1
|
Hazlett MF, Hall VL, Patel E, Halvorsen A, Calakos N, West AE. The Perineuronal Net Protein Brevican Acts in Nucleus Accumbens Parvalbumin-Expressing Interneurons of Adult Mice to Regulate Excitatory Synaptic Inputs and Motivated Behaviors. Biol Psychiatry 2024; 96:694-707. [PMID: 38346480 PMCID: PMC11315813 DOI: 10.1016/j.biopsych.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/13/2024] [Accepted: 02/07/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Experience-dependent functional adaptation of nucleus accumbens (NAc) circuitry underlies the development and expression of reward-motivated behaviors. Parvalbumin-expressing GABAergic (gamma-aminobutyric acidergic) interneurons (PVINs) within the NAc are required for this process. Perineuronal nets (PNNs) are extracellular matrix structures enriched around PVINs that arise during development and have been proposed to mediate brain circuit stability. However, their function in the adult NAc is largely unknown. Here, we studied the developmental emergence and adult regulation of PNNs in the NAc of male and female mice and examined the cellular and behavioral consequences of reducing the PNN component brevican in NAc PVINs. METHODS We characterized the expression of PNN components in mouse NAc using immunofluorescence and RNA in situ hybridization. We lowered brevican in NAc PVINs of adult mice using an intersectional viral and genetic method and quantified the effects on synaptic inputs to NAc PVINs and reward-motivated learning. RESULTS PNNs around NAc PVINs were developmentally regulated and appeared during adolescence. In the adult NAc, PVIN PNNs were also dynamically regulated by cocaine. Transcription of the gene that encodes brevican was regulated in a cell type- and isoform-specific manner in the NAc, with the membrane-tethered form of brevican being highly enriched in PVINs. Lowering brevican in NAc PVINs of adult mice decreased their excitatory inputs and enhanced both short-term novel object recognition and cocaine-induced conditioned place preference. CONCLUSIONS Regulation of brevican in NAc PVINs of adult mice modulates their excitatory synaptic drive and sets experience thresholds for the development of motivated behaviors driven by rewarding stimuli.
Collapse
Affiliation(s)
- Mariah F Hazlett
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Victoria L Hall
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Esha Patel
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Aaron Halvorsen
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Nicole Calakos
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina; Department of Neurology, Duke University Medical Center, Durham, North Carolina; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina; Duke Institute for Brain Sciences, Duke University Medical Center, Durham, North Carolina
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
2
|
Parmar P, Spahic H, Lechner C, St Pierre M, Carlin K, Nugent M, Chavez-Valdez R. Neonatal hypoxia-ischemia alters the events governing the hippocampal critical period of postnatal synaptic plasticity leading to deficits in working memory in mice. Neurobiol Dis 2024:106722. [PMID: 39486775 DOI: 10.1016/j.nbd.2024.106722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/04/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
The postnatal critical period of synaptic plasticity (CPsp) is characterized by profound neural network refinement, which is shaped by synaptic activity and sculpted by maturation of the GABAergic network. Even after therapeutic hypothermia (TH), neonatal hypoxia-ischemia (HI) impairs two triggers for the initiation of the CPsp in the hippocampus: i) PSA-NCAM developmental decline and ii) parvalbumin (PV) + interneuron (IN) maturation. Thus, we investigated whether neonatal HI despite TH disturbs other events governing the onset, consolidation and closure of the postnatal CPsp in the hippocampus. We induced cerebral HI in P10 C57BL6 mice with right carotid ligation and 45 m of hypoxia (FiO2 = 0.08), followed by normothermia (36 °C, NT) or TH (31 °C) for 4 h with anesthesia-exposed shams as controls. ELISA, immunoblotting and immunohistochemistry were performed at 24 h (P11), 5 days (P15), 8 days (P18) and 30 days (P40) after HI injury. We specifically assessed: i) BDNF levels and TrkB activation, controlling the CPsp, ii) Otx2 and NPTX2 immunoreactivity (IR), engaging CPsp onset and iii) NogoR1, Lynx1 IR, PNN formation and myelination (MBP) mediating CPsp closure. Pups aged to P40 also received a battery of tests assessing working memory. Here, we documented deficits in hippocampal BDNF levels and TrkB activation at P18 in response to neonatal HI even with TH. Neonatal HI impaired in the CA1 the developmental increase in PV, Otx2, and NPTX2 between P11 and P18, the colocalization of Otx2 and PV at P18 and P40, the accumulation of NPTX2 in PV+ dendrites at P18 and P40, and the expression of NogoR and Lynx1 at P40. Furthermore, neonatal HI decreased BDNF and impaired PNN development and myelination (MBP) at P40. Most of these abnormalities were insensitive to TH and correlated with memory deficits. Neonatal HI appears to disrupt many of the molecular and structural events initiating and consolidating the postnatal hippocampal CPsp, perhaps due to the early and delayed deficits in TrkB activation leading to memory deficits.
