1
|
Mostajo-Radji MA, Leon WRM, Breevoort A, Gonzalez-Ferrer J, Schweiger HE, Lehrer J, Zhou L, Schmitz MT, Perez Y, Mukhtar T, Robbins A, Chu J, Andrews MG, Sullivan FN, Tejera D, Choy EC, Paredes MF, Teodorescu M, Kriegstein AR, Alvarez-Buylla A, Pollen AA. Fate plasticity of interneuron specification. iScience 2025; 28:112295. [PMID: 40264797 PMCID: PMC12013500 DOI: 10.1016/j.isci.2025.112295] [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: 10/15/2024] [Revised: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Neuronal subtype generation in the mammalian central nervous system is governed by competing genetic programs. The medial ganglionic eminence (MGE) produces two major cortical interneuron (IN) populations, somatostatin (Sst) and parvalbumin (Pvalb), which develop on different timelines. The extent to which external signals influence these identities remains unclear. Pvalb-positive INs are crucial for cortical circuit regulation but challenging to model in vitro. We grafted mouse MGE progenitors into diverse 2D and 3D co-culture systems, including mouse and human cortical, MGE, and thalamic models. Strikingly, only 3D human corticogenesis models promoted efficient, non-autonomous Pvalb differentiation, characterized by upregulation of Pvalb maturation markers, downregulation of Sst-specific markers, and the formation of perineuronal nets. Additionally, lineage-traced postmitotic Sst-positive INs upregulated Pvalb when grafted onto human cortical models. These findings reveal unexpected fate plasticity in MGE-derived INs, suggesting that their identities can be dynamically shaped by the environment.
Collapse
Affiliation(s)
- Mohammed A. Mostajo-Radji
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Walter R. Mancia Leon
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arnar Breevoort
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jesus Gonzalez-Ferrer
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hunter E. Schweiger
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Julian Lehrer
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Li Zhou
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew T. Schmitz
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yonatan Perez
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tanzila Mukhtar
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ash Robbins
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Julia Chu
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Madeline G. Andrews
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Dario Tejera
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric C. Choy
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mercedes F. Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mircea Teodorescu
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Arnold R. Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alex A. Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
2
|
Padmashri R, Dunaevsky A. Targeting GABA Polarity During Cortical Development Improves Circuit and Sensory Deficits in Fragile X Mice. Biol Psychiatry 2025; 97:420-421. [PMID: 39919885 PMCID: PMC12004231 DOI: 10.1016/j.biopsych.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025]
Affiliation(s)
- Ragunathan Padmashri
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Anna Dunaevsky
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
3
|
Bromley-Coolidge S, Iruegas D, Appel B. Cspg4 sculpts oligodendrocyte precursor cell morphology. Differentiation 2024; 140:100819. [PMID: 39566199 PMCID: PMC11637897 DOI: 10.1016/j.diff.2024.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024]
Abstract
The extracellular matrix (ECM) provides critical biochemical and structural cues that regulate neural development. Chondroitin sulfate proteoglycans (CSPGs), a major ECM component, have been implicated in modulating oligodendrocyte precursor cell (OPC) proliferation, migration, and maturation, but their specific roles in oligodendrocyte lineage cell (OLC) development and myelination in vivo remain poorly understood. Here, we use zebrafish as a model system to investigate the spatiotemporal dynamics of ECM deposition and CSPG localization during central nervous system (CNS) development, with a focus on their relationship to OLCs. We demonstrate that ECM components, including CSPGs, are dynamically expressed in distinct spatiotemporal patterns coinciding with OLC development and myelination. We found that zebrafish lacking cspg4 function produced normal numbers of OLCs, which appeared to undergo proper differentiation. However, OPC morphology in mutant larvae was aberrant. Nevertheless, the number and length of myelin sheaths produced by mature oligodendrocytes were unaffected. These data indicate that Cspg4 regulates OPC morphogenesis in vivo, supporting the role of the ECM in neural development.
Collapse
Affiliation(s)
- Samantha Bromley-Coolidge
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80445, USA
| | - Diego Iruegas
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80445, USA
| | - Bruce Appel
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80445, USA.
| |
Collapse
|
4
|
Liu L, Li T, Chang J, Xia X, Ju J. Microglia inversely regulate the level of perineuronal nets with the treatment of lipopolysaccharide and valproic acid. Neurosci Lett 2024; 842:137992. [PMID: 39304021 DOI: 10.1016/j.neulet.2024.137992] [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: 06/22/2024] [Revised: 08/27/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Perineuronal nets (PNNs) are extracellular matrix which mostly surround the inhibitory neurons. They are changed in several brain diseases, such as autism spectrum disorder, but the mechanism of PNNs degradation is still unclear. In this study, we investigated the role of microglial cells in regulating PNNs levels. Specifically, 1 day or 3 days after a single dose of lipopolysaccharide (LPS, 0.25 mg/kg) increased the density of microglia and further reduced the density of PNNs in both hippocampus CA1 and visual cortex. Minocycline, an inhibitor of microglia activation, took effect time-dependently. Minocycline for 7 days before a single LPS injection (0.25 mg/kg) inhibited microglia increase and PNNs loss, but minocycline for 3 days did not work. Finally, in a valproic acid (VPA)-treated autism mouse model, microglia were reduced while PNNs+ cells were increased in both hippocampus CA1 and visual cortex. In summary, the microglia are involved in the balanced level of PNNs, while in the autism model, the altered level of PNNs might be due to the microglia hypofunction.
Collapse
Affiliation(s)
- Luping Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China; School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Tianxiang Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jinlong Chang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dali University, Dali 671003, China
| | - Xiaojiao Xia
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Jun Ju
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China; Brain Research Centre and Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
5
|
Lev-Ram V, Lemieux SP, Deerinck TJ, Bushong EA, Perez AJ, Pritchard DR, Toyama BH, Park SKR, McClatchy DB, Savas JN, Whitney M, Adams SR, Ellisman MH, Yates J, Tsien RY. Do Perineuronal Nets Stabilize the Engram of a Synaptic Circuit? Cells 2024; 13:1627. [PMID: 39404392 PMCID: PMC11476018 DOI: 10.3390/cells13191627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024] Open
Abstract
Perineuronal nets (PNNs), a specialized form of extra cellular matrix (ECM), surround numerous neurons in the CNS and allow synaptic connectivity through holes in its structure. We hypothesize that PNNs serve as gatekeepers that guard and protect synaptic territory and thus may stabilize an engram circuit. We present high-resolution and 3D EM images of PNN-engulfed neurons in mice brains, showing that synapses occupy the PNN holes and that invasion of other cellular components is rare. PNN constituents in mice brains are long-lived and can be eroded faster in an enriched environment, while synaptic proteins have a high turnover rate. Preventing PNN erosion by using pharmacological inhibition of PNN-modifying proteases or matrix metalloproteases 9 (MMP9) knockout mice allowed normal fear memory acquisition but diminished long-term memory stabilization, supporting the above hypothesis.
Collapse
Affiliation(s)
- Varda Lev-Ram
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Sakina Palida Lemieux
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Thomas J. Deerinck
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
| | - Eric A. Bushong
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
| | - Alex J. Perez
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
| | - Denise R. Pritchard
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Brandon H. Toyama
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sung Kyu R. Park
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
| | - Daniel B. McClatchy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
| | - Jeffrey N. Savas
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60208, USA
| | - Michael Whitney
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Stephen R. Adams
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, University of California San Diego, La Jolla, CA 92093, USA; (T.J.D.); (A.J.P.); (M.H.E.)
- Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - John Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA; (S.K.R.P.); (D.B.M.); (J.N.S.); (J.Y.III)
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Roger Y. Tsien
- Department of Pharmacology, University of California San Diego, CA 92093, USA; (S.P.L.); (D.R.P.); (M.W.); (S.R.A.)
- Department of Chemistry & Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Bromley-Coolidge S, Iruegas D, Appel B. Cspg4 sculpts oligodendrocyte precursor cell morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607226. [PMID: 39149260 PMCID: PMC11326215 DOI: 10.1101/2024.08.08.607226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The extracellular matrix (ECM) provides critical biochemical and structural cues that regulate neural development. Chondroitin sulfate proteoglycans (CSPGs), a major ECM component, have been implicated in modulating oligodendrocyte precursor cell (OPC) proliferation, migration, and maturation, but their specific roles in oligodendrocyte lineage cell (OLC) development and myelination in vivo remain poorly understood. Here, we use zebrafish as a model system to investigate the spatiotemporal dynamics of ECM deposition and CSPG localization during central nervous system (CNS) development, with a focus on their relationship to OLCs. We demonstrate that ECM components, including CSPGs, are dynamically expressed in distinct spatiotemporal patterns coinciding with OLC development and myelination. We found that zebrafish lacking cspg4 function produced normal numbers of OLCs, which appeared to undergo proper differentiation. However, OPC morphology in mutant larvae was aberrant. Nevertheless, the number and length of myelin sheaths produced by mature oligodendrocytes were unaffected. These data indicate that Cspg4 regulates OPC morphogenesis in vivo, supporting the role of the ECM in neural development.
Collapse
Affiliation(s)
- Samantha Bromley-Coolidge
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA, 80445
| | - Diego Iruegas
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA, 80445
| | - Bruce Appel
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA, 80445
| |
Collapse
|
7
|
Kuczynski-Noyau L, Karmann S, Alberton P, Martinez-Corral I, Nampoothiri S, Sauvé F, Lhomme T, Quarta C, Apte SS, Bouret S, Aszodi A, Rasika S, Ciofi P, Dam J, Prévot V, Mattot V. A plastic aggrecan barrier modulated by peripheral energy state gates metabolic signal access to arcuate neurons. Nat Commun 2024; 15:6701. [PMID: 39112471 PMCID: PMC11306556 DOI: 10.1038/s41467-024-50798-9] [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: 03/12/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
The hypothalamic arcuate nucleus (ARH) contains neurons vital for maintaining energy homeostasis that sense and respond to changes in blood-borne metabolic hormones. Despite its juxtaposition to the median eminence (ME), a circumventricular organ lacking a blood-brain barrier and thus exposed to circulating molecules, only a few ventral ARH neurons perceive these extravasating metabolic signals due to a poorly understood ME/ARH diffusion barrier. Here, we show in male mice that aggrecan, a perineural-net proteoglycan deposited by orexigenic ARH neurons, creates a peculiar ventrodorsal diffusion gradient. Fasting enhances aggrecan deposition more dorsally, reinforcing the diffusion barrier, particularly around neurons adjacent to fenestrated capillary loops that enter the ARH. The disruption of aggrecan deposits results in unregulated diffusion of blood-borne molecules into the ARH and impairs food intake. Our findings reveal the molecular nature and plasticity of the ME/ARH diffusion barrier, and indicate its physiological role in hypothalamic metabolic hormone sensing.
