Development of Schwann Cells: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , ,

Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. To achieve differentiation of Schwann cells from stem cell sources in vitro, cultures are manipulated using molecular factors to emulate developmental signaling events which lead to development of Schwann cells in vivo. Therefore, knowledge of molecular determinants in embryonal development of the Schwann cell fate is key to develop and refine in vitro differentiation protocols. The following sections briefly summarize the current knowledge on developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo.

  • glia
  • neural crest
  • Schwann cells
  • development
  • Schwann cell precursor
  • differentiation
  • peripheral nervous system

1. Introduction

Schwann cells are the most abundant cell type in the peripheral nerves and the best studied type of peripheral glia cells [1][2][3]. They are most well known for their function of wrapping myelin sheaths around peripheral axons, but the continuously increasing knowledge about Schwann cell biology has revealed a plethora of additional roles besides myelination and neurotrophic support [4], such as regulation of sensory perception [5][6], synaptic communication [7][8], and immune response [9][10]. They are also important in nerve repair, thus contributing to the regenerative ability of the peripheral nervous system (PNS) [11][12]. Therefore, models for exploring the physiology and pathophysiology of Schwann cells in vitro are of profound interest [13]. At the same time, Schwann cells are highly interesting for tissue engineering, since they play a critical role in nerve regeneration upon injury and are affected in peripheral neuropathies such as Charcot-Marie-Tooth disease and Guillain-Barré syndrome [11][14][15][16]. Therefore, the potential of Schwann cells regarding regenerative cell transplantation has been intensely explored in rodent models, with encouraging results. However, for potential clinical applications in human injury and disease, safe and reproducible methods for obtaining large quantities of human Schwann cells are needed. Primary human Schwann cells can be isolated from peripheral nerves, but obtaining a sufficient amount of cells to enable treatment is challenging. In addition, laborious methods are required to establish cultures, and these have a finite capacity for proliferation and passaging [17][18]. As such, rodent Schwann cells have mainly been used for in vitro studies. These are relatively easy to culture, do not cease proliferating in vitro, and can therefore be expanded to high numbers. They also achieve higher degrees of maturation in culture and are able to efficiently myelinate cocultured neurons, while human Schwann cells show only a poor myelination capacity in coculture assays [17][18][19][20]. As these differences hamper work with human Schwann cells and as data from non-human Schwann cells might not be translatable, alternative human in vitro models are important, to obtain valid information regarding Schwann cell development, pathology, and therapeutic potential. Advancements in stem cell technology have made in vitro derivation from multipotent cell sources a promising alternative [21][22][23], and some of these techniques have been used to differentiate Schwann cells in vitro, including human embryonic stem cells (hESC) and human induced pluripotent stem cells (hiPSC), as well as human and non-human adult tissue-derived stem cell types. However, many of these methods still suffer from drawbacks such as incomplete differentiation efficiency, variable reproducibility, or a lack of maturity of the derived cells.To establish and optimize such protocols, detailed knowledge of signaling mechanisms in embryonal development of the Schwann cell lineage is required. Therefore, the following paragraphs briefly summarize the current knowledge regarding key signaling events along the developing Schwann cell fate.

2. In Vivo Development of Schwann Cells

2.1. Development of Neural Crest and Schwann Cell Precursors

The Schwann cell lineage arises from the neural crest, a multipotent stem cell population specific to vertebrate embryonal development (Figure 1A). Of ectodermal origin, the neural crest is induced during gastrulation [24][25] and subsequently specified from the neural plate border territory by signaling from adjacent developing structures, i.e., the juxtaposed non-neural ectoderm, neural plate, and underlying mesoderm. After closure of the neural tube, premigratory neural crest cells undergo an epithelial-to-mesenchymal transition (EMT), allowing them to delaminate from the neuroepithelium and to migrate through the embryonal tissue. Dorsoventrally emigrating neural crest cells associate with developing peripheral nerves and are then termed Schwann cell precursors [26][27]. Both neural crest cells and Schwann cell precursors reach their target tissues through specific migration pathways and differentiate towards their terminal fate once settled in the target tissues, where they often undergo a mesenchymal-to-epithelial transition [28]. During this migration phase, Schwann cell precursors move along the peripheral nerves.
Figure 1. Development of the Schwann cell lineage. (A) During neurulation, the neural plate border (b) develops between the neural plate (a) and the non-neural ectoderm (c), along neighboring structures, including the notochord (d) and paraxial mesoderm (e). After the neural plate folds to build the neural tube (f), the neural plate borders merge and form the neural crest. Late neural crest cells (g) detach and migrate. (B) Differentiation along the Schwann cell lineage progresses through several intermediate stages, namely Schwann cell precursors and immature Schwann cells. While Schwann cell precursors are multipotent and can still differentiate into other cell types, such as melanocytes or neurons, immature Schwann cells are more fate restricted and differentiate into mature myelinating and non-myelinating Schwann cells, terminal Schwann cells of the neuromuscular junctions, as well as endoneurial fibroblasts. According to current knowledge, terminal Schwann cells segregate during differentiation prior to endoneurial fibroblasts and other Schwann cell subtypes. Figure from Hörner et al. 2022 under CC BY 4.0 license.
Neural crest induction and specification are primarily mediated by four major developmental pathways, i.e., Wnt, FGF, BMP, and Notch signaling (for review, see Prasad et al., 2019 [29]). Wnt signaling is crucial for important aspects of neural crest induction, i.e., formation of the neural plate border and subsequent activation of neural crest-specifying transcription factors (reviewed by Ji et al., 2019 [30]). FGF signaling has been especially implicated in the early phases of neural crest specification [24][31]. BMP signaling appears to be critical after initial neural crest induction [32]. It is required at an intermediate activity level, which is provided by gradients of laterally secreted BMP activating signals and medially secreted BMP antagonizing signals [33][34][35].