Collapse
Affiliation(s)
- Pritika Parmar
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Harisa Spahic
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles Lechner
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Mark St Pierre
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | | | - Michael Nugent
- Department of Neuroscience, The Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Raul Chavez-Valdez
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA; Neuroscience Intensive Care Nursery Program, Johns Hopkins University- School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Zhang N, Song B, Bai P, Du L, Chen L, Xu Y, Zeng T. Perineuronal nets' role in metabolism. Am J Physiol Endocrinol Metab 2024; 327:E411-E421. [PMID: 39140971 DOI: 10.1152/ajpendo.00154.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Perineuronal nets (PNNs), specialized extracellular matrix (ECM) structures that envelop neurons, have recently been recognized as key players in the regulation of metabolism. This review explores the growing body of knowledge concerning PNNs and their role in metabolic control, drawing insights from recent research and relevant studies. The pivotal role of PNNs in the context of energy balance and whole body blood glucose is examined. This review also highlights novel findings, including the effects of astroglia, microglia, sex and gonadal hormones, nutritional regulation, circadian rhythms, and age on PNNs dynamics. These findings illuminate the complex and multifaceted role of PNNs in metabolic health.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Beite Song
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Peng Bai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Li Du
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Xu
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, People's Republic of China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
4
|
Colodete DA, Grace AA, Guimarães FS, Gomes FV. Degradation of Perineuronal Nets in the Ventral Hippocampus of Adult Rats Recreates an Adolescent-Like Phenotype of Stress Susceptibility. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100338. [PMID: 39099729 PMCID: PMC11295568 DOI: 10.1016/j.bpsgos.2024.100338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/08/2024] [Accepted: 05/12/2024] [Indexed: 08/06/2024] Open
Abstract
Background Psychiatric disorders often emerge during late adolescence/early adulthood, a period with increased susceptibility to socioenvironmental factors that coincides with incomplete parvalbumin interneuron (PVI) development. Stress during this period causes functional loss of PVIs in the ventral hippocampus (vHip), which has been associated with dopamine system overdrive. This vulnerability persists until the appearance of perineuronal nets (PNNs) around PVIs. We assessed the long-lasting effects of adolescent or adult stress on behavior, ventral tegmental area dopamine neuron activity, and the number of PVIs and their associated PNNs in the vHip. Additionally, we tested whether PNN removal in the vHip of adult rats, proposed to reset PVIs to a juvenile-like state, would recreate an adolescent-like phenotype of stress susceptibility. Methods Male rats underwent a 10-day stress protocol during adolescence or adulthood. Three to 4 weeks poststress, we evaluated behaviors related to anxiety, sociability, and cognition, ventral tegmental area dopamine neuron activity, and the number of PV+ and PNN+ cells in the vHip. Furthermore, adult animals received intra-vHip infusion of ChABC (chondroitinase ABC) to degrade PNNs before undergoing stress. Results Unlike adult stress, adolescent stress induced anxiety responses, reduced sociability, cognitive deficits, ventral tegmental area dopamine system overdrive, and decreased PV+ and PNN+ cells in the vHip. However, intra-vHip ChABC infusion caused the adult stress to produce changes similar to the ones observed after adolescent stress. Conclusions Our findings underscore adolescence as a period of heightened vulnerability to the long-lasting impact of stress and highlight the protective role of PNNs against stress-induced damage in PVIs.