Collapse
Affiliation(s)
- Laura Kuczynski-Noyau
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Sixtine Karmann
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Paolo Alberton
- Department for Orthopaedics and Trauma Surgery Musculoskeletal University Center Munich (MUM) University Hospital, LMU, Munich, Germany
- Division of Hand, Plastic and Aesthetic Surgery LMU University Hospital, LMU, Munich, Germany
| | - Ines Martinez-Corral
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Sreekala Nampoothiri
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Florent Sauvé
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Tori Lhomme
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Carmelo Quarta
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland, OH, USA
| | - Sébastien Bouret
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Attila Aszodi
- Department for Orthopaedics and Trauma Surgery Musculoskeletal University Center Munich (MUM) University Hospital, LMU, Munich, Germany
| | - Sowmyalakshmi Rasika
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Philippe Ciofi
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Julie Dam
- Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Vincent Prévot
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France
| | - Virginie Mattot
- Univ. Lille, INSERM, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, Lille, France.
| |
Collapse
|
8
|
Barbugian F, Cadamuro F, Nicotra F, Riccardi C, Russo L. Plasma-treated collagen functionalized with chondroitin sulfate as bioactive and nanostructured extracellular matrix mimics. Nanomedicine (Lond) 2024; 19:799-810. [PMID: 38385248 DOI: 10.2217/nnm-2023-0310] [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] [Indexed: 02/23/2024] Open
Abstract
Aim: Cell microenvironment contains a plethora of information that influences cell modulation. Indeed, the extracellular matrix plays a central role in tissue development. Reproducing the cell-extracellular matrix crosstalk able to recapitulate both physical and biochemical signals is crucial to obtain functional tissue models or regenerative strategies. Materials & methods: Here, a combined method is proposed to easily functionalize collagen surface films, tailoring morphological properties. Oxygen nonthermal plasma treatment and glyco-conjugation with chondroitin sulfate are used to modify surface properties. Results: It results in higher adhesion, proliferation and morphological organization of U87 glioblastoma cells. Conclusion: Our finding suggests new promising strategies for the development of collagen-based biomaterials, which can be employed for advanced in vitro models.
Collapse
Affiliation(s)
- Federica Barbugian
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Francesca Cadamuro
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Francesco Nicotra
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| | - Claudia Riccardi
- Department of Physics, University of Milano-Bicocca, Milan, 20126, Italy
| | - Laura Russo
- School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, 20854, Italy
| |
Collapse
|
9
|
Carceller H, Gramuntell Y, Klimczak P, Nacher J. Perineuronal Nets: Subtle Structures with Large Implications. Neuroscientist 2023; 29:569-590. [PMID: 35872660 DOI: 10.1177/10738584221106346] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Perineuronal nets (PNNs) are specialized structures of the extracellular matrix that surround the soma and proximal dendrites of certain neurons in the central nervous system, particularly parvalbumin-expressing interneurons. Their appearance overlaps the maturation of neuronal circuits and the closure of critical periods in different regions of the brain, setting their connectivity and abruptly reducing their plasticity. As a consequence, the digestion of PNNs, as well as the removal or manipulation of their components, leads to a boost in this plasticity and can play a key role in the functional recovery from different insults and in the etiopathology of certain neurologic and psychiatric disorders. Here we review the structure, composition, and distribution of PNNs and their variation throughout the evolutive scale. We also discuss methodological approaches to study these structures. The function of PNNs during neurodevelopment and adulthood is discussed, as well as the influence of intrinsic and extrinsic factors on these specialized regions of the extracellular matrix. Finally, we review current data on alterations in PNNs described in diseases of the central nervous system (CNS), focusing on psychiatric disorders. Together, all the data available point to the PNNs as a promising target to understand the physiology and pathologic conditions of the CNS.
Collapse
Affiliation(s)
- Héctor Carceller
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
10
|
Engel K, Lee HN, Tewari BP, Lewkowicz AP, Ireland DDC, Manangeeswaran M, Verthelyi D. Neonatal Zika virus infection causes transient perineuronal net degradation. Front Cell Neurosci 2023; 17:1187425. [PMID: 37496706 PMCID: PMC10366369 DOI: 10.3389/fncel.2023.1187425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023] Open
Abstract
Perineuronal nets (PNNs) form a specialized extracellular matrix that predominantly surrounds parvalbumin (PV)-expressing GABAergic inhibitory interneurons and help regulate neuronal activity. Their formation early in the postnatal period is regulated by neuronal signaling and glial activation raising concerns that part of the long-term effects ascribed to perinatal viral infections could be mediated by altered PNN formation. Previously, we developed a model of neonatal Zika virus (ZIKV) infection where mice have lifelong neurological sequelae that includes motor disfunction and reduced anxiety coupled with a persistent low-grade expression in proinflammatory markers despite resolving the acute infection. Here, we demonstrate that ZIKV infection to P1 neonatal mice results in a reduction of PNN formation during the acute disease with significant reduction in Wisteria floribunda agglutinin (WFA) staining at the peak of infection [15 days post infection (dpi)] that persisted after the symptoms resolved (30 dpi). At 60 dpi, when there is residual inflammation in the CNS, the number of WFA+ cells and the level of WFA staining as well as levels of aggrecan and brevican in the brains of convalescent mice were not different from those in uninfected controls, however, there was increased frequency of PNNs with an immature phenotype. Over time the impact of the perinatal infection became less evident and there were no clear differences in PNN morphology between the groups at 1 year post infection. Of note, the reduction in PNNs during acute ZIKV infection was not associated with decreased mRNA levels of aggrecan or brevican, but increased levels of degraded aggrecan and brevican indicating increased PNN degradation. These changes were associated with increased expression of matrix metalloproteinase 12 (MMP12) and MMP19, but not MMP9, a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) or ADAMTS5. Together our findings indicate that infection at the time of PNN development interferes with PNN formation, but the nets can reform once the infection and inflammation subside.
Collapse
Affiliation(s)
- Kaliroi Engel
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ha-Na Lee
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Bhanu P. Tewari
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Aaron P. Lewkowicz
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Derek D. C. Ireland
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Mohanraj Manangeeswaran
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Daniela Verthelyi
- Laboratory of Immunology, Center of Excellence in Infectious Disease and Inflammation, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
11
|
Diethorn EJ, Gould E. Development of the hippocampal CA2 region and the emergence of social recognition. Dev Neurobiol 2023; 83:143-156. [PMID: 37326250 PMCID: PMC10529477 DOI: 10.1002/dneu.22919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/08/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023]
Abstract
Social memories formed in early life, like those for family and unrelated peers, are known to contribute to healthy social interactions throughout life, although how the developing brain supports social memory remains relatively unexplored. The CA2 subregion of the hippocampus is involved in social memory function, but most literature on this subject is restricted to studies of adult rodents. Here, we review the current literature on the embryonic and postnatal development of hippocampal subregion CA2 in mammals, with a focus on the emergence of its unusual molecular and cellular characteristics, including its notably high expression of plasticity-suppressing molecules. We also consider the connectivity of the CA2 with other brain areas, including intrahippocampal regions, such as the dentate gyrus, CA3, and CA1 regions, and extrahippocampal regions, such as the hypothalamus, ventral tegmental area, basal forebrain, raphe nuclei, and the entorhinal cortex. We review developmental milestones of CA2 molecular, cellular, and circuit-level features that may contribute to emerging social recognition abilities for kin and unrelated conspecifics in early life. Lastly, we consider genetic mouse models related to neurodevelopmental disorders in humans in order to survey evidence about whether atypical formation of the CA2 may contribute to social memory dysfunction.
Collapse
Affiliation(s)
- Emma J Diethorn
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
12
|
Lev-Ram V, Lemieux SP, Deerinck TJ, Bushong EA, Toyama BH, Perez A, Pritchard DR, Park SKR, McClatchy DB, Savas JN, Taylor SS, Ellisman MH, Yates J, Tsien RY. Do perineuronal nets stabilize the engram of a synaptic circuit? BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.09.536164. [PMID: 37066274 PMCID: PMC10104172 DOI: 10.1101/2023.04.09.536164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Perineuronal nets (PNN), a specialized form of ECM (?), surround numerous neurons in the CNS and allow synaptic connectivity through holes in its structure. We hypothesis that PNNs serve as gatekeepers that guard and protect synaptic territory, and thus may stabilize an engram circuit. We present high-resolution, and 3D EM images of PNN- engulfed neurons showing that synapses occupy the PNN holes, and that invasion of other cellular components are rare. PNN constituents are long-lived and can be eroded faster in an enriched environment, while synaptic proteins have high turnover rate. Preventing PNN erosion by using pharmacological inhibition of PNN-modifying proteases or MMP9 knockout mice allowed normal fear memory acquisition but diminished remote-memory stabilization, supporting the above hypothesis. Significance In this multidisciplinary work, we challenge the hypothesis that the pattern of holes in the perineuronal nets (PNN) hold the code for very-long-term memories. The scope of this work might lead us closer to the understanding of how we can vividly remember events from childhood to death bed. We postulate that the PNN holes hold the code for the engram. To test this hypothesis, we used three independent experimental strategies; high-resolution 3D electron microscopy, Stable Isotop Labeling in Mammals (SILAM) for proteins longevity, and pharmacologically and genetically interruption of memory consolidation in fear conditioning experiments. All of these experimental results did not dispute the PNN hypothesis.
Collapse
|
13
|
Gray DT, Khattab S, Meltzer J, McDermott K, Schwyhart R, Sinakevitch I, Härtig W, Barnes CA. Retrosplenial cortex microglia and perineuronal net densities are associated with memory impairment in aged rhesus macaques. Cereb Cortex 2023; 33:4626-4644. [PMID: 36169578 PMCID: PMC10110451 DOI: 10.1093/cercor/bhac366] [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: 05/06/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Synapse loss and altered plasticity are significant contributors to memory loss in aged individuals. Microglia, the innate immune cells of the brain, play critical roles in maintaining synapse function, including through a recently identified role in regulating the brain extracellular matrix. This study sought to determine the relationship between age, microglia, and extracellular matrix structure densities in the macaque retrosplenial cortex. Twenty-nine macaques ranging in age from young adult to aged were behaviorally characterized on 3 distinct memory tasks. Microglia, parvalbumin (PV)-expressing interneurons and extracellular matrix structures, known as perineuronal nets (PNNs), were immuno- and histochemically labeled. Our results indicate that microglia densities increase in the retrosplenial cortex of aged monkeys, while the proportion of PV neurons surrounded by PNNs decreases. Aged monkeys with more microglia had fewer PNN-associated PV neurons and displayed slower learning and poorer performance on an object recognition task. Stepwise regression models using age and the total density of aggrecan, a chondroitin sulfate proteoglycan of PNNs, better predicted memory performance than did age alone. Together, these findings indicate that elevated microglial activity in aged brains negatively impacts cognition in part through mechanisms that alter PNN assembly in memory-associated brain regions.
