Skip to main content

Advertisement

Log in

Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

The extracellular matrix (ECM) plays diverse roles in several physiological and pathological conditions. In the brain, the ECM is unique both in its composition and in functions. Furthermore, almost all the cells in the central nervous system contribute to different aspects of this intricate structure. Brain ECM, enriched with proteoglycans and other small proteins, aggregate into distinct structures around neurons and oligodendrocytes. These special structures have cardinal functions in the normal functioning of the brain, such as learning, memory, and synapse regulation. In this review, we have compiled the current knowledge about the structure and function of important ECM molecules in the brain and their proteolytic remodeling by matrix metalloproteinases and other enzymes, highlighting the special structures they form. In particular, the proteoglycans in brain ECM, which are essential for several vital functions, are emphasized in detail.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Abbreviations

AMPAR:

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor

AP-1:

Activator protein 1

ASD:

Autism spectrum disorders

Bral:

Brain-specific hyaluronan-binding protein

CAM:

Cell adhesion molecule

Cbln:

Cerebellin

CNS:

Central nervous system

CREB:

cAMP-response element-binding

CRP:

Complement regulatory protein

CSPG:

Chondroitin sulfate proteoglycan

DCC:

Deleted in colorectal cancer

ECD:

Extracellular domain

ECM:

Extracellular matrix

GAG:

Glycosaminoglycan

HA:

Hyaluronic acid

HAPLN1:

Hyaluronan and proteoglycan link protein 1

HAS:

Hyaluronan synthase

HSPG:

Heparin sulfate proteoglycan

LTD:

Long-term depression

LTP:

Long-term potentiation

L-VDCC:

l-type voltage-dependent Ca2+ channels

MMP:

Matrix metalloproteinase

NMDAR:

N-methyl-d-aspartate receptor

Narp:

Neuronal activity-regulated pentraxin

NF-186:

Neurofascin-186

NGC:

Neuroglycan C

NrCAM:

Neuron–glia-related cell adhesion molecule

PNN:

Perineuronal net

PSI:

Phosphacan short isoform

PTR:

Proteoglycan tandem repeat

RPTP:

Receptor-type protein-tyrosine phosphatase

SGGL:

Sulfoglucuronyl glycolipid

SNAP-25:

Synaptosomal nerve-associated protein 25

TIMP:

Tissue inhibitor of MMPs

TNC:

Tenascin-C

TNR:

Tenascin-R

tPA:

Tissue plasminogen activator

References

  1. Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK (2016) Extracellular matrix structure. Adv Drug Deliv Rev 97:4–27. https://doi.org/10.1016/j.addr.2015.11.001

    Article  CAS  PubMed  Google Scholar 

  2. Yue B (2014) Biology of the extracellular matrix: an overview. J Glaucoma 23:S20–S23. https://doi.org/10.1097/IJG.0000000000000108

    Article  PubMed  Google Scholar 

  3. Reichardt L (1991) Extracellular matrix molecules and their receptors: functions in neural development. Annu Rev Neurosci 14:531–570. https://doi.org/10.1146/annurev.neuro.14.1.531

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Naba A, Clauser KR, Ding H et al (2016) The extracellular matrix: tools and insights for the “omics” era. Matrix Biol 49:10–24. https://doi.org/10.1016/j.matbio.2015.06.003

    Article  CAS  PubMed  Google Scholar 

  5. Mouw JK, Ou G, Weaver VM (2014) Extracellular matrix assembly: a multiscale deconstruction. Nat Rev Mol Cell Biol 15:771–785. https://doi.org/10.1038/nrm3902

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Afratis NA, Nikitovic D, Multhaupt HAB et al (2017) Syndecans—key regulators of cell signaling and biological functions. FEBS J 284:27–41. https://doi.org/10.1111/febs.13940

    Article  CAS  PubMed  Google Scholar 

  7. O’Toole EA (2001) Extracellular matrix and keratinocyte migration. Clin Exp Dermatol 26:525–530

    Article  Google Scholar 

  8. Pickup MW, Mouw JK, Weaver VM (2014) The extracellular matrix modulates the hallmarks of cancer. EMBO Rep 15(12):1243–1253. https://doi.org/10.15252/embr.201439246

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Walker C, Mojares E, del Río Hernández A (2018) Role of extracellular matrix in development and cancer progression. Int J Mol Sci 19:3028. https://doi.org/10.3390/ijms19103028

    Article  CAS  PubMed Central  Google Scholar 

  10. Afratis N, Gialeli C, Nikitovic D et al (2012) Glycosaminoglycans: key players in cancer cell biology and treatment. FEBS J 279:1177–1197. https://doi.org/10.1111/j.1742-4658.2012.08529.x

    Article  CAS  PubMed  Google Scholar 

  11. Druso JE, Fischbach C (2018) Biophysical properties of extracellular matrix: linking obesity and cancer. Trends Cancer 4:271–273. https://doi.org/10.1016/j.trecan.2018.02.001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Urciuolo F, Garziano A, Imparato G et al (2016) Biophysical properties of dermal building-blocks affect extra cellular matrix assembly in 3D endogenous macrotissue. Biofabrication 8:015010. https://doi.org/10.1088/1758-5090/8/1/015010

    Article  CAS  PubMed  Google Scholar 

  13. Koláčná L, Bakešová J, Varga F et al (2007) Biochemical and biophysical aspects of collagen nanostructure in the extracellular matrix. Physiol Res 56:51–60

    Google Scholar 

  14. Bonnans C, Chou J, Werb Z (2014) Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 15:786–801. https://doi.org/10.1038/nrm3904

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hubmacher D, Apte SS (2013) The biology of the extracellular matrix: novel insights. Curr Opin Rheumatol 25:65

    Article  CAS  Google Scholar 

  16. Wolman M (1962) Symposium on the ground substance of the central nervous system. Introduction. World Neurol 3:97

    CAS  PubMed  Google Scholar 

  17. Hess A (1953) The ground substance of the central nervous system revealed by histochemical staining. J Comp Neurol 98:69–91. https://doi.org/10.1002/cne.900980106

    Article  CAS  PubMed  Google Scholar 

  18. Lei Y, Han H, Yuan F et al (2017) The brain interstitial system: anatomy, modeling, in vivo measurement, and applications. Prog Neurobiol 157:230–246. https://doi.org/10.1016/j.pneurobio.2015.12.007

    Article  PubMed  Google Scholar 

  19. Zimmermann DR, Dours-Zimmermann MT (2008) Extracellular matrix of the central nervous system: from neglect to challenge. Histochem Cell Biol 130:635–653. https://doi.org/10.1007/s00418-008-0485-9

    Article  CAS  PubMed  Google Scholar 

  20. Cragg B (1979) Brain extracellular space fixed for electron microscopy. Neurosci Lett 15:301–306. https://doi.org/10.1016/0304-3940(79)96130-5

    Article  CAS  PubMed  Google Scholar 

  21. Rasband MN, Peles E (2016) The nodes of Ranvier: molecular assembly and maintenance. Cold Spring Harb Perspect Biol 8:1–16. https://doi.org/10.1101/cshperspect.a020495

    Article  Google Scholar 

  22. Bandtlow CE, Zimmermann DR (2000) Proteoglycans in the developing brain: new conceptual insights for old proteins. Physiol Rev 80:1267–1290. https://doi.org/10.1152/physrev.2000.80.4.1267

    Article  CAS  PubMed  Google Scholar 

  23. Hook M, Kjellen L, Johansson S, Robinson J (1984) Cell-surface glycosaminoglycans. Annu Rev Biochem 53:847–869. https://doi.org/10.1146/annurev.bi.53.070184.004215

    Article  CAS  PubMed  Google Scholar 

  24. Schwartz NB, Domowicz MS (2018) Proteoglycans in brain development and pathogenesis. FEBS Lett. https://doi.org/10.1002/1873-3468.13026

    Article  PubMed  Google Scholar 

  25. Aono S, Oohira A (2006) Chondroitin sulfate proteoglycans in the brain. Adv Pharmacol 53:323–336. https://doi.org/10.1016/S1054-3589(05)53015-1

    Article  CAS  PubMed  Google Scholar 

  26. Galtrey CM, Fawcett JW (2007) The role of chondroitin sulfate proteoglycans in regeneration and plasticity in the central nervous system. Brain Res Rev 4:1–18. https://doi.org/10.1016/j.brainresrev.2006.09.006

    Article  CAS  Google Scholar 

  27. Dyck SM, Karimi-Abdolrezaee S (2015) Chondroitin sulfate proteoglycans: key modulators in the developing and pathologic central nervous system. Exp Neurol 269:169–187. https://doi.org/10.1016/j.expneurol.2015.04.006

  28. Pantazopoulos H, Woo TUW, Lim MP et al (2010) Extracellular matrix-glial abnormalities in the amygdala and entorhinal cortex of subjects diagnosed with schizophrenia. Arch Gen Psychiatry. https://doi.org/10.1001/archgenpsychiatry.2009.196

    Article  PubMed  PubMed Central  Google Scholar 

  29. Berretta S (2012) Extracellular matrix abnormalities in schizophrenia. Neuropharmacology. https://doi.org/10.1016/j.neuropharm.2011.08.010

    Article  PubMed  Google Scholar 

  30. Bonneh-Barkay D, Wiley CA (2009) Brain extracellular matrix in neurodegeneration. Brain Pathol 19:573–585. https://doi.org/10.1111/j.1750-3639.2008.00195.x

    Article  CAS  PubMed  Google Scholar 

  31. Avram S, Shaposhnikov S, Buiu C, Mernea M (2014) Chondroitin sulfate proteoglycans: structure-function relationship with implication in neural development and brain disorders. Biomed Res Int. https://doi.org/10.1155/2014/642798

    Article  PubMed  PubMed Central  Google Scholar 

  32. Brittis PA, Canning DR, Silver J (1992) Chondroitin sulfate as a regulator of neuronal patterning in the retina. Science. https://doi.org/10.1126/science.1738848

    Article  PubMed  Google Scholar 

  33. Laabs T, Carulli D, Geller HM, Fawcett JW (2005) Chondroitin sulfate proteoglycans in neural development and regeneration. Curr Opin Neurobiol 15:116–120. https://doi.org/10.1016/j.conb.2005.01.014

    Article  CAS  PubMed  Google Scholar 

  34. Sugahara K, Mikami T (2007) Chondroitin/dermatan sulfate in the central nervous system. Curr Opin Struct Biol 17:536–545

