Neural crest cells are crucial in development, not least because of their remarkable multipotency. Early findings stimulated two hypotheses for how fate specification and commitment from fully multipotent neural crest cells might occur, progressive fate restriction (PFR) and direct fate restriction, differing in whether partially restricted intermediates were involved. Initially hotly debated, they remain unreconciled, although PFR has become favoured. However, testing of a PFR hypothesis of zebrafish pigment cell development refutes this view. We propose a novel ‘cyclical fate restriction’ hypothesis, based upon a more dynamic view of transcriptional states, reconciling the experimental evidence underpinning the traditional hypotheses.

Neural crest cells (NCCs; see Glossary, Box 1) are vital for vertebrate development, and are a key model system for developmental biology. They are ectodermally derived, undergoing delamination (see Glossary, Box 1) and pausing in the premigratory ‘staging area’ near the dorsal neural tube (Marusich and Weston, 1991), before migrating extensively throughout the body. They generate diverse cell types, including most of the peripheral nervous system (PNS), all body pigment cells and skeletogenic cell types (so called ‘ectomesenchymal cell fates’) (Le Douarin and Kalcheim, 1999). NCCs can be divided into cranial and trunk populations, which differ in both their migration pathways and fate repertoire, with skeletogenic fates generally confined to the head. The isolation and in vitro characterisation of neural crest-derived stem cells, known as neural crest stem cells (NCSCs; see Glossary, Box 1), has added further interest to NCCs and has provided a controlled experimental paradigm for defining the molecular basis for fate specification and differentiation (see Glossary, Box 1).

Box 1. Glossary

Delamination. The process of NCCs undergoing epithelial-mesenchymal transition, exiting the neural epithelium to become mesenchymal.

Differentiation. The process of acquiring the specific morphological and transcriptional markers characteristic of an individual cell type (fate). We envisage differentiation as a continuum, a dynamic process in which a cell activates (or maintains, likely at elevated levels) expression of fate-specific transcription factors, gradually activating the transcriptional programme that results in adoption of the differentiated phenotype.

Fate commitment. The process whereby a cell stabilises (‘locks in’) its fate choice, i.e. the terminal stage of fate restriction. This is generally considered to involve loss of multipotency, resulting from epigenetic modification of the genome to ‘fix’ a specific transcriptional programme (ensuring unipotency). It is generally considered the terminal state of differentiation, but the realisation that some stem cells (e.g. neural stem cells in the central nervous system) adopt a distinctive differentiated phenotype means that caution needs to be exercised – a differentiated (‘specialised’) phenotype does not necessarily imply fate commitment or unipotency. Traditionally, demonstration of fate commitment requires a technically challenging transplantation experiment, but we consider that it could probably be assessed by examination of the transcriptome of a cell (provided that sequencing is sufficiently deep).

Fate potential (potency). The capacity of a progenitor cell to generate a defined differentiated cell type. In theory, this could be revealed in clonal cell culture, if we assume that culture conditions are suitable for all potential cell types and that all can be simultaneously distinguished by markers. In reality, clone size and other stochastic factors may vary the combination of cell types generated. Traditionally considered impossible to assess definitively in vivo, single-cell RNA sequencing may allow a glimpse into cell fate potential (see ‘Fate specification’ below).

Fate restriction. The process whereby a multipotent progenitor cell adopts (i.e. becomes restricted to) a specific fate. Cells may be described as partially fate restricted, when they can adopt one of a subset of fates, but are unable to adopt (i.e. restricted from adopting) others. However, confusingly, in a clonal study, a cell is considered a fate-restricted precursor when all its progeny adopt a single fate, but it should be noted that, although consistent with fate commitment (i.e. unipotency), it does not prove fate commitment, because limited environmental signals, small clone sizes and other factors can limit the clone's ability to display its full potency.

Fate specification. Multipotent cells are defined as showing fate specification as soon as fate-specific markers are detectable. It is crucial to remember that this expression is labile, and does not imply commitment. However, this term is limited by our ability to detect more than one marker simultaneously, with traditional whole-mount in situ hybridisation or immunofluorescence studies rarely allowing more than two or three markers to be assessed, and usually only one. Where considered, such studies may show co-expression of markers of different cell types, indicating that fate specification is clearly not the same as fate commitment (Petratou et al., 2021, 2018). New techniques, including single-cell RNA sequencing and in situ sequencing now allow assessment of ten to thousands of mRNA transcripts, revolutionising our ability to detect, and to distinguish, fate specification from fate commitment. We consider that expression of one or more fate-specific markers indicates at least the potential to differentiate into that cell type, and thus that at least a minimal estimate of the fate potential of a cell might be deduced from sufficiently deep transcriptional profiling.

Fully multipotent. A progenitor or stem cell is fully multipotent when it is still able to adopt any of its characteristic derivative fates. The term pluripotency was used in some of the early NC literature to distinguish cells generating all NC derivatives (i.e. pluripotent NCCs), but as the stem cell biology field has blossomed, so the meaning of this term has become more widely accepted to mean ‘capable of generating all embryonic (as opposed to extra-embryonic) cells’.

Migratory. NCCs moving around the body, usually on defined migratory pathways in the trunk and tail, but more broadly in the head. Such cells are not usually visibly differentiated (e.g. melanised) in mouse or chick, but in fish and amphibians they are often partially differentiated (e.g. melanised or displaying other pigments).

Multipotent. A progenitor or stem cell is multipotent when it is still able to adopt two or more of its characteristic derivative fates.

Neural crest cells(NCCs). Any of the numerous mesenchymal cells generated by the delamination of dorsal neural tube cells during embryonic (somitogenesis stages) development.

Neural crest stem cells(NCSCs). Originally isolated from embryonic mammals as a subset of NCCs, these are NC-derived cells that undergo extensive self-renewal and retain the potential to differentiate into one or more NC-derived cell types. Such cells can be derived from many post-migratory locations in embryos and even in adults, including skin and the PNS, reflecting the widespread maintenance of cells functioning in homeostasis (Delfino-Machin et al., 2007). Such cells in different locations normally generate only a subset of fates, and so may be named accordingly, but in at least some cases their potency has been shown to be considerably broader when environmental influences are changed by cell culture (e.g. Nishimura et al., 2002, 2010), leading to a much wider potency in vitro (Watanabe et al., 2016). This highlights a key inadequacy of the naming conventions within the literature. Long-term persistence of such cells, often incorporated within a stem cell definition, has been less thoroughly investigated, but the ready isolation from adult tissues implies that this feature exists here too. The term is also used more loosely for early NCCs in their fully multipotent form. Note that direct demonstration of these properties has not been performed in a zebrafish context, so that the use of the term NCSC is somewhat provisional.

Pre-delamination. NCCs still in the neural plate/dorsal neural tube.

Premigratory. NCCs sitting adjacent to the dorsal neural tube, a position designated the ‘staging area’ (Marusich and Weston, 1991), prior to migration.

Post-migratory. NCCs in their terminal positions. Such cells may initially be undifferentiated, but will often soon become (fully) differentiated, contributing to the physiological functions of the relevant organ or tissue.

Transdifferentiation. Transition of a cell of one differentiated morphology into the differentiated morphology characteristic of a distinct cell type, without dedifferentiation into an undifferentiated progenitor state.

Unipotent. A progenitor or stem cell is unipotent when it is stably committed to adopting one of its characteristic derivative fates (i.e. fate committed).

Our interest here is in fate restriction (see Glossary, Box 1), the process whereby NCCs become committed to individual fates. Early in vivo labelling and transplant experiments in avian embryos demonstrated the remarkable fate potential, or potency (see Glossary, Box 1), of NCCs, with heterotopic transplantation showing that the potential of NCC populations was even greater than actually exhibited in vivo (Le Douarin, 1986). Early discussions debated two extreme hypotheses for the potency of premigratory NCCs: (1) NCCs are initially homogeneous, fully multipotent cells (see Glossary, Box 1); or (2) the neural crest (NC) is a heterogeneous mixture of predetermined unipotent cells. Of course, it was acknowledged that the NC might consist of a mixture of these, perhaps with some fates specified independently, whereas others are derived from (nearly fully) multipotent progenitors (Fraser and Fraser, 1991; Vogel and Weston, 1988; see also Weston and Thiery, 2015).

Closely entwined with the issue of multipotency, was the question of when and how fate choices were made: if NCCs are fully multipotent, then fate restriction was likely to depend upon instructive cues received during migration or at their destination, but if unipotent, then their migration would likely be targeted to appropriate locations (Fraser and Bronner-Fraser, 1991; Vogel and Weston, 1988). Alternatively, cells might migrate randomly, with appropriate cell types selected for survival by regional trophic factors (Le Douarin, 1986). Clonal analysis of chick and mouse NCCs in primary cultures led to the conclusion that they were multipotent (see Glossary, Box 1) (Dupin et al., 1990; Ito et al., 1993; Ito and Sieber-Blum, 1993; Sieber-Blum, 1989; Sieber-Blum and Cohen, 1980). However, this still left the question of how fully multipotent cells became committed to single fates (i.e. unipotent; see Glossary, Box 1), and in particular whether or not cells of intermediate potency were involved. Work in the 1980s resulted in two distinct hypotheses, progressive fate restriction (PFR) and direct fate restriction (DFR), for how this might work (Fig. 1A-F).

Fig. 1.

Progressive and direct fate restriction hypotheses for NCC development. (A,B) Schemes show progressive (A) and direct (B) fate-restriction of NCCs, as deduced from mouse and chick studies. (A) A multipotent trunk NCC progenitor (multicoloured cell in green, purple, pink and black) produces a heterogeneous population of intermediate progenitors, here shown as bipotent sensory (purple and green) and autonomic ganglial (green and pink), but also tripotent shared autonomic ganglial and melanocyte progenitor (green, pink and black), prior to generating fate-restricted derivatives. (B) A multipotent NCC progenitor generates single-fate restricted cells: sensory neuroblast (Sn; purple); autonomic neuroblast (An; pink), glioblast (Gb; green); melanoblast (Mb; black) during or after migration. (C-F) Progressive and direct fate restriction placed in an anatomical context. (C,D) NCCs induced at the lateral border of the neural plate (NP) are considered to be fully multipotent in both hypotheses. Distinctions between the hypotheses become clear at later stages, perhaps from delamination, but especially during NCC migration. (E) Under the progressive fate restriction hypothesis, intermediates of a wide-range of partially and fully restricted potencies are rapidly segregated in premigratory NCCs [perhaps even beginning pre-delamination (see Glossary, Box 1)]. Migrating progenitors adopt routes appropriate to their potency, with melanocyte progenitors (black) in mouse and chick utilising exclusively the dorsolateral migration pathway between the epidermis and somites, and peripheral ganglial progenitors (purple and green, and pink and green) utilising the medial migration pathway between the somites and the neural tube, notochord and dorsal aorta. Ganglial progenitors accumulate in nascent ganglia, where neuronal and glial differentiation occur. Environmental cues, shown as shaded ovals, are considered to reinforce fate-restriction decisions, for example by controlling accessibility to dorsolateral migration pathway, restricting it to melanoblasts (black circle), or influencing aggregation of ganglial progenitors and the subsequent differentiation of both neuronal and glial fates (purple, pink and green circles). (F) Under the direct fate restriction hypothesis, delaminated premigratory and migrating NCCs retain full multipotency, until exposed to differentiation cues (coloured ovals) in the migratory/post-migratory environment, triggering direct differentiation into specific cell types in response to environment differentiation signals (green, purple, pink and black circles). For simplicity, the figure focuses on derivatives of trunk NCCs, although the original progressive fate restriction hypotheses also emphasised the derivation of ectomesenchymal fates (e.g. cartilage) from the cranial NC; in Weston's hypothesis this was seen as the first fate to segregate, whereas in Le Douarin's hypothesis cartilage-generating progenitors were of diverse potencies and hence likely to persist longer. Mesoderm is indicated in E and F only, and is simplified as somites (Som), without indicating segregation of sclerotome and dermomyotome; neural tube (NT), notochord (Nc), dorsal aorta (DA) and epidermis (Ep) are also indicated to delineate key dorsolateral and medial NC migration pathways. A, autonomic neuron; G, glial cell; M, melanocyte; S, sensory neuron.

Fig. 1.