Finally, Notch signaling is relevant in neural crest induction, migration, differentiation, and inhibition of neurogenesis [36][37][38][39], but its exact role in these processes and the apparent variations between species remain to be elucidated [29]. Although a wealth of in vivo and in vitro studies in different species have established these pathways and their crosstalk as principal regulators of neural crest development, new insights about their precise spatiotemporal activities are still emerging. In addition, other pathways have been suggested to play a role in neural crest development, such as retinoic acid signaling [40][41] and the Hippo/YAP pathway. For example, recent findings suggest that Hippo/YAP promotes neural crest fate and migration, as well as fate specification of neural crest cells (reviewed by Zhao et al., 2021 [42]): YAP activation could benefit the EMT and migration of neural crest cells, and studies have suggested the crosstalk of YAP signaling with that of BMP and Wnt [43], retinoic acid [44], and Notch [45] during neural crest development.
Overall, an astounding variety of cell types of neural, as well as mesenchymal, fates are generated from the neural crest [46]. Besides Schwann cells, these also include other peripheral glia such as enteric glia and ganglionic satellite glia, as well as neurons of the peripheral nervous system (PNS), including sensory neurons, postganglionic sympathetic and parasympathetic neurons, and enteric neurons. Furthermore, melanocytes, endocrine cells, and a variety of mesenchymal cells, including cranial bone and connective tissue, are also neural crest descendants. Some of these fates depend on the neural crest anteroposterior axial identity (cranial, vagal, trunk, sacral), as they contribute to different tissue types, suggesting there is already heterogeneity of differentiation competence in the neural crest population [47][48]. While neuron and glia cells differentiate from the neural crest at all axial levels, the major part of Schwann cells and PNS neurons are derived from trunk neural crest [27][49][50][51][52][53].
The development of neural crest cells into mature Schwann cells proceeds through intermediate stages: first, the previously mentioned Schwann cell precursors, which are found in the early embryonic nerve; and then the immature Schwann cells in late embryonic and perinatal nerves (Schwann cell precursor development has been reviewed in detail elsewhere [1][49][54][55]) (Figure 1B). The extrinsic signals inducing and specifying these stages are still unknown [54][56], but a key factor is neuregulin-1 (Nrg1), specifically the axon-derived splicing isoform Nrg1 type III [57][58][59]. In the Schwann cell precursor stage, membrane-bound Nrg1 type III on juxtaposed sensory and motor axons acts as a crucial survival factor through ErbB family receptor tyrosine kinases, mainly ErbB2/3 heterodimers [60][61]. The transition from neural crest to Schwann cell precursors has been connected to the acquisition of typical glia features. In particular, they enter developing nerves, associate with axons, and depend on axonal signals for survival. At the same time, glial and Schwann cell lineage specific markers are upregulated [1]. Nevertheless, Schwann cell precursors retain a high degree of multipotency, and become primed towards immature Schwann cells or other fates in a gradual manner [55][62].
Notably, several neural crest-derived cell types outside the Schwann cell lineage are derived from both the neural crest and Schwann cell precursors. One example is the melanocytes, which differentiate either directly from neural crest cells or from Schwann cell precursors, following their migration along nerves [63][64]. A similar mechanism might possibly be true for enteric glia, as both neural crest cells and Schwann cell precursors were found to populate the gut to differentiate [65]. To date, further cell types which can develop from Schwann cell precursors include endoneurial fibroblasts [66]; postganglionic parasympathetic neurons [67][68]; enteric neurons [69]; neuroendocrine chromaffin cells of the adrenal medulla [70] and Zuckerkandl organ [71], a small population of sympathetic neurons in paraganglia [71]; dental pulp mesenchymal stromal cells [72]; and even chondrocytes and osteocytes [73]; as well as, possibly, recently discovered immune-regulating splenic glia [74][75][76]. This remarkable developmental potential of multipotent Schwann cell precursors clearly indicates that no terminal commitment to the Schwann cell lineage has happened at this stage. Indeed, recent work using single-cell transcriptomics has confirmed that early Schwann cell precursors are highly similar to late migratory neural crest cells and exhibit multipotency towards various cell fates, while late Schwann cell precursors that progress towards the Schwann cell fate become gradually more restricted [62].

2.2. Commitment to Schwann Cell Lineage and Differentiation of Subtypes

Commitment to immature Schwann cells follows the multipotent Schwann cell precursor stage (Figure 1B). After migratory Schwann cell precursors have reached their target tissue, they either detach from nerves and assume other cell fates or further develop into post-migratory immature Schwann cells in a continued association with axons [55][77][78]. This is accompanied by upregulation of glial and Schwann cell markers (e.g., Krox20, P0, S100b, Oct6) and downregulation of earlier markers (e.g., Ap2α, Pax3, Sox2) [3][26][79][80][81]. At the same time, these cells start expressing basal lamina proteins, as well as growth factors, which act on differentiating neurons but also as autocrine survival factors [1][52][82][83]. During this period, immature Schwann cells initiate radial sorting of axons, whereby groups of immature Schwann cells bundle several axons together, before segregating the larger axons designated for myelination; these processes are orchestrated by axonal Nrg1 [84]. Furthermore, Notch signaling seems to be involved in the transition to immature Schwann cells, through boosting Nrg1 signaling; indeed, activation of Notch1 receptor through Jagged1 expressed on adjacent neurons maintains high expression levels of ErbB2 receptors in differentiating Schwann cells. At later stages, Notch inhibits myelination and is suppressed through Krox20 at the onset of the myelination program [85]. While immature Schwann cells are initially highly proliferative, more than Schwann cell precursors [86], they gradually exit the cell cycle during maturation, when Schwann cells assume mature myelinating and non-myelinating phenotypes. This mainly occurs postnatally in rodents [3][86][87].