    Article  CAS  Google Scholar 

  35. Celio MR, Spreafico R, De Biasi S, Vitellaro-Zuccarello L (1998) Perineuronal nets: past and present. Trends Neurosci 21:510–515. https://doi.org/10.1016/S0166-2236(98)01298-3

    Article  CAS  PubMed  Google Scholar 

  36. Kjellen L (2002) Proteoglycans: structures and interactions. Annu Rev Biochem. https://doi.org/10.1146/annurev.biochem.60.1.443

    Article  Google Scholar 

  37. Silbert JE, Sugumaran G (2002) Biosynthesis of chondroitin/dermatan sulfate. IUBMB life (International Union Biochem Mol Biol Life) 54:177–186. https://doi.org/10.1080/15216540214923

    Article  CAS  Google Scholar 

  38. Akita K, von Holst A, Furukawa Y et al (2008) Expression of multiple chondroitin/dermatan sulfotransferases in the neurogenic regions of the embryonic and adult central nervous system implies that complex chondroitin sulfates have a role in neural stem cell maintenance. Stem Cells 26:798–809. https://doi.org/10.1634/stemcells.2007-0448

    Article  CAS  PubMed  Google Scholar 

  39. Gama CI, Tully SE, Sotogaku N et al (2006) Sulfation patterns of glycosaminoglycans encode molecular recognition and activity. Nat Chem Biol 2:467–473. https://doi.org/10.1038/nchembio810

    Article  CAS  PubMed  Google Scholar 

  40. Sherman LS, Back SA (2008) A “GAG” reflex prevents repair of the damaged CNS. Trends Neurosci 31:44–52

    Article  CAS  Google Scholar 

  41. Beller JA, Kulengowski B, Kobraei EM et al (2013) Comparison of sensory neuron growth cone and filopodial responses to structurally diverse aggrecan variants, in vitro. Exp Neurol 247:143–157. https://doi.org/10.1016/J.EXPNEUROL.2013.02.012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Miyata S, Nishimura Y, Hayashi N, Oohira A (2005) Construction of perineuronal net-like structure by cortical neurons in culture. Neuroscience 136:95–104. https://doi.org/10.1016/j.neuroscience.2005.07.031

    Article  CAS  PubMed  Google Scholar 

  43. Maeda N (2010) Structural variation of chondroitin sulfate and its roles in the central nervous system. Cent Nerv Syst Agents Med Chem 10:22–31. https://doi.org/10.2174/187152410790780136

    Article  CAS  PubMed  Google Scholar 

  44. Yamaguchi Y (2000) Lecticans: organizers of the brain extracellular matrix. Cell Mol Life Sci 57:276–289

    Article  CAS  Google Scholar 

  45. Iozzo RV (1998) Matrix proteoglycans: from molecular design to cellular function. Annu Rev Biochem 67:609–652. https://doi.org/10.1146/annurev.biochem.67.1.609

    Article  CAS  PubMed  Google Scholar 

  46. Sandy JD, Flannery CR, Boynton RE, Neame PJ (1990) Isolation and characterization of disulfide-bonded peptides from the three globular domains of aggregating cartilage proteoglycan. J Biol Chem 265:21108–21113. https://doi.org/10.1109/CEIDP.1994.592029

    Article  CAS  PubMed  Google Scholar 

  47. Neame PJ, Christner JE, Baker JR (1987) Cartilage proteoglycan aggregates. The link protein and proteoglycan amino-terminal globular domains have similar structures. J Biol Chem 262:17768–17778

    CAS  PubMed  Google Scholar 

  48. Yamaguchi Y (2000) Lecticans: organizers of the brain extracellular matrix. Cell Mol Life Sci 57:276–289. https://doi.org/10.1007/PL00000690

    Article  CAS  PubMed  Google Scholar 

  49. Binette F, Cravens J, Kahoussi B et al (1994) Link protein is ubiquitously expressed in non-cartilaginous tissues where it enhances and stabilizes the interaction of proteoglycans with hyaluronic acid. J Biol Chem 269:19116–19122

    CAS  PubMed  Google Scholar 

  50. Faissner A (1997) The tenascin gene family in axon growth and guidance. Cell Tissue Res 290:331–341. https://doi.org/10.1007/s004410050938

    Article  CAS  PubMed  Google Scholar 

  51. Aspberg A, Binkert C, Ruoslahti E (1995) The versican C-type lectin domain recognizes the adhesion protein tenascin-R. Proc Natl Acad Sci USA 92:10590–10594

    Article  CAS  Google Scholar 

  52. Aspberg A, Miura R, Bourdoulous S et al (1997) The C-type lectin domains of lecticans, a family of aggregating chondroitin sulfate proteoglycans, bind tenascin-R by protein-protein interactions independent of carbohydrate moiety. Proc Natl Acad Sci USA 94:10116–10121. https://doi.org/10.1073/pnas.94.19.10116

    Article  CAS  PubMed  Google Scholar 

  53. Hagihara K, Miura R, Kosaki R et al (1999) Immunohistochemical evidence for the brevican-tenascin-R interaction: colocalization in perineuronal nets suggests a physiological role for the interaction in the adult rat brain. J Comp Neurol 410:256–264. https://doi.org/10.1002/(SICI)1096-9861(19990726)410:2%3c256:AID-CNE7%3e3.0.CO;2-5

    Article  CAS  PubMed  Google Scholar 

  54. Ujita M, Shinomura T, Ito K et al (1994) Expression and binding activity of the carboxyl-terminal portion of the core protein of PG-M, a large chondroitin sulfate proteoglycan. J Biol Chem 269:27603–27609

    CAS  PubMed  Google Scholar 

  55. Miura R, Aspberg A, Ethell IM et al (1999) The proteoglycan lectin domain binds sulfated cell surface glycolipids and promotes cell adhesion. J Biol Chem 274:11431–11438. https://doi.org/10.1074/jbc.274.16.11431

    Article  CAS  PubMed  Google Scholar 

  56. Jungalwala FB (1994) Expression and biological functions of sulfoglucuronyl glycolipids (SGGLs) in the nervous system—a review. Neurochem Res 19:945–957. https://doi.org/10.1007/BF00968704

    Article  CAS  PubMed  Google Scholar 

  57. Poduslo S, Miller K (1985) Levels of sulfatide synthesis distinguish oligodendroglia in different stages of maturation. Neurochem Res 10:1285–1297

    Article  CAS  Google Scholar 

  58. Grumet M, Milev P, Sakurai T et al (1994) Interactions with tenascin and differential effects on cell adhesion of neurocan and phosphacan, two major chondroitin sulfate proteoglycans of nervous tissue. J Biol Chem 269:12142–12146

    CAS  PubMed  Google Scholar 

  59. Grumet M, Flaccus A, Margolis RU (1993) Functional characterization of chondroitin sulfate proteoglycans of brain: interactions with neurons and neural cell adhesion molecules. Rockefeller University Press, New York

    Google Scholar 

  60. Rauch U, Clement A, Retzler C et al (1997) Mapping of a defined neurocan binding site to distinct domains of tenascin-C. J Biol Chem 272:26905–26912. https://doi.org/10.1074/jbc.272.43.26905

    Article  CAS  PubMed  Google Scholar 

  61. Friedlander DR, Milev P, Karthikeyan L et al (1994) The neuronal chondroitin sulfate proteoglycan neurocan binds to the neural cell adhesion molecules Ng-CAM/L1/NILE and N-CAM, and inhibits neuronal adhesion and neurite outgrowth. J Cell Biol 125:669–680. https://doi.org/10.1083/jcb.125.3.669

    Article  CAS  PubMed  Google Scholar 

  62. Milev P, Fischer D, Haring M et al (1997) The fibrinogen-like globe of tenascin-C mediates its interactions with neurocan and phosphacan/protein-tyrosine phosphatase-ζ/β. J Biol Chem 272:15501–15509. https://doi.org/10.1074/jbc.272.24.15501

    Article  CAS  PubMed  Google Scholar 

  63. Xu Y, Fisher GJ (2012) Receptor type protein tyrosine phosphatases (RPTPs)—roles in signal transduction and human disease. J. Cell Commun, Signal

    Book  Google Scholar 

  64. Fischer EH, Charbonneau H, Tonks NK (1991) Protein tyrosine phosphatases: a diverse family of intracellular and transmembrane enzymes. Science 253(80):401–406. https://doi.org/10.1126/science.1650499

    Article  CAS  PubMed  Google Scholar 

  65. Shitara K, Yamada H, Watanabe K et al (1994) Brain-specific receptor-type protein-tyrosine phosphatase RPTPβ is a chondroitin sulfate proteoglycan in vivo. J Biol Chem 269:20189–20193. https://doi.org/10.1111/bjc.12074

    Article  CAS  PubMed  Google Scholar 

  66. Krueger NX, Saito H (1992) A human transmembrane protein-tyrosine-phosphatase, PTP zeta, is expressed in brain and has an N-terminal receptor domain homologous to carbonic anhydrases. Proc Natl Acad Sci USA 89:7417–7421. https://doi.org/10.1073/pnas.89.16.7417

    Article  CAS  PubMed  Google Scholar 

  67. Maeda N, Noda M (1998) Involvement of receptor-like protein tyrosine phosphatase ζ/RPTPβ and its ligand pleiotrophin/heparin-binding growth-associated molecule (HB-GAM) in neuronal migration. J Cell Biol 142:203–216. https://doi.org/10.1083/jcb.142.1.203

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Saito H (1993) Structural diversity of eukaryotic protein tyrosine phosphatases: functional and evolutionary implications. Semin Cell Dev Biol. https://doi.org/10.1006/scel.1993.1045

    Article  Google Scholar 

  69. Milev P, Friedlander DR, Sakurai T et al (1994) Interactions of the chondroitin sulfate proteoglycan phosphacan, the extracellular domain of a receptor-type protein tyrosine phosphatase, with neurons, glia, and neural cell adhesion molecules. J Cell Biol 127:1703–1715. https://doi.org/10.1083/jcb.127.6.1703

    Article  CAS  PubMed  Google Scholar 

  70. Garwood J, Heck N, Reichardt F, Faissner A (2003) Phosphacan short isoform, a novel non-proteoglycan variant of phosphacan/receptor protein tyrosine phosphatase-β, interacts with neuronal receptors and promotes neurite outgrowth. J Biol Chem 278:24164–24173. https://doi.org/10.1074/jbc.M211721200