Collapse
Affiliation(s)
- Daniel T Gray
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States
| | - Salma Khattab
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States
| | - Jeri Meltzer
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States
| | - Kelsey McDermott
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States
| | - Rachel Schwyhart
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig 04103, Germany
| | - Irina Sinakevitch
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig 04103, Germany
| | - Carol A Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States
- Departments of Psychology, Neurology and Neuroscience, University of Arizona, Tucson, AZ 85721, United States
| |
Collapse
|
14
|
Ueno H, Takahashi Y, Murakami S, Wani K, Miyazaki T, Matsumoto Y, Okamoto M, Ishihara T. Component-Specific Reduction in Perineuronal Nets in Senescence-Accelerated Mouse Strains. IBRO Neurosci Rep 2023. [DOI: 10.1016/j.ibneur.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
15
|
Catale C, Martini A, Piscitelli RM, Senzasono B, Iacono LL, Mercuri NB, Guatteo E, Carola V. Early-life social stress induces permanent alterations in plasticity and perineuronal nets in the mouse anterior cingulate cortex. Eur J Neurosci 2022; 56:5763-5783. [PMID: 36117291 DOI: 10.1111/ejn.15825] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/13/2022] [Accepted: 09/15/2022] [Indexed: 12/29/2022]
Abstract
Child maltreatment disrupts trajectories of brain development, but the underlying pathways are unclear. Stressful stimuli in early life interfere with maturation of local inhibitory circuitry and deposition of perineuronal nets (PNNs), specialized extracellular matrix structures involved in the closure of critical periods of development. Alterations in cortical PNN and parvalbumin (PV) following early-life stress (ELS) have been detected in human and animal studies. Aberrations in the anterior cingulate cortex (ACC) are the most consistent neuroimaging findings in maltreated people, but the molecular mechanisms linking ELS with ACC dysfunctions are unknown. Here, we employed a mouse model of early social threat to test whether ELS experienced in a sensitive period for ACC maturation could induce long-term aberrations of PNN and PV development in the ACC, with consequences on plasticity and ACC-dependent behavior. We found that ELS increased PNN but not PV expression in the ACC of young adult mice. This was associated with reduced frequency of inhibitory postsynaptic currents and long-term potentiation impairments and expression of intense object phobia. Our findings provide information on the long-term effects of ELS on ACC functionality and PNN formation and present evidence for a novel neurobiological pathway underlying the impact of early adversity on the brain.
Collapse
Affiliation(s)
- Clarissa Catale
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Alessandro Martini
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Rosa Maria Piscitelli
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Motor Science and Wellness, Parthenope University of Naples, Naples, Italy
| | | | - Luisa Lo Iacono
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Rome, Italy
| | - Nicola B Mercuri
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Ezia Guatteo
- Division of Experimental Neuroscience, Experimental Neurology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Motor Science and Wellness, Parthenope University of Naples, Naples, Italy
| | - Valeria Carola
- Division of Experimental Neuroscience, Neurobiology of Behavior Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
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: 47] [Impact Index Per Article: 15.7] [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
|
17
|
Dard RF, Leprince E, Denis J, Rao Balappa S, Suchkov D, Boyce R, Lopez C, Giorgi-Kurz M, Szwagier T, Dumont T, Rouault H, Minlebaev M, Baude A, Cossart R, Picardo MA. The rapid developmental rise of somatic inhibition disengages hippocampal dynamics from self-motion. eLife 2022; 11:e78116. [PMID: 35856497 PMCID: PMC9363116 DOI: 10.7554/elife.78116] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Early electrophysiological brain oscillations recorded in preterm babies and newborn rodents are initially mostly driven by bottom-up sensorimotor activity and only later can detach from external inputs. This is a hallmark of most developing brain areas, including the hippocampus, which, in the adult brain, functions in integrating external inputs onto internal dynamics. Such developmental disengagement from external inputs is likely a fundamental step for the proper development of cognitive internal models. Despite its importance, the developmental timeline and circuit basis for this disengagement remain unknown. To address this issue, we have investigated the daily evolution of CA1 dynamics and underlying circuits during the first two postnatal weeks of mouse development using two-photon calcium imaging in non-anesthetized pups. We show that the first postnatal week ends with an abrupt shift in the representation of self-motion in CA1. Indeed, most CA1 pyramidal cells switch from activated to inhibited by self-generated movements at the end of the first postnatal week, whereas the majority of GABAergic neurons remain positively modulated throughout this period. This rapid switch occurs within 2 days and follows the rapid anatomical and functional surge of local somatic GABAergic innervation. The observed change in dynamics is consistent with a two-population model undergoing a strengthening of inhibition. We propose that this abrupt developmental transition inaugurates the emergence of internal hippocampal dynamics.
Collapse
Affiliation(s)
- Robin F Dard
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Erwan Leprince
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Julien Denis
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Shrisha Rao Balappa
- Turing Centre for Living systems, Aix-Marseille University, Université de Toulon, CNRS, CPT (UMR 7332)MarseilleFrance
| | - Dmitrii Suchkov
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Richard Boyce
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Catherine Lopez
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Marie Giorgi-Kurz
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Tom Szwagier
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
- Mines ParisTech, PSL Research UniversityParisFrance
| | - Théo Dumont
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
- Mines ParisTech, PSL Research UniversityParisFrance
| | - Hervé Rouault
- Turing Centre for Living systems, Aix-Marseille University, Université de Toulon, CNRS, CPT (UMR 7332)MarseilleFrance
| | - Marat Minlebaev
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Agnès Baude
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Rosa Cossart
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Michel A Picardo
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| |
Collapse
|
18
|
Ueno H, Takahashi Y, Murakami S, Wani K, Matsumoto Y, Okamoto M, Ishihara T. Fingolimod increases parvalbumin-positive neurons in adult mice. IBRO Neurosci Rep 2022; 13:96-106. [PMID: 36590091 PMCID: PMC9795291 DOI: 10.1016/j.ibneur.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, it has been shown that central nervous system agents, such as antidepressants and antiepileptic drugs, reopen a critical period in mature animals. Fingolimod, which is used for the treatment of multiple sclerosis, also restores neuroplasticity. In this study, we investigated the effects of parvalbumin (PV)-positive neurons and perineuronal nets (PNN) on fingolimod administration with respect to neuroplasticity. Fingolimod was chronically administered intraperitoneally to mature mice. PV-positive neurons and PNN in the hippocampus, prefrontal cortex, and somatosensory cortex were analyzed. An increase in PV-positive neurons was observed in the hippocampus, prefrontal cortex, and somatosensory cortex of the fingolimod-treated mice. An increase in Wisteria floribunda agglutinin-positive PNN was confirmed in mice treated with fingolimod in the somatosensory cortex only. Fingolimod increased the density of PV-positive neurons in the brains of mature mice. The results indicate that fingolimod may change the critical period in mature animals.
Collapse
Key Words
- CNS, central nervous system
- ECM, extracellular matrix
- Fingolimod
- GAD67, anti-glutamic acid decarboxylase
- GFAP, glial fibrillary acidic protein
- Hippocampus
- IL, infralimbic cortex
- NIH, National Institutes of Health, PBS, phosphate-buffered saline
- PL, prelimbic cortex
- PNN, perineuronal net
- PV neurons, parvalbumin-expressing interneurons
- Parvalbumin
- Perineuronal nets
- Prefrontal cortex
- Somatosensory cortex
- WFA, Wisteria floribunda agglutinin
- dAC, dorsal anterior cingulate cortex
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, Kurashiki 701–0193, Japan,Correspondence to: Department of Medical Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, Okayama 701–0193, Japan, 193.
| | - Yu Takahashi
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700–8558, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700–8558, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| |
Collapse
|
19
|
Browne CA, Conant K, Lasek AW, Nacher J. Editorial: Perineuronal Nets as Therapeutic Targets for the Treatment of Neuropsychiatric Disorders. Front Synaptic Neurosci 2022; 14:889800. [PMID: 35782789 PMCID: PMC9240763 DOI: 10.3389/fnsyn.2022.889800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Caroline A. Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, MD, United States
- *Correspondence: Caroline A. Browne
| | - Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Amy W. Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
20
|
Delalande JM, Nagy N, McCann CJ, Natarajan D, Cooper JE, Carreno G, Dora D, Campbell A, Laurent N, Kemos P, Thomas S, Alby C, Attié-Bitach T, Lyonnet S, Logan MP, Goldstein AM, Davey MG, Hofstra RMW, Thapar N, Burns AJ. TALPID3/KIAA0586 Regulates Multiple Aspects of Neuromuscular Patterning During Gastrointestinal Development in Animal Models and Human. Front Mol Neurosci 2022; 14:757646. [PMID: 35002618 PMCID: PMC8733242 DOI: 10.3389/fnmol.2021.757646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/10/2021] [Indexed: 12/26/2022] Open
Abstract
TALPID3/KIAA0586 is an evolutionary conserved protein, which plays an essential role in protein trafficking. Its role during gastrointestinal (GI) and enteric nervous system (ENS) development has not been studied previously. Here, we analyzed chicken, mouse and human embryonic GI tissues with TALPID3 mutations. The GI tract of TALPID3 chicken embryos was shortened and malformed. Histologically, the gut smooth muscle was mispatterned and enteric neural crest cells were scattered throughout the gut wall. Analysis of the Hedgehog pathway and gut extracellular matrix provided causative reasons for these defects. Interestingly, chicken intra-species grafting experiments and a conditional knockout mouse model showed that ENS formation did not require TALPID3, but was dependent on correct environmental cues. Surprisingly, the lack of TALPID3 in enteric neural crest cells (ENCC) affected smooth muscle and epithelial development in a non-cell-autonomous manner. Analysis of human gut fetal tissues with a KIAA0586 mutation showed strikingly similar findings compared to the animal models demonstrating conservation of TALPID3 and its necessary role in human GI tract development and patterning.
Collapse
Affiliation(s)
- Jean Marie Delalande
- Centre for Immunobiology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Dipa Natarajan
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Julie E Cooper
- Developmental Biology and Cancer Program, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Gabriela Carreno
- Developmental Biology and Cancer Program, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Alison Campbell
- Department of Paediatric Surgery, Christchurch Hospital, Christchurch, New Zealand
| | - Nicole Laurent
- Génétique et Anomalies du Développement, Université de Bourgogne, Service d'Anatomie Pathologique, Dijon, France
| | - Polychronis Kemos
- Centre for Immunobiology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sophie Thomas
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France
| | - Caroline Alby
- Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Tania Attié-Bitach
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France.,Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France.,Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stanislas Lyonnet
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France.,Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France.,Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Malcolm P Logan
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Megan G Davey
- Division of Developmental Biology, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Division of Neurogastroenterology and Motility, Department of Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom.,Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Inc., Cambridge, MA, United States
| |
Collapse
|
21
|
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
|
22
|
Ciccarelli A, Weijers D, Kwan W, Warner C, Bourne J, Gross CT. Sexually dimorphic perineuronal nets in the rodent and primate reproductive circuit. J Comp Neurol 2021; 529:3274-3291. [PMID: 33950531 DOI: 10.1002/cne.25167] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022]
Abstract
Perineuronal nets are extracellular glycoprotein structures that have been found on some neurons in the central nervous system and that have been shown to regulate their structural plasticity. Until now work on perineuronal nets has been focused on their role in cortical structures where they are selectively expressed on parvalbumin-positive neurons and are reported to restrict the experience-dependent plasticity of inhibitory afferents. Here, we examined the expression of perineuronal nets subcortically, showing that they are expressed in several discrete structures, including nuclei that comprise the brain network controlling reproductive behaviors (e.g., mounting, lordosis, aggression, and social defense). In particular, perineuronal nets were found in the posterior dorsal division of the medial amygdala, the medial preoptic nucleus, the posterior medial bed nucleus of the stria terminalis, the ventrolateral ventromedial hypothalamus and adjacent tuberal nucleus, and the ventral premammillary nucleus in both the mouse and primate brain. Comparison of perineuronal nets in male and female mice revealed a significant sexually dimorphic expression, with expression found prominently on estrogen receptor expressing neurons in the medial amygdala. These findings suggest that perineuronal nets may be involved in regulating neural plasticity in the mammalian reproductive system.
