inhibitory synapses formed by somatostatin (SST)-positive and parvalbumin (PV)-positive interneurons onto pyramidal cells (PCs). Using optogenetic stimulation in brain slices, we observed that acute degradation of ECM constituents by hyaluronidase or chondroitinase-ABC did not affect basal inhibitory synaptic transmission. However, short-term plasticity, particularly burst-induced depression, was enhanced at PV→PC synapses following enzymatic treatments. Long-term plasticity experiments demonstrated that CSPGs are essential for NMDA-induced iLTP at SST→PC synapses, whereas the digestion of hyaluronic acid by hyaluronidase impaired iLTP at PV→PC synapses. This indicates a synapse-specific role of CSPGs and hyaluronic acid in regulating GABAergic plasticity. Additionally, we report the presence of cryptic GABAergic plasticity at PV→PC synapses induced by prolonged NMDA application, which became evident after CSPG digestion and was absent under control conditions. Our results underscore the differential impact of ECM degradation on inhibitory synaptic plasticity, highlighting the synapse-specific interplay between ECM components and specific GABAergic synapses. This offers new perspectives in studies on learning and critical period timing.
Collapse
Affiliation(s)
- Jadwiga Jabłońska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland
| | - Grzegorz Wiera
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland.
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 3a Chalubinskiego Str., 50-368 Wroclaw, Poland.
| |
Collapse
|
6
|
Borsdorf S, Zeug A, Wu Y, Mitroshina E, Vedunova M, Gaitonde SA, Bouvier M, Wehr MC, Labus J, Ponimaskin E. The cell adhesion molecule CD44 acts as a modulator of 5-HT7 receptor functions. Cell Commun Signal 2024; 22:563. [PMID: 39580460 PMCID: PMC11585102 DOI: 10.1186/s12964-024-01931-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Homo- and heteromerization of G protein-coupled receptors (GPCRs) plays an important role in the regulation of receptor functions. Recently, we demonstrated an interaction between the serotonin receptor 7 (5-HT7R), a class A GPCR, and the cell adhesion molecule CD44. However, the functional consequences of this interaction on 5-HT7R-mediated signaling remained enigmatic. METHODS Using a quantitative FRET (Förster resonance energy transfer) approach, we determined the affinities for the formation of homo- and heteromeric complexes of 5-HT7R and CD44. The impact of heteromerization on 5-HT7R-mediated cAMP signaling was assessed using a cAMP responsive luciferase assay and a FRET-based cAMP biosensor under basal conditions as well as upon pharmacological modulation of the 5-HT7R and/or CD44 with specific ligands. We also investigated receptor-mediated G protein activation using BRET (bioluminescence resonance energy transfer)-based biosensors in both, homo- and heteromeric conditions. Finally, we analyzed expression profiles for 5-HT7R and CD44 in the brain during development. RESULTS We found that homo- and heteromerization of the 5-HT7R and CD44 occur at similar extent. Functionally, heteromerization increased 5-HT7R-mediated cAMP production under basal conditions. In contrast, agonist-mediated cAMP production was decreased in the presence of CD44. Mechanistically, this might be explained by increased Gαs and decreased GαoB activation by 5-HT7R/CD44 heteromers. Unexpectedly, treatment of the heteromeric complex with the CD44 ligand hyaluronic acid boosted constitutive 5-HT7R-mediated cAMP signaling and receptor-mediated transcription, suggesting the existence of a transactivation mechanism. CONCLUSIONS Interaction with the hyaluronan receptor CD44 modulates both the constitutive activity of 5-HT7R as well as its agonist-mediated signaling. Heteromerization also results in the transactivation of 5-HT7R-mediated signaling via CD44 ligand.
Collapse
Affiliation(s)
- Saskia Borsdorf
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Yuxin Wu
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elena Mitroshina
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Maria Vedunova
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Supriya A Gaitonde
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, Canada
| | - Michael C Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Systasy Bioscience GmbH, Planegg-Martinsried, Germany
| | - Josephine Labus
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
7
|
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
|
8
|
Mishchenko TA, Klimenko MO, Guryev EL, Savelyev AG, Krysko DV, Gudkov SV, Khaydukov EV, Zvyagin AV, Vedunova MV. Enhancing glioma treatment with 3D scaffolds laden with upconversion nanoparticles and temozolomide in orthotopic mouse model. Front Chem 2024; 12:1445664. [PMID: 39498377 PMCID: PMC11532134 DOI: 10.3389/fchem.2024.1445664] [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: 06/07/2024] [Accepted: 09/23/2024] [Indexed: 11/07/2024] Open
Abstract
Targeted drug delivery for primary brain tumors, particularly gliomas, is currently a promising approach to reduce patient relapse rates. The use of substitutable scaffolds, which enable the sustained release of clinically relevant doses of anticancer medications, offers the potential to decrease the toxic burden on the patient's organism while also enhancing their quality of life and overall survival. Upconversion nanoparticles (UCNPs) are being actively explored as promising agents for detection and monitoring of tumor growth, and as therapeutic agents that can provide isolated therapeutic effects and enhance standard chemotherapy. Our study is focused on the feasibility of constructing scaffolds using methacrylated hyaluronic acid with additional impregnation of UCNPs and the chemotherapeutic drug temozolomide (TMZ) for glioma treatment. The designed scaffolds have been demonstrated their efficacy as a drug and UCNPs delivery system for gliomas. Using the aggressive orthotopic glioma model in vivo, it was found that the scaffolds possess the capacity to ameliorate neurological disorders in mice. Moreover, upon intracranial co-implantation of the scaffolds and glioma cells, the constructs disintegrate into distinct segments, augmenting the release of UCNPs into the surrounding tissue and concurrently reducing postoperative damage to brain tissue. The use of TMZ in the scaffold composition contributed to restraining glioma development and the reduction of tumor invasiveness. Our findings unveil promising prospects for the application of photopolymerizable biocompatible scaffolds in the realm of neuro-oncology.
Collapse
Affiliation(s)
- Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria O. Klimenko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Evgenii L. Guryev
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alexander G. Savelyev
- Laboratory of Laser Biomedicine, NRC “Kurchatov Institute”, Moscow, Russia
- D. I. Mendeleev Russian University of Chemical Technology, Moscow, Russia
| | - Dmitri V. Krysko
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Cell Death Investigation and Therapy Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Sergey V. Gudkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow, Russia
| | - Evgeny V. Khaydukov
- D. I. Mendeleev Russian University of Chemical Technology, Moscow, Russia
- Petrovsky National Research Center of Surgery, Moscow, Russia
- Department of Biomaterials and Bionanotechnology, Laboratory "Polymers for biology", Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Andrei V. Zvyagin
- Molecular Immunology Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, Moscow, Russia
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Sirius, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
9
|
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: 6] [Impact Index Per Article: 6.0] [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
|
10
|
Li X, Wu X, Lu T, Kuang C, Si Y, Zheng W, Li Z, Xue Y. Perineuronal Nets in the CNS: Architects of Memory and Potential Therapeutic Target in Neuropsychiatric Disorders. Int J Mol Sci 2024; 25:3412. [PMID: 38542386 PMCID: PMC10970535 DOI: 10.3390/ijms25063412] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 11/11/2024] Open
Abstract
The extracellular matrix (ECM) within the brain possesses a distinctive composition and functionality, influencing a spectrum of physiological and pathological states. Among its constituents, perineuronal nets (PNNs) are unique ECM structures that wrap around the cell body of many neurons and extend along their dendrites within the central nervous system (CNS). PNNs are pivotal regulators of plasticity in CNS, both during development and adulthood stages. Characterized by their condensed glycosaminoglycan-rich structures and heterogeneous molecular composition, PNNs not only offer neuroprotection but also participate in signal transduction, orchestrating neuronal activity and plasticity. Interfering with the PNNs in adult animals induces the reactivation of critical period plasticity, permitting modifications in neuronal connections and promoting the recovery of neuroplasticity following spinal cord damage. Interestingly, in the adult brain, PNN expression is dynamic, potentially modulating plasticity-associated states. Given their multifaceted roles, PNNs have emerged as regulators in the domains of learning, memory, addiction behaviors, and other neuropsychiatric disorders. In this review, we aimed to address how PNNs contribute to the memory processes in physiological and pathological conditions.
Collapse
Affiliation(s)
- Xue Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xianwen Wu
- Department of Laboratory Animal Sciences, Peking University Health Sciences Center, Beijing 100191, China;
| | - Tangsheng Lu
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Chenyan Kuang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang 050017, China;
| | - Yue Si
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wei Zheng
- Peking-Tsinghua Centre for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China;
| | - Zhonghao Li
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yanxue Xue
- National Institute on Drug Dependence, Peking University, Beijing 100191, China; (X.L.); (T.L.); (Y.S.); (Z.L.)
| |
Collapse
|
11
|
Cathomas F, Lin HY, Chan KL, Li L, Parise LF, Alvarez J, Durand-de Cuttoli R, Aubry AV, Muhareb S, Desland F, Shimo Y, Ramakrishnan A, Estill M, Ferrer-Pérez C, Parise EM, Wilk CM, Kaster MP, Wang J, Sowa A, Janssen WG, Costi S, Rahman A, Fernandez N, Campbell M, Swirski FK, Nestler EJ, Shen L, Merad M, Murrough JW, Russo SJ. Circulating myeloid-derived MMP8 in stress susceptibility and depression. Nature 2024; 626:1108-1115. [PMID: 38326622 PMCID: PMC10901735 DOI: 10.1038/s41586-023-07015-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/29/2023] [Indexed: 02/09/2024]
Abstract
Psychosocial stress has profound effects on the body, including the immune system and the brain1,2. Although a large number of pre-clinical and clinical studies have linked peripheral immune system alterations to stress-related disorders such as major depressive disorder (MDD)3, the underlying mechanisms are not well understood. Here we show that expression of a circulating myeloid cell-specific proteinase, matrix metalloproteinase 8 (MMP8), is increased in the serum of humans with MDD as well as in stress-susceptible mice following chronic social defeat stress (CSDS). In mice, we show that this increase leads to alterations in extracellular space and neurophysiological changes in the nucleus accumbens (NAc), as well as altered social behaviour. Using a combination of mass cytometry and single-cell RNA sequencing, we performed high-dimensional phenotyping of immune cells in circulation and in the brain and demonstrate that peripheral monocytes are strongly affected by stress. In stress-susceptible mice, both circulating monocytes and monocytes that traffic to the brain showed increased Mmp8 expression following chronic social defeat stress. We further demonstrate that circulating MMP8 directly infiltrates the NAc parenchyma and controls the ultrastructure of the extracellular space. Depleting MMP8 prevented stress-induced social avoidance behaviour and alterations in NAc neurophysiology and extracellular space. Collectively, these data establish a mechanism by which peripheral immune factors can affect central nervous system function and behaviour in the context of stress. Targeting specific peripheral immune cell-derived matrix metalloproteinases could constitute novel therapeutic targets for stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hsiao-Yun Lin
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenny L Chan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long Li
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lyonna F Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johana Alvarez
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio V Aubry
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samer Muhareb
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fiona Desland
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yusuke Shimo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Molly Estill
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carmen Ferrer-Pérez
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C Matthias Wilk
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manuella P Kaster
- Department of Biochemistry, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Jun Wang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allison Sowa
- Microscopy CoRE and Advanced Bioimaging Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Janssen
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Microscopy CoRE and Advanced Bioimaging Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara Costi
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Adeeb Rahman
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicolas Fernandez
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Filip K Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Marc and Jennifer Lipschultz Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Brain and Body Research Center of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
12
|
Elder GA, Gama Sosa MA, De Gasperi R, Perez Garcia G, Perez GM, Abutarboush R, Kawoos U, Zhu CW, Janssen WGM, Stone JR, Hof PR, Cook DG, Ahlers ST. The Neurovascular Unit as a Locus of Injury in Low-Level Blast-Induced Neurotrauma. Int J Mol Sci 2024; 25:1150. [PMID: 38256223 PMCID: PMC10816929 DOI: 10.3390/ijms25021150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Blast-induced neurotrauma has received much attention over the past decade. Vascular injury occurs early following blast exposure. Indeed, in animal models that approximate human mild traumatic brain injury or subclinical blast exposure, vascular pathology can occur in the presence of a normal neuropil, suggesting that the vasculature is particularly vulnerable. Brain endothelial cells and their supporting glial and neuronal elements constitute a neurovascular unit (NVU). Blast injury disrupts gliovascular and neurovascular connections in addition to damaging endothelial cells, basal laminae, smooth muscle cells, and pericytes as well as causing extracellular matrix reorganization. Perivascular pathology becomes associated with phospho-tau accumulation and chronic perivascular inflammation. Disruption of the NVU should impact activity-dependent regulation of cerebral blood flow, blood-brain barrier permeability, and glymphatic flow. Here, we review work in an animal model of low-level blast injury that we have been studying for over a decade. We review work supporting the NVU as a locus of low-level blast injury. We integrate our findings with those from other laboratories studying similar models that collectively suggest that damage to astrocytes and other perivascular cells as well as chronic immune activation play a role in the persistent neurobehavioral changes that follow blast injury.