    Article  CAS  PubMed  Google Scholar 

  71. Peles E, Schlessinger J, Grumet M (1998) Multi-ligand interactions with receptor-like protein tyrosine phosphatase β: implications for intercellular signaling. Trends Biochem Sci 23:121–124. https://doi.org/10.1016/S0968-0004(98)01195-5

    Article  CAS  PubMed  Google Scholar 

  72. Ohyama K, Ikeda E, Kawamura K et al (2004) Receptor-like protein tyrosine phosphatase ζ/RPTP β is expressed on tangentially aligned neurons in early mouse neocortex. Dev Brain Res 148:121–127. https://doi.org/10.1016/j.devbrainres.2003.10.012

    Article  CAS  Google Scholar 

  73. Hayashi N, Miyata S, Yamada M et al (2005) Neuronal expression of the chondroitin sulfate proteoglycans receptor-type protein-tyrosine phosphatase β and phosphacan. Neuroscience 131:331–348. https://doi.org/10.1016/j.neuroscience.2004.11.017

    Article  CAS  PubMed  Google Scholar 

  74. Dobbertin A, Rhodes KE, Garwood J et al (2003) Regulation of RPTPβ/phosphacan expression and glycosaminoglycan epitopes in injured brain and cytokine-treated glia. Mol Cell Neurosci 24:951–971. https://doi.org/10.1016/S1044-7431(03)00257-4

    Article  CAS  PubMed  Google Scholar 

  75. Chow JPH, Fujikawa A, Shimizu H et al (2008) Metalloproteinase- and γ-secretase-mediated cleavage of protein-tyrosine phosphatase receptor type Z. J Biol Chem. https://doi.org/10.1074/jbc.M802976200

    Article  PubMed  PubMed Central  Google Scholar 

  76. Watanabe E, Maeda N, Matsui F et al (1995) Neuroglycan C, a novel membrane-spanning chondroitin sulfate proteoglycan that is restricted to the brain. J Biol Chem 270:26876–26882. https://doi.org/10.1074/jbc.270.45.26876

    Article  CAS  PubMed  Google Scholar 

  77. Yasuda Y, Tokita Y, Aono S et al (1998) Cloning and chromosomal mapping of the human gene of neuroglycan C (NGC), a neural transmembrane chondroitin sulfate proteoglycan with an EGF module. Neurosci Res 32:313–322. https://doi.org/10.1016/S0168-0102(98)00098-4

    Article  CAS  PubMed  Google Scholar 

  78. Aono S, Tokita Y, Yasuda Y et al (2006) Expression and identification of a new splice variant of neuroglycan C, a transmembrane chondroitin sulfate proteoglycan, in the human brain. J Neurosci Res 83:110–118. https://doi.org/10.2337/dc13-0560

    Article  CAS  PubMed  Google Scholar 

  79. Aono S, Keino H, Ono T et al (2000) Genomic organization and expression pattern of mouse neuroglycan C in the cerebellar development. J Biol Chem 275:337–342. https://doi.org/10.1074/jbc.275.1.337

    Article  CAS  PubMed  Google Scholar 

  80. Aono S, Tokita Y, Shuo T et al (2004) Glycosylation site for chondroitin sulfate on the neural part-time proteoglycan, neuroglycan C. J Biol Chem 279:46536–46541. https://doi.org/10.1074/jbc.M403263200

    Article  CAS  PubMed  Google Scholar 

  81. Kinugasa Y, Ishiguro H, Tokita Y et al (2004) Neuroglycan C, a novel member of the neuregulin family. Biochem Biophys Res Commun 321:1045–1049. https://doi.org/10.1016/j.bbrc.2004.07.066

    Article  CAS  PubMed  Google Scholar 

  82. Nakanishi K, Aono S, Hirano K et al (2006) Identification of neurite outgrowth-promoting domains of neuroglycan C, a brain-specific chondroitin sulfate proteoglycan, and involvement of phosphatidylinositol 3-kinase and protein kinase C signaling pathways in neuritogenesis. J Biol Chem. https://doi.org/10.1074/jbc.M601498200

    Article  PubMed  Google Scholar 

  83. Nörenberg U, Hubert M, Rathjen FG (1996) Structural and functional characterization of tenascin-R (restrictin), an extracellular matrix glycoprotein of glial cells and neurons. Int J Dev Neurosci 14:217–231. https://doi.org/10.1016/0736-5748(96)00009-3

    Article  PubMed  Google Scholar 

  84. Kammerer RA, Schulthess T, Landwehr R et al (1998) Tenascin-C hexabrachion assembly is a sequential two-step process initiated by coiled-coil α-helices. J Biol Chem 273:10602–10608. https://doi.org/10.1074/jbc.273.17.10602

    Article  CAS  PubMed  Google Scholar 

  85. Valcourt U, Alcaraz LB, Exposito JY et al (2015) Tenascin-X: beyond the architectural function. Cell Adhes Migr 9:154–165. https://doi.org/10.4161/19336918.2014.994893

    Article  CAS  Google Scholar 

  86. Faissner A, Kruse J, Chiquet-Ehrismann R, Mackie E (1988) The high-molecular-weight J1 glycoproteins are immunochemically related to tenascin. Differentiation 37:104–114. https://doi.org/10.1111/j.1432-0436.1988.tb00802.x

    Article  CAS  PubMed  Google Scholar 

  87. Chiquet-Ehrismann R, Tucker RP (2011) Tenascins and the importance of adhesion modulation. Cold Spring Harb Perspect Biol 3:1–19. https://doi.org/10.1101/cshperspect.a004960

    Article  CAS  Google Scholar 

  88. Tucker RP, Drabikowski K, Hess JF et al (2006) Phylogenetic analysis of the tenascin gene family: evidence of origin early in the chordate lineage. BMC Evol Biol 6:60. https://doi.org/10.1186/1471-2148-6-60

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Götz B, Scholze A, Clement A et al (1996) Tenascin-C contains distinct adhesive, anti-adhesive, and neurite outgrowth promoting sites for neurons. J Cell Biol. https://doi.org/10.1083/jcb.132.4.681

    Article  PubMed  Google Scholar 

  90. Joester A, Faissner A (2001) The structure and function of tenascins in the nervous system. Matrix Biol 20:13–22. https://doi.org/10.1016/S0945-053X(00)00136-0

    Article  CAS  PubMed  Google Scholar 

  91. Pas J, Wyszko E, Rolle K et al (2006) Analysis of structure and function of tenascin-C. Int J Biochem Cell Biol 38:1594–1602. https://doi.org/10.1016/j.biocel.2006.03.017

    Article  CAS  PubMed  Google Scholar 

  92. Lundell A, Olin AI, Mörgelin M et al (2004) Structural basis for interactions between tenascins and lectican C-type lectin domains: evidence for a crosslinking role for tenascins. Structure 12:1495–1506. https://doi.org/10.1016/j.str.2004.05.021

    Article  CAS  PubMed  Google Scholar 

  93. Brückner G, Grosche J, Schmidt S et al (2000) Postnatal development of perineuronal nets in wild-type mice and in a mutant deficient in tenascin-R. J Comp Neurol 428:616–629. https://doi.org/10.1002/1096-9861(20001225)428:4%3c616:AID-CNE3%3e3.0.CO;2-K

    Article  PubMed  Google Scholar 

  94. Testa D, Prochiantz A, Di Nardo AA (2018) Perineuronal nets in brain physiology and disease. Semin Cell Dev Biol. https://doi.org/10.1016/j.semcdb.2018.09.011

    Article  PubMed  Google Scholar 

  95. Spicer AP, Joo A, Bowling RA (2003) A hyaluronan binding link protein gene family whose members are physically linked adjacent to chrondroitin sulfate proteoglycan core protein genes. The missing links. J Biol Chem 278:21083–21091. https://doi.org/10.1074/jbc.M213100200

    Article  CAS  PubMed  Google Scholar 

  96. Oohashi T, Edamatsu M, Bekku Y, Carulli D (2015) The hyaluronan and proteoglycan link proteins: organizers of the brain extracellular matrix and key molecules for neuronal function and plasticity. Exp Neurol 274:134–144. https://doi.org/10.1016/j.expneurol.2015.09.010

    Article  CAS  PubMed  Google Scholar 

  97. Bekku Y, Su WD, Hirakawa S et al (2003) Molecular cloning of Bral2, a novel brain-specific link protein, and immunohistochemical colocalization with brevican in perineuronal nets. Mol Cell Neurosci. https://doi.org/10.1016/S1044-7431(03)00133-7

    Article  PubMed  Google Scholar 

  98. Deepa SS, Carulli D, Galtrey C et al (2006) Composition of perineuronal net extracellular matrix in rat brain: a different disaccharide composition for the net-associated proteoglycans. J Biol Chem 281:17789–17800. https://doi.org/10.1074/jbc.M600544200

    Article  CAS  PubMed  Google Scholar 

  99. Dityatev A, Rusakov DA (2011) Molecular signals of plasticity at the tetrapartite synapse. Curr Opin Neurobiol 21:353–359. https://doi.org/10.1016/j.conb.2010.12.006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Carulli D, Pizzorusso T, Kwok JCF et al (2010) Animals lacking link protein have attenuated perineuronal nets and persistent plasticity. Brain 133:2331–2347. https://doi.org/10.1093/brain/awq145

    Article  PubMed  Google Scholar 

  101. Bekku Y, Saito M, Moser M et al (2012) Bral2 is indispensable for the proper localization of brevican and the structural integrity of the perineuronal net in the brainstem and cerebellum. J Comp Neurol 520:1721–1736. https://doi.org/10.1002/cne.23009

    Article  CAS  PubMed  Google Scholar 

  102. Bekku Y, Rauch U, Ninomiya Y, Oohashi T (2009) Brevican distinctively assembles extracellular components at the large diameter nodes of Ranvier in the CNS. J Neurochem 108:1266–1276. https://doi.org/10.1111/j.1471-4159.2009.05873.x

    Article  CAS  PubMed  Google Scholar 

  103. Bekku Y, Vargova L, Goto Y et al (2010) Bral1: its role in diffusion barrier formation and conduction velocity in the CNS. J Neurosci 30:3113–3123. https://doi.org/10.1523/JNEUROSCI.5598-09.2010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Oohashi T, Hirakawa S, Bekku Y et al (2002) Bral1, a brain-specific link protein, colocalizing with the versican V2 isoform at the nodes of Ranvier in developing and adult mouse central nervous systems. Mol Cell Neurosci. https://doi.org/10.1006/mcne.2001.1061