The two known major subtypes of mature Schwann cells are myelinating Schwann cells and non-myelinating Remak Schwann cells. While the first associate with one individual axon through radial sorting and build up the myelin sheath wrapping a segment of this axon, the latter associate with several small diameter axons, and ensheath them in so-called Remak bundles, without producing myelin. Terminal (or perisynaptic) Schwann cells are a third distinct subtype of Schwann cells. They are associated with neuromuscular junctions, and although they are non-myelinating, terminal Schwann cells differ considerably from Remak Schwann cells in terms of their marker expression, appearance, and function [7][88][89]. Besides these, it is likely that other glial subtypes that have not been well characterized so far also arise from Schwann cell precursors and might differentiate through the immature Schwann cell stage as well [79], such as the glia cells of cutaneous sensory corpuscles [90][91]. While some factors that promote the development of the mature myelinating Schwann cell subtype are known, the differential mechanisms that specify the mature non-myelinating fates, including Remak and terminal Schwann cells, are largely unclear.
Myelinating Schwann cells develop through a further intermediate state, termed a promyelinating Schwann cell, which establishes a 1:1 relationship with an axon sorted for myelination [84]. Axonal sorting and maturation of myelinating Schwann cells is dependent on axonal contact and Nrg1 expression levels [92]. Myelination and terminal differentiation appear to involve activation of the adhesion G-protein coupled receptor GPR126 by basal lamina components such as laminin-211 [93][94][95][96]. GPR126 might signal through multiple downstream effectors, one being the myelination onset regulator Krox20 [97] through cAMP-dependent PKA activation [98]. Furthermore, the axonal Nrg1 expression level seems to tune the regulation of myelination fate. Indeed, while higher Nrg1 levels are required for myelination, lower Nrg1 levels may induce Remak differentiation [92]. At the same time, the Nrg1 expression level is proportional to the axon diameter, so that large caliber axons are sorted for myelination and small caliber axons are ensheathed by Remak bundles [99]. In addition, GABA-B receptors were found to regulate development towards non-myelinating Schwann cells through negative modulation of adenylate cyclase and downregulation of myelin-related proteins [100][101][102]. Furthermore, GPR126 is also involved in Remak Schwann cell differentiation [95]. However, it is unknown how GPR126 functions mechanistically in this context and how its role differs from that in differentiation of myelinating Schwann cells. As for terminal Schwann cells, to date, no differential specifying molecular factors are known, and even the developmental time window when they diverge from the other subtypes remained unknown until recently. Using single-cell transcriptomics, Kastriti and colleagues analyzed developmental transitions and fate choices along the neural crest and Schwann cell lineages. This revealed that immature Schwann cells develop into four terminally differentiated cell types: terminal, non-myelinating (Remak), and myelinating Schwann cells, as well as endoneurial fibroblasts. Of these, the terminal Schwann cell fate is the first to diverge from the others; thus, even before the path of endoneurial fibroblasts splits from myelinating and Remak Schwann cells (Figure 1B) [62].
Even after terminal differentiation into mitotically quiescent mature Schwann cell subtypes, the Schwann cell linage retains a remarkably plastic reprogramming potential, and terminal differentiation is reversible. Predominantly as a reaction to nerve injury, mature Schwann cells of all subtypes can re-enter the cell cycle, de-differentiate, and assume what is called the repair phenotype. This is distinct from other mature, as well as developmental, Schwann cell phenotypes [12][103]. Repair Schwann cells are crucial for nerve repair and account for the outstanding regeneration ability of the PNS [11][104]. Following the resolution of nerve damage, repair Schwann cells re-differentiate into mature myelinating and non-myelinating Schwann cells. During this re-differentiation process, prior mature myelinating Schwann cells can assume a mature non-myelinating phenotype and vice versa [105][106]. Notably, they might even re-differentiate into non-glial cells, including melanocytes [64], dental pulp mesenchymal stem cells [72], or enteric neurons [107]. This remarkable plasticity of developing and mature descendants of the Schwann cell lineage is controlled by transcription factor networks and chromatin remodeling [108][109][110] and makes Schwann cells exceptionally interesting for in vitro modeling studies on nerve injury, disease, and regeneration. However, their extensive developmental multipotency and adult plasticity also add to the challenges in deriving, characterizing, and maintaining Schwann cells in vitro.

This entry is adapted from the peer-reviewed paper 10.3390/cells11233753

References

  1. Jessen, K.R.; Mirsky, R. Schwann Cell Precursors; Multipotent Glial Cells in Embryonic Nerves. Front. Mol. Neurosci. 2019, 12, 69.
  2. Reed, C.B.; Feltri, M.L.; Wilson, E.R. Peripheral glia diversity. J. Anat. 2021, 241, 1219–1234.
  3. Gerber, D.; Pereira, J.A.; Gerber, J.; Tan, G.; Dimitrieva, S.; Yángüez, E.; Suter, U. Transcriptional profiling of mouse peripheral nerves to the single-cell level to build a sciatic nerve ATlas (SNAT). eLife 2021, 10, e58591.
  4. Sardella-Silva, G.; Mietto, B.S.; Ribeiro-Resende, V.T. Four Seasons for Schwann Cell Biology, Revisiting Key Periods: Development, Homeostasis, Repair, and Aging. Biomolecules 2021, 11, 1887.