Collapse
Affiliation(s)
- Débora A.E. Colodete
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry, and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Francisco S. Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Banovac I, Prkačin MV, Kirchbaum I, Trnski-Levak S, Bobić-Rasonja M, Sedmak G, Petanjek Z, Jovanov-Milosevic N. Morphological and Molecular Characteristics of Perineuronal Nets in the Human Prefrontal Cortex-A Possible Link to Microcircuitry Specialization. Mol Neurobiol 2024:10.1007/s12035-024-04306-1. [PMID: 38958887 DOI: 10.1007/s12035-024-04306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Perineuronal nets (PNNs) are a type of extracellular matrix (ECM) that play a significant role in synaptic activity and plasticity of interneurons in health and disease. We researched PNNs' regional and laminar representation and molecular composition using immunohistochemistry and transcriptome analysis of Brodmann areas (BA) 9, 14r, and 24 in 25 human postmortem brains aged 13-82 years. The numbers of VCAN- and NCAN-expressing PNNs, relative to the total number of neurons, were highest in cortical layers I and VI while WFA-binding (WFA+) PNNs were most abundant in layers III-V. The ECM glycosylation pattern was the most pronounced regional difference, shown by a significantly lower proportion of WFA+ PNNs in BA24 (3.27 ± 0.69%) compared to BA9 (6.32 ± 1.73%; P = 0.0449) and BA14 (5.64 ± 0.71%; P = 0.0278). The transcriptome of late developmental and mature stages revealed a relatively stable expression of PNN-related transcripts (log2-transformed expression values: 6.5-8.5 for VCAN and 8.0-9.5 for NCAN). Finally, we propose a classification of PNNs that envelop GABAergic neurons in the human cortex. The significant differences in PNNs' morphology, distribution, and molecular composition strongly suggest an involvement of PNNs in specifying distinct microcircuits in particular cortical regions and layers.
Collapse
Affiliation(s)
- Ivan Banovac
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Matija Vid Prkačin
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Ivona Kirchbaum
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Sara Trnski-Levak
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Mihaela Bobić-Rasonja
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Department of Biology, University of Zagreb School of Medicine, Šalata 3, HR-10000, Zagreb, Croatia
| | - Goran Sedmak
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
| | - Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia
| | - Natasa Jovanov-Milosevic
- Department of Anatomy and Clinical Anatomy, University of Zagreb School of Medicine, Šalata 11, HR-10000, Zagreb, Croatia.
- Croatian Institute for Brain Research, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine University of Zagreb, Šalata 12, HR-10000, Zagreb, Croatia.
- Department of Biology, University of Zagreb School of Medicine, Šalata 3, HR-10000, Zagreb, Croatia.
| |
Collapse
|
6
|
Guillotin S, Fulzele A, Vallet A, de Peredo AG, Mouton‐Barbosa E, Cestac P, Andrieu S, Burlet‐Schiltz O, Delcourt N, Schmidt E. Cerebrospinal fluid proteomic profile of frailty: Results from the PROLIPHYC cohort. Aging Cell 2024; 23:e14168. [PMID: 38698559 PMCID: PMC11258431 DOI: 10.1111/acel.14168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/21/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Frailty is a clinical state reflecting a decrease in physiological reserve capacities, known to affect numerous biological pathways and is associated with health issues, including neurodegenerative diseases. However, how global protein expression is affected in the central nervous system in frail subject remains underexplored. In this post hoc cross-sectional biomarker analysis, we included 90 adults (52-85 years) suspected of normal pressure hydrocephalus (NPH) and presenting with markers of neurodegenerative diseases. We investigated the human proteomic profile of cerebrospinal fluid associated with frailty defined by an established cumulated frailty index (FI, average = 0.32), not enriched for neurology clinical features. Using a label-free quantitative proteomic approach, we identified and quantified 999 proteins of which 13 were positively associated with frailty. Pathway analysis with the top positively frailty-associated proteins revealed enrichment for proteins related to inflammation and immune response. Among the 60 proteins negatively associated with frailty, functional pathways enriched included neurogenesis, synaptogenesis and neuronal guidance. We constructed a frailty prediction model using ridge regression with 932 standardized proteins. Our results showed that the "proteomic model" could become an equivalent predictor of FI in order to study chronological age. This study represents the first comprehensive exploration of the proteomic profile of frailty within cerebrospinal fluid. It sheds light on the physiopathology of frailty, particularly highlighting processes of neuroinflammation and inhibition of neurogenesis. Our findings unveil a range of biological mechanisms that are dysregulated in frailty, in NPH subjects at risk of neurodegenerative impairment, offering new perspectives on frailty phenotyping and prediction.