Progressive and direct fate restriction hypotheses for NCC development. (A,B) Schemes show progressive (A) and direct (B) fate-restriction of NCCs, as deduced from mouse and chick studies. (A) A multipotent trunk NCC progenitor (multicoloured cell in green, purple, pink and black) produces a heterogeneous population of intermediate progenitors, here shown as bipotent sensory (purple and green) and autonomic ganglial (green and pink), but also tripotent shared autonomic ganglial and melanocyte progenitor (green, pink and black), prior to generating fate-restricted derivatives. (B) A multipotent NCC progenitor generates single-fate restricted cells: sensory neuroblast (Sn; purple); autonomic neuroblast (An; pink), glioblast (Gb; green); melanoblast (Mb; black) during or after migration. (C-F) Progressive and direct fate restriction placed in an anatomical context. (C,D) NCCs induced at the lateral border of the neural plate (NP) are considered to be fully multipotent in both hypotheses. Distinctions between the hypotheses become clear at later stages, perhaps from delamination, but especially during NCC migration. (E) Under the progressive fate restriction hypothesis, intermediates of a wide-range of partially and fully restricted potencies are rapidly segregated in premigratory NCCs [perhaps even beginning pre-delamination (see Glossary, Box 1)]. Migrating progenitors adopt routes appropriate to their potency, with melanocyte progenitors (black) in mouse and chick utilising exclusively the dorsolateral migration pathway between the epidermis and somites, and peripheral ganglial progenitors (purple and green, and pink and green) utilising the medial migration pathway between the somites and the neural tube, notochord and dorsal aorta. Ganglial progenitors accumulate in nascent ganglia, where neuronal and glial differentiation occur. Environmental cues, shown as shaded ovals, are considered to reinforce fate-restriction decisions, for example by controlling accessibility to dorsolateral migration pathway, restricting it to melanoblasts (black circle), or influencing aggregation of ganglial progenitors and the subsequent differentiation of both neuronal and glial fates (purple, pink and green circles). (F) Under the direct fate restriction hypothesis, delaminated premigratory and migrating NCCs retain full multipotency, until exposed to differentiation cues (coloured ovals) in the migratory/post-migratory environment, triggering direct differentiation into specific cell types in response to environment differentiation signals (green, purple, pink and black circles). For simplicity, the figure focuses on derivatives of trunk NCCs, although the original progressive fate restriction hypotheses also emphasised the derivation of ectomesenchymal fates (e.g. cartilage) from the cranial NC; in Weston's hypothesis this was seen as the first fate to segregate, whereas in Le Douarin's hypothesis cartilage-generating progenitors were of diverse potencies and hence likely to persist longer. Mesoderm is indicated in E and F only, and is simplified as somites (Som), without indicating segregation of sclerotome and dermomyotome; neural tube (NT), notochord (Nc), dorsal aorta (DA) and epidermis (Ep) are also indicated to delineate key dorsolateral and medial NC migration pathways. A, autonomic neuron; G, glial cell; M, melanocyte; S, sensory neuron.

The PFR hypothesis was proposed independently by Weston and Le Douarin (Baroffio et al., 1988, 1991; Le Douarin, 1986; Weston, 1982, 1983, 1991). Noting the evidence for heterogeneity of marker expression in even premigratory or early migratory (see Glossary, Box 1) NCCs (e.g. Barald, 1988a,b; Barbu et al., 1986; Ciment and Weston, 1982, 1985; Henion et al., 1995; Kahane and Kalcheim, 1994; Tessarollo et al., 1993; Wehrle-Haller and Weston, 1995), Weston proposed that segregation of developmentally restricted subpopulations occurs progressively and in a specific sequence, with very early segregation of ectomesenchymal, and then primary sensory neuron, fates (Fig. 1A,C,E); at least some of these subpopulations have become distinct in the premigratory NCCs (Weston, 1991). Le Douarin's group built on the pioneering studies of Sieber-Blum and Cohen using single cell clones of NC in culture, which showed that primary NCCs were generally not unipotent, but also indicated considerable heterogeneity in vitro (Sieber-Blum and Cohen, 1980). Le Douarin and colleagues showed that early migrating NCCs generate both clones consistent with fully multipotent cells, and a broad range of clone sizes and cell-type compositions interpreted as showing PFR during migration (Baroffio et al., 1988, 1991). They proposed that different developmental fates form by progressive restriction of fully multipotent progenitors via partially restricted cell types, publishing an early version of the now classic textbook figure (Gilbert and Barresi, 2016). Numerous studies using these 2D cultures (Calloni et al., 2007, 2009; Lahav et al., 1998, 1996; Trentin et al., 2004), but also more recent 3D cultures of NCSCs (Mohlin et al., 2019), have demonstrated the multipotency, but also the apparent heterogeneity, of many premigratory and migrating chick and mammalian NCCs (reviewed by Dupin et al., 2018).

The DFR hypothesis was proposed based upon clonal analysis of NCC fates in vivo using iontophoretic labelling (whereby locally applied electrical current is used to drive a charged fluorescent dye into a single cell or small groups of cells) of chick trunk dorsal neural tube and premigratory NCCs (Bronner-Fraser and Fraser, 1989, 1988, 1991). These experiments revealed that most labelled NCCs generated heterogeneous clones with multiple derivative cell types, with some including all the fates that could be distinguished, although a significant proportion consisted of only a single cell type. These authors proposed that NCCs were homogeneous multipotent cells, and that fate choices were imposed upon them by environmental cues late in or after migration (Bronner-Fraser and Fraser, 1989, 1991) (Fig. 1B,D,F). Clonal heterogeneity in cell culture and in vivo would then be explained by statistical effects of clone sizes and inconsistencies of environmental cues encountered.

Strong support for the DFR hypothesis resulted from the isolation of rat NCSCs by Anderson's group (Stemple and Anderson, 1992). An elegant series of studies defined the key extracellular signals driving NCSC fate specification and differentiation, and demonstrated that these acted instructively, rather than simply selecting out a subset of cells pre-specified to individual fates (Kim et al., 2003; Lo et al., 1997; Morrison et al., 2000; Perez et al., 1999; Shah et al., 1996, 1994). Although these studies were limited in the fates assessed, they reinforced a key idea of the DFR hypothesis – that single cells choose directly between multiple fates, with environmental signals instructing the fate adopted.

The PFR and DFR hypotheses are distinguished by whether (PFR) or not (DFR) they transition through cells of reduced potency before adopting individual fates. A second distinction concerns when, and especially where, fate choices begin to be made: late in migration (DFR), or beginning in or adjacent to the neural tube (PFR). It will be apparent already that the PFR hypothesis matches the way we view development in general, readily integrating with Waddington's influential epigenetic landscape model (Waddington, 1940), but therein lies the importance and the excitement of this field: is it possible that NCCs differentiate in a different way, perhaps associated with their remarkable potential (Buitrago-Delgado et al., 2015)? These contrasting hypotheses were hotly debated throughout the 1990s. The debate was then largely forgotten, primarily because PFR became the accepted (‘textbook’) hypothesis, but also, in part, because the meaning of the term ‘multipotency’, originally used to mean ‘full multipotency’, has tended to drift towards a more generic ‘at least bipotent’. This is unfortunate, because it loses the essence of the debate – even in a PFR hypothesis most cells are at least bipotent! In recent years, certain key studies using single-cell resolution in vivo have reopened the discussion, but still most work has assumed a PFR interpretation.

This century, the PFR hypothesis has become dominant. Even the initial studies acknowledged that the data underpinning the DFR hypothesis did not rule out a PFR hypothesis (Anderson, 1989; Bronner-Fraser and Fraser, 1989; Fraser and Bronner-Fraser, 1991). In a later review of peripheral neuron development, Anderson concluded that segregation of sensory and autonomic lineages probably occurred prior to delamination (Anderson, 2000).

Delaminating chick NCCs were already fate restricted, but also emerged in a reproducible manner, filling more ventral locations (sympathetic ganglia) first, and then progressively occupying more dorsal ones [e.g. ventral root, dorsal root ganglia (DRG) and the skin] (Erickson et al., 1992; Kitamura et al., 1992; Reedy et al., 1998; Serbedzija et al., 1989). This was shown particularly convincingly by studies controlling carefully for time of delamination and for labelling of single cells (Krispin et al., 2010b; Nitzan et al., 2013a). Similarly, iontophoretic labelling of single premigratory NCCs in zebrafish showed that most were apparently already fate restricted (Dutton et al., 2001; Raible and Eisen, 1994; Schilling and Kimmel, 1994). Given the extensive evidence for variably multipotent (full to bipotent) migrating NCCs from primary chick culture noted before, the fate restriction demonstrated in chick in vivo is somewhat unexpected, but may reflect the combined impact of clone sizes and anatomical confinement of migration in vivo. Early fate restriction does not prove absence of multipotency (e.g. if migration is highly constrained), and thus does not strictly distinguish between the PFR and DFR hypotheses, but was consistent with early fate specification and fate-specific migration behaviour to target the appropriate locations.

This idea of fate restriction occurring prior to NCC migration was tested in mouse using the R26R-Confetti system to label a large sample of NCC clones with clonally distinguishable combinations of different coloured fluorescent proteins (Baggiolini et al., 2015). The authors combined clonal analysis of NCCs labelled genetically prior to delamination and in premigratory stages, with sophisticated statistical modelling to take account of proliferation rates and the relative size of the target site, and concluded that mouse NCCs at these stages show strong evidence for retained multipotency, in contrast to chick. However, in the context of our discussion, Baggiolini and colleagues defined ‘multipotent’ as ‘fated to form at least two cell types’, so that their data could be interpreted within a classic PFR hypothesis. The apparent contrasts between model systems is striking, and could be taken to indicate that there are species-specific differences in the timing of fate determination and/or regularity of migration.

Studies of fate specification have been highly limited by the number of markers that can be assessed simultaneously, making it impossible to make any authoritative statement of potency, even where fate specification was apparent, and reinforcing the difficulty of interpreting in vivo clonal studies showing apparent fate restriction (Box 1). However, with single-cell RNA-sequencing (scRNA-seq) offering a near-complete transcriptome, we would expect the potency of a cell to be reflected in the range of fate-specific markers that are expressed. In this context, a tour de force scRNA-seq study of mouse NC development apparently strongly reinforces the PFR hypothesis (Soldatov et al., 2019). Characterising NCCs expressing a fluorescent marker prior to delamination, NC fate specification towards skeletal and neural fates displayed an apparent pattern of sequential binary fate decisions during migration. Consistent with the conclusions of early segregation of sensory and autonomic lineages proposed previously (Anderson, 2000; Greenwood et al., 1999; Henion and Weston, 1997; Le Douarin, 1986; Perez et al., 1999; Sieber-Blum and Cohen, 1980; White et al., 2001; Ziller et al., 1983, 1987; Zirlinger et al., 2002), Soldatov and colleagues identified early segregation of sensory neuron precursors, followed by segregation of autonomic and mesenchymal progenitors. Mesenchymal fate segregation appeared not to be the primary decision, in contrast to early proposals (Weston, 1991), but in strong agreement with the apparent diversity of clones generating skeletal fates in clonal cell 2D and 3D cultures of chick NCCs (Calloni et al., 2007, 2009; Mohlin et al., 2019). This study clearly proposes a scheme highly consistent with the PFR hypothesis. However, it should be noted that (1) single-cell isolation methods usually focus (appropriately) on preserving the changing fate-specification signatures induced by in vivo environmental signals, rather than necessarily trying to assess the cells' fate potential, and (2) significant challenges remain in the reconstruction of developmental trajectories, even for state-of-the-art bioinformatics algorithms, as is evidenced by the necessary reliance on implicit notions, such as pseudotime, and the abundance of different algorithms available that attempt to make optimal choices of the trees describing bifurcation events, further complicated by the recent demonstration of non-binary fate choices and the dual origin of some cell types (Farrell et al., 2018).

A definitive test of the fate restriction mechanisms has been difficult to achieve. The classic textbook figure of PFR (Gilbert and Barresi, 2016) includes numerous intermediate cell types with various degrees of multipotency (see above), whereas the best-characterised examples in vivo are usually merely bipotent, with experimental support for well-defined progenitors with intermediate potency in vivo being limited (see above). Consequently, opportunities to challenge the PFR model experimentally have been lacking. One exception is the pigment cell system in fish, for which multiple bipotent progenitors have been suggested in vivo, but a multipotent intermediate has also been hypothesised (Bagnara et al., 1979).

The chromatoblast and bipotent pigment cell progenitors

Mammals only have a single pigment cell type (melanocyte), but most vertebrates (including zebrafish and medaka) have two or more pigment cell types (collectively known as chromatophores), including melanocytes (black), xanthophores (yellow), iridophores (iridescent, usually blue or silver), leucophores (white or cream) and others (Fujii, 1993; Schartl et al., 2016). The genetic accessibility of these cells has ensured that pigment cells are a well-studied ‘model-within-a-model’ for NC development (reviewed by Hashimoto et al., 2021). Here, we confine our attention to the proposed cell intermediates within pigment cell development in fish, which together formulate a widely accepted PFR model of pigment cell development.

Bagnara and colleagues proposed a common ‘chromatophore stem cell’ that gives rise to all (NC-derived) pigment cell types, but not to other NC derivatives (Bagnara et al., 1979). For the purposes of this Hypothesis article, we call this partially restricted progenitor a ‘chromatoblast’. Although the chromatoblast idea was rather speculative, being based, in part, on the transdifferentiation (see Glossary, Box 1) of pigment cell types in prolonged cell culture (e.g. Ide, 1986; Ide and Hama, 1976), the recent demonstration that pigment cell transdifferentiation contributes to normal metamorphic development in zebrafish (Lewis et al., 2019) strengthens the concept.