    Article  PubMed  Google Scholar 

  105. Brückner G, Brauer K, Härtig W et al (1993) Perineuronal nets provide a polyanionic, glia-associated form of microenvironment around certain neurons in many parts of the rat brain. Glia 8:183–200. https://doi.org/10.1002/glia.440080306

    Article  PubMed  Google Scholar 

  106. Itano N, Sawai T, Yoshida M et al (1999) Three isoforms of mammalian hyaluronan synthases have distinct enzymatic properties. J Biol Chem 274:25085–25092. https://doi.org/10.1074/jbc.274.35.25085

    Article  CAS  PubMed  Google Scholar 

  107. Kwok JCF, Carulli D, Fawcett JW (2010) In vitro modeling of perineuronal nets: hyaluronan synthase and link protein are necessary for their formation and integrity. J Neurochem 114:1447–1459. https://doi.org/10.1111/j.1471-4159.2010.06878.x

    Article  CAS  PubMed  Google Scholar 

  108. Carulli D, Rhodes KE, Brown DJ et al (2006) Composition of perineuronal nets in the adult rat cerebellum and the cellular origin of their components. J Comp Neurol 494:559–577. https://doi.org/10.1002/cne.20822

    Article  CAS  PubMed  Google Scholar 

  109. Golgi C (1893) Intorno all'origine del quarto nervo cerebrale (patetico o trocleare) e di una questione di Isto - fisiologia generale che a questo argomento si ricollega. Rend Mat Acc Lincei s. 5 2(1):379–389

    Google Scholar 

  110. Golgi C (1898) Intorno alla struttura delle cellule nervose. XXV. Sulla struttura delle cellule nervose dei ganglin spinali. Boll Soc Med Chirurgica di Pavia 1:655–665

    Google Scholar 

  111. Härtig W, Brauer K, Brückner G (1992) Wisteria floribunda agglutinin-labelled nets surround parvalbumin-containing neurons. NeuroReport. https://doi.org/10.1097/00001756-199210000-00012

    Article  PubMed  Google Scholar 

  112. Giamanco KAA, Matthews RTT (2012) Deconstructing the perineuronal net: cellular contributions and molecular composition of the neuronal extracellular matrix. Neuroscience 218:367–384. https://doi.org/10.1016/j.neuroscience.2012.05.055

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Tsien RY (2013) Very long-term memories may be stored in the pattern of holes in the perineuronal net. Proc Natl Acad Sci. https://doi.org/10.1073/pnas.1310158110

    Article  PubMed  Google Scholar 

  114. Lorenzo Bozzelli P, Alaiyed S, Kim E et al (2018) Proteolytic remodeling of perineuronal nets: effects on synaptic plasticity and neuronal population dynamics. Neural Plast. https://doi.org/10.1155/2018/5735789

    Article  PubMed  PubMed Central  Google Scholar 

  115. Sorg BA, Berretta S, Blacktop JM et al (2016) Casting a wide net: role of perineuronal nets in neural plasticity. J Neurosci. https://doi.org/10.1523/JNEUROSCI.2351-16.2016

    Article  PubMed  PubMed Central  Google Scholar 

  116. Lasek AW, Chen H, Chen WY (2017) Releasing addiction memories trapped in perineuronal nets. Trends Genet 34:197–208. https://doi.org/10.1016/j.tig.2017.12.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Morishita H, Hensch TK (2008) Critical period revisited: impact on vision. Curr Opin Neurobiol 18:101–107. https://doi.org/10.1016/j.conb.2008.05.009

    Article  CAS  PubMed  Google Scholar 

  118. Lorenzo Bozzelli P, Alaiyed S, Kim E et al (2018) Proteolytic remodeling of perineuronal nets: effects on synaptic plasticity and neuronal population dynamics. Neural Plast 2018:1–13. https://doi.org/10.1155/2018/5735789

    Article  CAS  Google Scholar 

  119. Wen TH, Binder DK, Ethell IM, Razak KA (2018) The Perineuronal ‘Safety’ Net? Perineuronal net abnormalities in neurological disorders. Front Mol Neurosci. https://doi.org/10.3389/fnmol.2018.00270

    Article  PubMed  PubMed Central  Google Scholar 

  120. Giamanco KA, Morawski M, Matthews RT (2010) Perineuronal net formation and structure in aggrecan knockout mice. Neuroscience 170:1314–1327. https://doi.org/10.1016/j.neuroscience.2010.08.032

    Article  CAS  PubMed  Google Scholar 

  121. Matthews RT, Kelly GM, Zerillo CA et al (2002) Aggrecan glycoforms contribute to the molecular heterogeneity of perineuronal nets. J Neurosci 22:7536–7547

    Article  CAS  Google Scholar 

  122. Ruoslahti E (1996) Brain extracellular matrix. Glycobiology 6:489–492. https://doi.org/10.1093/glycob/6.5.489

    Article  CAS  PubMed  Google Scholar 

  123. Matsumoto K, Shionyu M, Go M et al (2003) Distinct interaction of versican/PG-M with hyaluronan and link protein. J Biol Chem 278:41205–41212. https://doi.org/10.1074/jbc.M305060200

    Article  CAS  PubMed  Google Scholar 

  124. Blundell CD, Almond A, Mahoney DJ et al (2005) Towards a structure for a TSG-6·hyaluronan complex by modeling and NMR, spectroscopy: insights into other members of the link module superfamily. J Biol Chem 280:18189–18201. https://doi.org/10.1074/jbc.M414343200

    Article  CAS  PubMed  Google Scholar 

  125. Seyfried NT, McVey GF, Almond A et al (2005) Expression and purification of functionally active hyaluronan-binding domains from human cartilage link protein, aggrecan and versican: formation of ternary complexes with defined hyaluronan oligosaccharides. J Biol Chem 280:5435–5448. https://doi.org/10.1074/jbc.M411297200

    Article  CAS  PubMed  Google Scholar 

  126. Rutka JT, Apodaca G, Stern R, Rosenblum M (1988) The extracellular matrix of the central and peripheral nervous systems: structure and function. J Neurosurg 69:155–170. https://doi.org/10.3171/jns.1988.69.2.0155

    Article  CAS  PubMed  Google Scholar 

  127. van’t Spijker HM, Kwok JCF (2017) A sweet talk: the molecular systems of perineuronal nets in controlling neuronal communication. Front Integr Neurosci. https://doi.org/10.3389/fnint.2017.00033

    Article  Google Scholar 

  128. Karetko M, Skangiel-Kramska J (2009) Diverse functions of perineuronal nets. Acta Neurobiol Exp (Wars) 69:564–577

    Google Scholar 

  129. Pantazopoulos H, Berretta S (2016) In sickness and in health: perineuronal nets and synaptic plasticity in psychiatric disorders. Neural Plast 2016:9847696

    Article  Google Scholar 

  130. Belichenko PV, Hagberg B, Dahlström A (1997) Morphological study of neocortical areas in Rett syndrome. Acta Neuropathol 93:50–61. https://doi.org/10.1007/s004010050582

    Article  CAS  PubMed  Google Scholar 

  131. Hensch TK (2005) Critical period plasticity in local cortical circuits. Nat Rev Neurosci 6:877–888

    Article  CAS  Google Scholar 

  132. Sorg BA, Berretta S, Blacktop JM et al (2016) Casting a wide net: role of perineuronal nets in neural plasticity. J Neurosci 36:11459–11468. https://doi.org/10.1523/JNEUROSCI.2351-16.2016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Lensjø KK, Lepperød ME, Dick G et al (2017) Removal of perineuronal nets unlocks juvenile plasticity through network mechanisms of decreased inhibition and increased gamma activity. J Neurosci 37:1269–1283. https://doi.org/10.1523/JNEUROSCI.2504-16.2016

    Article  PubMed  PubMed Central  Google Scholar 

  134. van’t Spijker HM, Kwok JCF (2017) A sweet talk: the molecular systems of perineuronal nets in controlling neuronal communication. Front Integr Neurosci 11:33. https://doi.org/10.3389/fnint.2017.00033

    Article  CAS  Google Scholar 

  135. Frischknecht R, Heine M, Perrais D et al (2009) Brain extracellular matrix affects AMPA receptor lateral mobility and short-term synaptic plasticity. Nat Neurosci 12:897–904. https://doi.org/10.1038/nn.2338

    Article  CAS  PubMed  Google Scholar 

  136. Deepa SS, Umehara Y, Higashiyama S et al (2002) Specific molecular interactions of oversulfated chondroitin sulfate E with various heparin-binding growth factors: implications as a physiological binding partner in the brain and other tissues. J Biol Chem 277:43707–43716. https://doi.org/10.1074/jbc.M207105200

    Article  CAS  PubMed  Google Scholar 

  137. Caroni P, Donato F, Muller D (2012) Structural plasticity upon learning: regulation and functions. Nat Rev Neurosci 13:478–490

    Article  CAS  Google Scholar 

  138. Tsien RY (2013) Very long-term memories may be stored in the pattern of holes in the perineuronal net. Proc Natl Acad Sci 110:12456–12461. https://doi.org/10.1073/pnas.1310158110

    Article  PubMed  Google Scholar 

  139. Cohen LD, Zuchman R, Sorokina O et al (2013) Metabolic turnover of synaptic proteins: kinetics, interdependencies and implications for synaptic maintenance. PLoS One 8:e63191. https://doi.org/10.1371/journal.pone.0063191

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Happel MFK, Niekisch H, Castiblanco Rivera LL et al (2014) Enhanced cognitive flexibility in reversal learning induced by removal of the extracellular matrix in auditory cortex. Proc Natl Acad Sci 111:2800–2805. https://doi.org/10.1073/pnas.1310272111

    Article  CAS  PubMed  Google Scholar 

  141. Bradshaw KP, Figueroa Velez DX, Habeeb M, Gandhi SP (2018) Precocious deposition of perineuronal nets on Parvalbumin inhibitory neurons transplanted into adult visual cortex. Sci Rep 8:7480. https://doi.org/10.1038/s41598-018-25735-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Wen TH, Afroz S, Reinhard SM et al (2017) Genetic reduction of matrix metalloproteinase-9 promotes formation of perineuronal nets around parvalbumin-expressing interneurons and normalizes auditory cortex responses in developing fmr1 knock-out mice. Cereb Cortex. https://doi.org/10.1093/cercor/bhx258