Collapse
Affiliation(s)
- Gregory A. Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Georgina Perez Garcia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Rania Abutarboush
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Carolyn W. Zhu
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William G. M. Janssen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James R. Stone
- Department of Radiology and Medical Imaging, University of Virginia, 480 Ray C Hunt Drive, Charlottesville, VA 22903, USA;
| | - Patrick R. Hof
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA 98108, USA;
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
| |
Collapse
|
13
|
Naik A, Jensen V, Bakketun CB, Enger R, Hrabetova S, Hrabe J. BubbleDrive, a low-volume incubation chamber for acute brain slices. Sci Rep 2023; 13:20005. [PMID: 37973847 PMCID: PMC10654715 DOI: 10.1038/s41598-023-45949-9] [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: 05/14/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
Acute brain slices are a common and useful preparation in experimental neuroscience. A wide range of incubation chambers for brain slices exists but only a few are designed with very low volumes of the bath solution in mind. Such chambers are necessary when high-cost chemicals are to be added to the solution or when small amounts of substances released by the slice are to be collected for analysis. The principal challenge in designing a very low-volume incubation chamber is maintaining good oxygenation and flow without mechanically disturbing or damaging the slices. We designed and validated BubbleDrive, a 3D-printed incubation chamber with a minimum volume of 1.5 mL which can hold up to three coronal mouse slices from one hemisphere. It employs the carbogen gas bubbles to drive the flow circulation in a consistent and reproducible manner, and without disturbing the brain slices. The BubbleDrive design and construction were successfully validated by comparison to a conventional large-volume incubation chamber in several experimental designs involving measurements of extracellular diffusion parameters, the electrophysiology of neuronal and astrocytic networks, and the effectiveness of slice incubation with hyaluronidase enzyme.
Collapse
Affiliation(s)
- Aditi Naik
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
- Neural and Behavioral Science Graduate Program, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA
| | - Vidar Jensen
- Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Cecilie Bugge Bakketun
- Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Rune Enger
- Letten Centre, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Sabina Hrabetova
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA.
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA.
| | - Jan Hrabe
- Department of Cell Biology, State University of New York Downstate Health Sciences University, Brooklyn, NY, USA.
- Translational Neuroscience Laboratories, Center for Biomedical Imaging and Neuromodulation, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
14
|
Verissimo DCA, Camillo-Andrade AC, Santos MDM, Sprengel SL, Zanine SC, Borba LAB, Carvalho PC, da G. Fischer JDS. Proteomics reveals differentially regulated pathways when comparing grade 2 and 4 astrocytomas. PLoS One 2023; 18:e0290087. [PMID: 37967105 PMCID: PMC10651032 DOI: 10.1371/journal.pone.0290087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/25/2023] [Indexed: 11/17/2023] Open
Abstract
Astrocytic tumors are known for their high progression capacity and high mortality rates; in this regard, proteins correlated to prognosis can aid medical conduct. Although several genetic changes related to progression from grade 2 to grade 4 astrocytoma are already known, mRNA copies do not necessarily correlate with protein abundance and therefore could shadow further comprehension about this tumor's biology. This motivates us to seek for complementary strategies to study tumor progression at the protein level. Here we compare the proteomic profile of biopsies from patients with grade 2 (diffuse, n = 6) versus grade 4 astrocytomas (glioblastomas, n = 10) using shotgun proteomics. Data analysis performed with PatternLab for proteomics identified 5,206 and 6,004 proteins in the 2- and 4-grade groups, respectively. Our results revealed seventy-four differentially abundant proteins (p < 0.01); we then shortlist those related to greater malignancy. We also describe molecular pathways distinctly activated in the two groups, such as differences in the organization of the extracellular matrix, decisive both in tumor invasiveness and in signaling for cell division, which, together with marked contrasts in energy metabolism, are determining factors in the speed of growth and dissemination of these neoplasms. The degradation pathways of GABA, enriched in the grade 2 group, is consistent with a favorable prognosis. Other functions such as platelet degranulation, apoptosis, and activation of the MAPK pathway were correlated to grade 4 tumors and, consequently, unfavorable prognoses. Our results provide an important survey of molecular pathways involved in glioma pathogenesis for these histopathological groups.
Collapse
Affiliation(s)
- Denildo C. A. Verissimo
- Laboratory for Structural and Computational Proteomics—Carlos Chagas Institute, Fiocruz Paraná, Curitiba, PR, Brazil
- Clinical Hospital of the Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Amanda C. Camillo-Andrade
- Laboratory for Structural and Computational Proteomics—Carlos Chagas Institute, Fiocruz Paraná, Curitiba, PR, Brazil
| | - Marlon D. M. Santos
- Laboratory for Structural and Computational Proteomics—Carlos Chagas Institute, Fiocruz Paraná, Curitiba, PR, Brazil
| | - Sergio L. Sprengel
- Clinical Hospital of the Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Simone C. Zanine
- Clinical Hospital of the Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Luis A. B. Borba
- Clinical Hospital of the Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Paulo C. Carvalho
- Laboratory for Structural and Computational Proteomics—Carlos Chagas Institute, Fiocruz Paraná, Curitiba, PR, Brazil
| | - Juliana de S. da G. Fischer
- Laboratory for Structural and Computational Proteomics—Carlos Chagas Institute, Fiocruz Paraná, Curitiba, PR, Brazil
| |
Collapse
|
15
|
Chen MH, Lin HC, Chao T, Lee VSY, Hou CL, Wang TJ, Chen JR. Hyaluronic Acid Conjugated with 17β-Estradiol Effectively Alleviates Estropause-Induced Cognitive Deficits in Rats. Int J Mol Sci 2023; 24:15569. [PMID: 37958552 PMCID: PMC10649161 DOI: 10.3390/ijms242115569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Women are at a higher risk of cognitive impairments and Alzheimer's disease (AD), particularly after the menopause, when the estrous cycle becomes irregular and diminishes. Numerous studies have shown that estrogen deficiency, especially estradiol (E2) deficiency, plays a key role in this phenomenon. Recently, a novel polymeric drug, hyaluronic acid-17β-estradiol conjugate (HA-E2), has been introduced for the delivery of E2 to brain tissues. Studies have indicated that HA-E2 crosses the blood-brain barrier (BBB) and facilitates a prolonged E2 release profile while lowering the risk of estrogen-supplement-related side effects. In this study, we used ovariohysterectomy (OHE) rats, a postmenopausal cognitive deficit model, to explore the effect of a 2-week HA-E2 treatment (210 ng/kg body weight, twice a week) on the cholinergic septo-hippocampal innervation system, synaptic transmission in hippocampal pyramidal neurons and cognitive improvements. Our study revealed an 11% rise in choline acetyltransferase (ChAT) expression in both the medial septal nucleus (MS nucleus) and the hippocampus, along with a 14-18% increase in dendritic spine density in hippocampal pyramidal neurons, following HA-E2 treatment in OHE rats. These enhancements prompted the recovery of cognitive functions such as spatial learning and memory. These findings suggest that HA-E2 may prevent and improve estrogen-deficiency-induced cognitive impairment and AD.
Collapse
Affiliation(s)
- Mu-Hsuan Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| | - Hsiao-Chun Lin
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| | - Tzu Chao
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| | - Viola Szu-Yuan Lee
- Basic Research Division, Holy Stone Healthcare Co., Ltd., Taipei 11493, Taiwan; (V.S.-Y.L.); (C.-L.H.)
| | - Chia-Lung Hou
- Basic Research Division, Holy Stone Healthcare Co., Ltd., Taipei 11493, Taiwan; (V.S.-Y.L.); (C.-L.H.)
| | - Tsyr-Jiuan Wang
- Department of Nursing, National Taichung University of Science and Technology, No. 193, Section 1, Sanmin Rd., Taichung 403027, Taiwan
| | - Jeng-Rung Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, No. 145, Xingda Rd., Taichung 402202, Taiwan; (M.-H.C.); (H.-C.L.); (T.C.)
| |
Collapse
|
16
|
Kotla NG, Mohd Isa IL, Larrañaga A, Maddiboyina B, Swamy SK, Sivaraman G, Vemula PK. Hyaluronic Acid-Based Bioconjugate Systems, Scaffolds, and Their Therapeutic Potential. Adv Healthc Mater 2023; 12:e2203104. [PMID: 36972409 DOI: 10.1002/adhm.202203104] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/04/2023] [Indexed: 03/29/2023]
Abstract
In recent years, the development of hyaluronic acid or hyaluronan (HA) based scaffolds, medical devices, bioconjugate systems have expanded into a broad range of research and clinical applications. Research findings over the last two decades suggest that the abundance of HA in most mammalian tissues with distinctive biological roles and chemical simplicity for modifications have made it an attractive material with a rapidly growing global market. Besides its use as native forms, HA has received much interest on so-called "HA-bioconjugates" and "modified-HA systems". In this review, the importance of chemical modifications of HA, underlying rationale approaches, and various advancements of bioconjugate derivatives with their potential physicochemical, and pharmacological advantages are summarized. This review also highlights the current and emerging HA-based conjugates of small molecules, macromolecules, crosslinked systems, and surface coating strategies with their biological implications, including their potentials and key challenges discussed in detail.