  5. Abdo, H.; Calvo-Enrique, L.; Lopez, J.M.; Song, J.; Zhang, M.-D.; Usoskin, D.; El Manira, A.; Adameyko, I.; Hjerling-Leffler, J.; Ernfors, P. Specialized cutaneous Schwann cells initiate pain sensation. Science 2019, 365, 695–699.
  6. Rinwa, P.; Calvo-Enrique, L.; Zhang, M.-D.; Nyengaard, J.R.; Karlsson, P.; Ernfors, P. Demise of nociceptive Schwann cells causes nerve retraction and pain hyperalgesia. Pain 2021, 162, 1816–1827.
  7. Ko, C.-P.; Robitaille, R. Perisynaptic Schwann Cells at the Neuromuscular Synapse: Adaptable, Multitasking Glial Cells. Cold Spring Harb. Perspect. Biol. 2015, 7, a020503.
  8. Darabid, H.; St-Pierre-See, A.; Robitaille, R. Purinergic-Dependent Glial Regulation of Synaptic Plasticity of Competing Terminals and Synapse Elimination at the Neuromuscular Junction. Cell Rep. 2018, 25, 2070–2082.
  9. Zhang, S.H.; Shurin, G.V.; Khosravi, H.; Kazi, R.; Kruglov, O.; Shurin, M.R.; Bunimovich, Y.L. Immunomodulation by Schwann cells in disease. Cancer Immunol. Immunother. 2020, 69, 245–253.
  10. Meyer zu Hörste, G.; Hu, W.; Hartung, H.-P.; Lehmann, H.C.; Kieseier, B.C. The immunocompetence of Schwann cells. Muscle Nerve 2008, 37, 3–13.
  11. Jessen, K.R.; Mirsky, R. The repair Schwann cell and its function in regenerating nerves. J. Physiol. 2016, 594, 3521–3531.
  12. Gomez-Sanchez, J.A.; Pilch, K.S.; van der Lans, M.; Fazal, S.V.; Benito, C.; Wagstaff, L.J.; Mirsky, R.; Jessen, K.R. After Nerve Injury, Lineage Tracing Shows That Myelin and Remak Schwann Cells Elongate Extensively and Branch to Form Repair Schwann Cells, Which Shorten Radically on Remyelination. J. Neurosci. 2017, 37, 9086–9099.
  13. Negro, S.; Pirazzini, M.; Rigoni, M. Models and methods to study Schwann cells. J. Anat. 2022, 241, 1235–1258.
  14. McGonigal, R.; Campbell, C.I.; Barrie, J.A.; Yao, D.; Cunningham, M.E.; Crawford, C.L.; Rinaldi, S.; Rowan, E.G.; Willison, H.J. Schwann cell nodal membrane disruption triggers bystander axonal degeneration in a Guillain-Barré syndrome mouse model. J. Clin. Investig. 2022, 132, 158524.
  15. Murakami, T.; Sunada, Y. Schwann Cell and the Pathogenesis of Charcot-Marie-Tooth Disease. Adv. Exp. Med. Biol. 2019, 1190, 301–321.
  16. Rodella, U.; Negro, S.; Scorzeto, M.; Bergamin, E.; Jalink, K.; Montecucco, C.; Yuki, N.; Rigoni, M. Schwann cells are activated by ATP released from neurons in an in vitro cellular model of Miller Fisher syndrome. Dis. Model. Mech. 2017, 10, 597–603.
  17. Monje, P.V.; Sant, D.; Wang, G. Phenotypic and Functional Characteristics of Human Schwann Cells as Revealed by Cell-Based Assays and RNA-SEQ. Mol. Neurobiol. 2018, 55, 6637–6660.
  18. Monje, P.V. The properties of human Schwann cells: Lessons from in vitro culture and transplantation studies. Glia 2020, 68, 797–810.
  19. Monje, P.V. Schwann Cell Cultures: Biology, Technology and Therapeutics. Cells 2020, 9, 1848.
  20. Morrissey, T.K.; Levi, A.D.; Nuijens, A.; Sliwkowski, M.X.; Bunge, R.P. Axon-induced mitogenesis of human Schwann cells involves heregulin and p185erbB2. Proc. Natl. Acad. Sci. USA 1995, 92, 1431–1435.
  21. Huang, Z.; Powell, R.; Phillips, J.B.; Haastert-Talini, K. Perspective on Schwann Cells Derived from Induced Pluripotent Stem Cells in Peripheral Nerve Tissue Engineering. Cells 2020, 9, 2497.
  22. Lehmann, H.C.; Höke, A. Use of engineered Schwann cells in peripheral neuropathy: Hopes and hazards. Brain Res. 2016, 1638, 97–104.
  23. Ma, M.-S.; Boddeke, E.; Copray, S. Pluripotent stem cells for Schwann cell engineering. Stem Cell Rev. Rep. 2015, 11, 205–218.
  24. Betters, E.; Charney, R.M.; Garcia-Castro, M.I. Early specification and development of rabbit neural crest cells. Dev. Biol. 2018, 444 (Suppl. S1), 181–192.
  25. Basch, M.L.; Bronner-Fraser, M.; García-Castro, M.I. Specification of the neural crest occurs during gastrulation and requires Pax7. Nature 2006, 441, 218–222.
  26. Jessen, K.R.; Mirsky, R. The origin and development of glial cells in peripheral nerves. Nat. Rev. Neurosci. 2005, 6, 671–682.
  27. Le Douarin, N.M.; Teillet, M.-A.M. Experimental analysis of the migration and differentiation of neuroblasts of the autonomic nervous system and of neurectodermal mesenchymal derivatives, using a biological cell marking technique. Dev. Biol. 1974, 41, 162–184.