Collapse
Affiliation(s)
- Sophie Guillotin
- Aging‐MAINTAIN Research Team, Center for Epidemiology and Research in POPulation Health (CERPOP)University of ToulouseToulouseFrance
- Poison Control CenterToulouse University HospitalToulouseFrance
| | - Amit Fulzele
- Institute of Pharmacology and Structural Biology (IPBS)University of Toulouse, CNRS, University of Toulouse III (Paul Sabatier (UT3)ToulouseFrance
- Present address:
Institute of Molecular BiologyUniversity of MainzMainzGermany
| | - Alexandra Vallet
- Biological Tissue and Surface Engineering DepartmentINSERM U1059 Sainbiose, Ecole Des Mines of Saint‐EtienneSaint‐EtienneFrance
| | - Anne Gonzalez de Peredo
- Institute of Pharmacology and Structural Biology (IPBS)University of Toulouse, CNRS, University of Toulouse III (Paul Sabatier (UT3)ToulouseFrance
| | - Emmanuelle Mouton‐Barbosa
- Institute of Pharmacology and Structural Biology (IPBS)University of Toulouse, CNRS, University of Toulouse III (Paul Sabatier (UT3)ToulouseFrance
| | - Philippe Cestac
- Aging‐MAINTAIN Research Team, Center for Epidemiology and Research in POPulation Health (CERPOP)University of ToulouseToulouseFrance
- Department of Clinical PharmacyToulouse University HospitalToulouseFrance
| | - Sandrine Andrieu
- Aging‐MAINTAIN Research Team, Center for Epidemiology and Research in POPulation Health (CERPOP)University of ToulouseToulouseFrance
- Department of Epidemiology and Public HealthToulouse University HospitalToulouseFrance
- IHU HealthAgeToulouseFrance
| | - Odile Burlet‐Schiltz
- Institute of Pharmacology and Structural Biology (IPBS)University of Toulouse, CNRS, University of Toulouse III (Paul Sabatier (UT3)ToulouseFrance
| | - Nicolas Delcourt
- Poison Control CenterToulouse University HospitalToulouseFrance
- Toulouse NeuroImaging Center (ToNIC)University of Toulouse, INSERM UPSToulouseFrance
| | - Eric Schmidt
- Toulouse NeuroImaging Center (ToNIC)University of Toulouse, INSERM UPSToulouseFrance
- Department of NeurosurgeryToulouse University HospitalToulouseFrance
| |
Collapse
|
7
|
Melrose J. CNS/PNS proteoglycans functionalize neuronal and astrocyte niche microenvironments optimizing cellular activity by preserving membrane polarization dynamics, ionic microenvironments, ion fluxes, neuronal activation, and network neurotransductive capacity. J Neurosci Res 2024; 102:e25361. [PMID: 39034899 DOI: 10.1002/jnr.25361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 07/23/2024]
Abstract
Central and peripheral nervous system (CNS/PNS) proteoglycans (PGs) have diverse functional roles, this study examined how these control cellular behavior and tissue function. The CNS/PNS extracellular matrix (ECM) is a dynamic, responsive, highly interactive, space-filling, cell supportive, stabilizing structure maintaining tissue compartments, ionic microenvironments, and microgradients that regulate neuronal activity and maintain the neuron in an optimal ionic microenvironment. The CNS/PNS contains a high glycosaminoglycan content (60% hyaluronan, HA) and a diverse range of stabilizing PGs. Immobilization of HA in brain tissues by HA interactive hyalectan PGs preserves tissue hydration and neuronal activity, a paucity of HA in brain tissues results in a pro-convulsant epileptic phenotype. Diverse CS, KS, and HSPGs stabilize the blood-brain barrier and neurovascular unit, provide smart gel neurotransmitter neuron vesicle storage and delivery, organize the neuromuscular junction basement membrane, and provide motor neuron synaptic plasticity, and photoreceptor and neuron synaptic functions. PG-HA networks maintain ionic fluxes and microgradients and tissue compartments that contribute to membrane polarization dynamics essential to neuronal activation and neurotransduction. Hyalectans form neuroprotective perineuronal nets contributing to synaptic plasticity, memory, and cognitive learning. Sialoglycoprotein associated with cones and rods (SPACRCAN), an HA binding CSPG, stabilizes the inter-photoreceptor ECM. HSPGs pikachurin and eyes shut stabilize the photoreceptor synapse aiding in phototransduction and neurotransduction with retinal bipolar neurons crucial to visual acuity. This is achieved through Laminin G motifs in pikachurin, eyes shut, and neurexins that interact with the dystroglycan-cytoskeleton-ECM-stabilizing synaptic interconnections, neuronal interactive specificity, and co-ordination of regulatory action potentials in neural networks.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
- Sydney Medical School, Northern, The University of Sydney Faculty of Medicine and Health, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| |
Collapse
|
8
|