Collapse
Affiliation(s)
- Niranjan G Kotla
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, 560065, India
| | - Isma Liza Mohd Isa
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, 56000, Malaysia
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Faculty of Engineering, University of the Basque Country (UPV/EHU), Bilbao, 48013, Spain
| | - Balaji Maddiboyina
- Department of Medical Writing, Freyr Solutions, Hyderabad, Telangana, 500081, India
| | - Samantha K Swamy
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, 9037, Norway
| | - Gandhi Sivaraman
- Department of Chemistry, Gandhigram Rural Institute (Deemed to be University), Gandhigram, Tamil Nadu, 624302, India
| | - Praveen K Vemula
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, 560065, India
| |
Collapse
|
17
|
Rike WA, Stern S. Proteins and Transcriptional Dysregulation of the Brain Extracellular Matrix in Parkinson's Disease: A Systematic Review. Int J Mol Sci 2023; 24:ijms24087435. [PMID: 37108598 PMCID: PMC10138539 DOI: 10.3390/ijms24087435] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The extracellular matrix (ECM) of the brain is a dynamic structure made up of a vast network of bioactive macromolecules that modulate cellular events. Structural, organizational, and functional changes in these macromolecules due to genetic variation or environmental stressors are thought to affect cellular functions and may result in disease. However, most mechanistic studies to date usually focus on the cellular aspects of diseases and pay less attention to the relevance of the processes governing the dynamic nature of the extracellular matrix in disease pathogenesis. Thus, due to the ECM's diversified biological roles, increasing interest in its involvement in disease, and the lack of sufficient compiled evidence regarding its relationship with Parkinson's disease (PD) pathology, we aimed to compile the existing evidence to boost the current knowledge on the area and provide refined guidance for the future research. Here, in this review, we gathered postmortem brain tissue and induced pluripotent stem cell (iPSC)-related studies from PubMed and Google Scholar to identify, summarize and describe common macromolecular alterations in the expression of brain ECM components in Parkinson's disease (PD). A literature search was conducted up until 10 February 2023. The overall hits from the database and manual search for proteomic and transcriptome studies were 1243 and 1041 articles, respectively. Following a full-text review, 10 articles from proteomic and 24 from transcriptomic studies were found to be eligible for inclusion. According to proteomic studies, proteins such as collagens, fibronectin, annexins, and tenascins were recognized to be differentially expressed in Parkinson's disease. Transcriptomic studies displayed dysregulated pathways including ECM-receptor interaction, focal adhesion, and cell adhesion molecules in Parkinson's disease. A limited number of relevant studies were accessed from our search, indicating that much work remains to be carried out to better understand the roles of the ECM in neurodegeneration and Parkinson's disease. However, we believe that our review will elicit focused primary studies and thus support the ongoing efforts of the discovery and development of diagnostic biomarkers as well as therapeutic agents for Parkinson's disease.
Collapse
Affiliation(s)
- Wote Amelo Rike
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
18
|
Tewari BP, Woo AM, Prim CE, Chaunsali L, Kimbrough IF, Engel K, Browning JL, Campbell SL, Sontheimer H. Perineuronal nets support astrocytic ion and glutamate homeostasis at tripartite synapses. RESEARCH SQUARE 2023:rs.3.rs-2501039. [PMID: 36778342 PMCID: PMC9915772 DOI: 10.21203/rs.3.rs-2501039/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Perineuronal nets (PNNs) are dense, negatively charged extracellular matrices that cover the cell body of fast-spiking inhibitory neurons. Synapses can be embedded and stabilized by PNNs believed to prevent synaptic plasticity. We find that in cortical fast-spiking interneurons synaptic terminals localize to perforations in the PNNs, 95% of which contain either excitatory or inhibitory synapses or both. The majority of terminals also colocalize with astrocytic processes expressing Kir4.1 as well as glutamate (Glu) and GABA transporters, hence can be considered tripartite synapses. In the adult brain, degradation of PNNs does not alter axonal terminals but causes expansion of astrocytic coverage of the neuronal somata. However, loss of PNNs impairs astrocytic transmitter and K+ uptake and causes spillage of synaptic Glu into the extrasynaptic space. This data suggests a hitherto unrecognized role of PNNs, to synergize with astrocytes to contain synaptically released signals.
Collapse
Affiliation(s)
- Bhanu P. Tewari
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - AnnaLin M. Woo
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Courtney E. Prim
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Lata Chaunsali
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ian F. Kimbrough
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kaliroi Engel
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | | | | | - Harald Sontheimer
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
19
|
Mueller-Buehl C, Wegrzyn D, Bauch J, Faissner A. Regulation of the E/I-balance by the neural matrisome. Front Mol Neurosci 2023; 16:1102334. [PMID: 37143468 PMCID: PMC10151766 DOI: 10.3389/fnmol.2023.1102334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
In the mammalian cortex a proper excitatory/inhibitory (E/I) balance is fundamental for cognitive functions. Especially γ-aminobutyric acid (GABA)-releasing interneurons regulate the activity of excitatory projection neurons which form the second main class of neurons in the cortex. During development, the maturation of fast-spiking parvalbumin-expressing interneurons goes along with the formation of net-like structures covering their soma and proximal dendrites. These so-called perineuronal nets (PNNs) represent a specialized form of the extracellular matrix (ECM, also designated as matrisome) that stabilize structural synapses but prevent the formation of new connections. Consequently, PNNs are highly involved in the regulation of the synaptic balance. Previous studies revealed that the formation of perineuronal nets is accompanied by an establishment of mature neuronal circuits and by a closure of critical windows of synaptic plasticity. Furthermore, it has been shown that PNNs differentially impinge the integrity of excitatory and inhibitory synapses. In various neurological and neuropsychiatric disorders alterations of PNNs were described and aroused more attention in the last years. The following review gives an update about the role of PNNs for the maturation of parvalbumin-expressing interneurons and summarizes recent findings about the impact of PNNs in different neurological and neuropsychiatric disorders like schizophrenia or epilepsy. A targeted manipulation of PNNs might provide an interesting new possibility to indirectly modulate the synaptic balance and the E/I ratio in pathological conditions.
Collapse
|
20
|
Righes Marafiga J, Calcagnotto ME. Electrophysiology of Dendritic Spines: Information Processing, Dynamic Compartmentalization, and Synaptic Plasticity. ADVANCES IN NEUROBIOLOGY 2023; 34:103-141. [PMID: 37962795 DOI: 10.1007/978-3-031-36159-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
For many years, synaptic transmission was considered as information transfer between presynaptic neuron and postsynaptic cell. At the synaptic level, it was thought that dendritic arbors were only receiving and integrating all information flow sent along to the soma, while axons were primarily responsible for point-to-point information transfer. However, it is important to highlight that dendritic spines play a crucial role as postsynaptic components in central nervous system (CNS) synapses, not only integrating and filtering signals to the soma but also facilitating diverse connections with axons from many different sources. The majority of excitatory connections from presynaptic axonal terminals occurs on postsynaptic spines, although a subset of GABAergic synapses also targets spine heads. Several studies have shown the vast heterogeneous morphological, biochemical, and functional features of dendritic spines related to synaptic processing. In this chapter (adding to the relevant data on the biophysics of spines described in Chap. 1 of this book), we address the up-to-date functional dendritic characteristics assessed through electrophysiological approaches, including backpropagating action potentials (bAPs) and synaptic potentials mediated in dendritic and spine compartmentalization, as well as describing the temporal and spatial dynamics of glutamate receptors in the spines related to synaptic plasticity.
Collapse
Affiliation(s)
- Joseane Righes Marafiga
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Maria Elisa Calcagnotto
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
21
|
Amoah DK. Advances in the understanding and enhancement of the human cognitive functions of learning and memory. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Learning and memory are among the key cognitive functions that drive the human experience. As such, any defective condition associated with these cognitive domains could affect our navigation through everyday life. For years, researchers have been working toward having a clear understanding of how learning and memory work, as well as ways to improve them. Many advances have been made, as well as some challenges that have also been faced in the process. That notwithstanding, there are prospects with regards to the frontier of the enhancement of learning and memory in humans. This review article selectively highlights four broad areas of focus in research into the understanding and enhancement of learning and memory. Brain stimulation, effects of sleep, effects of stress and emotion, and synaptic plasticity are the main focal areas of this review, in terms of some pivotal research works, findings and theories.
Collapse
Affiliation(s)
- Daniel Kofi Amoah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra LG 25, Ghana
| |
Collapse
|
22
|
Tewari BP, Chaunsali L, Prim CE, Sontheimer H. A glial perspective on the extracellular matrix and perineuronal net remodeling in the central nervous system. Front Cell Neurosci 2022; 16:1022754. [PMID: 36339816 PMCID: PMC9630365 DOI: 10.3389/fncel.2022.1022754] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
A structural scaffold embedding brain cells and vasculature is known as extracellular matrix (ECM). The physical appearance of ECM in the central nervous system (CNS) ranges from a diffused, homogeneous, amorphous, and nearly omnipresent matrix to highly organized distinct morphologies such as basement membranes and perineuronal nets (PNNs). ECM changes its composition and organization during development, adulthood, aging, and in several CNS pathologies. This spatiotemporal dynamic nature of the ECM and PNNs brings a unique versatility to their functions spanning from neurogenesis, cell migration and differentiation, axonal growth, and pathfinding cues, etc., in the developing brain, to stabilizing synapses, neuromodulation, and being an active partner of tetrapartite synapses in the adult brain. The malleability of ECM and PNNs is governed by both intrinsic and extrinsic factors. Glial cells are among the major extrinsic factors that facilitate the remodeling of ECM and PNN, thereby acting as key regulators of diverse functions of ECM and PNN in health and diseases. In this review, we discuss recent advances in our understanding of PNNs and how glial cells are central to ECM and PNN remodeling in normal and pathological states of the CNS.
Collapse
|
23
|
Fell CW, Hagelkruys A, Cicvaric A, Horrer M, Liu L, Li JSS, Stadlmann J, Polyansky AA, Mereiter S, Tejada MA, Kokotović T, Achuta VS, Scaramuzza A, Twyman KA, Morrow MM, Juusola J, Yan H, Wang J, Burmeister M, Choudhury B, Andersen TL, Wirnsberger G, Holmskov U, Perrimon N, Žagrović B, Monje FJ, Moeller JB, Penninger JM, Nagy V. FIBCD1 is an endocytic GAG receptor associated with a novel neurodevelopmental disorder. EMBO Mol Med 2022; 14:e15829. [PMID: 35916241 PMCID: PMC9449597 DOI: 10.15252/emmm.202215829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/21/2022] Open
Abstract
Whole-exome sequencing of two patients with idiopathic complex neurodevelopmental disorder (NDD) identified biallelic variants of unknown significance within FIBCD1, encoding an endocytic acetyl group-binding transmembrane receptor with no known function in the central nervous system. We found that FIBCD1 preferentially binds and endocytoses glycosaminoglycan (GAG) chondroitin sulphate-4S (CS-4S) and regulates GAG content of the brain extracellular matrix (ECM). In silico molecular simulation studies and GAG binding analyses of patient variants determined that such variants are loss-of-function by disrupting FIBCD1-CS-4S association. Gene knockdown in flies resulted in morphological disruption of the neuromuscular junction and motor-related behavioural deficits. In humans and mice, FIBCD1 is expressed in discrete brain regions, including the hippocampus. Fibcd1 KO mice exhibited normal hippocampal neuronal morphology but impaired hippocampal-dependent learning. Further, hippocampal synaptic remodelling in acute slices from Fibcd1 KO mice was deficient but restored upon enzymatically modulating the ECM. Together, we identified FIBCD1 as an endocytic receptor for GAGs in the brain ECM and a novel gene associated with an NDD, revealing a critical role in nervous system structure, function and plasticity.
