Semin Thromb Hemost 2021; 47(03): 308-315
DOI: 10.1055/s-0041-1724117
Review Article

Three-Dimensional Human Neural Stem Cell Models to Mimic Heparan Sulfate Proteoglycans and the Neural Niche

Ian W. Peall
1   Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
,
Rachel K. Okolicsanyi
1   Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
,
Lyn R. Griffiths
1   Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
,
Larisa M. Haupt
1   Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
› Institutsangaben

Abstract

Heparan sulfate proteoglycans (HSPGs) are a diverse family of polysaccharides, consisting of a core protein with glycosaminoglycan (GAG) side chains attached. The heterogeneous GAG side-chain carbohydrates consist of repeating disaccharides, with each side chain possessing a specific sulfation pattern. It is the variable sulfation pattern that allows HSPGs to interact with numerous ligands including growth factors, cytokines, chemokines, morphogens, extracellular matrix (ECM) glycoproteins, collagens, enzymes, and lipases. HSPGs are classified according to their localization within an individual cell, and include the membrane bound syndecans (SDCs) and glypicans (GPCs), with perlecan, agrin, and type-XVIII collagen secreted into the ECM. The stem cell niche is defined as the environment that circumscribes stem cells when they are in their naïve state, and includes the ECM, which provides a complex contribution to various biological processes during development and throughout life. These contributions include facilitating cell adhesion, proliferation, migration, differentiation, specification, and cell survival. In contrast, HSPGs play an anticoagulant role in thrombosis through being present on the luminal surface of cells, while also playing roles in the stimulation and inhibition of angiogenesis, highlighting their varied and systemic roles in cellular control. To fully understand the complexities of cell-cell and cell–matrix interactions, three-dimensional (3D) models such as hydrogels offer researchers exciting opportunities, such as controllable 3D in vitro environments, that more readily mimic the in vivo/in situ microenvironment. This review examines our current knowledge of HSPGs in the stem cell niche, human stem cell models, and their role in the development of 3D models that mimic the in vivo neural ECM.

Authors' contributions

The concept of this manuscript was developed and drafted by I.W.P., R.K.O., and L.M.H. I.W.P. wrote the manuscript with R.K.O., L.R.G., and L.M.H. providing supervision in the structure, design and revision of the manuscript. All authors approved the final version.