  28. Simões-Costa, M.; Bronner, M.E. Establishing neural crest identity: A gene regulatory recipe. Development 2015, 142, 242–257.
  29. Prasad, M.S.; Charney, R.M.; García-Castro, M.I. Specification and formation of the neural crest: Perspectives on lineage segregation. Genesis 2019, 57, e23276.
  30. Ji, Y.; Hao, H.; Reynolds, K.; McMahon, M.; Zhou, C.J. Wnt Signaling in Neural Crest Ontogenesis and Oncogenesis. Cells 2019, 8, 1173.
  31. Lunn, J.S.; Fishwick, K.J.; Halley, P.A.; Storey, K.G. A spatial and temporal map of FGF/Erk1/2 activity and response repertoires in the early chick embryo. Dev. Biol. 2007, 302, 536–552.
  32. Steventon, B.; Araya, C.; Linker, C.; Kuriyama, S.; Mayor, R. Differential requirements of BMP and Wnt signalling during gastrulation and neurulation define two steps in neural crest induction. Development 2009, 136, 771–779.
  33. Anderson, R.M.; Stottmann, R.W.; Choi, M.; Klingensmith, J. Endogenous bone morphogenetic protein antagonists regulate mammalian neural crest generation and survival. Dev. Dyn. 2006, 235, 2507–2520.
  34. Tribulo, C.; Aybar, M.J.; Nguyen, V.H.; Mullins, M.C.; Mayor, R. Regulation of Msx genes by a Bmp gradient is essential for neural crest specification. Development 2003, 130, 6441–6452.
  35. Marchant, L.; Linker, C.; Ruiz, P.; Guerrero, N.; Mayor, R. The inductive properties of mesoderm suggest that the neural crest cells are specified by a BMP gradient. Dev. Biol. 1998, 198, 319–329.
  36. Noisa, P.; Lund, C.; Kanduri, K.; Lund, R.; Lähdesmäki, H.; Lahesmaa, R.; Lundin, K.; Chokechuwattanalert, H.; Otonkoski, T.; Tuuri, T.; et al. Notch signaling regulates the differentiation of neural crest from human pluripotent stem cells. J. Cell Sci. 2014, 127, 2083–2094.
  37. De Bellard, M.E.; Ching, W.; Gossler, A.; Bronner-Fraser, M. Disruption of segmental neural crest migration and ephrin expression in delta-1 null mice. Dev. Biol. 2002, 249, 121–130.
  38. Endo, Y.; Osumi, N.; Wakamatsu, Y. Bimodal functions of Notch-mediated signaling are involved in neural crest formation during avian ectoderm development. Development 2002, 129, 863–873.
  39. Morrison, S.J.; Perez, S.E.; Qiao, Z.; Verdi, J.M.; Hicks, C.; Weinmaster, G.; Anderson, D.J. Transient Notch Activation Initiates an Irreversible Switch from Neurogenesis to Gliogenesis by Neural Crest Stem Cells. Cell 2000, 101, 499–510.
  40. Rekler, D.; Kalcheim, C. Completion of neural crest cell production and emigration is regulated by retinoic-acid-dependent inhibition of BMP signaling. eLife 2022, 11, e72723.
  41. Martínez-Morales, P.L.; Del Diez Corral, R.; Olivera-Martínez, I.; Quiroga, A.C.; Das, R.M.; Barbas, J.A.; Storey, K.G.; Morales, A.V. FGF and retinoic acid activity gradients control the timing of neural crest cell emigration in the trunk. J. Cell Biol. 2011, 194, 489–503.
  42. Zhao, X.; Le, T.P.; Erhardt, S.; Findley, T.O.; Wang, J. Hippo-Yap Pathway Orchestrates Neural Crest Ontogenesis. Front. Cell Dev. Biol. 2021, 9, 706623.
  43. Kumar, D.; Nitzan, E.; Kalcheim, C. YAP promotes neural crest emigration through interactions with BMP and Wnt activities. Cell Commun. Signal. 2019, 17, 69.
  44. Hindley, C.J.; Condurat, A.L.; Menon, V.; Thomas, R.; Azmitia, L.M.; Davis, J.A.; Pruszak, J. The Hippo pathway member YAP enhances human neural crest cell fate and migration. Sci. Rep. 2016, 6, 23208.
  45. Manderfield, L.J.; Aghajanian, H.; Engleka, K.A.; Lim, L.Y.; Liu, F.; Jain, R.; Li, L.; Olson, E.N.; Epstein, J.A. Hippo signaling is required for Notch-dependent smooth muscle differentiation of neural crest. Development 2015, 142, 2962–2971.
  46. Dupin, E.; Calloni, G.W.; Coelho-Aguiar, J.M.; Le Douarin, N.M. The issue of the multipotency of the neural crest cells. Dev. Biol. 2018, 444, S47–S59.
  47. Rothstein, M.; Bhattacharya, D.; Simoes-Costa, M. The molecular basis of neural crest axial identity. Dev. Biol. 2018, 444 (Suppl. S1), 170–180.
  48. Abzhanov, A.; Tzahor, E.; Lassar, A.B.; Tabin, C.J. Dissimilar regulation of cell differentiation in mesencephalic (cranial) and sacral (trunk) neural crest cells in vitro. Development 2003, 130, 4567–4579.
  49. Solovieva, T.; Bronner, M. Schwann cell precursors: Where they come from and where they go. Cells Dev. 2021, 166, 203686.
  50. Rocha, M.; Beiriger, A.; Kushkowski, E.E.; Miyashita, T.; Singh, N.; Venkataraman, V.; Prince, V.E. From head to tail: Regionalization of the neural crest. Development 2020, 147, dev193888.