Collapse
Affiliation(s)
- Alessandro Ciccarelli
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome
| | - Dilys Weijers
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome
| | - William Kwan
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Claire Warner
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - James Bourne
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Cornelius T Gross
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome
| |
Collapse
|
23
|
Mascio G, Bucci D, Notartomaso S, Liberatore F, Antenucci N, Scarselli P, Imbriglio T, Caruso S, Gradini R, Cannella M, Di Menna L, Bruno V, Battaglia G, Nicoletti F. Perineuronal nets are under the control of type-5 metabotropic glutamate receptors in the developing somatosensory cortex. Transl Psychiatry 2021; 11:109. [PMID: 33597513 PMCID: PMC7889908 DOI: 10.1038/s41398-021-01210-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
mGlu5 metabotropic glutamate receptors are highly functional in the early postnatal life, and regulate developmental plasticity of parvalbumin-positive (PV+) interneurons in the cerebral cortex. PV+ cells are enwrapped by perineuronal nets (PNNs) at the closure of critical windows of cortical plasticity. Changes in PNNs have been associated with neurodevelopmental disorders. We found that the number of Wisteria Fluoribunda Agglutinin (WFA)+ PNNs and the density of WFA+/PV+ cells were largely increased in the somatosensory cortex of mGlu5-/- mice at PND16. An increased WFA+ PNN density was also observed after pharmacological blockade of mGlu5 receptors in the first two postnatal weeks. The number of WFA+ PNNs in mGlu5-/- mice was close to a plateau at PND16, whereas continued to increase in wild-type mice, and there was no difference between the two genotypes at PND21 and PND60. mGlu5-/- mice at PND16 showed increases in the transcripts of genes involved in PNN formation and a reduced expression and activity of type-9 matrix metalloproteinase in the somatosensory cortex suggesting that mGlu5 receptors control both PNN formation and degradation. Finally, unilateral whisker stimulation from PND9 to PND16 enhanced WFA+ PNN density in the contralateral somatosensory cortex only in mGlu5+/+ mice, whereas whisker trimming from PND9 to PND16 reduced WFA+ PNN density exclusively in mGlu5-/- mice, suggesting that mGlu5 receptors shape the PNN response to sensory experience. These findings disclose a novel undescribed mechanism of PNN regulation, and lay the groundwork for the study of mGlu5 receptors and PNNs in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Giada Mascio
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | - Domenico Bucci
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | | | | | - Nico Antenucci
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | | | | | - Stefano Caruso
- grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Roberto Gradini
- grid.7841.aDepartment of Experimental Medicine, Sapienza University, Rome, Italy
| | - Milena Cannella
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | - Luisa Di Menna
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy
| | - Valeria Bruno
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giuseppe Battaglia
- grid.419543.e0000 0004 1760 3561IRCCS Neuromed, Pozzilli, Italy ,grid.7841.aDepartment of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy. .,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.
| |
Collapse
|
24
|
Edwards JA, Risch M, Hoke KL. Dynamics of perineuronal nets over amphibian metamorphosis. J Comp Neurol 2020; 529:1768-1778. [PMID: 33067799 DOI: 10.1002/cne.25055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 09/18/2020] [Accepted: 10/07/2020] [Indexed: 11/10/2022]
Abstract
Extracellular matrix materials known as perineuronal nets (PNNs) have been shown to have remarkable consequences for the maturation of neural circuits and stabilization of behavior. It has been proposed that, due to the possibly long-lived biochemical nature of their components, PNNs may be an important substrate by which long-term memories are stored in the central nervous system. However, little empirical evidence exists that shows that PNNs are themselves stable once established. Thus, the question of their temporal dynamics remains unresolved. We leverage the dramatic morphological and behavioral transformations that occur during amphibian metamorphosis to show that PNNs can be highly dynamic in nature. We used established lectin histochemistry to show that PNNs undergo drastic reconstruction during the metamorphic transition. Pre-metamorphic tadpoles have abundant lectin-labeled pericellular material, which we interpret to be PNNs, surrounding neurons throughout the central nervous system. During the metamorphic transition, these structures degrade, and begin to reform in the months following metamorphosis. We show that PNN sizes and staining intensity further change over metamorphosis, suggesting compositional rearrangement. We found PNNs in brain regions with putative homology to regions in mammals with known PNN function, and in other shared regions where PNN function is unknown. Our results suggest that PNNs are susceptible to remodeling by endogenous mechanisms during development. Interpreting the roles of PNNs in circuit maturation and stability requires understanding their temporal relationship with the neurons and synapses they surround. Our work provides further impetus to investigate this relationship in tandem with developmental and behavioral studies.
Collapse
Affiliation(s)
- Jacob A Edwards
- Department of Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Makayla Risch
- Department of Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Kim L Hoke
- Department of Biology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
25
|
Alonge KM, Logsdon AF, Murphree TA, Banks WA, Keene CD, Edgar JS, Whittington D, Schwartz MW, Guttman M. Quantitative analysis of chondroitin sulfate disaccharides from human and rodent fixed brain tissue by electrospray ionization-tandem mass spectrometry. Glycobiology 2020; 29:847-860. [PMID: 31361007 DOI: 10.1093/glycob/cwz060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023] Open
Abstract
Chondroitin sulfates (CS) are long, negatively charged, unbranched glycosaminoglycan (GAG) chains attached to CS-proteoglycan (CSPG) core proteins that comprise the glycan component in both loose interstitial extracellular matrices (ECMs) and in rigid, structured perineuronal net (PNN) scaffolds within the brain. As aberrant CS-PNN formations have been linked to a range of pathological states, including Alzheimer's disease (AD) and schizophrenia, the analysis of CS-GAGs in brain tissue at the disaccharide level has great potential to enhance disease diagnosis and prognosis. Two mass-spectrometry (MS)-based approaches were adapted to detect CS disaccharides from minute fixed tissue samples with low picomolar sensitivity and high reproducibility. The first approach employed a straightforward, quantitative direct infusion (DI)-tandem mass spectrometry (MS/MS) technique to determine the percentages of Δ4S- and Δ6S-CS disaccharides within the 4S/6S-CS ratio, while the second used a comprehensive liquid chromatography (LC)-MS/MS technique to determine the relative percentages of Δ0S-, Δ4S-, Δ6S-, Δ4S6S-CS and Δ2S6S-CS disaccharides, with internal validation by full chondroitin lyase activity. The quantitative accuracy of the five primary biologically relevant CS disaccharides was validated using a developmental time course series in fixed rodent brain tissue. We then analyzed the CS disaccharide composition in formalin-fixed human brain tissue, thus providing the first quantitative report of CS sulfation patterns in the human brain. The ability to comprehensively analyze the CS disaccharide composition from fixed brain tissue provides a means with which to identify alterations in the CS-GAG composition in relation to the onset and/or progression of neurological diseases.
Collapse
Affiliation(s)
- Kimberly M Alonge
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, USA
| | - Aric F Logsdon
- Department of Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Taylor A Murphree
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - William A Banks
- Department of Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA, USA
| | - J Scott Edgar
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Dale Whittington
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, USA
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Uriarte N, Ferreño M, Méndez D, Nogueira J. Reorganization of perineuronal nets in the medial Preoptic Area during the reproductive cycle in female rats. Sci Rep 2020; 10:5479. [PMID: 32214157 PMCID: PMC7096482 DOI: 10.1038/s41598-020-62163-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/09/2020] [Indexed: 01/01/2023] Open
Abstract
Perineuronal nets (PNNs) are aggregations of extracellular matrix associated with specific neuronal populations in the central nervous system, suggested to play key roles in neural development, synaptogenesis and experience-dependent synaptic plasticity. Pregnancy and lactation are characterized by a dramatic increase in neuroplasticity. However, dynamic changes in the extracellular matrix associated with maternal circuits have been mostly overlooked. We analyzed the structure of PNNs in an essential nucleus of the maternal circuit, the medial preoptic area (mPOA), during the reproductive cycle of rats, using the Wisteria floribunda (WFA) label. PNNs associated to neurons in the mPOA start to assemble halfway through gestation and become highly organized prior to parturition, fading through the postpartum period. This high expression of PNNs during pregnancy appears to be mediated by the influence of estrogen, progesterone and prolactin, since a hormonal simulated-gestation treatment induced the expression of PNNs in ovariectomized females. We found that PNNs associated neurons in the mPOA express estrogen receptor α and progesterone receptors, supporting a putative role of reproductive hormones in the signaling mechanisms that trigger the assembly of PNNs in the mPOA. This is the first report of PNNs presence and remodeling in mPOA during adulthood induced by physiological variables.
Collapse
Affiliation(s)
- Natalia Uriarte
- Laboratorio de Neurociencias, Facultad de Ciencias, Universidad de la República, Montevideo, 11400, Uruguay
| | - Marcela Ferreño
- Laboratorio de Neurociencias, Facultad de Ciencias, Universidad de la República, Montevideo, 11400, Uruguay
| | - Diego Méndez
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, 11800, Uruguay
| | - Javier Nogueira
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, 11800, Uruguay.
| |
Collapse
|
27
|
Raghunathan R, Sethi MK, Klein JA, Zaia J. Proteomics, Glycomics, and Glycoproteomics of Matrisome Molecules. Mol Cell Proteomics 2019; 18:2138-2148. [PMID: 31471497 PMCID: PMC6823855 DOI: 10.1074/mcp.r119.001543] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/26/2019] [Indexed: 12/21/2022] Open
Abstract
The most straightforward applications of proteomics database searching involve intracellular proteins. Although intracellular gene products number in the thousands, their well-defined post-translational modifications (PTMs) makes database searching practical. By contrast, cell surface and extracellular matrisome proteins pass through the secretory pathway where many become glycosylated, modulating their physicochemical properties, adhesive interactions, and diversifying their functions. Although matrisome proteins number only a few hundred, their high degree of complex glycosylation multiplies the number of theoretical proteoforms by orders of magnitude. Given that extracellular networks that mediate cell-cell and cell-pathogen interactions in physiology depend on glycosylation, it is important to characterize the proteomes, glycomes, and glycoproteomes of matrisome molecules that exist in a given biological context. In this review, we summarize proteomics approaches for characterizing matrisome molecules, with an emphasis on applications to brain diseases. We demonstrate the availability of methods that should greatly increase the availability of information on matrisome molecular structure associated with health and disease.