Reinhard J, Mueller-Buehl C, Wiemann S, Roll L, Luft V, Shabani H, Rathbun DL, Gan L, Kuo CC, Franzen J, Joachim SC, Faissner A. Neural extracellular matrix regulates visual sensory motor integration. iScience 2024; 27:108846. [PMID: 38318351 PMCID: PMC10839651 DOI: 10.1016/j.isci.2024.108846] [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: 08/08/2023] [Revised: 10/12/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Visual processing depends on sensitive and balanced synaptic neurotransmission. Extracellular matrix proteins in the environment of cells are key modulators in synaptogenesis and synaptic plasticity. In the present study, we provide evidence that the combined loss of the four extracellular matrix components, brevican, neurocan, tenascin-C, and tenascin-R, in quadruple knockout mice leads to severe retinal dysfunction and diminished visual motion processing in vivo. Remarkably, impaired visual motion processing was accompanied by a developmental loss of cholinergic direction-selective starburst amacrine cells. Additionally, we noted imbalance of inhibitory and excitatory synaptic signaling in the quadruple knockout retina. Collectively, the study offers insights into the functional importance of four key extracellular matrix proteins for retinal function, visual motion processing, and synaptic signaling.
Collapse
Affiliation(s)
- Jacqueline Reinhard
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Cornelius Mueller-Buehl
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Susanne Wiemann
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Veronika Luft
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Hamed Shabani
- Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard-Karls-University Tuebingen, 72076 Tuebingen, Germany
| | - Daniel L. Rathbun
- Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard-Karls-University Tuebingen, 72076 Tuebingen, Germany
| | - Lin Gan
- Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Chao-Chung Kuo
- Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Julia Franzen
- Interdisciplinary Centre for Clinical Research Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr University Bochum, 44892 Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, 44780 Bochum, Germany
| |
Collapse
|
9
|
Hu R, He K, Chen B, Chen Y, Zhang J, Wu X, Shi M, Wu L, Ma R. Electroacupuncture promotes the repair of the damaged spinal cord in mice by mediating neurocan-perineuronal net. CNS Neurosci Ther 2024; 30:e14468. [PMID: 37950551 PMCID: PMC10805400 DOI: 10.1111/cns.14468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/06/2023] [Accepted: 08/29/2023] [Indexed: 11/12/2023] Open
Abstract
AIMS This study aimed to investigate the effect of perineuronal net (PNN) and neurocan (NCAN) on spinal inhibitory parvalbumin interneuron (PV-IN), and the mechanism of electroacupuncture (EA) in promoting spinal cord injury (SCI) repair through neurocan in PNN. METHODS A mouse model of SCI was established. Sham-operated mice or SCI model mice were treated with chondroitin sulfate ABC (ChABC) enzyme or control vehicle for 2 weeks (i.e., sham+veh group, sham+ChABC group, SCI+veh group, and SCI+ChABC group, respectively), and then spinal cord tissues were taken from the T10 lesion epicenter for RNA sequencing (RNA-seq). MSigDB Hallmark and C5 databases for functional analysis, analysis strategies such as differential expression gene analysis (DEG), Kyoto Encyclopedia of Genes and Genomes (KEGG), gene set enrichment analysis (GSEA), and protein-protein interaction (PPI). According to the results of RNA-seq analysis, the expression of NCAN was knocked down or overexpressed by virus intervention, or/and EA intervention. Polymerase chain reaction (PCR), immunofluorescence, western blot, electrophysiological, and behavioral tests were performed. RESULTS After the successful establishment of SCI model, the motor dysfunction of lower limbs, and the expression of PNN core glycan protein at the epicenter of SCI were reduced. RNA-seq and PCR showed that PNN core proteoglycans except NCAN showed the same expression trend in normal and injured spinal cord treated with ChABC. KEGG and GSEA showed that PNN is mainly associated with inhibitory GABA neuronal function in injured spinal cord tissue, and PPI showed that NCAN in PNN can be associated with inhibitory neuronal function through parvalbumin (PV). Calcium imaging showed that local parvalbumin interneuron (PV-IN) activity decreased after PNN destruction, whether due to ChABC treatment or surgical bruising of the spinal cord. Overexpression of neurocan in injured spinal cord can enhance local PV-IN activity. PCR and western blot suggested that overexpression or knockdown of neurocan could up-regulate or down-regulate the expression of GAD. At the same time, the activity of PV-IN in the primary motor cortex (M1) and the primary sensory cortex of lower (S1HL) extremity changed synchronously. In addition, overexpression of neurocan improved the electrical activity of the lower limb and promoted functional repair of the paralyzed hind limb. EA intervention reversed the down-regulation of neurocan, enhanced the expression of PNN in the lesioned area, M1 and S1HL. CONCLUSION Neurocan in PNN can regulate the activity of PV-IN, and EA can promote functional recovery of mice with SCI by upregulating neurocan expression in PNN.