  51. Mehrotra, P.; Tseropoulos, G.; Bronner, M.E.; Andreadis, S.T. Adult tissue-derived neural crest-like stem cells: Sources, regulatory networks, and translational potential. Stem Cells Transl. Med. 2020, 9, 328–341.
  52. Monk, K.R.; Feltri, M.L.; Taveggia, C. New insights on Schwann cell development. Glia 2015, 63, 1376–1393.
  53. Le Douarin, N.; Kalcheim, C. The Neural Crest, 2nd ed.; Cambridge University Press: Cambridge, UK, 1999; ISBN 9780511897948.
  54. Fledrich, R.; Kungl, T.; Nave, K.-A.; Stassart, R.M. Axo-glial interdependence in peripheral nerve development. Development 2019, 146, dev151704.
  55. Furlan, A.; Adameyko, I. Schwann cell precursor: A neural crest cell in disguise? Dev. Biol. 2018, 444 (Suppl. S1), 25–35.
  56. Jessen, K.R.; Mirsky, R.; Lloyd, A.C. Schwann Cells: Development and Role in Nerve Repair. Cold Spring Harb. Perspect. Biol. 2015, 7, a020487.
  57. Leimeroth, R.; Lobsiger, C.; Lüssi, A.; Taylor, V.; Suter, U.; Sommer, L. Membrane-bound neuregulin1 type III actively promotes Schwann cell differentiation of multipotent Progenitor cells. Dev. Biol. 2002, 246, 245–258.
  58. Meyer, D.; Yamaai, T.; Garratt, A.; Riethmacher-Sonnenberg, E.; Kane, D.; Theill, L.E.; Birchmeier, C. Isoform-specific expression and function of neuregulin. Development 1997, 124, 3575–3586.
  59. Dong, Z.; Brennan, A.; Liu, N.; Yarden, Y.; Lefkowitz, G.; Mirsky, R.; Jessen, K.R. Neu differentiation factor is a neuron-glia signal and regulates survival, proliferation, and maturation of rat schwann cell precursors. Neuron 1995, 15, 585–596.
  60. Grinspan, J.B.; Marchionni, M.A.; Reeves, M.; Coulaloglou, M.; Scherer, S.S. Axonal interactions regulate Schwann cell apoptosis in developing peripheral nerve: Neuregulin receptors and the role of neuregulins. J. Neurosci. J. Soc. Neurosci. 1996, 16, 6107–6118.
  61. Syroid, D.E.; Maycox, P.R.; Burrola, P.G.; Liu, N.; Wen, D.; Lee, K.F.; Lemke, G.; Kilpatrick, T.J. Cell death in the Schwann cell lineage and its regulation by neuregulin. Proc. Natl. Acad. Sci. USA 1996, 93, 9229–9234.
  62. Kastriti, M.E.; Faure, L.; Von Ahsen, D.; Bouderlique, T.G.; Boström, J.; Solovieva, T.; Jackson, C.; Bronner, M.; Meijer, D.; Hadjab, S.; et al. Schwann cell precursors represent a neural crest-like state with biased multipotency. EMBO J. 2022, 41, e108780.
  63. Kalcheim, C.; Kumar, D. Cell fate decisions during neural crest ontogeny. Int. J. Dev. Biol. 2017, 61, 195–203.
  64. Adameyko, I.; Lallemend, F.; Aquino, J.B.; Pereira, J.A.; Topilko, P.; Müller, T.; Fritz, N.; Beljajeva, A.; Mochii, M.; Liste, I.; et al. Schwann cell precursors from nerve innervation are a cellular origin of melanocytes in skin. Cell 2009, 139, 366–379.
  65. Muppirala, A.N.; Limbach, L.E.; Bradford, E.F.; Petersen, S.C. Schwann cell development: From neural crest to myelin sheath. Wiley Interdiscip. Rev. Dev. Biol. 2021, 10, e398.
  66. Joseph, N.M.; Mukouyama, Y.; Mosher, J.T.; Jaegle, M.; Crone, S.A.; Dormand, E.-L.; Lee, K.-F.; Meijer, D.; Anderson, D.J.; Morrison, S.J. Neural crest stem cells undergo multilineage differentiation in developing peripheral nerves to generate endoneurial fibroblasts in addition to Schwann cells. Development 2004, 131, 5599–5612.
  67. Dyachuk, V.; Furlan, A.; Shahidi, M.K.; Giovenco, M.; Kaukua, N.; Konstantinidou, C.; Pachnis, V.; Memic, F.; Marklund, U.; Müller, T.; et al. Neurodevelopment. Parasympathetic neurons originate from nerve-associated peripheral glial progenitors. Science 2014, 345, 82–87.
  68. Espinosa-Medina, I.; Outin, E.; Picard, C.A.; Chettouh, Z.; Dymecki, S.; Consalez, G.G.; Coppola, E.; Brunet, J.-F. Neurodevelopment. Parasympathetic ganglia derive from Schwann cell precursors. Science 2014, 345, 87–90.
  69. Uesaka, T.; Nagashimada, M.; Enomoto, H. Neuronal Differentiation in Schwann Cell Lineage Underlies Postnatal Neurogenesis in the Enteric Nervous System. J. Neurosci. 2015, 35, 9879–9888.
  70. Furlan, A.; Dyachuk, V.; Kastriti, M.E.; Calvo-Enrique, L.; Abdo, H.; Hadjab, S.; Chontorotzea, T.; Akkuratova, N.; Usoskin, D.; Kamenev, D.; et al. Multipotent peripheral glial cells generate neuroendocrine cells of the adrenal medulla. Science 2017, 357, eaal3753.