Collapse
Affiliation(s)
- Christopher W Fell
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Astrid Hagelkruys
- VBC – Vienna BioCenter CampusIMBA, Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
| | - Ana Cicvaric
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and PharmacologyMedical University of ViennaViennaAustria
- Department of Psychiatry and Behavioral Sciences, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Marion Horrer
- VBC – Vienna BioCenter CampusIMBA, Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
| | - Lucy Liu
- Department of Genetics, Harvard Medical SchoolHoward Hughes Medical InstituteBostonMAUSA
| | - Joshua Shing Shun Li
- Department of Genetics, Harvard Medical SchoolHoward Hughes Medical InstituteBostonMAUSA
| | - Johannes Stadlmann
- VBC – Vienna BioCenter CampusIMBA, Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
- Institute of BiochemistryUniversity of Natural Resource and Life SciencesViennaAustria
| | - Anton A Polyansky
- Department of Structural and Computational Biology, Max Perutz LabsUniversity of ViennaViennaAustria
- MM Shemyakin and Yu A Ovchinnikov Institute of Bioorganic ChemistryRussian Academy of SciencesMoscowRussia
| | - Stefan Mereiter
- VBC – Vienna BioCenter CampusIMBA, Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
| | - Miguel Angel Tejada
- VBC – Vienna BioCenter CampusIMBA, Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
- Research Unit on Women's Health‐Institute of Health Research INCLIVAValenciaSpain
| | - Tomislav Kokotović
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Venkat Swaroop Achuta
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Angelica Scaramuzza
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- Department of NeurologyMedical University of ViennaViennaAustria
| | | | | | | | - Huifang Yan
- Department of PediatricsPeking University First HospitalBeijingChina
- Joint International Research Center of Translational and Clinical ResearchBeijingChina
| | - Jingmin Wang
- Department of PediatricsPeking University First HospitalBeijingChina
- Joint International Research Center of Translational and Clinical ResearchBeijingChina
| | - Margit Burmeister
- Michigan Neuroscience InstituteUniversity of MichiganAnn ArborMIUSA
- Departments of Computational Medicine & Bioinformatics, Psychiatry and Human GeneticsUniversity of MichiganAnn ArborMIUSA
| | - Biswa Choudhury
- Department of Cellular and Molecular MedicineUCSDLa JollaCAUSA
| | - Thomas Levin Andersen
- Clinical Cell Biology, Department of PathologyOdense University HospitalOdenseDenmark
- Pathology Research Unit, Department of Clinical Research and Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Gerald Wirnsberger
- VBC – Vienna BioCenter CampusIMBA, Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
- Apeiron Biologics AG, Vienna BioCenter CampusViennaAustria
| | - Uffe Holmskov
- Cancer and Inflammation Research, Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical SchoolHoward Hughes Medical InstituteBostonMAUSA
| | - Bojan Žagrović
- Department of Structural and Computational Biology, Max Perutz LabsUniversity of ViennaViennaAustria
| | - Francisco J Monje
- Department of Neurophysiology and Neuropharmacology, Centre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Jesper Bonnet Moeller
- Cancer and Inflammation Research, Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Danish Institute for Advanced StudyUniversity of Southern DenmarkOdenseDenmark
| | - Josef M Penninger
- VBC – Vienna BioCenter CampusIMBA, Institute of Molecular Biotechnology of the Austrian Academy of SciencesViennaAustria
- Department of Medical Genetics, Life Science InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of NeurologyMedical University of ViennaViennaAustria
| |
Collapse
|
24
|
Mishchenko TA, Klimenko MO, Kuznetsova AI, Yarkov RS, Savelyev AG, Sochilina AV, Mariyanats AO, Popov VK, Khaydukov EV, Zvyagin AV, Vedunova MV. 3D-printed hyaluronic acid hydrogel scaffolds impregnated with neurotrophic factors (BDNF, GDNF) for post-traumatic brain tissue reconstruction. Front Bioeng Biotechnol 2022; 10:895406. [PMID: 36091441 PMCID: PMC9453866 DOI: 10.3389/fbioe.2022.895406] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Brain tissue reconstruction posttraumatic injury remains a long-standing challenge in neurotransplantology, where a tissue-engineering construct (scaffold, SC) with specific biochemical properties is deemed the most essential building block. Such three-dimensional (3D) hydrogel scaffolds can be formed using brain-abundant endogenous hyaluronic acid modified with glycidyl methacrylate by employing our proprietary photopolymerisation technique. Herein, we produced 3D hyaluronic scaffolds impregnated with neurotrophic factors (BDNF, GDNF) possessing 600 kPa Young’s moduli and 336% swelling ratios. Stringent in vitro testing of fabricated scaffolds using primary hippocampal cultures revealed lack of significant cytotoxicity: the number of viable cells in the SC+BDNF (91.67 ± 1.08%) and SC+GDNF (88.69 ± 1.2%) groups was comparable to the sham values (p > 0.05). Interestingly, BDNF-loaded scaffolds promoted the stimulation of neuronal process outgrowth during the first 3 days of cultures development (day 1: 23.34 ± 1.46 µm; day 3: 37.26 ± 1.98 µm, p < 0.05, vs. sham), whereas GDNF-loaded scaffolds increased the functional activity of neuron-glial networks of cultures at later stages of cultivation (day 14) manifested in a 1.3-fold decrease in the duration coupled with a 2.4-fold increase in the frequency of Ca2+ oscillations (p < 0.05, vs. sham). In vivo studies were carried out using C57BL/6 mice with induced traumatic brain injury, followed by surgery augmented with scaffold implantation. We found positive dynamics of the morphological changes in the treated nerve tissue in the post-traumatic period, where the GDNF-loaded scaffolds indicated more favorable regenerative potential. In comparison with controls, the physiological state of the treated mice was improved manifested by the absence of severe neurological deficit, significant changes in motor and orienting-exploratory activity, and preservation of the ability to learn and retain long-term memory. Our results suggest in favor of biocompatibility of GDNF-loaded scaffolds, which provide a platform for personalized brain implants stimulating effective morphological and functional recovery of nerve tissue after traumatic brain injury.
Collapse
Affiliation(s)
- Tatiana A. Mishchenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria O. Klimenko
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alisa I. Kuznetsova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Roman S. Yarkov
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alexander G. Savelyev
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Anastasia V. Sochilina
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Alexandra O. Mariyanats
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
| | - Vladimir K. Popov
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
| | - Evgeny V. Khaydukov
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk-Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Andrei V. Zvyagin
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- MQ Photonics Centre, Macquarie University, Sydney, NSW, Australia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
- *Correspondence: Maria V. Vedunova,
| |
Collapse
|
25
|
Fawcett JW, Fyhn M, Jendelova P, Kwok JCF, Ruzicka J, Sorg BA. The extracellular matrix and perineuronal nets in memory. Mol Psychiatry 2022; 27:3192-3203. [PMID: 35760878 PMCID: PMC9708575 DOI: 10.1038/s41380-022-01634-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 02/06/2023]
Abstract
All components of the CNS are surrounded by a diffuse extracellular matrix (ECM) containing chondroitin sulphate proteoglycans (CSPGs), heparan sulphate proteoglycans (HSPGs), hyaluronan, various glycoproteins including tenascins and thrombospondin, and many other molecules that are secreted into the ECM and bind to ECM components. In addition, some neurons, particularly inhibitory GABAergic parvalbumin-positive (PV) interneurons, are surrounded by a more condensed cartilage-like ECM called perineuronal nets (PNNs). PNNs surround the soma and proximal dendrites as net-like structures that surround the synapses. Attention has focused on the role of PNNs in the control of plasticity, but it is now clear that PNNs also play an important part in the modulation of memory. In this review we summarize the role of the ECM, particularly the PNNs, in the control of various types of memory and their participation in memory pathology. PNNs are now being considered as a target for the treatment of impaired memory. There are many potential treatment targets in PNNs, mainly through modulation of the sulphation, binding, and production of the various CSPGs that they contain or through digestion of their sulphated glycosaminoglycans.
Collapse
Affiliation(s)
- James W Fawcett
- John van Geest Centre for Brain Repair, Department Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK.
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic.
| | - Marianne Fyhn
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Pavla Jendelova
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Jessica C F Kwok
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jiri Ruzicka
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Videnska 1083, Prague 4, Prague, Czech Republic
| | - Barbara A Sorg
- Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, USA
| |
Collapse
|
26
|
Chelyshev YA, Kabdesh IM, Mukhamedshina YO. Extracellular Matrix in Neural Plasticity and Regeneration. Cell Mol Neurobiol 2022; 42:647-664. [PMID: 33128689 PMCID: PMC11441266 DOI: 10.1007/s10571-020-00986-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) is a fundamental component of biological tissues. The ECM in the central nervous system (CNS) is unique in both composition and function. Functions such as learning, memory, synaptogenesis, and plasticity are regulated by numerous ECM molecules. The neural ECM acts as a non-specific physical barrier that modulates neuronal plasticity and axon regeneration. There are two specialized types of ECM in the CNS, diffuse perisynaptic ECM and condensed ECM, which selectively surround the perikaryon and initial part of dendritic trees in subtypes of neurons, forming perineuronal nets. This review presents the current knowledge about the role of important neuronal ECM molecules in maintaining the basic functions of a neuron, including electrogenesis and the ability to form neural circuits. The review mainly focuses on the role of ECM components that participate in the control of key events such as cell survival, axonal growth, and synaptic remodeling. Particular attention is drawn to the numerous molecular partners of the main ECM components. These regulatory molecules are integrated into the cell membrane or disposed into the matrix itself in solid or soluble form. The interaction of the main matrix components with molecular partners seems essential in molecular mechanisms controlling neuronal functions. Special attention is paid to the chondroitin sulfate proteoglycan 4, type 1 transmembrane protein, neural-glial antigen 2 (NG2/CSPG4), whose cleaved extracellular domain is such a molecular partner that it not only acts directly on neural and vascular cells, but also exerts its influence indirectly by binding to resident ECM molecules.
Collapse
Affiliation(s)
- Yurii A Chelyshev
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Ilyas M Kabdesh
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlevskaya St 18, Kazan, Tatarstan, Russia, 420008
| | - Yana O Mukhamedshina
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia.
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlevskaya St 18, Kazan, Tatarstan, Russia, 420008.
| |
Collapse
|
27
|
Dankovich TM, Rizzoli SO. The Synaptic Extracellular Matrix: Long-Lived, Stable, and Still Remarkably Dynamic. Front Synaptic Neurosci 2022; 14:854956. [PMID: 35350469 PMCID: PMC8957932 DOI: 10.3389/fnsyn.2022.854956] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/16/2022] [Indexed: 01/09/2023] Open
Abstract
In the adult brain, synapses are tightly enwrapped by lattices of the extracellular matrix that consist of extremely long-lived molecules. These lattices are deemed to stabilize synapses, restrict the reorganization of their transmission machinery, and prevent them from undergoing structural or morphological changes. At the same time, they are expected to retain some degree of flexibility to permit occasional events of synaptic plasticity. The recent understanding that structural changes to synapses are significantly more frequent than previously assumed (occurring even on a timescale of minutes) has called for a mechanism that allows continual and energy-efficient remodeling of the extracellular matrix (ECM) at synapses. Here, we review recent evidence for such a process based on the constitutive recycling of synaptic ECM molecules. We discuss the key characteristics of this mechanism, focusing on its roles in mediating synaptic transmission and plasticity, and speculate on additional potential functions in neuronal signaling.