    Article  Google Scholar 

  143. Cabungcal J-H, Steullet P, Morishita H et al (2013) Perineuronal nets protect fast-spiking interneurons against oxidative stress. Proc Natl Acad Sci 110:9130–9135. https://doi.org/10.1073/pnas.1300454110

    Article  PubMed  Google Scholar 

  144. Balmer TS (2016) Perineuronal nets enhance the excitability of fast-spiking neurons. eNeuro. https://doi.org/10.1523/ENEURO.0112-16.2016

    Article  PubMed  PubMed Central  Google Scholar 

  145. Rossier J, Bernard A, Cabungcal JH et al (2015) Cortical fast-spiking parvalbumin interneurons enwrapped in the perineuronal net express the metallopeptidases Adamts8, Adamts15 and Neprilysin. Mol Psychiatry 20:154–161. https://doi.org/10.1038/mp.2014.162

    Article  CAS  PubMed  Google Scholar 

  146. Dupret D, Pleydell-Bouverie B, Csicsvari J (2008) Inhibitory interneurons and network oscillations. Proc Natl Acad Sci 105:18079–18080. https://doi.org/10.1073/pnas.0810064105

    Article  PubMed  Google Scholar 

  147. Engel AK, Fries P, Singer W (2001) Dynamic predictions: oscillations and synchrony in top–down processing. Nat Rev Neurosci 2:704–716. https://doi.org/10.1038/35094565

    Article  CAS  PubMed  Google Scholar 

  148. Howard MW, Rizzuto DS, Caplan JB et al (2003) Gamma oscillations correlate with working memory load in humans. Cereb Cortex 13:1369–1374

    Article  Google Scholar 

  149. Sun ZY, Bozzelli PL, Caccavano A et al (2018) Disruption of perineuronal nets increases the frequency of sharp wave ripple events. Hippocampus 28:42–52. https://doi.org/10.1002/hipo.22804

    Article  CAS  PubMed  Google Scholar 

  150. Schüppel K, Brauer K, Härtig W et al (2002) Perineuronal nets of extracellular matrix around hippocampal interneurons resist destruction by activated microglia in trimethyltin-treated rats. Brain Res 958:448–453. https://doi.org/10.1016/S0006-8993(02)03569-2

    Article  PubMed  Google Scholar 

  151. Ferrer-Ferrer M, Dityatev A (2018) Shaping synapses by the neural extracellular matrix. Front Neuroanat 12:40. https://doi.org/10.3389/fnana.2018.00040

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Eroglu C (2009) The role of astrocyte-secreted matricellular proteins in central nervous system development and function. J Cell Commun Signal 3:167–176

    Article  Google Scholar 

  153. Christopherson KS, Ullian EM, Stokes CCA et al (2005) Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell 120:421–433. https://doi.org/10.1016/j.cell.2004.12.020

    Article  CAS  PubMed  Google Scholar 

  154. Xu J, Xiao N, Xia J (2010) Thrombospondin 1 accelerates synaptogenesis in hippocampal neurons through neuroligin 1. Nat Neurosci 13:22–24. https://doi.org/10.1038/nn.2459

    Article  CAS  PubMed  Google Scholar 

  155. Benton RL, Maddie MA, Worth CA et al (2008) Transcriptomic screening of microvascular endothelial cells implicates novel molecular regulators of vascular dysfunction after spinal cord injury. J Cereb Blood Flow Metab 28:1771–1785. https://doi.org/10.1038/jcbfm.2008.76

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Lin T-N, Kim G-M, Chen J-J et al (2003) Differential regulation of thrombospondin-1 and thrombospondin-2 after focal cerebral ischemia/reperfusion. Stroke 34:177–186. https://doi.org/10.1161/01.STR.0000047100.84604.BA

    Article  CAS  PubMed  Google Scholar 

  157. Liauw J, Hoang S, Choi M et al (2008) Thrombospondins 1 and 2 are necessary for synaptic plasticity and functional recovery after stroke. J Cereb Blood Flow Metab 28:1722–1732. https://doi.org/10.1038/jcbfm.2008.65

    Article  CAS  PubMed  Google Scholar 

  158. Eroglu C, Allen NJ, Susman MW et al (2009) Gabapentin receptor alpha2delta-1 is a neuronal thrombospondin receptor responsible for excitatory CNS synaptogenesis. Cell 139:380–392. https://doi.org/10.1016/j.cell.2009.09.025

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Favuzzi E, Marques-Smith A, Deogracias R et al (2017) Activity-dependent gating of parvalbumin interneuron function by the perineuronal net protein brevican. Neuron 95:639–655. https://doi.org/10.1016/J.NEURON.2017.06.028

    Article  CAS  PubMed  Google Scholar 

  160. Brakebusch C, Seidenbecher CI, Asztely F et al (2002) Brevican-deficient mice display impaired hippocampal CA1 long-term potentiation but show no obvious deficits in learning and memory. Mol Cell Biol 22:7417–7427. https://doi.org/10.1128/mcb.22.21.7417-7427.2002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Chang MC, Park JM, Pelkey KA et al (2010) Narp regulates homeostatic scaling of excitatory synapses on Parvalbumin interneurons. Nat Neurosci 13:1090–1097. https://doi.org/10.1038/nn.2621.Narp

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Pelkey KA, Barksdale E, Craig MT et al (2015) Pentraxins coordinate excitatory synapse maturation and circuit integration of parvalbumin interneurons. Neuron 85:1257–1272. https://doi.org/10.1016/j.neuron.2015.02.020

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Xiao MF, Xu D, Craig MT et al (2017) NPTX2 and cognitive dysfunction in Alzheimer’s Disease. Elife 6:1–27

    CAS  Google Scholar 

  164. Baudouin S, Scheiffele P (2010) SnapShot: neuroligin-neurexin complexes. Cell 141:908

    Article  Google Scholar 

  165. Kucukdereli H, Allen NJ, Lee AT et al (2011) Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc Natl Acad Sci 108:E440–E449. https://doi.org/10.1073/pnas.1104977108

    Article  PubMed  Google Scholar 

  166. Wei P, Pattarini R, Rong Y et al (2012) The Cbln family of proteins interact with multiple signaling pathways. J Neurochem 121:717–729. https://doi.org/10.1111/j.1471-4159.2012.07648.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Haddick PCG, Tom I, Luis E et al (2014) Defining the ligand specificity of the deleted in colorectal cancer (DCC) receptor. PLoS One 9:e84823. https://doi.org/10.1371/journal.pone.0084823

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Rong Y, Wei P, Parris J et al (2012) Comparison of Cbln1 and Cbln2 functions using transgenic and knockout mice. J Neurochem 120:528–540. https://doi.org/10.1111/j.1471-4159.2011.07604.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Hirai H, Pang Z, Bao D et al (2005) Cbln1 is essential for synaptic integrity and plasticity in the cerebellum. Nat Neurosci 8:1534–1541. https://doi.org/10.1038/nn1576

    Article  CAS  PubMed  Google Scholar 

  170. Kusnoor SV, Parris J, Muly EC et al (2010) Extracerebellar role for cerebellin 1: modulation of dendritic spine density and synapses in striatal medium spiny neurons. J Comp Neurol 518:2525–2537. https://doi.org/10.1002/cne.22350

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Knight D, Xie W, Boulianne GL (2011) Neurexins and neuroligins: recent insights from invertebrates. Mol Neurobiol 44:426. https://doi.org/10.1007/S12035-011-8213-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Bock HH, May P (2016) Canonical and non-canonical Reelin signaling. Front Cell Neurosci 10:166. https://doi.org/10.3389/fncel.2016.00166

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Leemhuis J, Bouché E, Frotscher M et al (2010) Reelin signals through apolipoprotein E receptor 2 and Cdc42 to increase growth cone motility and filopodia formation. J Neurosci 30:14759–14772. https://doi.org/10.1523/JNEUROSCI.4036-10.2010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Lee GH, D’Arcangelo G (2016) New insights into Reelin-mediated signaling pathways. Front Cell Neurosci 10:122. https://doi.org/10.3389/fncel.2016.00122

    Article  PubMed  PubMed Central  Google Scholar 

  175. Niu S, Renfro A, Quattrocchi CC et al (2004) Reelin promotes hippocampal dendrite development through the VLDLR/ApoER2-Dab1 pathway. Neuron 41:71–84. https://doi.org/10.1016/S0896-6273(03)00819-5

    Article  CAS  PubMed  Google Scholar 

  176. Niu S, Yabut O, D’Arcangelo G (2008) The Reelin signaling pathway promotes dendritic spine development in hippocampal neurons. J Neurosci 28:10339–10348. https://doi.org/10.1523/JNEUROSCI.1917-08.2008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Bosch C, Masachs N, Exposito-Alonso D et al (2016) Reelin regulates the maturation of dendritic spines, synaptogenesis and glial ensheathment of newborn granule cells. Cereb Cortex 26:4282–4298. https://doi.org/10.1093/cercor/bhw216

    Article  PubMed  PubMed Central  Google Scholar 

  178. Lane-Donovan C, Philips GT, Herz J (2014) More than cholesterol transporters: lipoprotein receptors in CNS function and neurodegeneration. Neuron 83:771–787. https://doi.org/10.1016/J.NEURON.2014.08.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Weeber EJ, Beffert U, Jones C et al (2002) Reelin and apoE receptors cooperate to enhance hippocampal synaptic plasticity and learning. J Biol Chem 277:39944–39952. https://doi.org/10.1074/jbc.M205147200

    Article  CAS  PubMed  Google Scholar 

  180. Chen Y, Beffert U, Ertunc M et al (2005) Reelin modulates NMDA receptor activity in cortical neurons. J Neurosci 25:8209–8216. https://doi.org/10.1523/JNEUROSCI.1951-05.2005

    Article  CAS  PubMed  Google Scholar 

  181. Campo CG, Sinagra M, Verrier D et al (2009) Reelin secreted by GABAergic neurons regulates glutamate receptor homeostasis. PLoS One 4:e5505. https://doi.org/10.1371/journal.pone.0005505

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Qiu S, Zhao LF, Korwek KM, Weeber EJ (2006) Differential reelin-induced enhancement of NMDA and AMPA receptor activity in the adult hippocampus. J Neurosci 26:12943–12955. https://doi.org/10.1523/JNEUROSCI.2561-06.2006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Knuesel I (2010) Reelin-mediated signaling in neuropsychiatric and neurodegenerative diseases. Prog Neurobiol 91:257–274