Publikationsverlauf

Artikel online veröffentlicht:
01. April 2021

© 2021. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Moreno MJ, Ling B, Stanimirovic DB. In vivo near-infrared fluorescent optical imaging for CNS drug discovery. Expert Opin Drug Discov 2020; 15 (08) 903-915
  • 2 Yu C, Griffiths LR, Haupt LM. Exploiting heparan sulfate proteoglycans in human neurogenesis-controlling lineage specification and fate. Front Integr Nuerosci 2017; 11: 28
  • 3 Gincberg G, Arien-Zakay H, Lazarovici P, Lelkes PI. Neural stem cells: therapeutic potential for neurodegenerative diseases. Br Med Bull 2012; 104: 7-19
  • 4 de Miguel-Beriain I. The ethics of stem cells revisited. Adv Drug Deliv Rev 2015; 82-83: 176-180
  • 5 Donnelly H, Salmeron-Sanchez M, Dalby MJ. Designing stem cell niches for differentiation and self-renewal. J R Soc Interface 2018; 15 (145) 20180388
  • 6 Häcker U, Nybakken K, Perrimon N. Heparan sulphate proteoglycans: the sweet side of development. Nat Rev Mol Cell Biol 2005; 6 (07) 530-541
  • 7 Kempermann G. Adult neurogenesis: an evolutionary perspective. Cold Spring Harb Perspect Biol 2015; 8 (02) a018986
  • 8 Kuhn HG, Toda T, Gage FH. Adult hippocampal neurogenesis: a coming-of-age story. J Neurosci 2018; 38 (49) 10401-10410
  • 9 Yamaguchi Y. Heparan sulfate proteoglycans in the nervous system: their diverse roles in neurogenesis, axon guidance, and synaptogenesis. Semin Cell Dev Biol 2001; 12 (02) 99-106
  • 10 Wang Q, Yang L, Alexander C, Temple S. The niche factor syndecan-1 regulates the maintenance and proliferation of neural progenitor cells during mammalian cortical development. PLoS One 2012; 7 (08) e42883
  • 11 Bespalov MM, Sidorova YA, Tumova S. et al. Heparan sulfate proteoglycan syndecan-3 is a novel receptor for GDNF, neurturin, and artemin. J Cell Biol 2011; 192 (01) 153-169
  • 12 Poulain FE, Yost HJ. Heparan sulfate proteoglycans: a sugar code for vertebrate development?. Development 2015; 142 (20) 3456-3467
  • 13 Bansal R, Kumar M, Murray K, Pfeiffer SE. Developmental and FGF-2-mediated regulation of syndecans (1-4) and glypican in oligodendrocytes. Mol Cell Neurosci 1996; 7 (04) 276-288
  • 14 Winkler S, Stahl RC, Carey DJ, Bansal R. Syndecan-3 and perlecan are differentially expressed by progenitors and mature oligodendrocytes and accumulate in the extracellular matrix. J Neurosci Res 2002; 69 (04) 477-487
  • 15 Ford-Perriss M, Turner K, Guimond S. et al. Localisation of specific heparan sulfate proteoglycans during the proliferative phase of brain development. Dev Dyn 2003; 227 (02) 170-184
  • 16 Choi SH, Kim YH, Quinti L, Tanzi RE, Kim DY. 3D culture models of Alzheimer's disease: a road map to a “cure-in-a-dish”. Mol Neurodegener 2016; 11 (01) 75
  • 17 Freudenberg U, Hermann A, Welzel PB. et al. A star-PEG-heparin hydrogel platform to aid cell replacement therapies for neurodegenerative diseases. Biomaterials 2009; 30 (28) 5049-5060
  • 18 Barros D, Conde-Sousa E, Gonçalves AM. et al. Engineering hydrogels with affinity-bound laminin as 3D neural stem cell culture systems. Biomater Sci 2019; 7 (12) 5338-5349
  • 19 Kreuger J, Kjellén L. Heparan sulfate biosynthesis: regulation and variability. J Histochem Cytochem 2012; 60 (12) 898-907
  • 20 Condomitti G, de Wit J. Heparan sulfate proteoglycans as emerging players in synaptic specificity. Front Mol Neurosci 2018; 11: 14
  • 21 Oikari LE, Okolicsanyi RK, Qin A, Yu C, Griffiths LR, Haupt LM. Cell surface heparan sulfate proteoglycans as novel markers of human neural stem cell fate determination. Stem Cell Res (Amst) 2016; 16 (01) 92-104
  • 22 Okolicsanyi RK, Oikari LE, Yu C, Griffiths LR, Haupt LM. Heparan sulfate proteoglycans as drivers of neural progenitors derived from human mesenchymal stem cells. Front Mol Neurosci 2018; 11: 134
  • 23 Tabata T, Takei Y. Morphogens, their identification and regulation. Development 2004; 131 (04) 703-712
  • 24 Multhaupt HA, Couchman JR. Heparan sulfate biosynthesis: methods for investigation of the heparanosome. J Histochem Cytochem 2012; 60 (12) 908-915
  • 25 Coulson-Thomas VJ. The role of heparan sulphate in development: the ectodermal story. Int J Exp Pathol 2016; 97 (03) 213-229
  • 26 Jen YH, Musacchio M, Lander AD. Glypican-1 controls brain size through regulation of fibroblast growth factor signaling in early neurogenesis. Neural Dev 2009; 4: 33
  • 27 Listik E, Toma L. Glypican-1 in human glioblastoma: implications in tumorigenesis and chemotherapy. Oncotarget 2020; 11 (09) 828-845
  • 28 Oikari LE, Okolicsanyi RK, Griffiths LR, Haupt LM. Data defining markers of human neural stem cell lineage potential. Data Brief 2016; 7: 206-215
  • 29 Oikari LE, Yu C, Okolicsanyi RK. et al. HSPGs glypican-1 and glypican-4 are human neuronal proteins characteristic of different neural phenotypes. J Neurosci Res 2020; 98 (08) 1619-1645
  • 30 White J, Dalton S. Cell cycle control of embryonic stem cells. Stem Cell Rev 2005; 1 (02) 131-138