Collapse
Affiliation(s)
- Rekha Raghunathan
- Molecular and Translational Medicine Program, Boston University, Boston, MA 02218; Department of Biochemistry, Boston University, Boston, MA 02218
| | - Manveen K Sethi
- Department of Biochemistry, Boston University, Boston, MA 02218
| | - Joshua A Klein
- Bioinformatics Program, Boston University, Boston, MA 02218
| | - Joseph Zaia
- Molecular and Translational Medicine Program, Boston University, Boston, MA 02218; Department of Biochemistry, Boston University, Boston, MA 02218; Bioinformatics Program, Boston University, Boston, MA 02218.
| |
Collapse
|
28
|
Domínguez S, Rey CC, Therreau L, Fanton A, Massotte D, Verret L, Piskorowski RA, Chevaleyre V. Maturation of PNN and ErbB4 Signaling in Area CA2 during Adolescence Underlies the Emergence of PV Interneuron Plasticity and Social Memory. Cell Rep 2019; 29:1099-1112.e4. [DOI: 10.1016/j.celrep.2019.09.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/31/2019] [Accepted: 09/13/2019] [Indexed: 12/28/2022] Open
|
29
|
Reichelt AC, Gibson GD, Abbott KN, Hare DJ. A high-fat high-sugar diet in adolescent rats impairs social memory and alters chemical markers characteristic of atypical neuroplasticity and parvalbumin interneuron depletion in the medial prefrontal cortex. Food Funct 2019; 10:1985-1998. [PMID: 30900711 DOI: 10.1039/c8fo02118j] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Brain plasticity is a multifaceted process that is dependent on both neurons and extracellular matrix (ECM) structures, including perineuronal nets (PNNs). In the medial prefrontal cortex (mPFC) PNNs primarily surround fast-spiking parvalbumin (PV)-containing GABAergic interneurons and are central to regulation of neuroplasticity. In addition to the development of obesity, high-fat and high-sugar (HFHS) diets are also associated with alterations in brain plasticity and emotional behaviours in humans. To examine the underlying involvement of PNNs and cortical plasticity in the mPFC in diet-evoked social behaviour deficits (in this case social recognition), we exposed adolescent (postnatal days P28-P56) rats to a HFHS-supplemented diet. At P56 HFHS-fed animals and age-matched controls fed standard chow were euthanized and co-localization of PNNs with PV neurons in the prelimbic (PrL) and infralimbic (IL) and anterior cingulate (ACC) sub regions of the PFC were examined by dual fluorescence immunohistochemistry. ΔFosB expression was also assessed as a measure of chronic activity and behavioural addiction marker. Consumption of the HFHS diet reduced the number of PV+ neurons and PNNs in the infralimbic (IL) region of the mPFC by -21.9% and -16.5%, respectively. While PV+ neurons and PNNs were not significantly decreased in the ACC or PrL, the percentage of PV+ and PNN co-expressing neurons was increased in all assessed regions of the mPFC in HFHS-fed rats (+33.7% to +41.3%). This shows that the population of PV neurons remaining are those surrounded by PNNs, which may afford some protection against HFHS diet-induced mPFC-dysregulation. ΔFosB expression showed a 5-10-fold increase (p < 0.001) in each mPFC region, supporting the hypothesis that a HFHS diet induces mPFC dysfunction and subsequent behavioural deficits. The data presented shows a potential neurophysiological mechanism and response to specific diet-evoked social recognition deficits as a result of hypercaloric intake in adolescence.
Collapse
Affiliation(s)
- Amy C Reichelt
- BrainsCAN and Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.
| | | | | | | |
Collapse
|
30
|
Pesarico AP, Bueno-Fernandez C, Guirado R, Gómez-Climent MÁ, Curto Y, Carceller H, Nacher J. Chronic Stress Modulates Interneuronal Plasticity: Effects on PSA-NCAM and Perineuronal Nets in Cortical and Extracortical Regions. Front Cell Neurosci 2019; 13:197. [PMID: 31133813 PMCID: PMC6524695 DOI: 10.3389/fncel.2019.00197] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/18/2019] [Indexed: 01/31/2023] Open
Abstract
Chronic stress has an important impact on the adult brain. However, most of the knowledge on its effects is focused on principal neurons and less on inhibitory neurons. Consequently, recent reports have begun to describe stress-induced alterations in the structure, connectivity and neurochemistry of interneurons. Some of these changes appear to be mediated by certain molecules particularly associated to interneurons, such as the polysialylated form of the neural cell adhesion molecule (PSA-NCAM) and components of the perineuronal nets (PNN), specialized regions of the extracellular matrix. These plasticity-related molecules modulate interneuronal structure and connectivity, particularly of parvalbumin expressing basket interneurons, both during development and adult life. These inhibitory neurons are specially affected after chronic stress and in some stress-related disorders, in which the expression of PSA-NCAM and certain components of PNN are also altered. For these reasons we have decided to study PSA-NCAM, PNN and parvalbumin expressing interneurons after 10 days of chronic restraint stress, a time point in which its behavioral consequences are starting to appear. We have focused initially on the medial prefrontal cortex (mPFC), basolateral amygdala (BLA) and hippocampus, regions affected by stress and stress-related psychiatric diseases, but we have also explored the habenula and the thalamic reticular nucleus (TRN) due to the important presence of PNN and their relationship with certain disorders. PSA-NCAM expression was increased by stress in the stratum lacunosum-moleculare of CA1. Increases in parvalbumin immunoreactive cells were detected in the mPFC and the BLA, but were not accompanied by increases in the number of parvalbumin expressing perisomatic puncta on the somata of principal neurons. The number of PNN was also increased in the mPFC and the habenula, although habenular PNN were not associated to parvalbumin cells. Increased expression of parvalbumin and components of PNN were also detected in the TRN after chronic restraint stress, revealing for the first time substantial effects on this region. Our study shows that, even a short chronic stress protocol, can induce consistent changes in interneuronal plasticity-related molecules in cortical and extracortical regions, which may represent initial responses of inhibitory circuits to counteract the effects of this aversive experience.
Collapse
Affiliation(s)
- Ana Paula Pesarico
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Clara Bueno-Fernandez
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Ramón Guirado
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - María Ángeles Gómez-Climent
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Yasmina Curto
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Hector Carceller
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
31
|
Spatiotemporal expression patterns of chondroitin sulfate proteoglycan mRNAs in the developing rat brain. Neuroreport 2019; 29:517-523. [PMID: 29271834 DOI: 10.1097/wnr.0000000000000957] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are pluripotent components of the extracellular matrix in the brain. Although previous studies have examined the developmental change in certain CSPGs in the whole brain, no known systematic studies have been carried out on the temporal or spatial expression of CSPGs. Here, we used quantitative real-time PCR to examine the CSPG mRNAs expression in the postnatal developing rat brain starting from postnatal day 5-42, mainly focusing on the parietal cortex, hippocampus, and corpus callosum. Results were further verified by immunohistochemistry. Our results showed that aggrecan, brevican, phosphacan, and NG2 generally showed upregulation across developmental stages in all three regions. Neurocan showed a rapid increase until postnatal day 10 in all three regions. Versican, however, showed a sharp decrease until postnatal day 10. Cross-region analysis showed higher expression of most CSPG members in the corpus callosum during later stages of development. Further immunohistochemistry staining confirmed our results by showing prominent CSPGs protein expression in the corpus callosum. In summary, our study reported specific temporal-expression and spatial-expression patterns of the CSPGs species. These results are consistent with the known roles of these members in neurodevelopment. The current study provided clues for the development of CSPGs as potential treatment targets in neurodevelopmental disorders.
Collapse
|
32
|
Alteration of parvalbumin expression and perineuronal nets formation in the cerebral cortex of aged mice. Mol Cell Neurosci 2019; 95:31-42. [DOI: 10.1016/j.mcn.2018.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/18/2018] [Accepted: 12/26/2018] [Indexed: 01/15/2023] Open
|
33
|
Cattaud V, Bezzina C, Rey CC, Lejards C, Dahan L, Verret L. Early disruption of parvalbumin expression and perineuronal nets in the hippocampus of the Tg2576 mouse model of Alzheimer's disease can be rescued by enriched environment. Neurobiol Aging 2018; 72:147-158. [DOI: 10.1016/j.neurobiolaging.2018.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022]
|
34
|
Moffitt JR, Bambah-Mukku D, Eichhorn SW, Vaughn E, Shekhar K, Perez JD, Rubinstein ND, Hao J, Regev A, Dulac C, Zhuang X. Molecular, spatial, and functional single-cell profiling of the hypothalamic preoptic region. Science 2018; 362:eaau5324. [PMID: 30385464 PMCID: PMC6482113 DOI: 10.1126/science.aau5324] [Citation(s) in RCA: 750] [Impact Index Per Article: 107.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/21/2018] [Indexed: 12/23/2022]
Abstract
The hypothalamus controls essential social behaviors and homeostatic functions. However, the cellular architecture of hypothalamic nuclei-including the molecular identity, spatial organization, and function of distinct cell types-is poorly understood. Here, we developed an imaging-based in situ cell-type identification and mapping method and combined it with single-cell RNA-sequencing to create a molecularly annotated and spatially resolved cell atlas of the mouse hypothalamic preoptic region. We profiled ~1 million cells, identified ~70 neuronal populations characterized by distinct neuromodulatory signatures and spatial organizations, and defined specific neuronal populations activated during social behaviors in male and female mice, providing a high-resolution framework for mechanistic investigation of behavior circuits. The approach described opens a new avenue for the construction of cell atlases in diverse tissues and organisms.
Collapse
Affiliation(s)
- Jeffrey R Moffitt
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Dhananjay Bambah-Mukku
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Stephen W Eichhorn
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Eric Vaughn
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Karthik Shekhar
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
| | - Julio D Perez
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Nimrod D Rubinstein
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Junjie Hao
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Aviv Regev
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
- Koch Institute of Integrative Cancer Biology, Department of Biology, MIT, Cambridge, MA 02139, USA
| | - Catherine Dulac
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA.
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
35
|
Marchese E, Corvino V, Di Maria V, Furno A, Giannetti S, Cesari E, Lulli P, Michetti F, Geloso MC. The Neuroprotective Effects of 17β-Estradiol Pretreatment in a Model of Neonatal Hippocampal Injury Induced by Trimethyltin. Front Cell Neurosci 2018; 12:385. [PMID: 30416427 PMCID: PMC6213803 DOI: 10.3389/fncel.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17β-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.