  71. Kastriti, M.E.; Kameneva, P.; Kamenev, D.; Dyachuk, V.; Furlan, A.; Hampl, M.; Memic, F.; Marklund, U.; Lallemend, F.; Hadjab, S.; et al. Schwann Cell Precursors Generate the Majority of Chromaffin Cells in Zuckerkandl Organ and Some Sympathetic Neurons in Paraganglia. Front. Mol. Neurosci. 2019, 12, 6.
  72. Kaukua, N.; Shahidi, M.K.; Konstantinidou, C.; Dyachuk, V.; Kaucka, M.; Furlan, A.; An, Z.; Wang, L.; Hultman, I.; Ahrlund-Richter, L.; et al. Glial origin of mesenchymal stem cells in a tooth model system. Nature 2014, 513, 551–554.
  73. Xie, M.; Kamenev, D.; Kaucka, M.; Kastriti, M.E.; Zhou, B.; Artemov, A.V.; Storer, M.; Fried, K.; Adameyko, I.; Dyachuk, V.; et al. Schwann cell precursors contribute to skeletal formation during embryonic development in mice and zebrafish. Proc. Natl. Acad. Sci. USA 2019, 116, 15068–15073.
  74. Erickson, A.G.; Kameneva, P.; Adameyko, I. The transcriptional portraits of the neural crest at the individual cell level. Semin. Cell Dev. Biol. 2022, in press.
  75. Lucas, T.A.; Zhu, L.; Buckwalter, M.S. Spleen glia are a transcriptionally unique glial subtype interposed between immune cells and sympathetic axons. Glia 2021, 69, 1799–1815.
  76. Barlow-Anacker, A.J.; Fu, M.; Erickson, C.S.; Bertocchini, F.; Gosain, A. Neural Crest Cells Contribute an Astrocyte-like Glial Population to the Spleen. Sci. Rep. 2017, 7, 45645.
  77. Belle, M.; Godefroy, D.; Couly, G.; Malone, S.A.; Collier, F.; Giacobini, P.; Chédotal, A. Tridimensional Visualization and Analysis of Early Human Development. Cell 2017, 169, 161–173.e12.
  78. Jessen, K.R.; Brennan, A.; Morgan, L.; Mirsky, R.; Kent, A.; Hashimoto, Y.; Gavrilovic, J. The schwann cell precursor and its fate: A study of cell death and differentiation during gliogenesis in rat embryonic nerves. Neuron 1994, 12, 509–527.
  79. Kastriti, M.E.; Adameyko, I. Specification, plasticity and evolutionary origin of peripheral glial cells. Curr. Opin. Neurobiol. 2017, 47, 196–202.
  80. D’Antonio, M.; Michalovich, D.; Paterson, M.; Droggiti, A.; Woodhoo, A.; Mirsky, R.; Jessen, K.R. Gene profiling and bioinformatic analysis of Schwann cell embryonic development and myelination. Glia 2006, 53, 501–515.
  81. Buchstaller, J.; Sommer, L.; Bodmer, M.; Hoffmann, R.; Suter, U.; Mantei, N. Efficient isolation and gene expression profiling of small numbers of neural crest stem cells and developing Schwann cells. J. Neurosci. 2004, 24, 2357–2365.
  82. Wanner, I.B.; Guerra, N.K.; Mahoney, J.; Kumar, A.; Wood, P.M.; Mirsky, R.; Jessen, K.R. Role of N-cadherin in Schwann cell precursors of growing nerves. Glia 2006, 54, 439–459.
  83. Meier, C.; Parmantier, E.; Brennan, A.; Mirsky, R.; Jessen, K.R. Developing Schwann Cells Acquire the Ability to Survive without Axons by Establishing an Autocrine Circuit Involving Insulin-Like Growth Factor, Neurotrophin-3, and Platelet-Derived Growth Factor-BB. J. Neurosci. 1999, 19, 3847–3859.
  84. Feltri, M.L.; Poitelon, Y.; Previtali, S.C. How Schwann Cells Sort Axons: New Concepts. Neuroscientist 2016, 22, 252–265.
  85. Woodhoo, A.; Alonso, M.B.D.; Droggiti, A.; Turmaine, M.; D’Antonio, M.; Parkinson, D.B.; Wilton, D.K.; Al-Shawi, R.; Simons, P.; Shen, J.; et al. Notch controls embryonic Schwann cell differentiation, postnatal myelination and adult plasticity. Nat. Neurosci. 2009, 12, 839–847.
  86. Stewart, H.J.; Morgan, L.; Jessen, K.R.; Mirsky, R. Changes in DNA synthesis rate in the Schwann cell lineage in vivo are correlated with the precursor—Schwann cell transition and myelination. Eur. J. Neurosci. 1993, 5, 1136–1144.
  87. Brown, M. Schwann cell proliferation in the postnatal mouse: Timing and topography. Exp. Neurol. 1981, 74, 170–186.
  88. Barik, A.; Li, L.; Sathyamurthy, A.; Xiong, W.-C.; Mei, L. Schwann Cells in Neuromuscular Junction Formation and Maintenance. J. Neurosci. 2016, 36, 9770–9781.
  89. Georgiou, J.; Charlton, M.P. Non-myelin-forming perisynaptic Schwann cells express protein zero and myelin-associated glycoprotein. Glia 1999, 27, 101–109.
  90. Suazo, I.; Vega, J.A.; García-Mesa, Y.; García-Piqueras, J.; García-Suárez, O.; Cobo, T. The Lamellar Cells of Vertebrate Meissner and Pacinian Corpuscles: Development, Characterization, and Functions. Front. Neurosci. 2022, 16, 790130.