    Article  CAS  Google Scholar 

  184. Folsom TD, Fatemi SH (2013) The involvement of Reelin in neurodevelopmental disorders. Neuropharmacology 68:122–135

    Article  CAS  Google Scholar 

  185. Oohashi T, Edamatsu M, Bekku Y, Carulli D (2015) The hyaluronan and proteoglycan link proteins: organizers of the brain extracellular matrix and key molecules for neuronal function and plasticity. Exp Neurol 274:134–144. https://doi.org/10.1016/j.expneurol.2015.09.010

    Article  CAS  PubMed  Google Scholar 

  186. Vecino E, Kwok JCF (2016) The extracellular matrix in the nervous system: the good and the bad aspects. In: Travascio F (ed) Composition and function of the extracellular matrix in the human body. InTech

  187. Bukalo O, Schachner M, Dityatev A (2001) Modification of extracellular matrix by enzymatic removal of chondroitin sulfate and by lack of tenascin-R differentially affects several forms of synaptic plasticity in the hippocampus. Neuroscience 104:359–369. https://doi.org/10.1016/S0306-4522(01)00082-3

    Article  CAS  PubMed  Google Scholar 

  188. Saghatelyan AK, Dityatev A, Schmidt S et al (2001) Reduced perisomatic inhibition, increased excitatory transmission, and impaired long-term potentiation in mice deficient for the extracellular matrix glycoprotein tenascin-R. Mol Cell Neurosci 17:226–240. https://doi.org/10.1006/MCNE.2000.0922

    Article  CAS  PubMed  Google Scholar 

  189. Freitag S, Schachner M, Morellini F (2003) Behavioral alterations in mice deficient for the extracellular matrix glycoprotein tenascin-R. Behav Brain Res 145:189–207. https://doi.org/10.1016/S0166-4328(03)00109-8

    Article  CAS  PubMed  Google Scholar 

  190. Gurevicius K, Gureviciene I, Valjakka A et al (2004) Enhanced cortical and hippocampal neuronal excitability in mice deficient in the extracellular matrix glycoprotein tenascin-R. Mol Cell Neurosci 25:515–523. https://doi.org/10.1016/J.MCN.2003.12.001

    Article  CAS  PubMed  Google Scholar 

  191. Nikonenko A, Schmidt S, Skibo G et al (2003) Tenascin-R-deficient mice show structural alterations of symmetric perisomatic synapses in the CA1 region of the hippocampus. J Comp Neurol 456:338–349. https://doi.org/10.1002/cne.10537

    Article  CAS  PubMed  Google Scholar 

  192. Ferhat L, Chevassus Au Louis N, Jorquera I et al (1996) Transient increase of tenascin-C in immature hippocampus: Astroglial and neuronal expression. J Neurocytol 25:53–66. https://doi.org/10.1007/BF02284785

    Article  CAS  PubMed  Google Scholar 

  193. Nakic M, Manahan-Vaughan D, Reymann KG, Schachner M (1998) Long-term potentiation in vivo increases rat hippocampal tenascin-C expression. J Neurobiol 37:393–404. https://doi.org/10.1002/(SICI)1097-4695(19981115)37:3%3c393:AID-NEU5%3e3.0.CO;2-9

    Article  CAS  PubMed  Google Scholar 

  194. Morellini F, Malyshev A, Volgushev M et al (2017) Impaired fear extinction due to a deficit in Ca2+ influx through L-type voltage-gated Ca2+ channels in mice deficient for tenascin-C. Front Integr Neurosci 11:16. https://doi.org/10.3389/fnint.2017.00016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Schweitzer B, Singh J, Fejtova A et al (2017) Hyaluronic acid based extracellular matrix regulates surface expression of GluN2B containing NMDA receptors. Sci Rep 7:10991. https://doi.org/10.1038/s41598-017-07003-3

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Kochlamazashvili G, Henneberger C, Bukalo O et al (2010) The extracellular matrix molecule hyaluronic acid regulates hippocampal synaptic plasticity by modulating postsynaptic L-type Ca2+ channels. Neuron 67:116–128. https://doi.org/10.1016/j.neuron.2010.05.030

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Arancibia-Carcamo IL, Attwell D (2014) The node of Ranvier in CNS pathology. Acta Neuropathol 128:161–175

    Article  Google Scholar 

  198. Susuki K, Chang K-J, Zollinger DR et al (2013) Three mechanisms assemble central nervous system nodes of Ranvier. Neuron 78:469–482. https://doi.org/10.1016/j.neuron.2013.03.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Susuki K, Rasband MN (2008) Molecular mechanisms of node of Ranvier formation. Curr Opin Cell Biol 20:616–623. https://doi.org/10.1016/j.ceb.2008.09.007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Faivre-Sarrailh C, Devaux JJ (2013) Neuro-glial interactions at the nodes of Ranvier: implication in health and diseases. Front Cell Neurosci. https://doi.org/10.3389/fncel.2013.00196

    Article  PubMed  PubMed Central  Google Scholar 

  201. Oohashi T, Hirakawa S, Bekku Y et al (2002) Bral1, a brain-specific link protein, colocalizing with the versican V2 isoform at the nodes of Ranvier in developing and adult mouse central nervous systems. Mol Cell Neurosci 19:43–57. https://doi.org/10.1006/mcne.2001.1061

    Article  CAS  PubMed  Google Scholar 

  202. Fields RD (2015) A new mechanism of nervous system plasticity: activity-dependent myelination. Nat Rev Neurosci 16:756–767

    Article  CAS  Google Scholar 

  203. Löffek S, Schilling O, Franzke C-W (2011) Biological role of matrix metalloproteinases: a critical balance. Eur Respir J 38:191–208. https://doi.org/10.1183/09031936.00146510

    Article  CAS  PubMed  Google Scholar 

  204. Lu P, Takai K, Weaver VM, Werb Z (2011) Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol 3:a005058. https://doi.org/10.1101/cshperspect.a005058

    Article  PubMed  PubMed Central  Google Scholar 

  205. Nagy V (2006) Matrix metalloproteinase-9 is required for hippocampal late-phase long-term potentiation and memory. J Neurosci 26:1923–1934. https://doi.org/10.1523/JNEUROSCI.4359-05.2006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Tsilibary E, Tzinia A, Radenovic L et al (2014) Neural ECM proteases in learning and synaptic plasticity, 1st edn. Elsevier B.V, Amsterdam

    Google Scholar 

  207. Bilousova TV, Dansie L, Ngo M et al (2008) Minocycline promotes dendritic spine maturation and improves behavioural performance in the fragile X mouse model. J Med Genet 46:94–102. https://doi.org/10.1136/jmg.2008.061796

    Article  CAS  PubMed  Google Scholar 

  208. Michaluk P, Wawrzyniak M, Alot P et al (2011) Influence of matrix metalloproteinase MMP-9 on dendritic spine morphology. J Cell Sci 124:3369–3380. https://doi.org/10.1242/jcs.090852

    Article  CAS  PubMed  Google Scholar 

  209. Szepesi Z, Hosy E, Ruszczycki B et al (2014) Synaptically released matrix metalloproteinase activity in control of structural plasticity and the cell surface distribution of GluA1-AMPA receptors. PLoS One 9:e98274. https://doi.org/10.1371/journal.pone.0098274

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Wang X, Bozdagi O, Nikitczuk JS et al (2008) Extracellular proteolysis by matrix metalloproteinase-9 drives dendritic spine enlargement and long-term potentiation coordinately. Proc Natl Acad Sci 105:19520–19525. https://doi.org/10.1073/pnas.0807248105

    Article  PubMed  Google Scholar 

  211. Okulski P, Jay TM, Jaworski J et al (2007) TIMP-1 abolishes MMP-9-dependent long-lasting long-term potentiation in the prefrontal cortex. Biol Psychiatry 62:359–362. https://doi.org/10.1016/j.biopsych.2006.09.012

    Article  CAS  PubMed  Google Scholar 

  212. Niedringhaus M, Chen X, Dzakpasu R, Conant K (2012) MMPs and soluble ICAM-5 increase neuronal excitability within in vitro networks of hippocampal neurons. PLoS One 7:e42631. https://doi.org/10.1371/journal.pone.0042631

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Szepesi Z, Bijata M, Ruszczycki B et al (2013) Matrix metalloproteinases regulate the formation of dendritic spine head protrusions during chemically induced long-term potentiation. PLoS One 8:e63314. https://doi.org/10.1371/journal.pone.0063314

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Lonskaya I, Partridge J, Lalchandani RR et al (2013) Soluble ICAM-5, a product of activity dependent proteolysis, increases mEPSC frequency and dendritic expression of GluA1. PLoS One 8:e69136. https://doi.org/10.1371/journal.pone.0069136

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Nedivi E, Hevroni D, Naot D et al (1993) Numerous candidate plasticity-related genes revealed by differential cDNA cloning. Nature 363:718–722. https://doi.org/10.1038/363718a0

    Article  CAS  PubMed  Google Scholar 

  216. Cao W, Duan J, Wang X et al (2014) Early enriched environment induces an increased conversion of proBDNF to BDNF in the adult rat’s hippocampus. Behav Brain Res 265:76–83. https://doi.org/10.1016/j.bbr.2014.02.022

    Article  CAS  PubMed  Google Scholar 

  217. Bayat M, Sharifi MD, Haghani M, Shabani M (2015) Enriched environment improves synaptic plasticity and cognitive deficiency in chronic cerebral hypoperfused rats. Brain Res Bull 119:34–40. https://doi.org/10.1016/j.brainresbull.2015.10.001

    Article  PubMed  Google Scholar 

  218. Rivera S, Tremblay E, Timsit S et al (1997) Tissue inhibitor of metalloproteinases-1 (TIMP-1) is differentially induced in neurons and astrocytes after seizures: evidence for developmental, immediate early gene, and lesion response. J Neurosci 17:4223–4235

    Article  CAS  Google Scholar 

  219. Jaworski J, Biedermann IW, Lapinska J et al (1999) Neuronal excitation-driven and AP-1-dependent activation of tissue inhibitor of metalloproteinases-1 gene expression in rodent hippocampus. J Biol Chem 274:28106–28112. https://doi.org/10.1074/jbc.274.40.28106