Collapse
Affiliation(s)
- Tal M. Dankovich
- University Medical Center Göttingen, Institute for Neuro- and Sensory Physiology, Göttingen, Germany
- International Max Planck Research School for Neuroscience, Göttingen, Germany
- *Correspondence: Tal M. Dankovich Silvio O. Rizzoli
| | - Silvio O. Rizzoli
- University Medical Center Göttingen, Institute for Neuro- and Sensory Physiology, Göttingen, Germany
- Biostructural Imaging of Neurodegeneration (BIN) Center & Multiscale Bioimaging Excellence Center, Göttingen, Germany
- *Correspondence: Tal M. Dankovich Silvio O. Rizzoli
| |
Collapse
|
28
|
Laham BJ, Gould E. How Stress Influences the Dynamic Plasticity of the Brain’s Extracellular Matrix. Front Cell Neurosci 2022; 15:814287. [PMID: 35145379 PMCID: PMC8821883 DOI: 10.3389/fncel.2021.814287] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse and structured extracellular matrix (ECM) comprise ∼20% of the brain’s volume and play important roles in development and adult plasticity. Perineuronal nets (PNNs), specialized ECM structures that surround certain types of neurons in the brain, emerge during the postnatal period, making their development and maintenance potentially sensitive to experience. Recent studies have shown that stress affects diffuse ECM as well as PNNs, and that such effects are dependent on life stage and brain region. Given that the ECM participates in synaptic plasticity, the generation of neuronal oscillations, and synchronous firing across brain regions, all of which have been linked to cognition and emotional regulation, ECM components may be candidate therapeutic targets for stress-induced neuropsychiatric disease. This review considers the influence of stress over diffuse and structured ECM during postnatal life with a focus on functional outcomes and the potential for translational relevance.
Collapse
|
29
|
Kandell R, Kudryashev JA, Kwon EJ. Targeting the Extracellular Matrix in Traumatic Brain Injury Increases Signal Generation from an Activity-Based Nanosensor. ACS NANO 2021; 15:20504-20516. [PMID: 34870408 PMCID: PMC8716428 DOI: 10.1021/acsnano.1c09064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Traumatic brain injury (TBI) is a critical public health concern and major contributor to death and long-term disability. After the initial trauma, a sustained secondary injury involving a complex continuum of pathophysiology unfolds, ultimately leading to the destruction of nervous tissue. One disease hallmark of TBI is ectopic protease activity, which can mediate cell death, extracellular matrix breakdown, and inflammation. We previously engineered a fluorogenic activity-based nanosensor for TBI (TBI-ABN) that passively accumulates in the injured brain across the disrupted vasculature and generates fluorescent signal in response to calpain-1 cleavage, thus enabling in situ visualization of TBI-associated calpain-1 protease activity. In this work, we hypothesized that actively targeting the extracellular matrix (ECM) of the injured brain would improve nanosensor accumulation in the injured brain beyond passive delivery alone and lead to increased nanosensor activation. We evaluated several peptides that bind exposed/enriched ECM constituents in the brain and discovered that nanomaterials modified with peptides that target hyaluronic acid (HA) displayed widespread distribution across the injury lesion, in particular colocalizing with perilesional and hippocampal neurons. Modifying TBI-ABN with HA-targeting peptide led to increases in activation in a ligand-valency-dependent manner, up to 6.6-fold in the injured cortex compared to a nontargeted nanosensor. This robust nanosensor activation enabled 3D visualization of injury-specific protease activity in a cleared and intact brain. In our work, we establish that targeting brain ECM with peptide ligands can be leveraged to improve the distribution and function of a bioresponsive imaging nanomaterial.
Collapse
Affiliation(s)
| | | | - Ester J. Kwon
- Department of Bioengineering, University of California−San Diego, La Jolla, California 92093, United States
| |
Collapse
|
30
|
Izgordu MS, Uzgur EI, Ulag S, Sahin A, Karademir Yilmaz B, Kilic B, Ekren N, Oktar FN, Gunduz O. Investigation of 3D-Printed Polycaprolactone-/Polyvinylpyrrolidone-Based Constructs. Cartilage 2021; 13:626S-635S. [PMID: 31893944 PMCID: PMC8804864 DOI: 10.1177/1947603519897302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The aim of this study is to evaluate the mechanical and biological performance of cartilage-like constructs produced by 3D printing. During the investigation, poly(ε-caprolactone) (PCL) and polyvinylpyrrolidone (PVP) were used as a matrix polymer and low-molecular-weight chitosan (CS), hyaluronic acid (HA), and alginic acid sodium salt (SA) were integrated separately with the polymer matrix to fabricate the constructs. Thermal, mechanical, morphology, and chemical properties and swelling, degradation, and biocompatibility behaviors were evaluated in detail. With the addition of 3 fillers, the melting temperature of the matrix increased with the addition of fillers, and PCL/3wt.%PVP/1wt.%HA had the highest melting temperature value. Mechanical characterization results demonstrated that the printed PCL/3wt.%PVP/1wt.%CS displayed the highest compressive strength of around 9.51 MPa. The compressive strength difference between the PCL/3wt.%PVP and PCL/3wt.%PVP/1wt.%CS was 5.38 MPa. Biocompatibility properties of the constructs were tested by mitochondrial dehydrogenase activity, and in vitro studies showed that the PCL/3wt.%PVP/1wt.%HA composite construct had more cell viability than the other constructs by making use of the mesenchymal stem cell line.
Collapse
Affiliation(s)
- Muhammet Sefa Izgordu
- Department of Bioengineering, Faculty of
Engineering, Marmara University, Istanbul, Turkey
| | - Evren Isa Uzgur
- Department of Bioengineering, Faculty of
Engineering, Marmara University, Istanbul, Turkey
| | - Songul Ulag
- Center for Nanotechnology &
Biomaterials Application and Research (NBUAM), Marmara University, Istanbul,
Turkey,Metallurgical and Materials Engineering,
Institute of Pure and Applied Sciences, Marmara University, Istanbul, Turkey
| | - Ali Sahin
- Genetic and Metabolic Diseases Research
Center (GEMHAM), Marmara University, Istanbul, Turkey,Department of Biochemistry, Faculty of
Medicine, Marmara University, Istanbul, Turkey
| | - Betul Karademir Yilmaz
- Genetic and Metabolic Diseases Research
Center (GEMHAM), Marmara University, Istanbul, Turkey,Department of Biochemistry, Faculty of
Medicine, Marmara University, Istanbul, Turkey
| | - Beyhan Kilic
- Center for Nanotechnology &
Biomaterials Application and Research (NBUAM), Marmara University, Istanbul,
Turkey,Department of Electrical Engineering,
Faculty of Electrical and Electronics, Yildiz Technical University, Istanbul,
Turkey
| | - Nazmi Ekren
- Center for Nanotechnology &
Biomaterials Application and Research (NBUAM), Marmara University, Istanbul,
Turkey,Department of Electrical and Electronics
Engineering, Faculty of Technology, Marmara University, Istanbul, Turkey
| | - Faik Nuzhet Oktar
- Department of Bioengineering, Faculty of
Engineering, Marmara University, Istanbul, Turkey,Center for Nanotechnology &
Biomaterials Application and Research (NBUAM), Marmara University, Istanbul,
Turkey
| | - Oguzhan Gunduz
- Center for Nanotechnology &
Biomaterials Application and Research (NBUAM), Marmara University, Istanbul,
Turkey,Department of Metallurgical and
Materials Engineering, Faculty of Technology, Marmara University, Istanbul,
Turkey,Oguzhan Gunduz, Department of Metallurgical
and Materials Engineering, Faculty of Technology, Marmara University, Metalurji
ve Malzeme Müh. Göztepe Kampüsü, Kadıköy, Istanbul, 34722, Turkey.
| |
Collapse
|
31
|
Zakusilo FT, Kerry O'Banion M, Gelbard HA, Seluanov A, Gorbunova V. Matters of size: Roles of hyaluronan in CNS aging and disease. Ageing Res Rev 2021; 72:101485. [PMID: 34634492 PMCID: PMC8903057 DOI: 10.1016/j.arr.2021.101485] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/29/2022]
Abstract
Involvement of extracellular matrix (ECM) components in aging and age-related neurodegeneration is not well understood. The role of hyaluronan (HA), a major extracellular matrix glycosaminoglycan, in malignancy and inflammation is gaining new understanding. In particular, the differential biological effects of high molecular weight (HMW-HA) and low molecular weight hyaluronan (LMW-HA), and the mechanism behind such differences are being uncovered. Tightly regulated in the brain, HA can have diverse effects on cellular development, growth and degeneration. In this review, we summarize the homeostasis and signaling of HA in healthy tissue, discuss its distribution and ontogeny in the central nervous system (CNS), summarize evidence for its involvement in age-related neurodegeneration and Alzheimer Disease (AD), and assess the potential of HA as a therapeutic target in the CNS.
Collapse
Affiliation(s)
- Frances Tolibzoda Zakusilo
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA; Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA; Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Harris A Gelbard
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA; Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY, USA
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| |
Collapse
|
32
|
Lin JZ, Duan MR, Lin N, Zhao WJ. The emerging role of the chondroitin sulfate proteoglycan family in neurodegenerative diseases. Rev Neurosci 2021; 32:737-750. [PMID: 33655733 DOI: 10.1515/revneuro-2020-0146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/07/2021] [Indexed: 02/05/2023]
Abstract
Chondroitin sulfate (CS) is a kind of linear polysaccharide that is covalently linked to proteins to form proteoglycans. Chondroitin sulfate proteoglycans (CSPGs) consist of a core protein, with one or more CS chains covalently attached. CSPGs are precisely regulated and they exert a variety of physiological functions by binding to adhesion molecules and growth factors. Widely distributed in the nervous system in human body, CSPGs contribute to the major component of extracellular matrix (ECM), where they play an important role in the development and maturation of the nervous system, as well as in the pathophysiological response to damage to the central nervous system (CNS). While there are more than 30 types of CSPGs, this review covers the roles of the most important ones, including versican, aggrecan, neurocan and NG2 in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and multiple sclerosis. The updated reports of the treatment of neurodegenerative diseases are involving CSPGs.
Collapse
Affiliation(s)
- Jia-Zhe Lin
- Neurosurgical Department, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Ming-Rui Duan
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Nuan Lin
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou 515041, Guangdong, China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China
| |
Collapse
|
33
|
Abstract
Perineuronal nets (PNNs) are condensed extracellular matrix (ECM) assemblies of
polyanionic chondroitin sulfate proteoglycans, hyaluronan, and tenascins that
primarily wrap around GABAergic parvalbumin (PV) interneurons. During
development, PNN formation terminates the critical period of neuroplasticity, a
process that can be reversed by experimental disruption of PNNs. Perineuronal
nets also regulate the intrinsic properties of the enclosed PV neurons thereby
maintaining their inhibitory activity. Recent studies have implicated PNNs in
central nervous system diseases as well as PV neuron dysfunction; consequently,
they have further been associated with altered inhibition, particularly in the
genesis of epilepsy. A wide range of seizure presentations in human and rodent
models exhibit ECM remodeling with PNN disruption due to elevated protease
activity. Inhibition of PNN proteolysis reduces seizure activity suggesting that
PNN degrading enzymes may be potential novel therapeutic targets.