Collapse
Affiliation(s)
- Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alfredo Furno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Cesari
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Paola Lulli
- Laboratorio di Biochimica Clinica e Biologia Molecolare, IRCCS Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Facoltà di Medicina e Chirurgia - IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
36
|
Santiago AN, Lim KY, Opendak M, Sullivan RM, Aoki C. Early life trauma increases threat response of peri-weaning rats, reduction of axo-somatic synapses formed by parvalbumin cells and perineuronal net in the basolateral nucleus of amygdala. J Comp Neurol 2018; 526:2647-2664. [PMID: 30136731 DOI: 10.1002/cne.24522] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/20/2018] [Accepted: 08/03/2018] [Indexed: 01/17/2023]
Abstract
Early life trauma is a risk factor for life-long disorders related to emotional processing, but knowledge underlying its enduring effect is incomplete. This study was motivated by the hypothesis that early life trauma increases amygdala-dependent threat responses via reduction in inhibition by parvalbumin (PV) interneurons and perineuronal nets (PNN) supporting PV cells, thus increasing excitability of the basolateral amygdala (BLA). From postnatal day (PN) 8-12, rat pups of both sexes were reared under normal bedding or under insufficient nest-building materials to induce maternal-to-infant maltreatment trauma (Scarcity-Adversity Model, SAM). At weaning age of PN23, the SAM group exhibited increased threat responses to predator odor. The SAM-induced increase in threat response was recapitulated in normally reared PN22-23 rats that were unilaterally depleted of PNN in the BLA by the enzymes, chondroitinase-ABC plus hyaluronidase at PN19-20. Light and electron microscopic analysis of the BLA revealed that anterior-to-mid levels of SAM group's BLAs exhibited decreased PNN intensity and decreased axo-somatic synapses between PV-to-principal pyramidal-like neurons and PV-to-PV. PV and PNN densities (cells/mm2 ) in the BLA of both control (CON) and SAM groups were still low at PN12 and SAM delayed the ontogenetic rise of PV intensity and PNN density. Moreover, PV cell density in the anterior-to-mid BLA correlated negatively with threat response of CON animals, but not for SAM animals. Thus, reduction of PNN-supported, PV-mediated somatic inhibition of pyramidal cells provides a mechanistic support for the enduring effect of early life maltreatment manifested as increasing innate threat response at weaning.
Collapse
Affiliation(s)
- Adrienne N Santiago
- Center for Neural Science, New York University, New York, New York.,Emotional Brain Institute, Nathan Kline Institute, New York University School of Medicine, New York, New York.,Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, New York
| | - Kayla Y Lim
- Center for Neural Science, New York University, New York, New York
| | - Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute, New York University School of Medicine, New York, New York.,Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, New York
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute, New York University School of Medicine, New York, New York.,Department of Child and Adolescent Psychiatry, NYU School of Medicine, New York, New York
| | - Chiye Aoki
- Center for Neural Science, New York University, New York, New York
| |
Collapse
|
37
|
Erasure of striatal chondroitin sulfate proteoglycan-associated extracellular matrix rescues aging-dependent decline of motor learning. Neurobiol Aging 2018; 71:61-71. [PMID: 30099347 DOI: 10.1016/j.neurobiolaging.2018.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 07/14/2018] [Accepted: 07/15/2018] [Indexed: 01/13/2023]
Abstract
Cognitive decline is a feature of aging. Accumulating evidence suggests that the brain extracellular matrix (ECM) is involved in the process of aging-dependent cognitive impairment and neurodegeneration by regulating synaptic neurotransmission and affecting neuroplasticity. Age-related changes in brain structure and cognition are not uniform across the whole brain. Being one of the most vulnerable brain regions to aging-dependent alterations, striatum is integral to several central nervous system functions, such as motor, cognition, and affective control. However, the striatal ECM is largely understudied. We first describe 2 major types of chondroitin sulfate proteoglycan (CSPG)-associated ECM in striatum: perineuronal nets and diffusive ECM. Both types of ECM accumulate in an aging-dependent manner. The accumulation of CSPG-associated ECM correlates with aging-dependent decline in striatum-related cognitive functions, including motor learning and working memory. Enzymatic depletion of CSPG-associated ECM in aged mice via chondroitinase ABC significantly improves motor learning, suggesting that changes in neural ECM CSPGs regulate striatal plasticity. Our study provides a greater understanding of the role of neural ECM underlying striatal plasticity, which is an important precursor to design appropriate therapeutic strategies for normal and pathologic aging.
Collapse
|
38
|
Ueno H, Fujii K, Suemitsu S, Murakami S, Kitamura N, Wani K, Aoki S, Okamoto M, Ishihara T, Takao K. Expression of aggrecan components in perineuronal nets in the mouse cerebral cortex. IBRO Rep 2018; 4:22-37. [PMID: 30135949 PMCID: PMC6084874 DOI: 10.1016/j.ibror.2018.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 01/27/2018] [Indexed: 11/18/2022] Open
Abstract
Specific regions of the cerebral cortex are highly plastic in an organism's lifetime. It is thought that perineuronal nets (PNNs) regulate plasticity, but labeling for Wisteria floribunda agglutinin (WFA), which is widely used to detect PNNs, is observed throughout the cortex. The aggrecan molecule-a PNN component-may regulate plasticity, and may also be involved in determining region-specific vulnerability to stress. To clarify cortical region-specific plasticity and vulnerability, we qualitatively analyzed aggrecan-positive and glycosylated aggrecan-positive PNNs in the mature mouse cerebral cortex. Our findings revealed the selective expression of both aggrecan-positive and glycosylated aggrecan-positive PNNs in the cortex. WFA-positive PNNs expressed aggrecan in a region-specific manner in the cortex. Furthermore, we observed variable distributions of PNNs containing WFA- and aggrecan-positive molecules. Together, our findings suggest that PNN components and their function differ depending on the cortical region, and that aggrecan molecules may be involved in determining region-specific plasticity and vulnerability in the cortex.
Collapse
Key Words
- Aggrecan
- Au1, primary auditory cortex
- AuD, secondary auditory cortex dorsal area
- AuV, secondary auditory cortex ventral area
- Brain region-specific
- Cg, cingulate cortex
- Chondroitin sulfate proteoglycan
- DIEnt, dorsintermed entorhinal cortex
- DLEnt, dorsolateral entorhinal cortex
- DLO, dorsolateral orbital cortex
- DP, dorsal peduncular cortex
- Ect, ectorhinal cortex
- Extracellular matrix
- FrA, frontal association cortex
- IL, infralimbic cortex
- LO, lateral orbital cortex
- LPtA, lateral parietal association cortex
- M1, primary motor cortex
- M2, secondary motor cortex
- MPtA, medial parietal association cortex
- PL, prelimbic cortex
- PRh, perirhinal cortex
- Perineuronal nets
- Plasticity
- RSD, retrosplenial dysgranular cortex
- RSGa, retrosplenial granular cortex a region
- RSGb, retrosplenial granular cortex b region
- RSGc, retrosplenial granular cortex c region
- S1BF, primary somatosensory cortex–barrel field
- S1Tr, primary somatosensory cortex–trunk region
- S2, secondary somatosensory cortex
- TeA, temporal association cortex
- V1B, primary visual cortex binocular area
- V1M, primary visual cortex monocular area
- V2L, secondary visual cortex lateral area
- V2ML, secondary visual cortex mediolateral area
- V2MM, secondary visual cortex–mediomedial area
- VO, ventral orbital cortex
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, Okayama, 701-0193, Japan
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, 700-8558, Japan
- Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Kazuki Fujii
- Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama, 930-0194, Japan
| | - Shunsuke Suemitsu
- Department of Psychiatry, Kawasaki Medical School, Okayama, 701-0192, Japan
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Okayama, 701-0192, Japan
| | - Naoya Kitamura
- Department of Psychiatry, Kawasaki Medical School, Okayama, 701-0192, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Okayama, 701-0192, Japan
| | - Shozo Aoki
- Department of Psychiatry, Kawasaki Medical School, Okayama, 701-0192, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Okayama, 701-0192, Japan
| | - Keizo Takao
- Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama, 930-0194, Japan
| |
Collapse
|
39
|
Perineuronal Nets in the Deep Cerebellar Nuclei Regulate GABAergic Transmission and Delay Eyeblink Conditioning. J Neurosci 2018; 38:6130-6144. [PMID: 29858484 DOI: 10.1523/jneurosci.3238-17.2018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/28/2018] [Accepted: 05/28/2018] [Indexed: 11/21/2022] Open
Abstract
Perineuronal nets (PNNs), composed mainly of chondroitin sulfate proteoglycans, are the extracellular matrix that surrounds cell bodies, proximal dendrites, and axon initial segments of adult CNS neurons. PNNs are known to regulate neuronal plasticity, although their physiological roles in cerebellar functions have yet to be elucidated. Here, we investigated the contribution of PNNs to GABAergic transmission from cerebellar Purkinje cells (PCs) to large glutamatergic neurons in the deep cerebellar nuclei (DCN) in male mice by recording IPSCs from cerebellar slices, in which PNNs were depleted with chondroitinase ABC (ChABC). We found that PNN depletion increased the amplitude of evoked IPSCs and enhanced the paired-pulse depression. ChABC treatment also facilitated spontaneous IPSCs and increased the miniature IPSC frequency without changing not only the amplitude but also the density of PC terminals, suggesting that PNN depletion enhances presynaptic GABA release. We also demonstrated that the enhanced GABAergic transmission facilitated rebound firing in large glutamatergic DCN neurons, which is expected to result in the efficient induction of synaptic plasticity at synapses onto DCN neurons. Furthermore, we tested whether PNN depletion affects cerebellar motor learning. Mice having received the enzyme into the interpositus nuclei, which are responsible for delay eyeblink conditioning, exhibited the conditioned response at a significantly higher rate than control mice. Therefore, our results suggest that PNNs of the DCN suppress GABAergic transmission between PCs and large glutamatergic DCN neurons and restrict synaptic plasticity associated with motor learning in the adult cerebellum.SIGNIFICANCE STATEMENT Perineuronal nets (PNNs) are one of the extracellular matrices of adult CNS neurons and implicated in regulating various brain functions. Here we found that enzymatic PNN depletion in the mouse deep cerebellar nuclei (DCN) reduced the paired-pulse ratio of IPSCs and increased the miniature IPSC frequency without changing the amplitude, suggesting that PNN depletion enhances GABA release from the presynaptic Purkinje cell (PC) terminals. Mice having received the enzyme in the interpositus nuclei exhibited a higher conditioned response rate in delay eyeblink conditioning than control mice. These results suggest that PNNs regulate presynaptic functions of PC terminals in the DCN and functional plasticity of synapses on DCN neurons, which influences the flexibility of adult cerebellar functions.