    Article  CAS  PubMed  Google Scholar 

  220. Knapska E, Lioudyno V, Kiryk A et al (2013) Reward learning requires activity of matrix metalloproteinase-9 in the central amygdala. J Neurosci 33:14591–14600. https://doi.org/10.1523/JNEUROSCI.5239-12.2013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Brown TE, Forquer MR, Cocking DL et al (2007) Role of matrix metalloproteinases in the acquisition and reconsolidation of cocaine-induced conditioned place preference. Learn Mem 14:214–223. https://doi.org/10.1101/lm.476207

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Wilczynski GM, Konopacki FA, Wilczek E et al (2008) Important role of matrix metalloproteinase 9 in epileptogenesis. J Cell Biol 180:1021–1035. https://doi.org/10.1083/jcb.200708213

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Meighan SE, Meighan PC, Choudhury P et al (2006) Effects of extracellular matrix-degrading proteases matrix metalloproteinases 3 and 9 on spatial learning and synaptic plasticity. J Neurochem 96:1227–1241. https://doi.org/10.1111/j.1471-4159.2005.03565.x

    Article  CAS  PubMed  Google Scholar 

  224. Kaliszewska A, Bijata M, Kaczmarek L, Kossut M (2012) Experience-dependent plasticity of the barrel cortex in mice observed with 2-DG brain mapping and c-Fos: effects of MMP-9 KO. Cereb Cortex 22:2160–2170. https://doi.org/10.1093/cercor/bhr303

    Article  PubMed  Google Scholar 

  225. Spolidoro M, Putignano E, Munaf C et al (2012) Inhibition of matrix metalloproteinases prevents the potentiation of nondeprived-eye responses after monocular deprivation in juvenile rats. Cereb Cortex 22:725–734. https://doi.org/10.1093/cercor/bhr158

    Article  CAS  PubMed  Google Scholar 

  226. Gorkiewicz T, Balcerzyk M, Kaczmarek L, Knapska E (2015) Matrix metalloproteinase 9 (MMP-9) is indispensable for long term potentiation in the central and basal but not in the lateral nucleus of the amygdala. Front Cell Neurosci 9:73. https://doi.org/10.3389/fncel.2015.00073

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Kaczmarek L, Lapinska-Dzwonek J, Szymczak S (2002) Matrix metalloproteinases in the adult brain physiology: a link between c-Fos, AP-1 and remodeling of neuronal connections? EMBO J 21:6643–6648

    Article  CAS  Google Scholar 

  228. Ganguly K, Rejmak E, Mikosz M et al (2013) Matrix metalloproteinase (MMP) 9 transcription in mouse brain induced by fear learning. J Biol Chem 288:20978–20991. https://doi.org/10.1074/jbc.M113.457903

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Dziembowska M, Milek J, Janusz A et al (2012) Activity-dependent local translation of matrix metalloproteinase-9. J Neurosci 32:14538–14547. https://doi.org/10.1523/JNEUROSCI.6028-11.2012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Janusz A, Milek J, Perycz M et al (2013) The fragile X mental retardation protein regulates matrix metalloproteinase 9 mRNA at synapses. J Neurosci 33:18234–18241. https://doi.org/10.1523/JNEUROSCI.2207-13.2013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Nagaoka A, Takehara H, Hayashi-Takagi A et al (2016) Abnormal intrinsic dynamics of dendritic spines in a fragile X syndrome mouse model in vivo. Sci Rep 6:26651. https://doi.org/10.1038/srep26651

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Gantois I, Khoutorsky A, Popic J et al (2017) Metformin ameliorates core deficits in a mouse model of fragile X syndrome. Nat Med 23:674–677. https://doi.org/10.1038/nm.4335

    Article  CAS  PubMed  Google Scholar 

  233. Conant K, Wang Y, Szklarczyk A et al (2010) Matrix metalloproteinase-dependent shedding of intercellular adhesion molecule-5 occurs with long-term potentiation. Neuroscience 166:508–521. https://doi.org/10.1016/j.neuroscience.2009.12.061

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Michaluk P, Mikasova L, Groc L et al (2009) Matrix metalloproteinase-9 controls NMDA receptor surface diffusion through integrin _1 signaling. J Neurosci 29:6007–6012. https://doi.org/10.1523/JNEUROSCI.5346-08.2009

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Michaluk P, Kaczmarek L (2007) Matrix metalloproteinase-9 in glutamate-dependent adult brain function and dysfunction. Cell Death Differ 14:1255–1258

    Article  CAS  Google Scholar 

  236. Tian L, Stefanidakis M, Ning L et al (2007) Activation of NMDA receptors promotes dendritic spine development through MMP-mediated ICAM-5 cleavage. J Cell Biol 178:687–700. https://doi.org/10.1083/jcb.200612097

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Lu B, Nagappan G, Lu Y (2015) BDNF and synaptic plasticity, cognitive function, and dysfunction. Handb Exp Pharmacol 220:223–250. https://doi.org/10.1007/978-3-642-45106-5_9

    Article  CAS  Google Scholar 

  238. Gawlak M, Górkiewicz T, Gorlewicz A et al (2009) High resolution in situ zymography reveals matrix metalloproteinase activity at glutamatergic synapses. Neuroscience 158:167–176. https://doi.org/10.1016/j.neuroscience.2008.05.045

    Article  CAS  PubMed  Google Scholar 

  239. Konopacki FA, Rylski M, Wilczek E et al (2007) Synaptic localization of seizure-induced matrix metalloproteinase-9 mRNA. Neuroscience 150:31–39. https://doi.org/10.1016/j.neuroscience.2007.08.026

    Article  CAS  PubMed  Google Scholar 

  240. Sbai O, Ferhat L, Bernard A et al (2008) Vesicular trafficking and secretion of matrix metalloproteinases-2, -9 and tissue inhibitor of metalloproteinases-1 in neuronal cells. Mol Cell Neurosci 39:549–568. https://doi.org/10.1016/J.MCN.2008.08.004

    Article  CAS  PubMed  Google Scholar 

  241. Ogata Y, Enghild JJ, Nagase H (1992) Matrix metalloproteinase 3 (stromelysin) activates the precursor for the human matrix metalloproteinase 9. J Biol Chem 267:3581–3584. https://doi.org/10.1108/20423891211271728

    Article  CAS  PubMed  Google Scholar 

  242. Ethell IM, Ethell DW (2007) Matrix metalloproteinases in brain development and remodeling: synaptic functions and targets. J Neurosci Res 85:2813–2823. https://doi.org/10.1002/jnr

    Article  CAS  PubMed  Google Scholar 

  243. Overall CM (2002) Molecular determinants of metalloproteinase substrate specificity: matrix metalloproteinase substrate binding domains, modules, and exosites. Mol Biotechnol 22:051–086. https://doi.org/10.1385/MB:22:1:051

    Article  CAS  Google Scholar 

  244. Cua RC, Lau LW, Keough MB et al (2013) Overcoming neurite-inhibitory chondroitin sulfate proteoglycans in the astrocyte matrix. Glia 61:972–984. https://doi.org/10.1002/glia.22489

    Article  PubMed  Google Scholar 

  245. Yuan W, Matthews RT, Sandy JD, Gottschall PE (2002) Association between protease-specific proteolytic cleavage of brevican and synaptic loss in the dentate gyrus of kainate-treated rats. Neuroscience 114:1091–1101. https://doi.org/10.1016/S0306-4522(02)00347-0

    Article  CAS  PubMed  Google Scholar 

  246. Tauchi R, Imagama S, Natori T et al (2012) The endogenous proteoglycan-degrading enzyme ADAMTS-4 promotes functional recovery after spinal cord injury. J Neuroinflammation 9:53. https://doi.org/10.1186/1742-2094-9-53

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Hamel MG, Ajmo JM, Leonardo CC et al (2008) Multimodal signaling by the ADAMTSs (a disintegrin and metalloproteinase with thrombospondin motifs) promotes neurite extension. Exp Neurol 210:428–440. https://doi.org/10.1016/j.expneurol.2007.11.014

    Article  CAS  PubMed  Google Scholar 

  248. Howell MD, Torres-Collado AX, Iruela-Arispe ML, Gottschall PE (2012) Selective decline of synaptic protein levels in the frontal cortex of cemale mice deficient in the extracellular metalloproteinase ADAMTS1. PLoS One 7:e47226. https://doi.org/10.1371/journal.pone.0047226

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Calabresi P, Napolitano M, Centonze D et al (2000) Tissue plasminogen activator controls multiple forms of synaptic plasticity and memory. Eur J Neurosci 12:1002–1012. https://doi.org/10.1046/j.1460-9568.2000.00991.x

    Article  CAS  PubMed  Google Scholar 

  250. Huang YY, Bach ME, Lipp HP et al (1996) Mice lacking the gene encoding tissue-type plasminogen activator show a selective interference with late-phase long-term potentiation in both Schaffer collateral and mossy fiber pathways. Proc Natl Acad Sci 93:8699–8704. https://doi.org/10.1073/pnas.93.16.8699

    Article  CAS  PubMed  Google Scholar 

  251. Samson AL, Medcalf RL (2006) Tissue-type plasminogen activator: a multifaceted modulator of neurotransmission and synaptic plasticity. Neuron 50:673–678. https://doi.org/10.1016/j.neuron.2006.04.013

    Article  CAS  PubMed  Google Scholar 

  252. Qian Z, Gilbert ME, Colicos MA et al (1993) Tissue-plasminogen activator is induced as an immediate-early gene during seizure, kindling and long-term potentiation. Nature 361:453–457. https://doi.org/10.1038/361453a0

    Article  CAS  PubMed  Google Scholar 

  253. Norris EH, Strickland S (2007) Modulation of NR2B-regulated contextual fear in the hippocampus by the tissue plasminogen activator system. Proc Natl Acad Sci 104:13473–13478. https://doi.org/10.1002/asna.18440211403

    Article  CAS  PubMed  Google Scholar 

  254. Pawlak R, Nagai N, Urano T et al (2002) Rapid, specific and active site-catalyzed effect of tissue-plasminogen activator on hippocampus-dependent learning in mice. Neuroscience 113:995–1001. https://doi.org/10.1016/S0306-4522(02)00166-5

    Article  CAS  PubMed  Google Scholar 

  255. Seeds NW, Basham ME, Haffke SP (1999) Neuronal migration is retarded in mice lacking the tissue plasminogen activator gene. Proc Natl Acad Sci 96:14118–14123