However, evidence of a chromatoblast has been hard to come by. Single-cell fate mapping of premigratory zebrafish NCCs has been inconclusive; most cells are apparently fate restricted, but the very small clone sizes characteristic of this species make interpretation of this result difficult (Dutton et al., 2001; Raible and Eisen, 1994; Schilling and Kimmel, 1994). All three zebrafish pigment cells are absent in colourless (sox10) mutants, but the peripheral nervous system is severely affected too, making the phenotype more consistent with the idea of a ‘non-ectomesenchymal progenitor’ rather than a chromatoblast (Dutton et al., 2001; Kelsh, 2006; Kelsh and Eisen, 2000); Sox10 has a similar role in medaka (Nagao et al., 2014). However, careful assessment of the zebrafish sox10 mutant phenotype has identified a subset of NCCs that are trapped in a premigratory position and co-express key pigment cell fate specification factors (including sox10, ltk, tfec); these cells were hypothesised to be the elusive chromatoblasts (Lopes et al., 2008; Petratou et al., 2018, 2021). scRNA-seq profiling, focused on adult and larval zebrafish, identified a ‘pigment cell precursor’ expressing sox10, mitfa and tfec, consistent with a putative chromatoblast (Howard et al., 2021; Saunders et al., 2019).

Several bipotent pigment cell progenitors have been inferred from the study of mutant phenotypes. Melanocytes are completely absent in nacre (mitfa) mutant zebrafish (Lister et al., 1999), consistent with the known role of Mitf in mammals as a master regulator of melanocyte development (Steingrimsson et al., 1994; Tassabehji et al., 1994). Alongside the absence of melanocytes, these zebrafish mutants display a substantial increase in iridophore numbers, leading to the proposal that both cell types derive from a bipotent ‘melanoiridoblast’. Lineage tracing of mitfa-expressing cells at 24 h post-fertilisation (hpf) indicated that these cells are post-mitotic and many develop as melanocytes or iridophores (Curran et al., 2010). Less is known about xanthophore fate specification, but studies of mutants for pax3, pax7 and sox5 in zebrafish and/or medaka have provided tantalising evidence for potential bipotent ‘melanoxanthoblasts’ and, especially, ‘xantholeucoblasts’ (Kimura et al., 2014; Minchin and Hughes, 2008; Nagao et al., 2014, 2018, 2016).

These studies lead, naturally, to a hypothesis of zebrafish pigment cell development that is explicitly a PFR hypothesis (Fig. 2). However, a study of zebrafish using sensitive NanoString detection of mRNA expression in individual NC-derived cells from several embryonic/early larval stages failed to detect cells showing signatures characteristic of a chromatoblast (mitfa, tfec and pax7, but not phox2b) or of bipotent pigment cell progenitors (e.g. mitfa and tfec, but not pax7, for melanoiridoblast) (Nikaido et al., 2021 preprint). Instead, progenitor cells that expressed the proposed chromatoblast fate-specification genes (e.g. mitfa, tfec, pax7, etc.) also expressed known fate-specification genes for neural fates (e.g. phox2b and sox10), leading to their interpretation as broadly multipotent intermediates (Nikaido et al., 2021 preprint).

Fig. 2.

A PFR hypothesis of zebrafish pigment cell development. The current working hypothesis of how zebrafish trunk NCCs generate the three distinct pigment cell types, shown as an adaptation of the general PFR hypothesis shown in Fig. 1. (A) It is assumed that the initial NC is fully multipotent, producing sensory and autonomic neurons, glia, and melanocytes, iridophores and xanthophores. A multipotent, but partially restricted progenitor of all the pigment cells (chromatoblast) has been proposed as an intermediate stage, as has a bipotent melanoiridoblast. A, autonomic neuron; An, autonomic neuroblast; G, glial cell; Gb, glioblast; I, iridophore; Ib, iridoblast; M, melanocyte; Mb, melanoblast; S, sensory neuron; Sn, sensory neuroblast; X, xanthophore; Xb, xanthoblast. (B) In an anatomical context, migrating pigment progenitors on the dorsolateral migration pathway are considered to be fate-specified melanoblasts and xanthoblasts, whereas cells on the medial pathway include both bipotent neural progenitors (indicated on left migration pathway) and pigment cell progenitors (right migration pathway), from which individual cell types emerge. Initially, migrating pigment progenitors show overlapping expression of marker genes consistent with melanoiridoblast status (Petratou et al., 2021, 2018). Note that the status of progenitors with respect to xanthophore fate has been less well-explored and is ignored here for the sake of simplicity. DA, dorsal aorta; Ep, epidermis; Nc, notochord; NT, neural tube; Som, somite.

Fig. 2.

A PFR hypothesis of zebrafish pigment cell development. The current working hypothesis of how zebrafish trunk NCCs generate the three distinct pigment cell types, shown as an adaptation of the general PFR hypothesis shown in Fig. 1. (A) It is assumed that the initial NC is fully multipotent, producing sensory and autonomic neurons, glia, and melanocytes, iridophores and xanthophores. A multipotent, but partially restricted progenitor of all the pigment cells (chromatoblast) has been proposed as an intermediate stage, as has a bipotent melanoiridoblast. A, autonomic neuron; An, autonomic neuroblast; G, glial cell; Gb, glioblast; I, iridophore; Ib, iridoblast; M, melanocyte; Mb, melanoblast; S, sensory neuron; Sn, sensory neuroblast; X, xanthophore; Xb, xanthoblast. (B) In an anatomical context, migrating pigment progenitors on the dorsolateral migration pathway are considered to be fate-specified melanoblasts and xanthoblasts, whereas cells on the medial pathway include both bipotent neural progenitors (indicated on left migration pathway) and pigment cell progenitors (right migration pathway), from which individual cell types emerge. Initially, migrating pigment progenitors show overlapping expression of marker genes consistent with melanoiridoblast status (Petratou et al., 2021, 2018). Note that the status of progenitors with respect to xanthophore fate has been less well-explored and is ignored here for the sake of simplicity. DA, dorsal aorta; Ep, epidermis; Nc, notochord; NT, neural tube; Som, somite.

In attempts to identify putative chromatoblasts in sox10 mutants, expression of leukocyte tyrosine kinase (ltk), which encodes a receptor crucial for iridophore fate specification, has been noted as characteristic of cells trapped in a putative pigment cell progenitor state (Lopes et al., 2008). In a direct test of the chromatoblast hypothesis, genetic fate mapping of these ltk-expressing cells showed that they generate all pigment cell types, but also peripheral glial and neuronal fates (Nikaido et al., 2021 preprint). Taking into account the single-cell NanoString data, it was proposed that these ltk-expressing cells were not chromatoblasts, but instead NC-derived highly multipotent progenitors (NC-HMPs), conflicting with the PFR hypothesis for pigment cell development (Nikaido et al., 2021 preprint) (Fig. 3). In the NanoString data, some cells from the NC-HMP cluster were, as expected, derived from earlier stages when premigratory NCCs are prominent (Nikaido et al., 2021 preprint); however, these multipotent cell clusters included many cells from later stages (early larval zebrafish; 3-5 days post-fertilisation), when most NCCs are considered to have differentiated. Consequently, these clusters were interpreted as including both widely multipotent NCCs from the earlier stages and also likely glial cells with retained multipotency from the later stages; such an interpretation was based in part on analogy to neural stem cells (Alvarez-Buylla et al., 2001; Obernier and Alvarez-Buylla, 2019; Than-Trong and Bally-Cuif, 2015), but also on studies of so-called adult ‘melanocyte stem cells’ (MSCs) in the zebrafish (Box 2).

Box 2. Adult NCSCs and MSCs

So-called MSCs, derived from the NC and set aside during embryonic development (Dooley et al., 2013), are the origin of numerous pigment cells in the adult. Fish show a prominent metamorphosis whereby body structure, including skin pigment pattern, is modified to generate the adult form. In zebrafish, de novo generated melanocytes, iridophores and some xanthophores, replace the embryonically derived early larval pattern, although many adult xanthophores seem to be generated through a process of dedifferentiation, proliferation and differentiation of embryonic xanthophores (Hultman and Johnson, 2010; Johnson et al., 1995; Mahalwar et al., 2014; McMenamin et al., 2014; Parichy et al., 1999, 2003; Quigley et al., 2004; Tryon et al., 2011; Walderich et al., 2016). These observations, plus those of regeneration of melanocytes after their chemical or physical ablation (Hultman et al., 2009; O'Reilly-Pol and Johnson, 2008; Yang and Johnson, 2006; Yang et al., 2004), suggested the presence of NC-derived stem cells, which normally remain quiescent until metamorphosis, but which can be activated for regeneration. The number, diversity and location of these cells remains poorly defined, although they are associated with the PNS, utilising the peripheral nerves to reach diverse locations in the skin (Budi et al., 2011; Dooley et al., 2013) and hypodermis (Iyengar et al., 2015). A breakthrough study identified a set of these stem cells residing in the DRG (Dooley et al., 2013), but indirect evidence indicates that they may be more widespread, associated with peripheral nerves (Camargo-Sosa et al., 2019). These stem cells are multipotent, with clones including all three pigment cell types, neurons and glia of the DRG and the PNS (Budi et al., 2011; Singh et al., 2016): originally named MSCs, a better name would therefore be adult NCSCs. Detailed studies of SCPs in birds and mammals identify them as an important source of melanocytes, as well as neurons and glia (Adameyko et al., 2009, 2012; Nitzan et al., 2013b), making it likely that these zebrafish adult NCSCs are their evolutionary equivalent.

Fig. 3.

Experimental test of the PFR hypothesis of zebrafish pigment cell development. (A) The original working hypothesis of pigment cell development, as shown in Fig. 2, with fully multipotent initial NC generating sensory and autonomic neurons, glia, and melanocytes, iridophores and xanthophores via multipotent, but partially restricted progenitors of all the pigment cells (chromatoblast) and a bipotent melanoiridoblast. (B) Revised hypothesis, based on findings of Nikaido et al. (2021 preprint). Owing to its unexpectedly broad multipotency, we propose the name ‘NC-derived highly multipotent progenitors’ (NC-HMPs) for the trunk NCCs; cells of intermediate potency were not detected. A, autonomic neuron; An, autonomic neuroblast; G, glial cell; Gb, glioblast; I, iridophore; Ib, iridoblast; M, melanocyte; Mb, melanoblast; S, sensory neuron; Sn, sensory neuroblast; X, xanthophore; Xb, xanthoblast.

Fig. 3.

Experimental test of the PFR hypothesis of zebrafish pigment cell development. (A) The original working hypothesis of pigment cell development, as shown in Fig. 2, with fully multipotent initial NC generating sensory and autonomic neurons, glia, and melanocytes, iridophores and xanthophores via multipotent, but partially restricted progenitors of all the pigment cells (chromatoblast) and a bipotent melanoiridoblast. (B) Revised hypothesis, based on findings of Nikaido et al. (2021 preprint). Owing to its unexpectedly broad multipotency, we propose the name ‘NC-derived highly multipotent progenitors’ (NC-HMPs) for the trunk NCCs; cells of intermediate potency were not detected. A, autonomic neuron; An, autonomic neuroblast; G, glial cell; Gb, glioblast; I, iridophore; Ib, iridoblast; M, melanocyte; Mb, melanoblast; S, sensory neuron; Sn, sensory neuroblast; X, xanthophore; Xb, xanthoblast.

We propose a novel, dynamic view to reconcile the observations underlying the PFR and DFR hypotheses, i.e. early apparent fate specification and in vivo clonal restriction, but late retention of multipotency. In testing the PFR hypothesis for pigment cell development, we have identified a group of premigratory NCCs that co-express key factors involved in cell fate specification of all pigment cell types, including ltk, tfec, mitfa, pax7 and sox10 (Nikaido et al., 2021 preprint). In normal development, these markers are transient, being strongly downregulated in the majority of cells as they adopt specific fates; this observation highlights repression of key fate-specification genes, including fate-specific transcription factors and receptors, as an important, but largely overlooked, mechanism underpinning fate specification (Petratou et al., 2021, 2018). Formation of individual cell types is blocked in sox10 mutants, leading to cells becoming trapped in an NC-HMP-like progenitor state (Dutton et al., 2001; Elworthy et al., 2003; Greenhill et al., 2011; Nikaido et al., 2021 preprint; Petratou et al., 2021, 2018)

We propose a ‘cyclical fate restriction’ (CFR) hypothesis in which NC-HMP progenitors are highly dynamic, cycling asynchronously through a series of sub-states, each of which is biased to adopt a single fate (Fig. 4). Here, we define a ‘sub-state’ as one in which the cell, which is not itself in equilibrium, is transiently biased (i.e. primed) to adopt a specific fate, such as a melanocyte, before moving into a state in which it is biased to a different fate, and so on. We use the term ‘cyclical’ because we envisage a process in which the cell repeatedly visits and transits through all these sub-states, until such time as it becomes committed to a single fate. It is important to note that this ‘fate-cycling’ process reflects changes in the transcriptome/proteome of the cell, and is considered to be independent of the cell cycle. Whether or not the process is strictly periodic, or more broadly simply involves the cell recurrently accessing these sub-states, is not our key concern here; experimental investigation to test for the recurrence of transcriptional profiles characteristic of the sub-states is highly demanding and remains to be investigated.

Fig. 4.