Collapse
|
40
|
McDonald AJ, Hamilton PG, Barnstable CJ. Perineuronal nets labeled by monoclonal antibody VC1.1 ensheath interneurons expressing parvalbumin and calbindin in the rat amygdala. Brain Struct Funct 2018; 223:1133-1148. [PMID: 29094304 PMCID: PMC5871560 DOI: 10.1007/s00429-017-1542-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 10/19/2017] [Indexed: 01/06/2023]
Abstract
Perineuronal nets (PNNs) are specialized condensations of extracellular matrix that ensheath particular neuronal subpopulations in the brain and spinal cord. PNNs regulate synaptic plasticity, including the encoding of fear memories by the amygdala. The present immunohistochemical investigation studied PNN structure and distribution, as well as the neurochemistry of their ensheathed neurons, in the rat amygdala using monoclonal antibody VC1.1, which recognizes a glucuronic acid 3-sulfate glycan associated with PNNs in the cerebral cortex. VC1.1+ PNNs surrounded the cell bodies and dendrites of a subset of nonpyramidal neurons in cortex-like portions of the amygdala (basolateral amygdalar complex, cortical nuclei, nucleus of the lateral olfactory tract, and amygdalohippocampal region). There was also significant neuropilar VC1.1 immunoreactivity, whose density varied in different amygdalar nuclei. Cell counts in the basolateral nucleus revealed that virtually all neurons ensheathed by VC1.1+ PNNs were parvalbumin-positive (PV+) interneurons, and these VC1.1+/PV+ cells constituted 60% of all PV+ interneurons, including all of the larger PV+ neurons. Approximately 70% of VC1.1+ neurons were calbindin-positive (CB+), and these VC1.1+/CB+ cells constituted about 40% of all CB+ neurons. Colocalization of VC1.1 with Vicia villosa agglutinin (VVA) binding, which stains terminal N-acetylgalactosamines, revealed that VC1.1+ PNNs were largely a subset of VVA+ PNNs. This investigation provides baseline data regarding PNNs in the rat which should be useful for future studies of their function in this species.
Collapse
Affiliation(s)
- Alexander J McDonald
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| | - Patricia G Hamilton
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Colin J Barnstable
- Department of Neural and Behavioral Sciences, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
41
|
Klein PM, Lu AC, Harper ME, McKown HM, Morgan JD, Beenhakker MP. Tenuous Inhibitory GABAergic Signaling in the Reticular Thalamus. J Neurosci 2018; 38:1232-1248. [PMID: 29273603 PMCID: PMC5792478 DOI: 10.1523/jneurosci.1345-17.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/20/2017] [Accepted: 11/03/2017] [Indexed: 11/21/2022] Open
Abstract
Maintenance of a low intracellular Cl- concentration ([Cl-]i) is critical for enabling inhibitory neuronal responses to GABAA receptor-mediated signaling. Cl- transporters, including KCC2, and extracellular impermeant anions ([A]o) of the extracellular matrix are both proposed to be important regulators of [Cl-]i Neurons of the reticular thalamic (RT) nucleus express reduced levels of KCC2, indicating that GABAergic signaling may produce excitation in RT neurons. However, by performing perforated patch recordings and calcium imaging experiments in rats (male and female), we find that [Cl-]i remains relatively low in RT neurons. Although we identify a small contribution of [A]o to a low [Cl-]i in RT neurons, our results also demonstrate that reduced levels of KCC2 remain sufficient to maintain low levels of Cl- Reduced KCC2 levels, however, restrict the capacity of RT neurons to rapidly extrude Cl- following periods of elevated GABAergic signaling. In a computational model of a local RT network featuring slow Cl- extrusion kinetics, similar to those we found experimentally, model RT neurons are predisposed to an activity-dependent switch from GABA-mediated inhibition to excitation. By decreasing the activity threshold required to produce excitatory GABAergic signaling, weaker stimuli are able to propagate activity within the model RT nucleus. Our results indicate the importance of even diminished levels of KCC2 in maintaining inhibitory signaling within the RT nucleus and suggest how this important activity choke point may be easily overcome in disorders such as epilepsy.SIGNIFICANCE STATEMENT Precise regulation of intracellular Cl- levels ([Cl-]i) preserves appropriate, often inhibitory, GABAergic signaling within the brain. However, there is disagreement over the relative contribution of various mechanisms that maintain low [Cl-]i We found that the Cl- transporter KCC2 is an important Cl- extruder in the reticular thalamic (RT) nucleus, despite this nucleus having remarkably low KCC2 immunoreactivity relative to other regions of the adult brain. We also identified a smaller contribution of fixed, impermeant anions ([A]o) to lowering [Cl-]i in RT neurons. Inhibitory signaling among RT neurons is important for preventing excessive activation of RT neurons, which can be responsible for generating seizures. Our work suggests that KCC2 critically restricts the spread of activity within the RT nucleus.
Collapse
Affiliation(s)
- Peter M Klein
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Adam C Lu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Megan E Harper
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Hannah M McKown
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Jessica D Morgan
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Mark P Beenhakker
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| |
Collapse
|
42
|
Lasek AW, Chen H, Chen WY. Releasing Addiction Memories Trapped in Perineuronal Nets. Trends Genet 2017; 34:197-208. [PMID: 29289347 DOI: 10.1016/j.tig.2017.12.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/28/2017] [Accepted: 12/06/2017] [Indexed: 12/20/2022]
Abstract
Drug addiction can be conceptualized at a basic level as maladaptive learning and memory. Addictive substances elicit changes in brain circuitry involved in reward, cognition, and emotional state, leading to the formation and persistence of strong drug-associated memories that lead to craving and relapse. Recently, perineuronal nets (PNNs), extracellular matrix (ECM) structures surrounding neurons, have emerged as regulators of learning, memory, and addiction behaviors. PNNs do not merely provide structural support to neurons but are dynamically remodeled in an experience-dependent manner by metalloproteinases. They function in various brain regions through constituent proteins such as brevican that are implicated in neural plasticity. Understanding the function of PNN components in memory processes may lead to new therapeutic approaches to treating addiction.
Collapse
Affiliation(s)
- Amy W Lasek
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Hu Chen
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Wei-Yang Chen
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
43
|
Ueno H, Suemitsu S, Murakami S, Kitamura N, Wani K, Okamoto M, Aoki S, Ishihara T. Postnatal development of GABAergic interneurons and perineuronal nets in mouse temporal cortex subregions. Int J Dev Neurosci 2017; 63:27-37. [PMID: 28859888 DOI: 10.1016/j.ijdevneu.2017.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/20/2017] [Accepted: 08/25/2017] [Indexed: 10/19/2022] Open
Abstract
In human neuropsychiatric disorders, there are functional and anatomical abnormalities of GABAergic interneurons in each temporal cortex subregion. Furthermore, accumulation of amyloid-β is observed in the temporal cortex in the early stages of Alzheimer's disease. Each subregion of the temporal cortex has an important role in coordinating the input and output of the hippocampus. When subregions of the temporal cortex are impaired, memory and learning ability decrease. GABAergic interneurons control excitatory neurons, forming the cortico-cortical and cortico-hippocampal networks. However, in temporal cortex subregions, details of the distribution and developmental processes of GABAergic interneurons and perineuronal nets (PNNs) have not been elucidated. Here we examined the development of GABAergic interneurons and PNNs in mouse temporal cortex subregions. Results indicate that temporal cortex GABAergic interneurons have developmental stages different to those of the primary sensory cortex. In addition, the density of PNNs in the temporal cortex is lower than that in the sensory cortex. Furthermore, we found that the Wisteria floribunda agglutinin-reactive extracellular matrix molecule is present in the upper level of layer 1 of the temporal cortex. These results support the idea that mouse temporal cortex subregions develop differently from other cortical regions and have region-specific characteristics after maturation. The present study results suggested that the structure of the temporal cortex is significantly different from the sensory cortex and that temporal cortex may be highly vulnerable to neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, 288, Matsushima, Kurashiki, Okayama, 701-0193, Japan; Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, kitaku, Okayama, 700-8558, Japan.
| | - Shunsuke Suemitsu
- Department of Psychiatry, Kawasaki Medical School, 577, Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, 577, Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Naoya Kitamura
- Department of Psychiatry, Kawasaki Medical School, 577, Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, 577, Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, 2-5-1, Shikata-cho, kitaku, Okayama, 700-8558, Japan.
| | - Shozo Aoki
- Department of Psychiatry, Kawasaki Medical School, 577, Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, 577, Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| |
Collapse
|
44
|
Perineuronal Nets Suppress Plasticity of Excitatory Synapses on CA2 Pyramidal Neurons. J Neurosci 2017; 36:6312-20. [PMID: 27277807 DOI: 10.1523/jneurosci.0245-16.2016] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/02/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Long-term potentiation of excitatory synapses on pyramidal neurons in the stratum radiatum rarely occurs in hippocampal area CA2. Here, we present evidence that perineuronal nets (PNNs), a specialized extracellular matrix typically localized around inhibitory neurons, also surround mouse CA2 pyramidal neurons and envelop their excitatory synapses. CA2 pyramidal neurons express mRNA transcripts for the major PNN component aggrecan, identifying these neurons as a novel source for PNNs in the hippocampus. We also found that disruption of PNNs allows synaptic potentiation of normally plasticity-resistant excitatory CA2 synapses; thus, PNNs play a role in restricting synaptic plasticity in area CA2. Finally, we found that postnatal development of PNNs on CA2 pyramidal neurons is modified by early-life enrichment, suggesting that the development of circuits containing CA2 excitatory synapses are sensitive to manipulations of the rearing environment. SIGNIFICANCE STATEMENT Perineuronal nets (PNNs) are thought to play a major role in restricting synaptic plasticity during postnatal development, and are altered in several models of neurodevelopmental disorders, such as schizophrenia and Rett syndrome. Although PNNs have been predominantly studied in association with inhibitory neurons throughout the brain, we describe a dense expression of PNNs around excitatory pyramidal neurons in hippocampal area CA2. We also provide insight into a previously unrecognized role for PNNs in restricting plasticity at excitatory synapses and raise the possibility of an early critical period of hippocampal plasticity that may ultimately reveal a key mechanism underlying learning and memory impairments of PNN-associated neurodevelopmental disorders.
Collapse
|
45
|
Noguchi A, Matsumoto N, Morikawa S, Tamura H, Ikegaya Y. Juvenile Hippocampal CA2 Region Expresses Aggrecan. Front Neuroanat 2017; 11:41. [PMID: 28539874 PMCID: PMC5423971 DOI: 10.3389/fnana.2017.00041] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 04/21/2017] [Indexed: 12/14/2022] Open
Abstract
Perineuronal nets (PNNs) are distributed primarily around inhibitory interneurons in the hippocampus, such as parvalbumin-positive interneurons. PNNs are also present around excitatory neurons in some brain regions and prevent plasticity in these neurons. A recent study demonstrated that PNNs also exist around mouse hippocampal pyramidal cells, which are the principle type of excitatory neurons, in the CA2 subregion and modulate the excitability and plasticity of these neurons. However, the development of PNNs in the CA2 region during postnatal maturation was not fully investigated. This study found that a main component of PNNs, aggrecan, existed in the pyramidal cell layer of the putative CA2 subarea prior to the appearance of the CA2 region, which was defined by the CA2 marker protein regulator of G protein signaling 14 (RGS14). We also found that aggrecan immunoreactivity was more evident in the anterior sections of the CA2 area than the posterior sections, which suggests that the function of CA2 PNNs varies along the anterior-posterior axis.