Collapse
Affiliation(s)
- Rong Hu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Kelin He
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
- Department of Acupuncture and MoxibustionThird Affiliated Hospital of Zhejiang Chinese Medical UniversityZhejiangChina
| | - Bowen Chen
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Yi Chen
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Jieqi Zhang
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Xingying Wu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Mengting Shi
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
| | - Lei Wu
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
- Department of Acupuncture and MoxibustionThird Affiliated Hospital of Zhejiang Chinese Medical UniversityZhejiangChina
| | - Ruijie Ma
- The Third School of Clinical Medicine (School of Rehabilitation Medicine), Key Laboratory of Acupuncture and Neurology of Zhejiang ProvinceZhejiang Chinese Medical UniversityZhejiangChina
- Department of Acupuncture and MoxibustionThird Affiliated Hospital of Zhejiang Chinese Medical UniversityZhejiangChina
| |
Collapse
|
10
|
Woo AM, Sontheimer H. Interactions between astrocytes and extracellular matrix structures contribute to neuroinflammation-associated epilepsy pathology. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1198021. [PMID: 39086689 PMCID: PMC11285605 DOI: 10.3389/fmmed.2023.1198021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/31/2023] [Indexed: 08/02/2024]
Abstract
Often considered the "housekeeping" cells of the brain, astrocytes have of late been rising to the forefront of neurodegenerative disorder research. Identified as crucial components of a healthy brain, it is undeniable that when astrocytes are dysfunctional, the entire brain is thrown into disarray. We offer epilepsy as a well-studied neurological disorder in which there is clear evidence of astrocyte contribution to diseases as evidenced across several different disease models, including mouse models of hippocampal sclerosis, trauma associated epilepsy, glioma-associated epilepsy, and beta-1 integrin knockout astrogliosis. In this review we suggest that astrocyte-driven neuroinflammation, which plays a large role in the pathology of epilepsy, is at least partially modulated by interactions with perineuronal nets (PNNs), highly structured formations of the extracellular matrix (ECM). These matrix structures affect synaptic placement, but also intrinsic neuronal properties such as membrane capacitance, as well as ion buffering in their immediate milieu all of which alters neuronal excitability. We propose that the interactions between PNNs and astrocytes contribute to the disease progression of epilepsy vis a vis neuroinflammation. Further investigation and alteration of these interactions to reduce the resultant neuroinflammation may serve as a potential therapeutic target that provides an alternative to the standard anti-seizure medications from which patients are so frequently unable to benefit.