CFR and CFR modelling. (A) In our new hypothesis, we propose that the fully multipotent NCC transitions to an NC-HMP (large multicoloured circle). Crucially, we envisage the HMP as fate-cycling through a series of sub-states (shown as cells spaced around the circle), each biased to adopt a single fate (indicated by expansion of one colour in the ‘rainbow’: neuroblast (Nb; pink), glioblast (Gb; green), iridoblast (Ib; blue), melanoblast (Mb; black), xanthoblast (Xb; yellow). Single fate specification occurs upon an NC-HMP encountering specific differentiation signals (pink, green, blue, black and yellow ovals), otherwise the multipotent progenitor continues cycling through the subsequent sub-states. Transition from one sub-state to the next is promoted by fate-specific transcription factors (TFs), including Mitfa and Tfec for pro-melanoblast and pro-iridoblast sub-states, respectively. G, glial cell; I, iridophore; M, melanocyte; N, neuron; X, xanthophore. (B) Initially (prior to delamination?), NC-HMPs show unbiased multipotency, indicated by even multicoloured shading. However, influenced by local environmental cues (specification factors; pink, green, black, blue and yellow shading) encountered before and/or during migration, cells become biased in their fate preferences, although not actually committed; such cells would appear to be fate-specified in a snap-shot view (e.g. by whole-mount in situ hybridisation). Depending upon the signalling environment, these biases may favour one or more fates (indicated by expansion of one or two colours in the ‘rainbow’ shading). In response to continuing fate specification signalling, cells exit the transcriptional fate-cycling phase and begin differentiation (unicoloured circles). DA, dorsal aorta; Ep, epidermis; Nc, notochord; NT, neural tube; Som, somite. (C) A simple mathematical model of a ‘cross-repressilator’ (Farjami et al., 2021) GRN, which exhibits different behaviours under different conditions. (Ca) Topology of a GRN in which each of the three TFs shown (1-3) mutually cross-represses the others; simulations show that this simple GRN readily displays behaviours matching key features of CFR. (Cb-Ce) Time courses of expression levels of TFs in the mathematical model, with Cb-Cd showing effects of increasing external (environmental) signal under conditions where it increases the production rates of all TFs equally; as the external signal intensifies, the GRN transitions from a non-cycling state with all TFs at very low levels (Cb), mimicking early NCCs, to a cycling state with each TF transiently and sequentially expressed at a comparatively high level (Cc), mimicking NC-HMPs, and, finally, to a stable state with one TF constantly expressed at a high level (Cd), mimicking the differentiated state. (Ce) TF levels in a cycling progenitor when the increasing level of external (environmental) signal increases production rates for TFs 1 and 3 (red, blue) but decreases that for TF 2 (green) compared with the simulation shown in Cc. Note that the cycling behaviour continues, but now the cell lingers sequentially in states favouring each of two (‘red’ and ‘blue’) fates, and the cell only very transiently lingers in a state favouring the third (‘green’) fate; this is one example of how the system can display a behaviour compatible with bias towards a subset of fates, while still retaining multipotency (cycling through all states). Note that the time courses and expression levels are illustrative, and in arbitrary units (a.u.). For simplicity, the model for an NC-HMP with just three fates is shown, but the model can be generalised to higher multipotency (Farjami et al., 2021).

Fig. 4.

CFR and CFR modelling. (A) In our new hypothesis, we propose that the fully multipotent NCC transitions to an NC-HMP (large multicoloured circle). Crucially, we envisage the HMP as fate-cycling through a series of sub-states (shown as cells spaced around the circle), each biased to adopt a single fate (indicated by expansion of one colour in the ‘rainbow’: neuroblast (Nb; pink), glioblast (Gb; green), iridoblast (Ib; blue), melanoblast (Mb; black), xanthoblast (Xb; yellow). Single fate specification occurs upon an NC-HMP encountering specific differentiation signals (pink, green, blue, black and yellow ovals), otherwise the multipotent progenitor continues cycling through the subsequent sub-states. Transition from one sub-state to the next is promoted by fate-specific transcription factors (TFs), including Mitfa and Tfec for pro-melanoblast and pro-iridoblast sub-states, respectively. G, glial cell; I, iridophore; M, melanocyte; N, neuron; X, xanthophore. (B) Initially (prior to delamination?), NC-HMPs show unbiased multipotency, indicated by even multicoloured shading. However, influenced by local environmental cues (specification factors; pink, green, black, blue and yellow shading) encountered before and/or during migration, cells become biased in their fate preferences, although not actually committed; such cells would appear to be fate-specified in a snap-shot view (e.g. by whole-mount in situ hybridisation). Depending upon the signalling environment, these biases may favour one or more fates (indicated by expansion of one or two colours in the ‘rainbow’ shading). In response to continuing fate specification signalling, cells exit the transcriptional fate-cycling phase and begin differentiation (unicoloured circles). DA, dorsal aorta; Ep, epidermis; Nc, notochord; NT, neural tube; Som, somite. (C) A simple mathematical model of a ‘cross-repressilator’ (Farjami et al., 2021) GRN, which exhibits different behaviours under different conditions. (Ca) Topology of a GRN in which each of the three TFs shown (1-3) mutually cross-represses the others; simulations show that this simple GRN readily displays behaviours matching key features of CFR. (Cb-Ce) Time courses of expression levels of TFs in the mathematical model, with Cb-Cd showing effects of increasing external (environmental) signal under conditions where it increases the production rates of all TFs equally; as the external signal intensifies, the GRN transitions from a non-cycling state with all TFs at very low levels (Cb), mimicking early NCCs, to a cycling state with each TF transiently and sequentially expressed at a comparatively high level (Cc), mimicking NC-HMPs, and, finally, to a stable state with one TF constantly expressed at a high level (Cd), mimicking the differentiated state. (Ce) TF levels in a cycling progenitor when the increasing level of external (environmental) signal increases production rates for TFs 1 and 3 (red, blue) but decreases that for TF 2 (green) compared with the simulation shown in Cc. Note that the cycling behaviour continues, but now the cell lingers sequentially in states favouring each of two (‘red’ and ‘blue’) fates, and the cell only very transiently lingers in a state favouring the third (‘green’) fate; this is one example of how the system can display a behaviour compatible with bias towards a subset of fates, while still retaining multipotency (cycling through all states). Note that the time courses and expression levels are illustrative, and in arbitrary units (a.u.). For simplicity, the model for an NC-HMP with just three fates is shown, but the model can be generalised to higher multipotency (Farjami et al., 2021).

A molecular model for CFR

Although various underlying molecular mechanisms biasing progenitor fate can be envisaged, we consider that an attractive one is focused on the expression levels of receptors for fate-specification factors (Fig. 5) (Kelsh, 2006; Weston, 1991). The biased sub-states would then be characterised by higher level expression of one or more fate-specification receptors, thus making them more sensitive to specific environmental fate-specification signals. For example, one sub-state might have higher expression of the Ltk receptor, and this sub-state would be primed to interact with environmental cues, such as ALKAL proteins (Fadeev et al., 2018), leading to differentiation into an iridophore (Lopes et al., 2008). Indeed, the heterogeneous expression of ltk is striking in both whole-mount in situ hybridisation and single cell NanoString profiling studies (Lopes et al., 2008).

Fig. 5.

Potential molecular basis for cyclical fate restriction. The CFR model expanded to show a plausible molecular mechanism underpinning the key features, although we note that other molecular interpretations would also be compatible with the concept we are proposing. NC-derived highly multipotent progenitors (NC-HMPs; multicoloured circles) express key transcription factors for different cell fate specification programmes [Phox2bb (autonomic neuron), Sox10 (glia), Tfec (iridophore), Mitfa (melanocyte) and Pax3/7 (xanthophore)] and enter fate-cycling phase under influence of environmental signals (not shown). During the cycling phase (centre), the NC-HMP cycles through a series of sub-states (multicoloured circles with a larger area coloured in pink, green, blue, black and yellow), each of them biased towards the specification of a single cell fate: autonomic neuroblast (An), glioblast (Gb), iridoblast (Ib), melanoblast (Mb) and xanthoblast (Xb), respectively. Transition to a new sub-state is promoted by increased activity of key transcription factors (hypothesised to be Phox2bb, Sox10, Tfec, Mitfa and Pax3/7) specific to each cell specification programme. Before entering the cycling phase, the NC-HMP has unbiased numbers of receptors [rectangles around cell surface coloured in pink (Bmpr), green (ErbB3 and Notch1), blue (Ltk), black (Wnt receptor; Wnt R) and yellow (Csfr1a)], each responsive to specific environmental cell fate specification signals [coloured areas: Bmp (pink), Neuregulins and Delta (green), ALK and LTK ligand ALKALs (blue), Wnt ligand (black) and Csf1a (yellow)]. Upon entering the fate-cycling phase, and as result of increased activity of transcription factors, the receptors specific to the sub-state are increased and those of other sub-states are decreased. When the cycling NC-HMP receives insufficient of the sub-state-specific environmental differentiation signal, it transitions to a new substate (red arrows); this is considered an emergent property of the GRN underlying the NC-HMP. In contrast, an NC-HMP exposed to sufficient sub-state-specific differentiation signal will activate the corresponding cell fate specification programme, and downregulate all other transcription factors and the receptors for other cell fate specification signals, thus exiting the cycling phase (black arrows), i.e. the cell has become committed to a single fate. These committed progenitors [single-coloured circles in pink (autonomic neurons), green (glioblast), blue (iridoblast), black (melanoblasts) and yellow (xanthoblast)] will differentiate (white arrows) into the respective cell type [autonomic neuron (A), glial cell (G), iridophore (I), melanocyte (M) or xanthophore (X), respectively].

Fig. 5.

Potential molecular basis for cyclical fate restriction. The CFR model expanded to show a plausible molecular mechanism underpinning the key features, although we note that other molecular interpretations would also be compatible with the concept we are proposing. NC-derived highly multipotent progenitors (NC-HMPs; multicoloured circles) express key transcription factors for different cell fate specification programmes [Phox2bb (autonomic neuron), Sox10 (glia), Tfec (iridophore), Mitfa (melanocyte) and Pax3/7 (xanthophore)] and enter fate-cycling phase under influence of environmental signals (not shown). During the cycling phase (centre), the NC-HMP cycles through a series of sub-states (multicoloured circles with a larger area coloured in pink, green, blue, black and yellow), each of them biased towards the specification of a single cell fate: autonomic neuroblast (An), glioblast (Gb), iridoblast (Ib), melanoblast (Mb) and xanthoblast (Xb), respectively. Transition to a new sub-state is promoted by increased activity of key transcription factors (hypothesised to be Phox2bb, Sox10, Tfec, Mitfa and Pax3/7) specific to each cell specification programme. Before entering the cycling phase, the NC-HMP has unbiased numbers of receptors [rectangles around cell surface coloured in pink (Bmpr), green (ErbB3 and Notch1), blue (Ltk), black (Wnt receptor; Wnt R) and yellow (Csfr1a)], each responsive to specific environmental cell fate specification signals [coloured areas: Bmp (pink), Neuregulins and Delta (green), ALK and LTK ligand ALKALs (blue), Wnt ligand (black) and Csf1a (yellow)]. Upon entering the fate-cycling phase, and as result of increased activity of transcription factors, the receptors specific to the sub-state are increased and those of other sub-states are decreased. When the cycling NC-HMP receives insufficient of the sub-state-specific environmental differentiation signal, it transitions to a new substate (red arrows); this is considered an emergent property of the GRN underlying the NC-HMP. In contrast, an NC-HMP exposed to sufficient sub-state-specific differentiation signal will activate the corresponding cell fate specification programme, and downregulate all other transcription factors and the receptors for other cell fate specification signals, thus exiting the cycling phase (black arrows), i.e. the cell has become committed to a single fate. These committed progenitors [single-coloured circles in pink (autonomic neurons), green (glioblast), blue (iridoblast), black (melanoblasts) and yellow (xanthoblast)] will differentiate (white arrows) into the respective cell type [autonomic neuron (A), glial cell (G), iridophore (I), melanocyte (M) or xanthophore (X), respectively].

The switch to a new sub-state would involve downregulation of that fate-specification receptor, and upregulation of another, such as a Frizzled receptor, which would prime the cell for a melanocyte fate. Importantly, we propose that the shift to the next sub-state depends upon activity of an appropriate, key, fate-specific transcription factor. We emphasise transcription factor activity, rather than expression levels, to make clear that regulation need not be at the level of transcription; such a view has been eloquently expounded by Goding and colleagues (Goding and Meyskens, 2006; Goding and Arnheiter, 2019). Cyclical changes in activity of the key fate-specification transcription factors would result in cyclical changes in the fate-specification receptors, and hence in bias of the sub-states. For example, for a sub-state to express high levels of Ltk, and thus be biased to become an iridophore, the cell would first need an increase in Tfec activity, because ltk expression in premigratory NCCs depends upon Tfec (Petratou et al., 2021). In the absence of that key transcription factor, however, cells are unable to enter the specific sub-state.

As the cell fate-cycles through the sub-states, its final fate depends partly upon how long the cell remains in that sub-state; longer duration increases the opportunity for receiving the appropriate fate-specification signal. In addition, cell fate depends on whether fate-specification signals (i.e. ligands) are present in sufficient quantities and for a sufficient length of time to drive fate specification, as shown in the ventral neural tube, for example (Sagner and Briscoe, 2019).