Collapse
Affiliation(s)
- Lata Chaunsali
- School of Neuroscience Graduate Program, Virginia Tech, Blacksburg, VA, USA.,Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Bhanu P Tewari
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Harald Sontheimer
- Glial Biology in Health, Disease, and Cancer Center, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| |
Collapse
|
34
|
Wilson ES, Litwa K. Synaptic Hyaluronan Synthesis and CD44-Mediated Signaling Coordinate Neural Circuit Development. Cells 2021; 10:2574. [PMID: 34685554 PMCID: PMC8533746 DOI: 10.3390/cells10102574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/20/2022] Open
Abstract
The hyaluronan-based extracellular matrix is expressed throughout nervous system development and is well-known for the formation of perineuronal nets around inhibitory interneurons. Since perineuronal nets form postnatally, the role of hyaluronan in the initial formation of neural circuits remains unclear. Neural circuits emerge from the coordinated electrochemical signaling of excitatory and inhibitory synapses. Hyaluronan localizes to the synaptic cleft of developing excitatory synapses in both human cortical spheroids and the neonatal mouse brain and is diminished in the adult mouse brain. Given this developmental-specific synaptic localization, we sought to determine the mechanisms that regulate hyaluronan synthesis and signaling during synapse formation. We demonstrate that hyaluronan synthase-2, HAS2, is sufficient to increase hyaluronan levels in developing neural circuits of human cortical spheroids. This increased hyaluronan production reduces excitatory synaptogenesis, promotes inhibitory synaptogenesis, and suppresses action potential formation. The hyaluronan receptor, CD44, promotes hyaluronan retention and suppresses excitatory synaptogenesis through regulation of RhoGTPase signaling. Our results reveal mechanisms of hyaluronan synthesis, retention, and signaling in developing neural circuits, shedding light on how disease-associated hyaluronan alterations can contribute to synaptic defects.
Collapse
Affiliation(s)
| | - Karen Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA;
| |
Collapse
|
35
|
Extracellular Metalloproteinases in the Plasticity of Excitatory and Inhibitory Synapses. Cells 2021; 10:cells10082055. [PMID: 34440823 PMCID: PMC8391609 DOI: 10.3390/cells10082055] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Long-term synaptic plasticity is shaped by the controlled reorganization of the synaptic proteome. A key component of this process is local proteolysis performed by the family of extracellular matrix metalloproteinases (MMPs). In recent years, considerable progress was achieved in identifying extracellular proteases involved in neuroplasticity phenomena and their protein substrates. Perisynaptic metalloproteinases regulate plastic changes at synapses through the processing of extracellular and membrane proteins. MMP9 was found to play a crucial role in excitatory synapses by controlling the NMDA-dependent LTP component. In addition, MMP3 regulates the L-type calcium channel-dependent form of LTP as well as the plasticity of neuronal excitability. Both MMP9 and MMP3 were implicated in memory and learning. Moreover, altered expression or mutations of different MMPs are associated with learning deficits and psychiatric disorders, including schizophrenia, addiction, or stress response. Contrary to excitatory drive, the investigation into the role of extracellular proteolysis in inhibitory synapses is only just beginning. Herein, we review the principal mechanisms of MMP involvement in the plasticity of excitatory transmission and the recently discovered role of proteolysis in inhibitory synapses. We discuss how different matrix metalloproteinases shape dynamics and turnover of synaptic adhesome and signal transduction pathways in neurons. Finally, we discuss future challenges in exploring synapse- and plasticity-specific functions of different metalloproteinases.
Collapse
|
36
|
Strackeljan L, Baczynska E, Cangalaya C, Baidoe-Ansah D, Wlodarczyk J, Kaushik R, Dityatev A. Microglia Depletion-Induced Remodeling of Extracellular Matrix and Excitatory Synapses in the Hippocampus of Adult Mice. Cells 2021; 10:1862. [PMID: 34440631 PMCID: PMC8393852 DOI: 10.3390/cells10081862] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix (ECM) plays a key role in synaptogenesis and the regulation of synaptic functions in the central nervous system. Recent studies revealed that in addition to dopaminergic and serotoninergic neuromodulatory systems, microglia also contribute to the regulation of ECM remodeling. In the present work, we investigated the physiological role of microglia in the remodeling of perineuronal nets (PNNs), predominantly associated with parvalbumin-immunopositive (PV+) interneurons, and the perisynaptic ECM around pyramidal neurons in the hippocampus. Adult mice were treated with PLX3397 (pexidartinib), as the inhibitor of colony-stimulating factor 1 receptor (CSF1-R), to deplete microglia. Then, confocal analysis of the ECM and synapses was performed. Although the elimination of microglia did not alter the overall number or intensity of PNNs in the CA1 region of the hippocampus, it decreased the size of PNN holes and elevated the expression of the surrounding ECM. In the neuropil area in the CA1 str. radiatum, the depletion of microglia increased the expression of perisynaptic ECM proteoglycan brevican, which was accompanied by the elevated expression of presynaptic marker vGluT1 and the increased density of dendritic spines. Thus, microglia regulate the homeostasis of pre- and postsynaptic excitatory terminals and the surrounding perisynaptic ECM as well as the fine structure of PNNs enveloping perisomatic-predominantly GABAergic-synapses.
Collapse
Affiliation(s)
- Luisa Strackeljan
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; (L.S.); (C.C.); (D.B.-A.)
| | - Ewa Baczynska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland; (E.B.); (J.W.)
| | - Carla Cangalaya
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; (L.S.); (C.C.); (D.B.-A.)
- Institut für Biochemie und Zellbiologie, Medical Faculty, Otto-von-Guericke-University, 39120 Magdeburg, Germany
- ESF International Graduate School on Analysis, Imaging and Modelling of Neuronal and Inflammatory Processes, 39120 Magdeburg, Germany
| | - David Baidoe-Ansah
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; (L.S.); (C.C.); (D.B.-A.)
| | - Jakub Wlodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland; (E.B.); (J.W.)
| | - Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; (L.S.); (C.C.); (D.B.-A.)
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; (L.S.); (C.C.); (D.B.-A.)
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany
| |
Collapse
|
37
|
Hughes EG, Stockton ME. Premyelinating Oligodendrocytes: Mechanisms Underlying Cell Survival and Integration. Front Cell Dev Biol 2021; 9:714169. [PMID: 34368163 PMCID: PMC8335399 DOI: 10.3389/fcell.2021.714169] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022] Open
Abstract
In the central nervous system, oligodendrocytes produce myelin sheaths that enwrap neuronal axons to provide trophic support and increase conduction velocity. New oligodendrocytes are produced throughout life through a process referred to as oligodendrogenesis. Oligodendrogenesis consists of three canonical stages: the oligodendrocyte precursor cell (OPC), the premyelinating oligodendrocyte (preOL), and the mature oligodendrocyte (OL). However, the generation of oligodendrocytes is inherently an inefficient process. Following precursor differentiation, a majority of premyelinating oligodendrocytes are lost, likely due to apoptosis. If premyelinating oligodendrocytes progress through this survival checkpoint, they generate new myelinating oligodendrocytes in a process we have termed integration. In this review, we will explore the intrinsic and extrinsic signaling pathways that influence preOL survival and integration by examining the intrinsic apoptotic pathways, metabolic demands, and the interactions between neurons, astrocytes, microglia, and premyelinating oligodendrocytes. Additionally, we will discuss similarities between the maturation of newly generated neurons and premyelinating oligodendrocytes. Finally, we will consider how increasing survival and integration of preOLs has the potential to increase remyelination in multiple sclerosis. Deepening our understanding of premyelinating oligodendrocyte biology may open the door for new treatments for demyelinating disease and will help paint a clearer picture of how new oligodendrocytes are produced throughout life to facilitate brain function.
Collapse
Affiliation(s)
- Ethan G Hughes
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Michael E Stockton
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
38
|
Pantazopoulos H, Katsel P, Haroutunian V, Chelini G, Klengel T, Berretta S. Molecular signature of extracellular matrix pathology in schizophrenia. Eur J Neurosci 2021; 53:3960-3987. [PMID: 33070392 PMCID: PMC8359380 DOI: 10.1111/ejn.15009] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023]
Abstract
Growing evidence points to a critical involvement of the extracellular matrix (ECM) in the pathophysiology of schizophrenia (SZ). Decreases of perineuronal nets (PNNs) and altered expression of chondroitin sulphate proteoglycans (CSPGs) in glial cells have been identified in several brain regions. GWAS data have identified several SZ vulnerability variants of genes encoding for ECM molecules. Given the potential relevance of ECM functions to the pathophysiology of this disorder, it is necessary to understand the extent of ECM changes across brain regions, their region- and sex-specificity and which ECM components contribute to these changes. We tested the hypothesis that the expression of genes encoding for ECM molecules may be broadly disrupted in SZ across several cortical and subcortical brain regions and include key ECM components as well as factors such as ECM posttranslational modifications and regulator factors. Gene expression profiling of 14 neocortical brain regions, caudate, putamen and hippocampus from control subjects (n = 14/region) and subjects with SZ (n = 16/region) was conducted using Affymetrix microarray analysis. Analysis across brain regions revealed widespread dysregulation of ECM gene expression in cortical and subcortical brain regions in SZ, impacting several ECM functional key components. SRGN, CD44, ADAMTS1, ADAM10, BCAN, NCAN and SEMA4G showed some of the most robust changes. Region-, sex- and age-specific gene expression patterns and correlation with cognitive scores were also detected. Taken together, these findings contribute to emerging evidence for large-scale ECM dysregulation in SZ and point to molecular pathways involved in PNN decreases, glial cell dysfunction and cognitive impairment in SZ.
Collapse
Affiliation(s)
- Harry Pantazopoulos
- Department of Neurobiology and Anatomical SciencesUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Pavel Katsel
- Department of PsychiatryThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of NeuroscienceThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Mental Illness Research Education ClinicalCenters of Excellence (MIRECC)JJ Peters VA Medical CenterBronxNYUSA
| | - Vahram Haroutunian
- Department of PsychiatryThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Department of NeuroscienceThe Icahn School of Medicine at Mount SinaiNew YorkNYUSA
- Mental Illness Research Education ClinicalCenters of Excellence (MIRECC)JJ Peters VA Medical CenterBronxNYUSA
| | - Gabriele Chelini
- Translational Neuroscience LaboratoryMclean HospitalBelmontMAUSA
- Department of PsychiatryHarvard Medical SchoolBostonMAUSA
| | - Torsten Klengel
- Department of PsychiatryHarvard Medical SchoolBostonMAUSA
- Translational Molecular Genomics LaboratoryMclean HospitalBelmontMAUSA
- Department of PsychiatryUniversity Medical Center GöttingenGöttingenGermany
| | - Sabina Berretta
- Translational Neuroscience LaboratoryMclean HospitalBelmontMAUSA
- Department of PsychiatryHarvard Medical SchoolBostonMAUSA
- Program in NeuroscienceHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
39
|
Ucar B. Natural biomaterials in brain repair: A focus on collagen. Neurochem Int 2021; 146:105033. [PMID: 33785419 DOI: 10.1016/j.neuint.2021.105033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials derived from natural resources have increasingly been used for versatile applications in the central nervous system (CNS). Thanks to their biocompatibility and biodegradability, natural biomaterials offer vast possibilities for future clinical repair strategies for the CNS. These materials can be used for diverse applications such as hydrogels to fill the tissue cavities, microparticles to deliver drugs across the blood-brain barrier, and scaffolds to transplant stem cells. In this review, various uses of prominent protein and polysaccharide biomaterials, with a special focus on collagen, in repair and regenerative applications for the brain are summarized together with their individual advantages and disadvantages.