Collapse
Affiliation(s)
- AnnaLin M. Woo
- Neuroscience Graduate Program, Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| | - Harald Sontheimer
- Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
11
|
Irala D, Wang S, Sakers K, Nagendren L, Ulloa-Severino FP, Bindu DS, Eroglu C. Astrocyte-Secreted Neurocan Controls Inhibitory Synapse Formation and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535448. [PMID: 37066164 PMCID: PMC10104008 DOI: 10.1101/2023.04.03.535448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Astrocytes strongly promote the formation and maturation of synapses by secreted proteins. To date, several astrocyte-secreted synaptogenic proteins controlling different stages of excitatory synapse development have been identified. However, the identities of astrocytic signals that induce inhibitory synapse formation remain elusive. Here, through a combination of in vitro and in vivo experiments, we identified Neurocan as an astrocyte-secreted inhibitory synaptogenic protein. Neurocan is a chondroitin sulfate proteoglycan that is best known as a protein localized to the perineuronal nets. However, Neurocan is cleaved into two after secretion from astrocytes. We found that the resulting N- and C-terminal fragments have distinct localizations in the extracellular matrix. While the N-terminal fragment remains associated with perineuronal nets, the Neurocan C-terminal fragment localizes to synapses and specifically controls cortical inhibitory synapse formation and function. Neurocan knockout mice lacking the whole protein or only its C-terminal synaptogenic region have reduced inhibitory synapse numbers and function. Through super-resolution microscopy and in vivo proximity labeling by secreted TurboID, we discovered that the synaptogenic domain of Neurocan localizes to somatostatin-positive inhibitory synapses and strongly regulates their formation. Together, our results unveil a mechanism through which astrocytes control circuit-specific inhibitory synapse development in the mammalian brain.
Collapse
|
12
|
Reduced inhibitory and excitatory input onto parvalbumin interneurons mediated by perineuronal net might contribute to cognitive impairments in a mouse model of sepsis-associated encephalopathy. Neuropharmacology 2023; 225:109382. [PMID: 36543316 DOI: 10.1016/j.neuropharm.2022.109382] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is commonly defined as diffuse brain dysfunction and can manifest as delirium to coma. Accumulating evidence has suggested that perineuronal net (PNN) plays an important role in the modulation of the synaptic plasticity of central nervous system. We here investigated the role of PNN in SAE induced by lipopolysaccharide (LPS) injection. Behavioral tests were performed by open field, Y-maze, and fear conditioning tests at the indicated time points. The densities of vesicular γ-aminobutyric acid transporter, vesicular glutamate transporter 1, PNN, and parvalbumin (PV) in the hippocampus were evaluated by immunofluorescence. Matrix metalloproteinases-9 (MMP-9) expression and its activity were detected by Western blot and gel zymography, respectively. Local field potential was recorded by in vivo electrophysiology. LPS-treated mice displayed significant cognitive impairments, coincided with activated MMP-9, decreased PNN and PV densities, reduced inhibitory and excitatory input onto PV interneurons enwrapped by PNN, and decreased gamma oscillations in hippocampal CA1. Notably, MMP-9 inhibitor SB-3CT treatment rescued most of these abnormalities. Taken together, our study demonstrates that active MMP-9 mediated PNN remodeling, leading to reduced inhibitory and excitatory input onto PV interneurons and abnormal gamma oscillations in hippocampal CA1, which consequently contributed to cognitive impairments after LPS injection.
Collapse
|
13
|
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: 3.0] [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
|
14
|
Lima R, Monteiro A, Salgado AJ, Monteiro S, Silva NA. Pathophysiology and Therapeutic Approaches for Spinal Cord Injury. Int J Mol Sci 2022; 23:ijms232213833. [PMID: 36430308 PMCID: PMC9698625 DOI: 10.3390/ijms232213833] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Spinal cord injury (SCI) is a disabling condition that disrupts motor, sensory, and autonomic functions. Despite extensive research in the last decades, SCI continues to be a global health priority affecting thousands of individuals every year. The lack of effective therapeutic strategies for patients with SCI reflects its complex pathophysiology that leads to the point of no return in its function repair and regeneration capacity. Recently, however, several studies started to uncover the intricate network of mechanisms involved in SCI leading to the development of new therapeutic approaches. In this work, we present a detailed description of the physiology and anatomy of the spinal cord and the pathophysiology of SCI. Additionally, we provide an overview of different molecular strategies that demonstrate promising potential in the modulation of the secondary injury events that promote neuroprotection or neuroregeneration. We also briefly discuss other emerging therapies, including cell-based therapies, biomaterials, and epidural electric stimulation. A successful therapy might target different pathologic events to control the progression of secondary damage of SCI and promote regeneration leading to functional recovery.
Collapse
Affiliation(s)
- Rui Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Andreia Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Nuno A. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
- Correspondence:
| |
Collapse
|