The CFR model is consistent with key biological observations of NCCs

Our hypothesis is consistent with heterogeneities in gene expression in premigratory NCCs, and apparent fate specification; as the NC-HMP fate-cycles through the various sub-states, it displays varying expression profiles, appearing to be specified when seen in the static snapshot view characteristic of almost all studies, but retaining multipotency. The CFR hypothesis helps to explain the embryonic origin of NCSCs (Hultman et al., 2009). We suggest that as NC-HMPs in these PNS locations become differentiated as neurons and glia, some satellite glia in the DRG [and likely also Schwann cell precursors (SCPs) in some or all of the PNS; Adameyko et al., 2009, 2012; Dooley et al., 2013; Kelsh and Barsh, 2011; Nitzan et al., 2013b; Parichy and Spiewak, 2015; Singh et al., 2016] retain their multipotency and are thus cryptic NCSCs; their entry into a quiescent state is driven by their exposure to the niche within the PNS. It is only after some process of re-activation (e.g. at metamorphosis) that they begin to generate pigment cells; re-activation will probably involve local removal of the quiescence-maintenance signals in the niche and re-entry into the sub-state fate-cycling mode, with the local niche signals controlling the cell types formed by biasing the time spent in each poised sub-state. We note that mouse MSCs in vivo appear to be lineally restricted to generate melanocytes (Nishimura et al., 2002, 2010), yet in vitro culture studies reveal a much wider potency (Watanabe et al., 2016), consistent with them also being intrinsically highly multipotent, but with their niche restricting the fates their progeny actually adopt in vivo.

Importantly, the CFR hypothesis provides a natural explanation for two paradoxes. First, it explains why NCCs adopt different fates even in a crowded premigratory position. Under the DFR hypothesis, where fate specification signals were received late in migration, it is easy to see how fates adopted would be locally appropriate. However, if fate specification occurs very early – prior to migration – then how cells make different decisions needs to be explained. In fish, NCCs in the premigratory position are likely exposed to high levels of Wnt and ALKAL signals, but only a subset become each of melanocytes and iridophores. In the chick, such heterogeneity of fate choice is attributed to differences in timing of delamination from the dorsal neural tube (Krispin et al., 2010a,b; Nitzan et al., 2013a), and this is plausible in the zebrafish too. However, the CFR hypothesis offers another intriguing explanation – that cells make different choices because they are only transiently in a receptive sub-state for each of the relevant fate-specification signals.

Second, our hypothesis provides an alternative explanation for the observations at embryonic stages that in some fate-specification mutants (e.g. mitfa) the absence of one cell type (melanocyte) is accompanied by elevated numbers of another (iridophore), previously interpreted under PFR as evidence for a bipotent cell. Under our CFR hypothesis, the explanation results from the preferential order of progression through the sub-states, such that if transition is blocked by a mutation, the cell pauses in a specific sub-state. For example, in mitfa mutants (Lister et al., 1999), we propose that cells cannot progress from a pro-iridophore sub-state to a pro-melanocyte sub-state [or, more accurately, cannot readily progress; it is likely that cells in such mutants are not permanently trapped in the specific prior sub-state, but simply their ‘dwell-time’ in that sub-state is prolonged. We envisage that the underlying gene regulatory network (GRN) allows alternative dynamic routes for exit from the prior sub-state, in a manner bypassing the subsequent sub-state]. Consequently, they spend longer in the former sub-state, making them more sensitive to pro-iridophore specification signals; consequently, more iridophores are formed.

It is worth considering carefully how our hypothesis compares with the original DFR hypothesis. Although the full transcriptional profile of the NC-HMP state remains to be defined, its potency apparently includes all pigment cell, peripheral neuron and glial fates, so our CFR hypothesis is, in terms of biological behaviour, closer to DFR (wherein migrating NCCs are homogeneous, fully multipotent progenitors), than to PFR. However, we note that the progenitor state in our model is distinct from the earliest NCCs, as these do not express sox10 initially (Lopes et al., 2008), whereas our single-cell study sorted cells using a reporter of sox10 expression (Nikaido et al., 2021 preprint). Furthermore, even amongst these sox10+ cells, our clustering identifies multiple clusters equivalent to NC-HMP progenitors, including ones without (‘early NC-HMP’) and with (‘late NC-HMP’) elevated ltk expression; conceivably, these might represent detection of distinct sub-states themselves. Careful analysis of marker expression in vivo by RNAscope also indicates a series of early progenitor states (Petratou et al., 2021), although their exact correspondence to the cell types defined by NanoString clustering remains to be determined. We speculate that our NC-HMPs may best correspond to a classic ‘trunk NCC’, and for that cell type might be similar to the original DFR. However, the key feature we are proposing, the dynamic nature of the GRN within these cells, makes our hypothesis distinctive. It also has some interesting consequences, as we will now begin to explore.

Modelling CFR – beyond bifurcations in fate specification

To formalise our conceptual model and to begin to explore its properties and feasibility, we took a mathematical modelling approach. Here, we consider a mathematical model based only on deterministic dynamics, although alternative models incorporating stochastic fluctuations as key drivers of transitions between sub-states are also attractive, having been proposed in other developmental contexts (e.g. Corson and Siggia, 2017). In these latter models, deterministic gene regulation is responsible for creating the relevant sub-states, and gene expression fluctuations allow for transitions between them. These models are attractive because they readily display features mimicking the biology, such as a ‘noisy’ and blurred state corresponding to a multipotent progenitor state, becoming more differentiated under the control of external signals capable of reshaping the basins of attraction of the sub-states. However, such stochastic models require a careful tuning of parameters balancing constraining deterministic dynamics with heterogeneity-inducing stochastic components, leading us to pursue the ‘extreme’ version of a fully deterministic system in the first instance.

Mathematical modelling of fate specification has mainly focused on mutual cross-repression between a pair of key fate-specific transcription factors, resulting in paired fate choices, e.g. macrophage versus neutrophil (Huang et al., 2007; Laslo et al., 2006). This approach may reinforce the impression that fate choice obligatorily proceeds through a series of bifurcating fate decisions, but this need not necessarily be the case. A simple expansion of the cross-repressive model to encompass multiple transcription factors driving multiple different fates reveals intrinsic cycling behaviour strikingly similar to that envisaged under, and hence providing theoretical support for, the CFR hypothesis (Fig. 4C) (Farjami et al., 2021). From a mathematical perspective, the emergence of cycling is a natural and generic consequence of negative-feedback loops imposed by cross-repression. Our modelling work indicates that the key features of the dynamics we observe appear over a wide range of model parameter values and indeed for different choices of the precise form of the mathematical equations describing the cross-repression.

The possibility of oscillatory dynamics within GRNs incorporating negative feedback was highlighted by the well-known synthetic ‘repressilator’ network constructed by Elowitz and Leibler (2000). In the context of the Notch signalling pathway, oscillations have been previously observed to result in important features that explain observed biology, such as sequential formation of somites, and the balance of neural stem cell maintenance and neuronal differentiation (Lewis, 2003; Monk, 2003; Ochi et al., 2020). We have developed a series of mathematical models of such cross-repression models (Fig. 4Ca) of cell differentiation from multipotent progenitors, exploring their outputs in simulations. Analytically, we have identified remarkable behaviours that mimic many aspects of the biology envisaged in the CFR hypothesis (Farjami et al., 2021). Particularly interesting is the effect of changing intrinsic cellular properties, such as production and degradation rates of specific regulatory transcription factors, which in turn can be viewed as a response to alterations in extrinsic fate-specification signals. Time spent in the vicinity of each of the successive stable sub-states expands, consistent with cells becoming (apparently) more specified and, in response to the correct signals, actually committed. Thus, transitions from a non-cycling, multipotent phase – analogous to the early NCC, prior to any fate specification (Fig. 4Cb) – into a dynamic phase cycling through all sub-states (Fig. 4Cc), and to eventual stable adoption of a fate (i.e. committed final fates) (Fig. 4Cd), can be displayed over wide ranges of model parameters (Farjami et al., 2021). We note that, in vivo, there is a considerable delay between the first expression of sox10 as part of NC induction and the first evidence of individual fate specification in response (e.g. in zebrafish, sox10 expression begins in the trunk at around 12 hpf, but the first downstream fate-specification response, mitfa transcription, is not detected until around 18 hpf; Dutton et al., 2001; Lister et al., 1999; Montero Balaguar et al., 2006); perhaps this delay reflects the time when the cells are in the non-cycling multipotent phase (Fig. 4Cb). When in the cycling phase, cells linger close to one sub-state (characterised by higher expression of relevant markers), but rapidly move on to another sub-state primed for another fate. This is consistent with the dynamicity and heterogeneity (for the appropriate markers, i.e. those undergoing cycling of expression) of the NC-HMP. Intriguingly, our modelling shows that the balance of probabilities of progenitors adopting different individual fates can be modified from equivalent (all fates equal) to highly biased [a few, or just one, fate(s) strongly favoured], by altering the relative production rates of specific transcription factors, as might happen, for example, in response to specific environmental signals (Fig. 4Ce). This recalls the evidence for strong and more subtle shifts in NCSC potency, depending upon their anatomical origin (White et al., 2001), which clearly indicates that their multipotency is ‘tweaked’ to alter the favoured fates and, indeed, that it becomes tuned by their environment. Thus, in contrast to early NCCs (most of which are considered to be NCSCs), NCSCs isolated from the sciatic nerve seem unable to contribute to sensory neurons (as opposed to sensory ganglial glia) in transplant studies. More subtly, their ability to generate sympathetic neurons decreases in favour of production of parasympathetic neurons, an effect associated with a decreased sensitivity to BMP2 (White et al., 2001).

Thus, we see from the model an indication of the type of mechanism that might underpin the behaviour proposed in the CFR hypothesis, with environmental factors influencing the apparent fate restriction of migrating and post-migratory (see Glossary, Box 1), progenitors. The effect of the environment is to change the balance of time spent in each of the sub-states, so that, for example, in cells in the skin, more time is spent in the sub-state favouring melanocyte specification. This, likely combined with local differences in the levels of fate-specification signals, would have the effect that, in vivo, these cells would be far more likely to differentiate into the favoured fate (e.g. melanocyte) rather than others, and thus would appear to be fate restricted. Indeed, as they migrate, local signals along the migration route will drive their cycling behaviour to biases that, by definition, become appropriate to that route (e.g. iridophores on the medial migration route). Importantly, depending upon ongoing signalling, dwell times may favour two (or possibly more) biased states. This combined with mRNA/protein perdurance may explain the ‘double specification’ observations, and the simultaneous partial activation of fate-specification programmes, seen in migrating NCCs (Petratou et al., 2021, 2018; Soldatov et al., 2019). In a snapshot view, this gives the impression of a PFR process, masking in vivo the underlying full multipotency that is revealed when cells are removed from those environmental signals.

The original suggestion in the DFR hypothesis that fate specification occurs in a post-migratory location, with local signals determining the fate chosen, is now replaced in the CFR hypothesis with the idea that fate maintenance (i.e. fate commitment; see Glossary, Box 1) is strengthened and stabilised by that post-migratory environment. This view suggests the extreme hypothesis that cells locked in the differentiated state by epigenetic reinforcement of this transcriptional programme might retain latent multipotency, which could be liberated when that epigenetic lockdown is released. This is, in essence, the mechanism likely to underlie transdifferentiation (e.g. Shen et al., 2000).

Moreover, mathematical modelling of the CFR hypothesis allows us to see how NCSCs (Box 2), which we propose are retained NC-HMP cells, might be held by local environmental conditions (the niche) in a (pseudostabilised) sub-state (as a glial cell in a DRG or in peripheral nerves), but might retain potency for other fates when environmental conditions change [e.g. on liberation from the niche, such as when the DRG niche becomes activating, or upon losing contact with peripheral dendrites (Adameyko et al., 2009), or, alternatively, when single cells are isolated under conditions in which environmental signals are diluted and cells ‘relax’ from their varied specified states]. For these ‘differentiated’ cells, multipotency can be readily reactivated by changing environmental signals and cause dedifferentiation, perhaps even back to the cycling progenitor state.

Could CFR apply more widely, to include neuronal fates?

Our work has focused on pigment cell fates, but several observations suggest that CFR applies not just to chromatophores, but more widely, at least to neural fates. First, sox10 expression is a prominent feature of the NC-HMP state that we propose is fate-cycling. Given the key roles for Sox10 in both NCC multipotency and glial fate specification and differentiation (Delfino-Machin et al., 2017; Dutton et al., 2001; Kelsh and Eisen, 2000; Kuhlbrodt et al., 1998; Liu et al., 2020; Paratore et al., 2001; Peirano and Wegner, 2000; Sonnenberg-Riethmacher et al., 2001), it seems likely that any multipotent NCC will have glial fates as an option. Second, our fate mapping of ltk-expressing cells, interpreted as the NC-HMPs and their progeny, showed the unexpected inclusion of peripheral neuronal and glial fates (Nikaido et al., 2021 preprint). Third, our modelling considerations, described above, and the dynamic view of differentiation as a potentially pseudo-stabilised state that results from them, readily encompasses the emerging view of glial cells as (including) NCSCs. In agreement with Dupin and colleagues (Dupin et al., 2018), we propose that multipotent NCSCs are retained in numerous locations, as observed for SCPs (Box 2). However, we take this one step further, proposing the radical view that these cells might be fully multipotent, albeit often constrained by their local stem cell niche. Fourth, various studies have shown a close link between melanocytes and the PNS, especially glial cell types (e.g. Adameyko et al., 2009; Dupin et al., 2000, 2003; Girdlestone and Weston, 1985; Kunisada et al., 2014; Motohashi et al., 2009; Real et al., 2006; Watanabe et al., 2016). Finally, scRNA-seq reveals the transcriptional similarity between glia and migratory multipotent cells (Soldatov et al., 2019).