  • 31 Mikami T, Kitagawa H. Sulfated glycosaminoglycans: their distinct roles in stem cell biology. Glycoconj J 2017; 34 (06) 725-735
  • 32 Seidlits SK, Liang J, Bierman RD. et al. Peptide-modified, hyaluronic acid-based hydrogels as a 3D culture platform for neural stem/progenitor cell engineering. J Biomed Mater Res A 2019; 107 (04) 704-718
  • 33 Morla S. Glycosaminoglycans and glycosaminoglycan mimetics in cancer and inflammation. Int J Mol Sci 2019; 20 (08) E1963
  • 34 Okolicsanyi RK, Camilleri ET, Oikari LE. et al. Human mesenchymal stem cells retain multilineage differentiation capacity including neural marker expression after extended in vitro expansion. PLoS One 2015; 10 (09) e0137255
  • 35 Okolicsanyi RK. Mesenchymal stem cells as mediators of the neuronal cell niche, in faculty of health. Accessed November 3, 2020 at: https://eprints.qut.edu.au/84485/1/Rachel_Okolicsanyi_Thesis.pdf
  • 36 Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126 (04) 663-676
  • 37 Bordoni M, Rey F, Fantini V. et al. From neuronal differentiation of iPSCs to 3D neural organoids: modeling of neurodegenerative diseases. Int J Mol Sci 2018; 19 (12) 3972
  • 38 Yan Y, Bejoy J, Marzano M, Li Y. The use of pluripotent stem cell-derived organoids to study extracellular matrix development during neural degeneration. Cells 2019; 8 (03) E242
  • 39 Kim YH, Choi SH, D'Avanzo C. et al. A 3D human neural cell culture system for modeling Alzheimer's disease. Nat Protoc 2015; 10 (07) 985-1006
  • 40 Faley SL, Neal EH, Wang JX. et al. iPSC-derived brain endothelium exhibits stable, long-term barrier function in perfused hydrogel scaffolds. Stem Cell Reports 2019; 12 (03) 474-487
  • 41 Donato R, Miljan EA, Hines SJ. et al. Differential development of neuronal physiological responsiveness in two human neural stem cell lines. BMC Neurosci 2007; 8: 36
  • 42 Duval K, Grover H, Han LH. et al. Modeling physiological events in 2D vs. 3D cell culture. Physiology (Bethesda) 2017; 32 (04) 266-277
  • 43 Korhonen P, Malm T, White AR. 3D human brain cell models: New frontiers in disease understanding and drug discovery for neurodegenerative diseases. Neurochem Int 2018; 120: 191-199
  • 44 Hunyadi A, Gaál B, Matesz C. et al. Distribution and classification of the extracellular matrix in the olfactory bulb. Brain Struct Funct 2020; 225 (01) 321-344
  • 45 Yu C, Zhu W, Sun B, Mei D, Gou M, Chen S. Modulating physical, chemical, and biological properties in 3D printing for tissue engineering applications. Appl Phys Rev 2018; 5 (04) 041107
  • 46 Brown TD, Dalton PD, Hutmacher DW. Melt electrospinning today: An opportune time for an emerging polymer process. Prog Polym Sci 2016; 56: 116-166
  • 47 Lam D, Enright HA, Peters SKG. et al. Optimizing cell encapsulation condition in ECM-Collagen I hydrogels to support 3D neuronal cultures. J Neurosci Methods 2020; 329: 108460
  • 48 Madl CM, LeSavage BL, Dewi RE, Lampe KJ, Heilshorn SC. Matrix remodeling enhances the differentiation capacity of neural progenitor cells in 3d hydrogels. Adv Sci (Weinh) 2019; 6 (04) 1801716
  • 49 Loessner D, Meinert C, Kaemmerer E. et al. Functionalization, preparation and use of cell-laden gelatin methacryloyl-based hydrogels as modular tissue culture platforms. Nat Protoc 2016; 11 (04) 727-746
  • 50 Leipzig ND, Shoichet MS. The effect of substrate stiffness on adult neural stem cell behavior. Biomaterials 2009; 30 (36) 6867-6878
  • 51 Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta 2014; 1840 (08) 2506-2519
  • 52 Binner M, Bray LJ, Friedrichs J, Freudenberg U, Tsurkan MV, Werner C. Cell-instructive starPEG-heparin-collagen composite matrices. Acta Biomater 2017; 53: 70-80
  • 53 Ylä-Outinen L, Harju V, Joki T. et al. Screening of hydrogels for human pluripotent stem cell-derived neural cells: hyaluronan-polyvinyl alcohol-collagen-based interpenetrating polymer network provides an improved hydrogel scaffold. Macromol Biosci 2019; 19 (07) e1900096
  • 54 Papadimitriou C, Celikkaya H, Cosacak MI. et al. 3D culture method for alzheimer's disease modeling reveals interleukin-4 rescues Aβ42-induced loss of human neural stem cell plasticity. Dev Cell 2018; 46 (01) 85-101.e8
  • 55 Nazemi Z, Nourbakhsh MS, Kiani S. et al. Co-delivery of minocycline and paclitaxel from injectable hydrogel for treatment of spinal cord injury. J Control Release 2020; 321: 145-158
  • 56 Lee KY, Mooney DJ. Alginate: properties and biomedical applications. Prog Polym Sci 2012; 37 (01) 106-126
  • 57 Tang Y, Yu P, Cheng L. Current progress in the derivation and therapeutic application of neural stem cells. Cell Death Dis 2017; 8 (10) e3108
  • 58 Kirschen GW, Ge S. Young at heart: insights into hippocampal neurogenesis in the aged brain. Behav Brain Res 2019; 369: 111934
  • 59 Hopkins AM, DeSimone E, Chwalek K, Kaplan DL. 3D in vitro modeling of the central nervous system. Prog Neurobiol 2015; 125: 1-25
  • 60 Grasman JM, Ferreira JA, Kaplan DL. Tissue models for neurogenesis and repair in 3D. Adv Funct Mater 2018; 28 (48) 1803822