Collapse
Affiliation(s)
- Buket Ucar
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
40
|
Wingert JC, Sorg BA. Impact of Perineuronal Nets on Electrophysiology of Parvalbumin Interneurons, Principal Neurons, and Brain Oscillations: A Review. Front Synaptic Neurosci 2021; 13:673210. [PMID: 34040511 PMCID: PMC8141737 DOI: 10.3389/fnsyn.2021.673210] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/14/2021] [Indexed: 12/11/2022] Open
Abstract
Perineuronal nets (PNNs) are specialized extracellular matrix structures that surround specific neurons in the brain and spinal cord, appear during critical periods of development, and restrict plasticity during adulthood. Removal of PNNs can reinstate juvenile-like plasticity or, in cases of PNN removal during early developmental stages, PNN removal extends the critical plasticity period. PNNs surround mainly parvalbumin (PV)-containing, fast-spiking GABAergic interneurons in several brain regions. These inhibitory interneurons profoundly inhibit the network of surrounding neurons via their elaborate contacts with local pyramidal neurons, and they are key contributors to gamma oscillations generated across several brain regions. Among other functions, these gamma oscillations regulate plasticity associated with learning, decision making, attention, cognitive flexibility, and working memory. The detailed mechanisms by which PNN removal increases plasticity are only beginning to be understood. Here, we review the impact of PNN removal on several electrophysiological features of their underlying PV interneurons and nearby pyramidal neurons, including changes in intrinsic and synaptic membrane properties, brain oscillations, and how these changes may alter the integration of memory-related information. Additionally, we review how PNN removal affects plasticity-associated phenomena such as long-term potentiation (LTP), long-term depression (LTD), and paired-pulse ratio (PPR). The results are discussed in the context of the role of PV interneurons in circuit function and how PNN removal alters this function.
Collapse
Affiliation(s)
- Jereme C Wingert
- Program in Neuroscience, Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| | - Barbara A Sorg
- Program in Neuroscience, Robert S. Dow Neurobiology Laboratories, Legacy Research Institute, Portland, OR, United States
| |
Collapse
|
41
|
Dembitskaya Y, Gavrilov N, Kraev I, Doronin M, Tang Y, Li L, Semyanov A. Attenuation of the extracellular matrix increases the number of synapses but suppresses synaptic plasticity through upregulation of SK channels. Cell Calcium 2021; 96:102406. [PMID: 33848733 DOI: 10.1016/j.ceca.2021.102406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 01/01/2023]
Abstract
The effect of brain extracellular matrix (ECM) on synaptic plasticity remains controversial. Here, we show that targeted enzymatic attenuation with chondroitinase ABC (ChABC) of ECM triggers the appearance of new glutamatergic synapses on hippocampal pyramidal neurons, thereby increasing the amplitude of field EPSPs while decreasing both the mean miniature EPSC amplitude and AMPA/NMDA ratio. Although the increased proportion of 'unpotentiated' synapses caused by ECM attenuation should promote long-term potentiation (LTP), surprisingly, LTP was suppressed. The upregulation of small conductance Ca2+-activated K+ (SK) channels decreased the excitability of pyramidal neurons, thereby suppressing LTP. A blockade of SK channels restored cell excitability and enhanced LTP; this enhancement was abolished by a blockade of Rho-associated protein kinase (ROCK), which is involved in the maturation of dendritic spines. Thus, targeting ECM elicits the appearance of new synapses, which can have potential applications in regenerative medicine. However, this process is compensated for by a reduction in postsynaptic neuron excitability, preventing network overexcitation at the expense of synaptic plasticity.
Collapse
Affiliation(s)
- Yulia Dembitskaya
- Department of Molecular Neurobiology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street 16/10, Moscow, 117997, Russia
| | - Nikolay Gavrilov
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, 603950, Russia
| | - Igor Kraev
- Electron Microscopy Suite, Faculty of Science, Technology, Engineering and Mathematics, Open University, Milton Keynes MK7 6AA, UK
| | - Maxim Doronin
- Department of Molecular Neurobiology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street 16/10, Moscow, 117997, Russia
| | - Yong Tang
- School of Acupuncture and Tuina and International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Li
- Department of Physiology, Jiaxing University College of Medicine, Zhejiang, 314033 China
| | - Alexey Semyanov
- Department of Molecular Neurobiology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya Street 16/10, Moscow, 117997, Russia; Department of Physiology, Jiaxing University College of Medicine, Zhejiang, 314033 China; Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya Str 19с1, Moscow, 119146, Russia.
| |
Collapse
|
42
|
An Extracellular Perspective on CNS Maturation: Perineuronal Nets and the Control of Plasticity. Int J Mol Sci 2021; 22:ijms22052434. [PMID: 33670945 PMCID: PMC7957817 DOI: 10.3390/ijms22052434] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
During restricted time windows of postnatal life, called critical periods, neural circuits are highly plastic and are shaped by environmental stimuli. In several mammalian brain areas, from the cerebral cortex to the hippocampus and amygdala, the closure of the critical period is dependent on the formation of perineuronal nets. Perineuronal nets are a condensed form of an extracellular matrix, which surrounds the soma and proximal dendrites of subsets of neurons, enwrapping synaptic terminals. Experimentally disrupting perineuronal nets in adult animals induces the reactivation of critical period plasticity, pointing to a role of the perineuronal net as a molecular brake on plasticity as the critical period closes. Interestingly, in the adult brain, the expression of perineuronal nets is remarkably dynamic, changing its plasticity-associated conditions, including memory processes. In this review, we aimed to address how perineuronal nets contribute to the maturation of brain circuits and the regulation of adult brain plasticity and memory processes in physiological and pathological conditions.
Collapse
|
43
|
MMP-9 Signaling Pathways That Engage Rho GTPases in Brain Plasticity. Cells 2021; 10:cells10010166. [PMID: 33467671 PMCID: PMC7830260 DOI: 10.3390/cells10010166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023] Open
Abstract
The extracellular matrix (ECM) has been identified as a critical factor affecting synaptic function. It forms a functional scaffold that provides both the structural support and the reservoir of signaling molecules necessary for communication between cellular constituents of the central nervous system (CNS). Among numerous ECM components and modifiers that play a role in the physiological and pathological synaptic plasticity, matrix metalloproteinase 9 (MMP-9) has recently emerged as a key molecule. MMP-9 may contribute to the dynamic remodeling of structural and functional plasticity by cleaving ECM components and cell adhesion molecules. Notably, MMP-9 signaling was shown to be indispensable for long-term memory formation that requires synaptic remodeling. The core regulators of the dynamic reorganization of the actin cytoskeleton and cell adhesion are the Rho family of GTPases. These proteins have been implicated in the control of a wide range of cellular processes occurring in brain physiology and pathology. Here, we discuss the contribution of Rho GTPases to MMP-9-dependent signaling pathways in the brain. We also describe how the regulation of Rho GTPases by post-translational modifications (PTMs) can influence these processes.
Collapse
|
44
|
Modulatory properties of extracellular matrix glycosaminoglycans and proteoglycans on neural stem cells behavior: Highlights on regenerative potential and bioactivity. Int J Biol Macromol 2021; 171:366-381. [PMID: 33422514 DOI: 10.1016/j.ijbiomac.2021.01.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/01/2021] [Accepted: 01/02/2021] [Indexed: 12/25/2022]
Abstract
Despite the poor regenerative capacity of the adult central nervous system (CNS) in mammals, two distinct regions, subventricular zone (SVZ) and the subgranular zone (SGZ), continue to generate new functional neurons throughout life which integrate into the pre-existing neuronal circuitry. This process is not fixed but highly modulated, revealing many intrinsic and extrinsic mechanisms by which this performance can be optimized for a given environment. The capacity for self-renewal, proliferation, migration, and multi-lineage potency of neural stem cells (NSCs) underlines the necessity of controlling stem cell fate. In this context, the native and local microenvironment plays a critical role, and the application of this highly organized architecture in the CNS has been considered as a fundamental concept in the generation of new effective therapeutic strategies in tissue engineering approaches. The brain extracellular matrix (ECM) is composed of biomacromolecules, including glycosaminoglycans, proteoglycans, and glycoproteins that provide various biological actions through biophysical and biochemical signaling pathways. Herein, we review predominantly the structure and function of the mentioned ECM composition and their regulatory impact on multiple and diversity of biological functions, including neural regeneration, survival, migration, differentiation, and final destiny of NSCs.
Collapse
|
45
|
Henneberger C, Bard L, Panatier A, Reynolds JP, Kopach O, Medvedev NI, Minge D, Herde MK, Anders S, Kraev I, Heller JP, Rama S, Zheng K, Jensen TP, Sanchez-Romero I, Jackson CJ, Janovjak H, Ottersen OP, Nagelhus EA, Oliet SHR, Stewart MG, Nägerl UV, Rusakov DA. LTP Induction Boosts Glutamate Spillover by Driving Withdrawal of Perisynaptic Astroglia. Neuron 2020; 108:919-936.e11. [PMID: 32976770 PMCID: PMC7736499 DOI: 10.1016/j.neuron.2020.08.030] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/14/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023]
Abstract
Extrasynaptic actions of glutamate are limited by high-affinity transporters expressed by perisynaptic astroglial processes (PAPs): this helps maintain point-to-point transmission in excitatory circuits. Memory formation in the brain is associated with synaptic remodeling, but how this affects PAPs and therefore extrasynaptic glutamate actions is poorly understood. Here, we used advanced imaging methods, in situ and in vivo, to find that a classical synaptic memory mechanism, long-term potentiation (LTP), triggers withdrawal of PAPs from potentiated synapses. Optical glutamate sensors combined with patch-clamp and 3D molecular localization reveal that LTP induction thus prompts spatial retreat of astroglial glutamate transporters, boosting glutamate spillover and NMDA-receptor-mediated inter-synaptic cross-talk. The LTP-triggered PAP withdrawal involves NKCC1 transporters and the actin-controlling protein cofilin but does not depend on major Ca2+-dependent cascades in astrocytes. We have therefore uncovered a mechanism by which a memory trace at one synapse could alter signal handling by multiple neighboring connections.