  91. Etxaniz, U.; Pérez-San Vicente, A.; Gago-López, N.; García-Dominguez, M.; Iribar, H.; Aduriz, A.; Pérez-López, V.; Burgoa, I.; Irizar, H.; Muñoz-Culla, M.; et al. Neural-competent cells of adult human dermis belong to the Schwann lineage. Stem Cell Rep. 2014, 3, 774–788.
  92. Taveggia, C.; Zanazzi, G.; Petrylak, A.; Yano, H.; Rosenbluth, J.; Einheber, S.; Xu, X.; Esper, R.M.; Loeb, J.A.; Shrager, P.; et al. Neuregulin-1 type III determines the ensheathment fate of axons. Neuron 2005, 47, 681–694.
  93. Petersen, S.C.; Luo, R.; Liebscher, I.; Giera, S.; Jeong, S.-J.; Mogha, A.; Ghidinelli, M.; Feltri, M.L.; Schöneberg, T.; Piao, X.; et al. The adhesion GPCR GPR126 has distinct, domain-dependent functions in Schwann cell development mediated by interaction with laminin-211. Neuron 2015, 85, 755–769.
  94. Mogha, A.; Benesh, A.E.; Patra, C.; Engel, F.B.; Schöneberg, T.; Liebscher, I.; Monk, K.R. Gpr126 functions in Schwann cells to control differentiation and myelination via G-protein activation. J. Neurosci. 2013, 33, 17976–17985.
  95. Monk, K.R.; Oshima, K.; Jörs, S.; Heller, S.; Talbot, W.S. Gpr126 is essential for peripheral nerve development and myelination in mammals. Development 2011, 138, 2673–2680.
  96. Monk, K.R.; Naylor, S.G.; Glenn, T.D.; Mercurio, S.; Perlin, J.R.; Dominguez, C.; Moens, C.B.; Talbot, W.S. A G protein-coupled receptor is essential for Schwann cells to initiate myelination. Science 2009, 325, 1402–1405.
  97. Topilko, P.; Schneider-Maunoury, S.; Levi, G.; Baron-Van Evercooren, A.; Chennoufi, A.B.; Seitanidou, T.; Babinet, C.; Charnay, P. Krox-20 controls myelination in the peripheral nervous system. Nature 1994, 371, 796–799.
  98. Glenn, T.D.; Talbot, W.S. Analysis of Gpr126 function defines distinct mechanisms controlling the initiation and maturation of myelin. Development 2013, 140, 3167–3175.
  99. Michailov, G.V.; Sereda, M.W.; Brinkmann, B.G.; Fischer, T.M.; Haug, B.; Birchmeier, C.; Role, L.; Lai, C.; Schwab, M.H.; Nave, K.-A. Axonal neuregulin-1 regulates myelin sheath thickness. Science 2004, 304, 700–703.
  100. Faroni, A.; Castelnovo, L.F.; Procacci, P.; Caffino, L.; Fumagalli, F.; Melfi, S.; Gambarotta, G.; Bettler, B.; Wrabetz, L.; Magnaghi, V. Deletion of GABA-B receptor in Schwann cells regulates remak bundles and small nociceptive C-fibers. Glia 2014, 62, 548–565.
  101. Procacci, P.; Ballabio, M.; Castelnovo, L.F.; Mantovani, C.; Magnaghi, V. GABA-B receptors in the PNS have a role in Schwann cells differentiation? Front. Cell. Neurosci. 2012, 6, 68.
  102. Magnaghi, V.; Ballabio, M.; Cavarretta, I.T.R.; Froestl, W.; Lambert, J.J.; Zucchi, I.; Melcangi, R.C. GABAB receptors in Schwann cells influence proliferation and myelin protein expression. Eur. J. Neurosci. 2004, 19, 2641–2649.
  103. Milichko, V.; Dyachuk, V. Novel Glial Cell Functions: Extensive Potency, Stem Cell-Like Properties, and Participation in Regeneration and Transdifferentiation. Front. Cell Dev. Biol. 2020, 8, 809.
  104. Boerboom, A.; Dion, V.; Chariot, A.; Franzen, R. Molecular Mechanisms Involved in Schwann Cell Plasticity. Front. Mol. Neurosci. 2017, 10, 38.
  105. Stierli, S.; Napoli, I.; White, I.J.; Cattin, A.-L.; Monteza Cabrejos, A.; Garcia Calavia, N.; Malong, L.; Ribeiro, S.; Nihouarn, J.; Williams, R.; et al. The regulation of the homeostasis and regeneration of peripheral nerve is distinct from the CNS and independent of a stem cell population. Development 2018, 145, dev170316.
  106. Aguayo, A.J.; Epps, J.; Charron, L.; Bray, G.M. Multipotentiality of Schwann cells in cross-anastomosed and grafted myelinated and unmyelinated nerves: Quantitative microscopy and radioautography. Brain Res. 1976, 104, 1–20.
  107. El-Nachef, W.N.; Bronner, M.E. De novo enteric neurogenesis in post-embryonic zebrafish from Schwann cell precursors rather than resident cell types. Development 2020, 147, dev186619.
  108. Fröb, F.; Wegner, M. The role of chromatin remodeling complexes in Schwann cell development. Glia 2020, 68, 1596–1603.
  109. Jacob, C. Chromatin-remodeling enzymes in control of Schwann cell development, maintenance and plasticity. Curr. Opin. Neurobiol. 2017, 47, 24–30.
  110. Ma, K.H.; Hung, H.A.; Svaren, J. Epigenomic Regulation of Schwann Cell Reprogramming in Peripheral Nerve Injury. J. Neurosci. 2016, 36, 9135–9147.
More
This entry is offline, you can click here to edit this entry!
Video Production Service