One key aspect of the PFR hypothesis when applied to the PNS is the ‘bipotent’ ganglial progenitors (Fig. 1A); we note that although these cells are generally labelled as ‘bipotent’, they give rise to multiple types of neurons and glia and hence might equally be considered more multipotent. As noted above, there is considerable evidence for early segregation of sensory and autonomic ganglial progenitors, with the choice of neurons and glia arising only later, suggesting that cells become at least strongly biased towards these fate combinations. This is readily accommodated within the CFR hypothesis by suggesting that the nascent ganglial niche drives cells into a fate-cycling state in which the pro-neuronal and pro-glial sub-states are dominant, biasing NC-HMPs in these regions to neuronal and glial fates (Fig. 4Ce). We note that, under the CFR hypothesis, these ‘neuroglial progenitors’ are only strongly biased towards appropriate neural fates; they retain full multipotency, explaining the NanoString data at later embryonic stages noted above. In the case of these progenitors, this cryptic multipotency is rather easier to envisage because the evidence for melanocyte derivation from peripheral nerve SCPs is so strong, and given that, in zebrafish, adult melanocyte stem cells (MSCs) also generate neurons and glia (Box 2) (Adameyko et al., 2009, 2012; Dooley et al., 2013; Kelsh and Barsh, 2011; Nitzan et al., 2013b; Parichy and Spiewak, 2015; Singh et al., 2016).

How do we reconcile the detailed dissection of mouse peripheral neural development using scRNA-seq, which provided strong support for a PFR hypothesis (Soldatov et al., 2019)? In many respects, their data can be readily reconciled with the CFR hypothesis. Although the authors were unable to resolve melanocyte development, Mitf expression was widespread amongst delaminating and migrating NC. They show that the earliest phase of Neurog2 expression, in premigratory NCCs, corresponds to fully multipotent cells, and not to committed sensory neuron progenitors. More broadly, their data show that these delaminated, premigratory NCCs exhibit low level expression of a wide range of markers of derived fates, including three other key neuron specification transcription factors: Phox2b, Neurog1 and Pou4f1. We propose these cells are likely equivalent to our NC-HMPs and might be fate-cycling in the manner we propose. Detecting this would be challenging using current bioinformatics tools because cycling behaviour would result in overlapping data that would be hard to analyse in terms of a well-defined pseudotime (Mao et al., 2017). Interestingly Soldatov and colleagues characterise a gradual process of fate decisions, initiating early in migration with co-activation of differentiation programmes, then gradually biased towards genetic programmes associated with specific fates, and becoming distinct in committed cells in post-migratory locations (Soldatov et al., 2019); this seems to be compatible with the gradual emergence of a committed differentiated state from multipotent progenitors that we are proposing.

Our CFR hypothesis provides a novel framework for thinking about the developmental mechanisms underpinning NCC fate specification and differentiation, reconciling data supporting both traditional hypotheses, which may also be relevant in other stem cell and developmental contexts. Indeed, it has been proposed that oscillatory dynamics might underpin stemness itself (Furusawa and Kaneko, 2012). Although our CFR hypothesis is still speculative, it is certainly time to think differently in order to resolve the contest between the historically proposed and irreconcilable mechanisms. The CFR hypothesis calls into question the validity of assuming repeated bifurcations as a necessary feature of progenitor cell development. This, in turn, highlights the need for improvements in scRNA-seq analysis algorithms that may be required in order to assess differentiation trajectories in more detail, including the ability to detect increasingly complex graph structures, including cycles, robustly. Recent developments in bioinformatics algorithms, such as the use of reversed graph embedding (Qiu et al., 2017) and partition-based graph abstraction (Wolf et al., 2019), are extremely welcome.

Recent results in single-cell analysis have supplied a growing number of examples of dedifferentiation and reversible differentiation, especially in the contexts of early development (Papatsenko et al., 2015) and regeneration (Lin et al., 2021). In this context, we consider that a simple two (sub-)state model of reversible differentiation, such as that proposed by Papatsenko et al. (2015), is a primitive example of a cyclical-type model, with cells having the option to continue fate-cycling by switching to the alternative state or exiting the cycle at the current state. In this respect, the multistate CFR model suggested here can be considered as a generalisation of this two-state model for a larger number of cell types. Obviously, the concept of recurrence does not necessarily require any prescribed ordering of sub-states and the dynamics may move between sub-states that have differing sensitivities to different signals, controlling transitions to other sub-states or the exit from the fate-cycling regime. The GRN dynamics in general is likely to include additional stochastic effects; in this case, fate-cycling would describe only the most probable pathway of transitions, assuming that each transcription factor becomes active relatively often. In the case of absence of such a transition-inducing transcription factor (e.g. in a mutant context), the cell would be delayed or prevented from moving on to the next sub-state and would become (transiently) trapped in a particular sub-state. A wide range of detailed alternatives can be envisaged, including, in another extreme case, these transitions all being spontaneous. We propose that the key feature of recurrence of specific sub-states is the distinctive feature of such systems, and justifies our use of the term ‘cyclical’.

The CFR hypothesis makes a series of testable predictions, which together are distinctive; some already have at least some experimental support, but now require comprehensive assessment. These predictions include:

  1. Co-expression of key fate-specification transcription factors, in early progenitor stages, reflecting their potency.

  2. Consequently, the process of differentiation and fate commitment is only partly about activation of expression of key transcription factors, and is also about their maintenance (and upregulation) while repressing those driving alternative fates.

  3. Expression of fate-specification receptors in premigratory NCCs should be fate-specification transcription factor dependent.

  4. Cyclical expression of some genes in NC-HMPs, likely including those encoding these fate specification signal receptors.

  5. Cyclical expression underpinned by a pattern of cross-repression between fate-specific transcription factors.

  6. Retained, but cryptic, multipotency in most NCCs, persisting at least until differentiation. This cryptic multipotency may underpin the setting aside of APSCs/NCSCs, which may be much more widespread within the peripheral nervous system than currently envisaged.

  7. This multipotency is most clearly revealed by transcriptional profiling studies in which conditions favour both highly sensitive detection of very low level gene expression, and ‘relaxation’ of the fate-specified state induced by environmental signals.

  8. Although many cells may undergo terminal differentiation, with that transcriptional state ‘locked in’ by epigenetic mechanisms (commitment), quiescent APSCs/NCSCs may retain propensity for a more dynamic, transcriptional state as a result of local factors forming their niche.

  9. Activated APSCs/NCSCs will have broad, but cryptic, multipotency, with local niche factors dictating the specific cell fates they generate in vivo.

In summary, the CFR hypothesis provides a novel and rich framework within which to consider the diverse and conflicting data surrounding NCC fate specification and differentiation. Its testing, and the resolution of this long-standing conflict over developmental mechanisms, will continue to provide exciting challenges. We propose that it may even be more widely applicable, for example in the context of haematopoietic and neural stem cells.

We wish to thank colleagues who have provided invaluable discussions and critique as we were developing the CFR concept and/or who have given insightful comments on drafts of the manuscript, in particular Heinz Arnheiter, Laure Bally-Cuif, Chaya Kalcheim, Alfonso Martinez-Arias, Adele Murrell, Jonathan Slack, David Tosh and Andrew Ward. R.N.K. would like to thank Jim Weston and Judith Eisen for the initial discussions (more than 25 years ago!) that triggered his interest in the question addressed here.

Funding

This work was funded by the Biotechnology and Biological Sciences Research Council (BB/ L00769X/1 to R.N.K.; BB/S015906/1 to R.N.K. and J.H.P.D.; BB/L007789/1 and BB/S01604X/1 to A.R.) and partnership grants from the Royal Society (IEC\R2\170199 to R.N.K.) and the Russian Foundation for Basic Research (Project No. 17-54-10014\19 to V.M.).