Collapse
Affiliation(s)
- Asako Noguchi
- Graduate School of Pharmaceutical Sciences, The University of TokyoTokyo, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of TokyoTokyo, Japan
| | - Shota Morikawa
- Life Science Tokyo Advanced Research Center (L-StaR), School of Pharmacy and Pharmaceutical Sciences, Hoshi UniversityTokyo, Japan
| | - Hideki Tamura
- Life Science Tokyo Advanced Research Center (L-StaR), School of Pharmacy and Pharmaceutical Sciences, Hoshi UniversityTokyo, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of TokyoTokyo, Japan.,Center for Information and Neural Networks, National Institute of Information and Communications TechnologyOsaka, Japan
| |
Collapse
|
46
|
Ueno H, Suemitsu S, Okamoto M, Matsumoto Y, Ishihara T. Sensory experience-dependent formation of perineuronal nets and expression of Cat-315 immunoreactive components in the mouse somatosensory cortex. Neuroscience 2017; 355:161-174. [PMID: 28495333 DOI: 10.1016/j.neuroscience.2017.04.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/29/2017] [Accepted: 04/27/2017] [Indexed: 10/19/2022]
Abstract
Perineuronal nets (PNNs) are structures of extracellular matrix molecules surrounding the cell bodies and proximal dendrites of certain neurons. While PNNs are present throughout the mouse cerebral cortex, recent studies have shown that the components differ among cortical sub-regions and layers, suggesting region-specific functions. Parvalbumin-expressing interneurons (PV neurons) may be important regulators of cortical plasticity during the early "critical period" that is sensitive to sensory input. Here we examined the distribution and developmental functions of PNN components associated with PV neurons in the somatosensory cortex during the critical period. Aggrecan, brevican, neurocan, phosphacan, and tenascin-R were identified as PNN components in the mouse somatosensory cortex. High-magnification analysis revealed that some lectin Wisteria floribunda agglutinin (WFA)-reactive molecules did not co-localize with monoclonal antibody Cat-315 recognition molecules around the cell body. During postnatal development, Cat-315-positive (Cat-315+) PNNs appeared later than PNNs binding to the lectin WFA (WFA+ PNNs). These WFA+ PNNs changed from granular-like to reticular-like structures during normal cortical development, while this transition was delayed by sensory deprivation. This study indicates that the formation of reticular-like WFA+ PNNs is dependent on sensory experience in the mouse somatosensory cortex. We suggest that Cat-315+ molecules and WFA expression in PNNs are involved in the early critical period of input-dependent cortical plasticity.
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki College of Allied Health Professions, Okayama 701-0194, Japan; Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Shunsuke Suemitsu
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701-0192, Japan.
| |
Collapse
|
47
|
Okuda A, Horii-Hayashi N, Sasagawa T, Shimizu T, Shigematsu H, Iwata E, Morimoto Y, Masuda K, Koizumi M, Akahane M, Nishi M, Tanaka Y. Bone marrow stromal cell sheets may promote axonal regeneration and functional recovery with suppression of glial scar formation after spinal cord transection injury in rats. J Neurosurg Spine 2017; 26:388-395. [DOI: 10.3171/2016.8.spine16250] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE
Transplantation of bone marrow stromal cells (BMSCs) is a theoretical potential as a therapeutic strategy in the treatment of spinal cord injury (SCI). Although a scaffold is sometimes used for retaining transplanted cells in damaged tissue, it is also known to induce redundant immunoreactions during the degradation processes. In this study, the authors prepared cell sheets made of BMSCs, which are transplantable without a scaffold, and investigated their effects on axonal regeneration, glial scar formation, and functional recovery in a completely transected SCI model in rats.
METHODS
BMSC sheets were prepared from the bone marrow of female Fischer 344 rats using ascorbic acid and were cryopreserved until the day of transplantation. A gelatin sponge (GS), as a control, or BMSC sheet was transplanted into a 2-mm-sized defect of the spinal cord at the T-8 level. Axonal regeneration and glial scar formation were assessed 2 and 8 weeks after transplantation by immunohistochemical analyses using anti-Tuj1 and glial fibrillary acidic protein (GFAP) antibodies, respectively. Locomotor function was evaluated using the Basso, Beattie, and Bresnahan scale.
RESULTS
The BMSC sheets promoted axonal regeneration at 2 weeks after transplantation, but there was no significant difference in the number of Tuj1-positive axons between the sheet- and GS-transplanted groups. At 8 weeks after transplantation, Tuj1-positive axons elongated across the sheet, and their numbers were significantly greater in the sheet group than in the GS group. The areas of GFAP-positive glial scars in the sheet group were significantly reduced compared with those of the GS group at both time points. Finally, hindlimb locomotor function was ameliorated in the sheet group at 4 and 8 weeks after transplantation.
CONCLUSIONS
The results of the present study indicate that an ascorbic acid–induced BMSC sheet is effective in the treatment of SCI and enables autologous transplantation without requiring a scaffold.
Collapse
Affiliation(s)
- Akinori Okuda
- 1Department of Orthopaedic Surgery, Nara Medical University, Kashihara
- 2Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara
| | - Noriko Horii-Hayashi
- 2Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara
| | - Takayo Sasagawa
- 2Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara
| | - Takamasa Shimizu
- 1Department of Orthopaedic Surgery, Nara Medical University, Kashihara
| | - Hideki Shigematsu
- 1Department of Orthopaedic Surgery, Nara Medical University, Kashihara
| | - Eiichiro Iwata
- 1Department of Orthopaedic Surgery, Nara Medical University, Kashihara
| | - Yasuhiko Morimoto
- 1Department of Orthopaedic Surgery, Nara Medical University, Kashihara
| | - Keisuke Masuda
- 1Department of Orthopaedic Surgery, Nara Medical University, Kashihara
| | - Munehisa Koizumi
- 3Spine and Spinal Cord Surgery Center, Nara Prefecture General Medical Center; and
| | - Manabu Akahane
- 4Department of Public Health, Health Management, and Policy, Nara Medical University, Kashihara, Nara, Japan
| | - Mayumi Nishi
- 2Department of Anatomy and Cell Biology, Faculty of Medicine, Nara Medical University, Kashihara
| | - Yasuhito Tanaka
- 1Department of Orthopaedic Surgery, Nara Medical University, Kashihara
| |
Collapse
|
48
|
Stamenkovic V, Stamenkovic S, Jaworski T, Gawlak M, Jovanovic M, Jakovcevski I, Wilczynski GM, Kaczmarek L, Schachner M, Radenovic L, Andjus PR. The extracellular matrix glycoprotein tenascin-C and matrix metalloproteinases modify cerebellar structural plasticity by exposure to an enriched environment. Brain Struct Funct 2017; 222:393-415. [PMID: 27089885 DOI: 10.1007/s00429-016-1224-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 04/04/2016] [Indexed: 02/05/2023]
Abstract
The importance of the extracellular matrix (ECM) glycoprotein tenascin-C (TnC) and the ECM degrading enzymes, matrix metalloproteinases (MMPs) -2 and -9, in cerebellar histogenesis is well established. This study aimed to examine whether there is a functional relationship between these molecules in regulating structural plasticity of the lateral deep cerebellar nucleus. To this end, starting from postnatal day 21, TnC- or MMP-9-deficient mice were exposed to an enriched environment (EE). We show that 8 weeks of exposure to EE leads to reduced lectin-based staining of perineuronal nets (PNNs), reduction in the size of GABAergic and increase in the number and size of glutamatergic synaptic terminals in wild-type mice. Conversely, TnC-deficient mice showed reduced staining of PNNs compared to wild-type mice maintained under standard conditions, and exposure to EE did not further reduce, but even slightly increased PNN staining. EE did not affect the densities of the two types of synaptic terminals in TnC-deficient mice, while the size of inhibitory, but not excitatory synaptic terminals was increased. In the time frame of 4-8 weeks, MMP-9, but not MMP-2, was observed to influence PNN remodeling and cerebellar synaptic plasticity as revealed by measurement of MMP-9 activity and colocalization with PNNs and synaptic markers. These findings were supported by observations on MMP-9-deficient mice. The present study suggests that TnC contributes to the regulation of structural plasticity in the cerebellum and that interactions between TnC and MMP-9 are likely to be important for these processes to occur.
Collapse
Affiliation(s)
- Vera Stamenkovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Stefan Stamenkovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Tomasz Jaworski
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Maciej Gawlak
- Laboratory of Physiology and Pathophysiology, Center for Preclinical Research and Technology, The Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Milos Jovanovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Igor Jakovcevski
- Experimental Neurophysiology, University Hospital Cologne, 50931, Cologne, Germany
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, 53175, Bonn, Germany
| | - Grzegorz M Wilczynski
- Laboratory of Neuromorphology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, 515041, People's Republic of China
| | - Lidija Radenovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Pavle R Andjus
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia.
| |
Collapse
|
49
|
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute Northern Sydney Local Health District, St. Leonards, NSW, Australia
- Sydney Medical School, Royal North Shore Hospital, The University of Sydney, Camperdown, NSW, Australia
- School of Biomedical Engineering, The University of New South Wales, Kensington, NSW, Australia
| |
Collapse
|
50
|
Insights from extracellular matrix studies in the hypothalamus: structural variations of perineuronal nets and discovering a new perifornical area of the anterior hypothalamus. Anat Sci Int 2016; 92:18-24. [PMID: 27714583 DOI: 10.1007/s12565-016-0375-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022]
Abstract
The hypothalamus controls metabolism, stress responses, and instinctive behaviors for individual survival and species preservation. Recent studies suggest that hypothalamic neurons retain plasticity throughout adulthood, which enables these neurons to respond to various kinds of changes in environment, nutrients, and fluctuating hormones. One of the mechanisms underlying the regulation of neural plasticity is the formation of a stable extracellular matrix (ECM) structure called perineuronal nets (PNNs). PNNs are large aggregates of heterogeneous ECM molecules such as chondroitin sulfate proteoglycans (CSPGs), hyaluronan, their link proteins, and tenascin-R. PNNs surround the cell body and proximal dendrites of a subset of neurons and limit adult neural plasticity. This review describes the CSPG-based ECM, including the PNNs, with a special focus on the hypothalamus of mice. We first provide an overview of PNNs in terms of their structure, molecular components, and functions, most of which have been demonstrated by extrahypothalamic studies. Second, we show the presence or absence of PNNs within individual hypothalamic regions and then describe non-PNN-formed ECM containing CSPGs that can be observed in particular hypothalamic regions. Finally, we will introduce a newly identified mouse hypothalamic area that we named the perifornical area of the anterior hypothalamus (PeFAH), which contains a cluster of PNN-positive neurons. PeFAH neurons express enkephalin and have bidirectional connections with the lateral septum. The anterior hypothalamus and lateral septum are thought to regulate defensive behaviors; therefore, the PeFAH neurons and PNNs around them could be involved in the regulation of defensive behaviors.
Collapse
|