    Article  CAS  Google Scholar 

  256. Frey U, Müller M, Kuhl D (1996) A different form of long-lasting potentiation revealed in tissue plasminogen activator mutant mice. J Neurosci 16:2057–2063

    Article  CAS  Google Scholar 

  257. Madani R, Hulo S, Toni N et al (1999) Enhanced hippocampal long-term potentiation and learning by increased neuronal expression of tissue-type plasminogen activator in transgenic mice. EMBO J 18:3007–3012. https://doi.org/10.1093/emboj/18.11.3007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Zhuo M, Holtzman DM, Li Y et al (2000) Role of tissue plasminogen activator receptor LRP in hippocampal long-term potentiation. J Neurosci 20:542–549

    Article  CAS  Google Scholar 

  259. Pang PT, Teng HK, Zaitsev E et al (2004) Cleavage of proBDNF by tPA/plasmin is essential for long-term hippocampal plasticity. Science 306(80):487–491. https://doi.org/10.1126/science.1100135

    Article  CAS  PubMed  Google Scholar 

  260. Tirosh O, Schwartz B, Zusman I et al (2004) Long-lived αMUPA transgenic mice exhibit increased mitochondrion-mediated apoptotic capacity. Ann N Y Acad Sci 1019:439–442. https://doi.org/10.1196/annals.1297.080

    Article  CAS  PubMed  Google Scholar 

  261. Miskin R, Masos T (1997) Transgenic mice overexpressing urokinase-type plasminogen activator in the brain exhibit reduced food consumption, body weight and size, and increased longevity. J Gerontol Ser A Biol Sci Med Sci 52:B118–B124. https://doi.org/10.1093/gerona/52A.2.B118

    Article  CAS  Google Scholar 

  262. Meiri N, Masos T, Rosenblum K et al (1994) Overexpression of urokinase-type plasminogen activator in transgenic mice is correlated with impaired learning. Proc Natl Acad Sci USA 91:3196–3200

    Article  CAS  Google Scholar 

  263. Rivera S, Khrestchatisky M, Kaczmarek L et al (2010) Metzincin proteases and their inhibitors: foes or friends in nervous system physiology? J Neurosci 30:15337–15357. https://doi.org/10.1523/JNEUROSCI.3467-10.2010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Baranger K, Rivera S, Liechti FD, Grandgirard D, Bigas J, Seco J, Tarrago T, Leib SL, Khrestchatisky M (2014) Endogenous and synthetic MMP inhibitors in CNS physiopathology. In: Prog Brain Res 214:313–351

    Article  Google Scholar 

  265. Agrawal S, Lau L, Yong V (2008) MMPs in the central nervous system: where the good guys go bad. Semin Cell Dev Biol 19:42–51. https://doi.org/10.1016/j.semcdb.2007.06.003

    Article  CAS  PubMed  Google Scholar 

  266. Hewson AK, Smith T, Leonard JP, Cuzner ML (1995) Suppression of experimental allergic encephalomyelitis in the Lewis rat by the matrix metalloproteinase inhibitor Ro31-9790. Inflamm Res 44:345–349. https://doi.org/10.1007/BF01796266

    Article  CAS  PubMed  Google Scholar 

  267. Brundula V, Rewcastle NB, Metz LM et al (2002) Targeting leukocyte MMPs and transmigration: minocycline as a potential therapy for multiple sclerosis. Brain 125:1297–1308. https://doi.org/10.1093/BRAIN/AWF133

    Article  PubMed  Google Scholar 

  268. Rempe RG, Hartz AMS, Bauer B (2016) Matrix metalloproteinases in the brain and blood-brain barrier: versatile breakers and makers. J Cereb Blood Flow Metab 36:1481–1507. https://doi.org/10.1177/0271678X16655551

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Suzuki Y, Nagai N, Umemura K et al (2007) Stromelysin-1 (MMP-3) is critical for intracranial bleeding after t-PA treatment of stroke in mice. J Thromb Haemost 5:1732–1739. https://doi.org/10.1111/j.1538-7836.2007.02628.x

    Article  CAS  PubMed  Google Scholar 

  270. Sumii T, Lo EH (2002) Involvement of matrix metalloproteinase in thrombolysis-associated hemorrhagic transformation after embolic focal ischemia in rats. Stroke 33:831–836

    Article  CAS  Google Scholar 

  271. Testero SA, Lee M, Staran RT et al (2011) Sulfonate-containing thiiranes as selective gelatinase inhibitors. ACS Med Chem Lett 2:177–181. https://doi.org/10.1021/ml100254e

    Article  CAS  PubMed  Google Scholar 

  272. Gu Z, Cui J, Brown S et al (2005) A highly specific inhibitor of matrix metalloproteinase-9 rescues laminin from proteolysis and neurons from apoptosis in transient focal cerebral ischemia. J Neurosci 25:6401–6408. https://doi.org/10.1523/JNEUROSCI.1563-05.2005

    Article  CAS  PubMed  Google Scholar 

  273. Cui J, Chen S, Zhang C et al (2012) Inhibition of MMP-9 by a selective gelatinase inhibitor protects neurovasculature from embolic focal cerebral ischemia. Mol Neurodegener 7:21. https://doi.org/10.1186/1750-1326-7-21

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Zhao B-Q, Wang S, Kim H-Y et al (2006) Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med 12:441–445. https://doi.org/10.1038/nm1387

    Article  CAS  PubMed  Google Scholar 

  275. Fagan SC, Waller JL, Nichols FT et al (2010) Minocycline to improve neurologic outcome in stroke (MINOS). Stroke 41:2283–2287. https://doi.org/10.1161/STROKEAHA.110.582601

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Minagar A, Alexander JS, Schwendimann RN et al (2008) Combination therapy with interferon beta-1a and doxycycline in multiple sclerosis. Arch Neurol 65:199–204. https://doi.org/10.1001/archneurol.2007.41

    Article  PubMed  Google Scholar 

  277. Groves MD, Puduvalli VK, Conrad CA et al (2006) Phase II trial of temozolomide plus marimastat for recurrent anaplastic gliomas: a relationship among efficacy, joint toxicity and anticonvulsant status. J Neurooncol 80:83–90. https://doi.org/10.1007/s11060-006-9160-y

    Article  CAS  PubMed  Google Scholar 

  278. Groves MD, Puduvalli VK, Hess KR et al (2002) Phase II trial of temozolomide plus the matrix metalloproteinase inhibitor, marimastat, in recurrent and progressive glioblastoma multiforme. J Clin Oncol 20:1383–1388. https://doi.org/10.1200/JCO.2002.20.5.1383

    Article  CAS  PubMed  Google Scholar 

  279. Prinomastat plus temozolomide following radiation therapy in treating patients with newly diagnosed glioblastoma multiforme—full text view—ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT00004200. Accessed 8 Apr 2019

  280. Rudek MA, New P, Mikkelsen T et al (2011) Phase I and pharmacokinetic study of COL-3 in patients with recurrent high-grade gliomas. J Neurooncol 105:375–381. https://doi.org/10.1007/s11060-011-0602-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  281. Vandenbroucke RE, Libert C (2014) Is there new hope for therapeutic matrix metalloproteinase inhibition? Nat Rev Drug Discov 13:904–927. https://doi.org/10.1038/nrd4390

    Article  CAS  PubMed  Google Scholar 

  282. Gantois I, Khoutorsky A, Popic J et al (2017) Metformin ameliorates core deficits in a mouse model of fragile X syndrome. Nat Med 23:674–677. https://doi.org/10.1038/nm.4335

    Article  CAS  PubMed  Google Scholar 

  283. Combined treatment of minocycline and lovastatin to treat individuals with fragile × syndrome—full text view—ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02680379. Accessed 8 Apr 2019

  284. Levin M, Udi Y, Solomonov I, Sagi I (2017) Next generation matrix metalloproteinase inhibitors—novel strategies bring new prospects. Biochim Biophys Acta Mol Cell Res 1864:1927–1939. https://doi.org/10.1016/j.bbamcr.2017.06.009

    Article  CAS  PubMed  Google Scholar 

  285. Pantazopoulos H, Berretta S (2016) In sickness and in health: perineuronal nets and synaptic plasticity in psychiatric disorders. Neural Plast. 2016:9847696

    Article  Google Scholar 

  286. Suttkus A, Rohn S, Weigel S et al (2014) Aggrecan, link protein and tenascin-R are essential components of the perineuronal net to protect neurons against iron-induced oxidative stress. Cell Death Dis 5:e1119. https://doi.org/10.1038/cddis.2014.25

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  287. Morawski M, Dityatev A, Hartlage-Rübsamen M et al (2014) Tenascin-R promotes assembly of the extracellular matrix of perineuronal nets via clustering of aggrecan. Philos Trans R Soc B Biol Sci 369:20140046. https://doi.org/10.1098/rstb.2014.0046

    Article  CAS  Google Scholar 

  288. Vitellaro-Zuccarello L, Bosisio P, Mazzetti S et al (2007) Differential expression of several molecules of the extracellular matrix in functionally and developmentally distinct regions of rat spinal cord. Cell Tissue Res 327:433–447. https://doi.org/10.1007/s00441-006-0289-y

    Article  CAS  PubMed  Google Scholar 

  289. Matthews RT, Kelly GM, Zerillo CA et al (2002) Aggrecan glycoforms contribute to the molecular heterogeneity of perineuronal nets. J Neurosci 22:7536–7547

    Article  CAS  Google Scholar 

  290. Mohan V, Talmi-Frank D, Arkadash V, Papo NSI (2016) Matrix metalloproteinase protein inhibitors: highlighting a new beginning for metalloproteinases in medicine. Met Med 3:31–47. https://doi.org/10.2147/MNM.S119588

    Article  Google Scholar 

Download references

Acknowledgements

Irit Sagi is an Incumbent of the Maurizio Pontecorvo Professorial Chair and has received funding from the Israeli Science Foundation (1226/13), the European Research Council AdG (THZCALORIMETRY—DLV-695437), and the USA-Israel Binational Science Foundation (712506-01). She is grateful for the Azrieli Foundation for its generous grant to conduct research on the extracellular matrix of the brain.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Irit Sagi.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Krishnaswamy, V.R., Benbenishty, A., Blinder, P. et al. Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights. Cell. Mol. Life Sci. 76, 3229–3248 (2019). https://doi.org/10.1007/s00018-019-03182-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-019-03182-6

Keywords

Navigation