Adameyko
,
I.
,
Lallemend
,
F.
,
Aquino
,
J. B.
,
Pereira
,
J. A.
,
Topilko
,
P.
,
Muller
,
T.
,
Fritz
,
N.
,
Beljajeva
,
A.
,
Mochii
,
M.
,
Liste
,
I.
et al. 
(
2009
).
Schwann cell precursors from nerve innervation are a cellular origin of melanocytes in skin
.
Cell
139
,
366
-
379
.
Adameyko
,
I.
,
Lallemend
,
F.
,
Furlan
,
A.
,
Zinin
,
N.
,
Aranda
,
S.
,
Kitambi
,
S. S.
,
Blanchart
,
A.
,
Favaro
,
R.
,
Nicolis
,
S.
,
Lubke
,
M.
et al. 
(
2012
).
Sox2 and Mitf cross-regulatory interactions consolidate progenitor and melanocyte lineages in the cranial neural crest
.
Development
139
,
397
-
410
.
Alvarez-Buylla
,
A.
,
Garcia-Verdugo
,
J. M.
and
Tramontin
,
A. D.
(
2001
).
A unified hypothesis on the lineage of neural stem cells
.
Nat. Rev. Neurosci.
2
,
287
-
293
.
Anderson
,
D. J.
(
1989
).
The neural crest cell lineage problem: neuropoiesis?
Neuron
3
,
1
-
12
.
Anderson
,
D. J.
(
2000
).
Genes, lineages and the neural crest: a speculative review
.
Philos. Trans. R. Soc. Lond. B Biol. Sci.
355
,
953
-
964
.
Baggiolini
,
A.
,
Varum
,
S.
,
Mateos
,
J. M.
,
Bettosini
,
D.
,
John
,
N.
,
Bonalli
,
M.
,
Ziegler
,
U.
,
Dimou
,
L.
,
Clevers
,
H.
,
Furrer
,
R.
et al. 
(
2015
).
Premigratory and migratory neural crest cells are multipotent in vivo
.
Cell Stem Cell
16
,
314
-
322
.
Bagnara
,
J. T.
,
Matsumoto
,
J.
,
Ferris
,
W.
,
Frost
,
S. K.
,
Turner
,
W. A.
, Jr
,
Tchen
,
T. T.
and
Taylor
,
J. D.
(
1979
).
Common origin of pigment cells
.
Science
203
,
410
-
415
.
Barald
,
K. F.
(
1988a
).
Antigen recognized by monoclonal antibodies to mesencephalic neural crest and to ciliary ganglion neurons is involved in the high affinity choline uptake mechanism in these cells
.
J. Neurosci. Res.
21
,
119
-
134
.
Barald
,
K. F.
(
1988b
).
Monoclonal antibodies made to chick mesencephalic neural crest cells and to ciliary ganglion neurons identify a common antigen on the neurons and a neural crest subpopulation
.
J. Neurosci. Res.
21
,
107
-
118
.
Barbu
,
M.
,
Ziller
,
C.
,
Rong
,
P.
and
Le Douarin
,
N.
(
1986
).
Heterogeneity in migrating neural crestc ells revealed bya monoclonal antibody
.
J. Neuroscience
6
,
2215
-
2225
.
Baroffio
,
A.
,
Dupin
,
E.
and
Le Douarin
,
N.
(
1988
).
Clone-forming ability and differentiation potential of migratory neural crest cells
.
Proc. Natl. Acad. Sci. U. S. A.
85
,
5325
-
5329
.
Baroffio
,
A.
,
Dupin
,
E.
and
Le Douarin
,
N. M.
(
1991
).
Common precursors for neural and mesectodermal derivatives in the cephalic neural crest
.
Development
112
,
301
-
305
.
Bronner-Fraser
,
M.
and
Fraser
,
S.
(
1989
).
Developmental potential of avian trunk neural crest cells in situ
.
Neuron
3
,
755
-
766
.
Bronner-Fraser
,
M.
and
Fraser
,
S. E.
(
1988
).
Cell lineage analysis reveals multipotency of some avian neural crest cells
.
Nature
335
,
161
-
164
.
Budi
,
E. H.
,
Patterson
,
L. B.
and
Parichy
,
D. M.
(
2011
).
Post-embryonic nerve-associated precursors to adult pigment cells: genetic requirements and dynamics of morphogenesis and differentiation
.
PLoS Genet.
7
,
e1002044
.
Buitrago-Delgado
,
E.
,
Nordin
,
K.
,
Rao
,
A.
,
Geary
,
L.
and
LaBonne
,
C.
(
2015
).
NEURODEVELOPMENT. Shared regulatory programs suggest retention of blastula-stage potential in neural crest cells
.
Science
348
,
1332
-
1335
.
Calloni
,
G. W.
,
Glavieux-Pardanaud
,
C.
,
Le Douarin
,
N. M.
and
Dupin
,
E.
(
2007
).
Sonic Hedgehog promotes the development of multipotent neural crest progenitors endowed with both mesenchymal and neural potentials
.
Proc. Natl. Acad. Sci. U. S. A.
104
,
19879
-
19884
.
Calloni
,
G. W.
,
Le Douarin
,
N. M.
and
Dupin
,
E.
(
2009
).
High frequency of cephalic neural crest cells shows coexistence of neurogenic, melanogenic, and osteogenic differentiation capacities
.
Proc. Natl. Acad. Sci. U. S. A.
106
,
8947
-
8952
.
Camargo-Sosa
,
K.
,
Colanesi
,
S.
,
Muller
,
J.
,
Schulte-Merker
,
S.
,
Stemple
,
D.
,
Patton
,
E. E.
and
Kelsh
,
R. N.
(
2019
).
Endothelin receptor Aa regulates proliferation and differentiation of Erb-dependent pigment progenitors in zebrafish
.
PLoS Genet.
15
,
e1007941
.
Ciment
,
G.
and
Weston
,
J. A.
(
1982
).
Early appearance in neural crest and crest-derived cells of an antigenic determinant present in avian neurons
.
Dev. Biol.
93
,
355
-
367
.
Ciment
,
G.
and
Weston
,
J. A.
(
1985
).
Segregation of developmental abilities in neural-crest-derived cells: identification of partially restricted intermediate cell types in the branchial arches of avian embryos
.
Dev. Biol.
111
,
73
-
83
.
Corson
,
F.
and
Siggia
,
E. D.
(
2017
).
Gene-free methodology for cell fate dynamics during development
.
Elife
6
,
e30743
.
Curran
,
K.
,
Lister
,
J. A.
,
Kunkel
,
G. R.
,
Prendergast
,
A.
,
Parichy
,
D. M.
and
Raible
,
D. W.
(
2010
).
Interplay between Foxd3 and Mitf regulates cell fate plasticity in the zebrafish neural crest
.
Dev. Biol.
344
,
107
-
118
.
Delfino-Machin
,
M.
,
Chipperfield
,
T. R.
,
Rodrigues
,
F. S.
and
Kelsh
,
R. N.
(
2007
).
The proliferating field of neural crest stem cells
.
Dev. Dyn.
236
,
3242
-
3254
.
Delfino-Machin
,
M.
,
Madelaine
,
R.
,
Busolin
,
G.
,
Nikaido
,
M.
,
Colanesi
,
S.
,
Camargo-Sosa
,
K.
,
Law
,
E. W.
,
Toppo
,
S.
,
Blader
,
P.
,
Tiso
,
N.
et al. 
(
2017
).
Sox10 contributes to the balance of fate choice in dorsal root ganglion progenitors
.
PLoS One
12
,
e0172947
.
Dooley
,
C. M.
,
Mongera
,
A.
,
Walderich
,
B.
and
Nusslein-Volhard
,
C.
(
2013
).
On the embryonic origin of adult melanophores: the role of ErbB and Kit signalling in establishing melanophore stem cells in zebrafish
.
Development
140
,
1003
-
1013
.
Dupin
,
E.
,
Baroffio
,
A.
,
Dulac
,
C.
,
Cameron-Curry
,
P.
and
Le Douarin
,
N.
(
1990
).
Schwann-cell differentiation in clonal cultures of the neural crest, as evidenced by the anti-Schwann cell myelin protein monoclonal antibody
.
Proc. Natl. Acad. Sci. U. S. A.
87
,
1119
-
1123
.
Dupin
,
E.
,
Glavieux
,
C.
,
Vaigot
,
P.
and
Le Douarin
,
N. M.
(
2000
).
Endothelin 3 induces the reversion of melanocytes to glia through a neural crest-derived glial-melanocytic progenitor
.
Proc. Natl. Acad. Sci. U. S. A.
97
,
7882
-
7887
.
Dupin
,
E.
,
Real
,
C.
,
Glavieux-Pardanaud
,
C.
,
Vaigot
,
P.
and
Le Douarin
,
N. M.
(
2003
).
Reversal of developmental restrictions in neural crest lineages: transition from Schwann cells to glial-melanocytic precursors in vitro
.
Proc. Natl. Acad. Sci. U. S. A.
100
,
5229
-
5233
.
Dupin
,
E.
,
Calloni
,
G. W.
,
Coelho-Aguiar
,
J. M.
and
Le Douarin
,
N. M.
(
2018
).
The issue of the multipotency of the neural crest cells
.
Dev. Biol.
444
Suppl. 1
,
S47
-
S59
.
Dutton
,
K. A.
,
Pauliny
,
A.
,
Lopes
,
S. S.
,
Elworthy
,
S.
,
Carney
,
T. J.
,
Rauch
,
J.
,
Geisler
,
R.
,
Haffter
,
P.
and
Kelsh
,
R. N.
(
2001
).
Zebrafish colourless encodes sox10 and specifies non-ectomesenchymal neural crest fates
.
Development
128
,
4113
-
4125
.
Elowitz
,
M. B.
and
Leibler
,
S.
(
2000
).
A synthetic oscillatory network of transcriptional regulators
.
Nature
403
,
335
-
338
.
Elworthy
,
S.
,
Lister
,
J. A.
,
Carney
,
T. J.
,
Raible
,
D. W.
and
Kelsh
,
R. N.
(
2003
).
Transcriptional regulation of mitfa accounts for the sox10 requirement in zebrafish melanophore development
.
Development
130
,
2809
-
2818
.
Erickson
,
C. A.
,
Duong
,
T. D.
and
Tosney
,
K. W.
(
1992
).
Descriptive and experimental analysis of the dispersion of neural crest cells along the dorsolateral path and their entry into ectoderm in the chick embryo
.
Dev. Biol.
151
,
251
-
272
.
Fadeev
,
A.
,
Mendoza-Garcia
,
P.
,
Irion
,
U.
,
Guan
,
J.
,
Pfeifer
,
K.
,
Wiessner
,
S.
,
Serluca
,
F.
,
Singh
,
A. P.
,
Nusslein-Volhard
,
C.
and
Palmer
,
R. H.
(
2018
).
ALKALs are in vivo ligands for ALK family receptor tyrosine kinases in the neural crest and derived cells
.
Proc. Natl. Acad. Sci. USA
115
,
E630
-
E638
.
Farjami
,
S.
,
Camargo Sosa
,
K.
,
Dawes
,
J. H. P.
,
Kelsh
,
R. N.
and
Rocco
,
A.
(
2021
).
Novel generic models for differentiating stem cells reveal oscillatory mechanisms
.
J. R Soc. Interface
18
,
20210442
.
Farrell
,
J. A.
,
Wang
,
Y.
,
Riesenfeld
,
S. J.
,
Shekhar
,
K.
,
Regev
,
A.
and
Schier
,
A. F.
(
2018
).
Single-cell reconstruction of developmental trajectories during zebrafish embryogenesis
.
Science
360
,
eaar3131
.
Fraser
,
S. E.
and
Bronner-Fraser
,
M.
(
1991
).
Migrating neural crest cells in the trunk of the avian embryo are multipotent
.
Development
112
,
913
-
920
.
Fujii
,
R.
(
1993
).
Cytophysiology of Fish Chromatophores
.
Int. Rev. Cytology
143
,
191
-
255
.
Furusawa
,
C.
and
Kaneko
,
K.
(
2012
).
A dynamical-systems view of stem cell biology
.
Science
338
,
215
-
217
.
Gilbert
,
S. F.
and
Barresi
,
M. J. F.
(
2016
).
Neural crest cells and axonal specificity
. In
Developmental Biology
, 11th edn, p.
468
.
Sunderland, Mass
:
Sinauer Associates, Inc
.
Girdlestone
,
J.
and
Weston
,
J. A.
(
1985
).
Identification of early neuronal subpopulations in avian neural crest cell cultures
.
Dev. Biol.
109
,
274
-
287
.
Goding
,
C.
and
Meyskens
,
F. L., Jr.
(
2006
).
Microphthalmic-associated transcription factor integrates melanocyte biology and melanoma progression
.
Clin. Cancer Res.
12
,
1069
-
1073
.
Goding
,
C. R.
and
Arnheiter
,
H.
(
2019
).
MITF - the first 25 years
.
Genes Dev.
33
,
983
-
1007
.
Greenhill
,
E. R.
,
Rocco
,
A.
,
Vibert
,
L.
,
Nikaido
,
M.
and
Kelsh
,
R. N.
(
2011
).
An iterative genetic and dynamical modelling approach identifies novel features of the gene regulatory network underlying melanocyte development
.
PLoS Genet.
7
,
e1002265
.
Greenwood
,
A. L.
,
Turner
,
E. E.
and
Anderson
,
D. J.
(
1999
).
Identification of dividing, determined sensory neuron precursors in the mammalian neural crest
.
Development
126
,
3545
-
3559
.
Hashimoto
,
H.
,
Goda
,
M.
and
Kelsh
,
R. N.
(
2021
).
Pigment Cell Development in Teleosts
. In
Pigments, Pigment Cells and Pigment Patterns
(ed.
H.
Hashimoto
,
M.
Goda
,
R.
Futahashi
,
R. N.
Kelsh
and
T.
Akiyama
), pp.
209
-
246
.
Singapore
:
Springer Nature Singapore Pte Ltd
.
Henion
,
P. D.
and
Weston
,
J. A.
(
1997
).
Timing and pattern of cell fate restrictions in the neural crest lineage
.
Development
124
,
4351
-
4359
.
Henion
,
P. D.
,
Garner
,
A. S.
,
Large
,
T. H.
and
Weston
,
J. A.
(
1995
).
trkC-mediated NT-3 signaling is required for the early development of a subpopulation of neurogenic neural crest cells
.
Dev. Biol.
172
,
602
-
613
.
Howard
,
A. G. T.
,
Baker
,
P. A.
,
Ibarra-Garcia-Padilla
,
R.
,
Moore
,
J. A.
,
Rivas
,
L. J.
,
Tallman
,
J. J.
,
Singleton
,
E. W.
,
Westheimer
,
J. L.
,
Corteguera
,
J. A.
and
Uribe
,
R. A.
(
2021
).
An atlas of neural crest lineages along the posterior developing zebrafish at single-cell resolution
.
Elife
10
,
e60005
.
Huang
,
S.
,
Guo
,
Y. P.
,
May
,
G.
and
Enver
,
T.
(
2007
).