Collapse
Affiliation(s)
- Christian Henneberger
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK; Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany.
| | - Lucie Bard
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Aude Panatier
- INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France; Université de Bordeaux, 33000 Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France
| | - James P Reynolds
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Olga Kopach
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | | | - Daniel Minge
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Michel K Herde
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Stefanie Anders
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Igor Kraev
- Life Sciences, The Open University, Milton Keynes MK7 6AA, UK
| | - Janosch P Heller
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sylvain Rama
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Kaiyu Zheng
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Thomas P Jensen
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | | | - Colin J Jackson
- Research School of Chemistry, Australian National University, Acton, ACT 2601, Australia
| | - Harald Janovjak
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria; EMBL Australia, Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, VIC 3800, Australia
| | - Ole Petter Ottersen
- Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway; Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Stephane H R Oliet
- INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France; Université de Bordeaux, 33000 Bordeaux, France
| | | | - U Valentin Nägerl
- Université de Bordeaux, 33000 Bordeaux, France; Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33000 Bordeaux, France.
| | - Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK.
| |
Collapse
|
46
|
Washington PM, Lee C, Dwyer MKR, Konofagou EE, Kernie SG, Morrison B. Hyaluronidase reduced edema after experimental traumatic brain injury. J Cereb Blood Flow Metab 2020; 40:2026-2037. [PMID: 31648593 PMCID: PMC7786840 DOI: 10.1177/0271678x19882780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cerebral edema and the subsequent increased intracranial pressure are associated with mortality and poor outcome following traumatic brain injury. Previous in vitro studies have shown that the Gibbs-Donnan effect, which describes the tendency of a porous, negatively charged matrix to attract positive ions and water, applies to brain tissue and that enzymatic reduction of the fixed charge density can prevent tissue swelling. We tested whether hyaluronidase, an enzyme that degrades the large, negatively charged glycosaminoglycan hyaluronan, could reduce brain edema after traumatic brain injury. In vivo, intracerebroventricular injection of hyaluronidase after controlled cortical impact in mice reduced edema in the ipsilateral hippocampus at 24 h by both the wet-weight/dry-weight method (78.15 ± 0.65% vs. 80.4 ± 0.46%; p < 0.01) and T2-weighted magnetic resonance imaging (13.88 ± 3.09% vs. 29.23 ± 6.14%; p < 0.01). Hyaluronidase did not adversely affect blood-brain-barrier-integrity measured by dynamic contrast-enhanced magnetic resonance imaging, nor did hyaluronidase negatively affect functional recovery after controlled cortical impact measured with the rotarod or Morris water maze tasks. Reduction of fixed charge density by hyaluronidase was confirmed in cortical explants in vitro (5.46 ± 1.15 µg/mg vs. 7.76 ± 1.87 µg/mg; p < 0.05). These data demonstrate that targeting the fixed charge density with hyaluronidase reduced edema in an in vivo mouse model of traumatic brain injury.
Collapse
Affiliation(s)
- Patricia M Washington
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Changhee Lee
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Mary Kate R Dwyer
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Steven G Kernie
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Barclay Morrison
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Barclay Morrison III, Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
47
|
Stoyanov S, Sun W, Düsedau HP, Cangalaya C, Choi I, Mirzapourdelavar H, Baidoe-Ansah D, Kaushik R, Neumann J, Dunay IR, Dityatev A. Attenuation of the extracellular matrix restores microglial activity during the early stage of amyloidosis. Glia 2020; 69:182-200. [PMID: 32865286 DOI: 10.1002/glia.23894] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022]
Abstract
In the advanced stages of Alzheimer's disease (AD), microglia are transformed to an activated phenotype with thickened and retracted processes, migrate to the site of amyloid-beta (Aβ) plaques, and proliferate. In the early stages of AD, it is still poorly understood whether the microglial function is altered and which factors may regulate these changes. Here, we focused on studying microglia in the retrosplenial cortex (RSC) in 3- to 4-month-old 5xFAD mice as a transgenic mouse model of AD. At this age, there are neither Aβ plaques, nor activation of microglia, nor dysregulation in the expression of genes encoding major extracellular matrix (ECM) molecules or extracellular proteases in the RSC. Still, histochemical evaluation of the fine structure of neural ECM revealed increased levels of Wisteria floribunda agglutinin labeling in holes of perineuronal nets and changes in the perimeter of ECM barriers around the holes in 5xFAD mice. Two-photon vital microscopy demonstrated normal morphology and resting motility of microglia but strongly diminished number of microglial cells that migrated to the photolesion site in 5xFAD mice. Enzymatic digestion of ECM by chondroitinase ABC (ChABC) ameliorated this defect. Accordingly, the characterization of cell surface markers by flow cytometry demonstrated altered expression of microglial CD45. Moreover, ChABC treatment reduced the invasion of myeloid-derived mononuclear cells into the RSC of 5xFAD mice. Hence, the migration of both microglia and myeloid cells is altered during the early stages of amyloidosis and can be restored at least partially by the attenuation of the ECM.
Collapse
Affiliation(s)
- Stoyan Stoyanov
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Weilun Sun
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Carla Cangalaya
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Biochemistry, Otto-von-Guericke University, Magdeburg, Germany
| | - Ilseob Choi
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - David Baidoe-Ansah
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Jens Neumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity, 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
|
48
|
Diverse Roles for Hyaluronan and Hyaluronan Receptors in the Developing and Adult Nervous System. Int J Mol Sci 2020; 21:ijms21175988. [PMID: 32825309 PMCID: PMC7504301 DOI: 10.3390/ijms21175988] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hyaluronic acid (HA) plays a vital role in the extracellular matrix of neural tissues. Originally thought to hydrate tissues and provide mechanical support, it is now clear that HA is also a complex signaling molecule that can regulate cell processes in the developing and adult nervous systems. Signaling properties are determined by molecular weight, bound proteins, and signal transduction through specific receptors. HA signaling regulates processes such as proliferation, differentiation, migration, and process extension in a variety of cell types including neural stem cells, neurons, astrocytes, microglia, and oligodendrocyte progenitors. The synthesis and catabolism of HA and the expression of HA receptors are altered in disease and influence neuroinflammation and disease pathogenesis. This review discusses the roles of HA, its synthesis and breakdown, as well as receptor expression in neurodevelopment, nervous system function and disease.
Collapse
|
49
|
Yang X. Chondroitin sulfate proteoglycans: key modulators of neuronal plasticity, long-term memory, neurodegenerative, and psychiatric disorders. Rev Neurosci 2020; 31:555-568. [PMID: 32126020 DOI: 10.1515/revneuro-2019-0117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022]
Abstract
The chondroitin sulfate proteoglycans (CSPGs) are large groups of heterogenous proteoglycans that are mainly expressed by reactive astrocytes in the central nervous system (CNS). They share similar core proteins and are post-transcriptionally modified by chondroitin sulfate glycosaminoglycans. CSPGs are the major components of the perineuronal nets (PNN) that regulate the opening and closure of the critical period. Mounting reports have documented the crucial roles of CSPGs in restricting neuronal plasticity, axonal growth, and pathfinding during development as well as axonal regeneration after CNS injury. Moreover, CSPGs and PNNs modulate long-term memory, which impairments frequently happened in several neurodegenerative and psychiatric disorders. This review will shortly introduce the expression patterns of CSPGs during development and after injury, the PNNs constitutions, the roles of CSPGs and PNNs in axonal regrowth, discuss the most recently identified roles of CSPGs and PNNs in mediating long-term memory and their correlation with brain disorders, and finally, propose a short perspective of future investigations. Hopefully, further explorations may validate the therapeutic potentials of PNNs and CSPGs.
Collapse
Affiliation(s)
- Xin Yang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, P.R. China
| |
Collapse
|
50
|
Soria FN, Paviolo C, Doudnikoff E, Arotcarena ML, Lee A, Danné N, Mandal AK, Gosset P, Dehay B, Groc L, Cognet L, Bezard E. Synucleinopathy alters nanoscale organization and diffusion in the brain extracellular space through hyaluronan remodeling. Nat Commun 2020; 11:3440. [PMID: 32651387 PMCID: PMC7351768 DOI: 10.1038/s41467-020-17328-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/18/2020] [Indexed: 01/18/2023] Open
Abstract
In recent years, exploration of the brain extracellular space (ECS) has made remarkable progress, including nanoscopic characterizations. However, whether ECS precise conformation is altered during brain pathology remains unknown. Here we study the nanoscale organization of pathological ECS in adult mice under degenerative conditions. Using electron microscopy in cryofixed tissue and single nanotube tracking in live brain slices combined with super-resolution imaging analysis, we find enlarged ECS dimensions and increased nanoscale diffusion after α-synuclein-induced neurodegeneration. These animals display a degraded hyaluronan matrix in areas close to reactive microglia. Furthermore, experimental hyaluronan depletion in vivo reduces dopaminergic cell loss and α-synuclein load, induces microgliosis and increases ECS diffusivity, highlighting hyaluronan as diffusional barrier and local tissue organizer. These findings demonstrate the interplay of ECS, extracellular matrix and glia in pathology, unraveling ECS features relevant for the α-synuclein propagation hypothesis and suggesting matrix manipulation as a disease-modifying strategy. The nanoscale organisation of the brain extracellular space can be studied in vivo. Here, the authors investigate how it changes in response to α-synuclein pathology, and identify interactions between microglia and the extracellular matrix.
Collapse
Affiliation(s)
- Federico N Soria
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33076, Bordeaux, France.,Centre National de la Recherche Scientifique, IMN, UMR 5293, 33076, Bordeaux, France.,Achucarro Basque Center for Neuroscience, Universidad del País Vasco (UPV/EHU), 48940, Leioa, Spain
| | - Chiara Paviolo
- Université de Bordeaux, Laboratoire Photonique Numérique et Nanosciences, UMR 5298, 33400, Talence, France.,Institut d'Optique & Centre National de la Recherche Scientifique, LP2N, UMR 5298, 33400, Talence, France
| | - Evelyne Doudnikoff
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33076, Bordeaux, France.,Centre National de la Recherche Scientifique, IMN, UMR 5293, 33076, Bordeaux, France
| | - Marie-Laure Arotcarena
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33076, Bordeaux, France.,Centre National de la Recherche Scientifique, IMN, UMR 5293, 33076, Bordeaux, France
| | - Antony Lee
- Université de Bordeaux, Laboratoire Photonique Numérique et Nanosciences, UMR 5298, 33400, Talence, France.,Institut d'Optique & Centre National de la Recherche Scientifique, LP2N, UMR 5298, 33400, Talence, France
| | - Noémie Danné
- Université de Bordeaux, Laboratoire Photonique Numérique et Nanosciences, UMR 5298, 33400, Talence, France.,Institut d'Optique & Centre National de la Recherche Scientifique, LP2N, UMR 5298, 33400, Talence, France
| | - Amit Kumar Mandal
- Université de Bordeaux, Laboratoire Photonique Numérique et Nanosciences, UMR 5298, 33400, Talence, France.,Institut d'Optique & Centre National de la Recherche Scientifique, LP2N, UMR 5298, 33400, Talence, France
| | - Philippe Gosset
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33076, Bordeaux, France.,Centre National de la Recherche Scientifique, IMN, UMR 5293, 33076, Bordeaux, France
| | - Benjamin Dehay
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33076, Bordeaux, France.,Centre National de la Recherche Scientifique, IMN, UMR 5293, 33076, Bordeaux, France
| | - Laurent Groc
- Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, 33076, Bordeaux, France.,Centre National de la Recherche Scientifique, IINS, UMR 5297, 33076, Bordeaux, France
| | - Laurent Cognet
- Université de Bordeaux, Laboratoire Photonique Numérique et Nanosciences, UMR 5298, 33400, Talence, France. .,Institut d'Optique & Centre National de la Recherche Scientifique, LP2N, UMR 5298, 33400, Talence, France.
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33076, Bordeaux, France. .,Centre National de la Recherche Scientifique, IMN, UMR 5293, 33076, Bordeaux, France.
| |
Collapse
|