Bifurcation dynamics in lineage-commitment in bipotent progenitor cells
.
Dev. Biol.
305
,
695
-
713
.
Hultman
,
K. A.
and
Johnson
,
S. L.
(
2010
).
Differential contribution of direct-developing and stem cell-derived melanocytes to the zebrafish larval pigment pattern
.
Dev. Biol.
337
,
425
-
431
.
Hultman
,
K. A.
,
Budi
,
E. H.
,
Teasley
,
D. C.
,
Gottlieb
,
A. Y.
,
Parichy
,
D. M.
and
Johnson
,
S. L.
(
2009
).
Defects in ErbB-dependent establishment of adult melanocyte stem cells reveal independent origins for embryonic and regeneration melanocytes
.
PLoS Genet.
5
,
e1000544
.
Ide
,
H.
(
1986
).
Transdifferentiation of amphibian chromatophores
.
Curr. Top. Dev. Biol.
20
,
79
-
87
.
Ide
,
H.
and
Hama
,
T.
(
1976
).
Transformation of amphibian iridophores into melanophores in clonal culture
.
Dev. Biol.
53
,
297
-
302
.
Ito
,
K.
and
Sieber-Blum
,
M.
(
1993
).
Pluripotent and developmentally restricted neural-crest-derived cells in posterior visceral arches
.
Dev. Biol.
156
,
191
-
200
.
Ito
,
K.
,
Morita
,
T.
and
Sieber-Blum
,
M.
(
1993
).
In vitro clonal analysis of mouse neural crest development
.
Dev. Biol.
157
,
517
-
525
.
Iyengar
,
S.
,
Kasheta
,
M.
and
Ceol
,
C. J.
(
2015
).
Poised regeneration of zebrafish melanocytes involves direct differentiation and concurrent replenishment of tissue-resident progenitor cells
.
Dev. Cell
33
,
631
-
643
.
Johnson
,
S. L.
,
Africa
,
D.
,
Walker
,
C.
and
Weston
,
J. A.
(
1995
).
Genetic control of adult pigment stripe development in zebrafish
.
Dev. Biol.
167
,
27
-
33
.
Kahane
,
N.
and
Kalcheim
,
C.
(
1994
).
Expression of trkC receptor mRNA during development of the avian nervous system
.
J. Neurobiol.
25
,
571
-
584
.
Kelsh
,
R. N.
(
2006
).
Sorting out Sox10 functions in neural crest development
.
BioEssays
28
,
788
-
798
.
Kelsh
,
R. N.
and
Barsh
,
G. S.
(
2011
).
A nervous origin for fish stripes
.
PLoS Genet.
7
,
e1002081
.
Kelsh
,
R. N.
and
Eisen
,
J. S.
(
2000
).
The zebrafish colourless gene regulates development of non-ectomesenchymal neural crest derivatives
.
Development
127
,
515
-
525
.
Kim
,
J.
,
Lo
,
L.
,
Dormand
,
E.
and
Anderson
,
D. J.
(
2003
).
SOX10 maintains multipotency and inhibits neuronal differentiation of neural crest stem cells
.
Neuron
38
,
17
-
31
.
Kimura
,
T.
,
Nagao
,
Y.
,
Hashimoto
,
H.
,
Yamamoto-Shiraishi
,
Y.
,
Yamamoto
,
S.
,
Yabe
,
T.
,
Takada
,
S.
,
Kinoshita
,
M.
,
Kuroiwa
,
A.
and
Naruse
,
K.
(
2014
).
Leucophores are similar to xanthophores in their specification and differentiation processes in medaka
.
Proc. Natl. Acad. Sci. U. S. A.
111
,
7343
-
7348
.
Kitamura
,
K.
,
Takiguchi-Hayashi
,
K.
,
Sezaki
,
M.
,
Yamamoto
,
H.
and
Takeuchi
,
T.
(
1992
).
Avian neural crest cells express a melanogenic trait during early migration from the neural tube: observations with the new monoclonal antibody, “MEBL-1”
.
Development
114
,
367
-
378
.
Krispin
,
S.
,
Nitzan
,
E.
and
Kalcheim
,
C.
(
2010a
).
The dorsal neural tube: a dynamic setting for cell fate decisions
.
Dev. Neurobiol.
70
,
796
-
812
.
Krispin
,
S.
,
Nitzan
,
E.
,
Kassem
,
Y.
and
Kalcheim
,
C.
(
2010b
).
Evidence for a dynamic spatiotemporal fate map and early fate restrictions of premigratory avian neural crest
.
Development
137
,
585
-
595
.
Kuhlbrodt
,
K.
,
Herbarth
,
B.
,
Sock
,
E.
,
Hermans-Borgmeyer
,
I.
and
Wegner
,
M.
(
1998
).
Sox10, a novel transcriptional modulator in glial cells
.
J. Neurosci.
18
,
237
-
250
.
Kunisada
,
T.
,
Tezulka
,
K.
,
Aoki
,
H.
and
Motohashi
,
T.
(
2014
).
The stemness of neural crest cells and their derivatives
.
Birth Defects Res. C Embryo Today
102
,
251
-
262
.
Lahav
,
R.
,
Ziller
,
C.
,
Dupin
,
E.
and
Le Douarin
,
N. M.
(
1996
).
Endothelin 3 promotes neural crest cell proliferation and mediates a vast increase in melanocyte number in culture
.
Proc. Natl. Acad. Sci. U. S. A.
93
,
3892
-
3897
.
Lahav
,
R.
,
Dupin
,
E.
,
Lecoin
,
L.
,
Glavieux
,
C.
,
Champeval
,
D.
,
Ziller
,
C.
and
Le Douarin
,
N. M.
(
1998
).
Endothelin 3 selectively promotes survival and proliferation of neural crest-derived glial and melanocytic precursors in vitro
.
Proc. Natl. Acad. Sci. U. S. A.
95
,
14214
-
14219
.
Laslo
,
P.
,
Spooner
,
C. J.
,
Warmflash
,
A.
,
Lancki
,
D. W.
,
Lee
,
H. J.
,
Sciammas
,
R.
,
Gantner
,
B. N.
,
Dinner
,
A. R.
and
Singh
,
H.
(
2006
).
Multilineage transcriptional priming and determination of alternate hematopoietic cell fates
.
Cell
126
,
755
-
766
.
Le Douarin
,
N. M.
(
1986
).
Cell line segregation during peripheral nervous system ontogeny
.
Science
231
,
1515
-
1522
.
Le Douarin
,
N. M.
and
Kalcheim
,
C.
(
1999
).
The Neural Crest
, 2nd edn
Cambridge
:
Cambridge University Press
.
Lewis
,
J.
(
2003
).
Autoinhibition with transcriptional delay: a simple mechanism for the zebrafish somitogenesis oscillator
.
Curr. Biol.
13
,
1398
-
1408
.
Lewis
,
V. M.
,
Saunders
,
L. M.
,
Larson
,
T. A.
,
Bain
,
E. J.
,
Sturiale
,
S. L.
,
Gur
,
D.
,
Chowdhury
,
S.
,
Flynn
,
J. D.
,
Allen
,
M. C.
,
Deheyn
,
D. D.
et al. 
(
2019
).
Fate plasticity and reprogramming in genetically distinct populations of Danio leucophores
.
Proc. Natl. Acad. Sci. U. S. A.
116
,
11806
-
11811
.
Lin
,
T. Y.
,
Gerber
,
T.
,
Taniguchi-Sugiura
,
Y.
,
Murawala
,
P.
,
Hermann
,
S.
,
Grosser
,
L.
,
Shibata
,
E.
,
Treutlein
,
B.
and
Tanaka
,
E. M.
(
2021
).
Fibroblast dedifferentiation as a determinant of successful regeneration
.
Dev. Cell
56
,
1541
-
1551.e1546
.
Lister
,
J. A.
,
Robertson
,
C. P.
,
Lepage
,
T.
,
Johnson
,
S. L.
and
Raible
,
D. W.
(
1999
).
nacre encodes a zebrafish microphthalmia-related protein that regulates neural-crest-derived pigment cell fate
.
Development
126
,
3757
-
3767
.
Liu
,
J. A.
,
Tai
,
A.
,
Hong
,
J.
,
Cheung
,
M. P. L.
,
Sham
,
M. H.
,
Cheah
,
K. S. E.
,
Cheung
,
C. W.
and
Cheung
,
M.
(
2020
).
Fbxo9 functions downstream of Sox10 to determine neuron-glial fate choice in the dorsal root ganglia through Neurog2 destabilization
.
Proc. Natl. Acad. Sci. U. S. A.
117
,
4199
-
4210
.
Lo
,
L.
,
Sommer
,
L.
and
Anderson
,
D. J.
(
1997
).
MASH1 maintains competence for BMP2-induced neuronal differentiation in post-migratory neural crest cells
.
Curr. Biol.
7
,
440
-
450
.
Lopes
,
S. S.
,
Yang
,
X.
,
Muller
,
J.
,
Carney
,
T. J.
,
McAdow
,
A. R.
,
Rauch
,
G. J.
,
Jacoby
,
A. S.
,
Hurst
,
L. D.
,
Delfino-Machin
,
M.
,
Haffter
,
P.
et al. 
(
2008
).
Leukocyte tyrosine kinase functions in pigment cell development
.
PLoS Genet.
4
,
e1000026
.
Mahalwar
,
P.
,
Walderich
,
B.
,
Singh
,
A. P.
and
Nusslein-Volhard
,
C.
(
2014
).
Local reorganization of xanthophores fine-tunes and colors the striped pattern of zebrafish
.
Science
345
,
1362
-
1364
.
Mao
,
Q.
,
Wang
,
L.
,
Tsang
,
I. W.
and
Sun
,
Y. J.
(
2017
).
Principal Graph and Structure Learning Based on Reversed Graph Embedding
.
Ieee T Pattern Anal
39
,
2227
-
2241
.
Marusich
,
M. F.
and
Weston
,
J. A.
(
1991
).
Development of the neural crest
.
Curr. Opin. Genet. Dev.
1
,
221
-
229
.
McMenamin
,
S. K.
,
Bain
,
E. J.
,
McCann
,
A. E.
,
Patterson
,
L. B.
,
Eom
,
D. S.
,
Waller
,
Z. P.
,
Hamill
,
J. C.
,
Kuhlman
,
J. A.
,
Eisen
,
J. S.
and
Parichy
,
D. M.
(
2014
).
Thyroid hormone-dependent adult pigment cell lineage and pattern in zebrafish
.
Science
345
,
1358
-
1361
.
Minchin
,
J. E.
and
Hughes
,
S. M.
(
2008
).
Sequential actions of Pax3 and Pax7 drive xanthophore development in zebrafish neural crest
.
Dev. Biol.
317
,
508
-
522
.
Mohlin
,
S.
,
Kunttas
,
E.
,
Persson
,
C. U.
,
Abdel-Haq
,
R.
,
Castillo
,
A.
,
Murko
,
C.
,
Bronner
,
M. E.
and
Kerosuo
,
L.
(
2019
).
Maintaining multipotent trunk neural crest stem cells as self-renewing crestospheres
.
Dev. Biol.
447
,
137
-
146
.
Monk
,
N. A. M.
(
2003
).
Oscillatory expression of Hes1, p53, and NF-kappaB driven by transcriptional time delays
.
Curr. Biol.
19
,
1409
-
1413
.
Montero-Balaguer
,
M.
,
Lang
,
M. R.
,
Sachdev
,
S. W.
,
Knappmeyer
,
C.
,
Stewart
,
R. A.
,
De La Guardia
,
A.
,
Hatzopoulos
,
A. K.
and
Knapik
,
E. W.
(
2006
).
The mother superior mutation ablates foxd3 activity in neural crest progenitor cells and depletes neural crest derivatives in zebrafish
.
Dev. Dyn.
235
,
3199
-
3212
.
Morrison
,
S. J.
,
Perez
,
S. E.
,
Qiao
,
Z.
,
Verdi
,
J. M.
,
Hicks
,
C.
,
Weinmaster
,
G.
and
Anderson
,
D. J.
(
2000
).
Transient Notch activation initiates an irreversible switch from neurogenesis to gliogenesis by neural crest stem cells
.
Cell
101
,
499
-
510
.
Motohashi
,
T.
,
Yamanaka
,
K.
,
Chiba
,
K.
,
Aoki
,
H.
and
Kunisada
,
T.
(
2009
).
Unexpected Multipotency of Melanoblasts Isolated from Murine Skin
.
Stem Cells
27
,
888
-
897
.
Nagao
,
Y.
,
Suzuki
,
T.
,
Shimizu
,
A.
,
Kimura
,
T.
,
Seki
,
R.
,
Adachi
,
T.
,
Inoue
,
C.
,
Omae
,
Y.
,
Kamei
,
Y.
,
Hara
,
I.
et al. 
(
2014
).
Sox5 functions as a fate switch in medaka pigment cell development
.
PLoS Genet.
10
,
e1004246
.
Nagao
,
Y.
,
Takada
,
H.
,
Miyadai
,
M.
,
Adachi
,
T.
,
Seki
,
R.
,
Kamei
,
Y.
,
Hara
,
I.
,
Taniguchi
,
Y.
,
Naruse
,
K.
,
Hibi
,
M.
et al. 
(
2018
).
Distinct interactions of Sox5 and Sox10 in fate specification of pigment cells in medaka and zebrafish
.
PLoS Genet.
14
,
e1007260
.
Nikaido
,
M.
,
Subkhankulova
,
T.
,
Kasianov
,
A.
,
Uroshlev
,
L.
,
Camargo Sosa
,
K.
,
Bavister
,
G.
,
Yang
,
X.
,
Rodrigues
,
F. S. L. M.
,
Carney
,
T. J.
,
Dawes
,
J. H. P.
et al. 
(
2021
).
Zebrafish pigment cells develop directly from highly multipotent progenitors
.
bioRxiv
.
Nishimura
,
E. K.
,
Jordan
,
S. A.
,
Oshima
,
H.
,
Yoshida
,
H.
,
Osawa
,
M.
,
Moriyama
,
M.
,
Jackson
,
I. J.
,
Barrandon
,
Y.
,
Miyachi
,
Y.
and
Nishikawa
,
S.
(
2002
).
Dominant role of the niche in melanocyte stem-cell fate determination
.
Nature
416
,
854
-
860
.
Nishimura
,
E. K.
,
Suzuki
,
M.
,
Igras
,
V.
,
Du
,
J.
,
Lonning
,
S.
,
Miyachi
,
Y.
,
Roes
,
J.
,
Beermann
,
F.
and
Fisher
,
D. E.
(
2010
).
Key roles for transforming growth factor beta in melanocyte stem cell maintenance
.
Cell Stem Cell
6
,
130
-
140
.
Nitzan
,
E.
,
Krispin
,
S.
,
Pfaltzgraff
,
E. R.
,
Klar
,
A.
,
Labosky
,
P. A.
and
Kalcheim
,
C.
(
2013a
).
A dynamic code of dorsal neural tube genes regulates the segregation between neurogenic and melanogenic neural crest cells
.
Development
140
,
2269
-
2279
.
Nitzan
,
E.
,
Pfaltzgraff
,
E. R.
,
Labosky
,
P. A.
and
Kalcheim
,
C.
(
2013b
).
Neural crest and Schwann cell progenitor-derived melanocytes are two spatially segregated populations similarly regulated by Foxd3
.
Proc. Natl. Acad. Sci. U. S. A.
110
,
12709
-
12714
.
Nord
,
H.
,
Dennhag
,
N.
,
Muck
,
J.
and
von Hofsten
,
J.
(
2016
).
Pax7 is required for establishment of the xanthophore lineage in zebrafish embryos
.
Mol. Biol. Cell
27
,
1853
-
1862
.
Obernier
,
K.
and
Alvarez-Buylla
,
A.
(
2019
).
Neural stem cells: origin, heterogeneity and regulation in the adult mammalian brain
.
Development
146
,
dev156059
.
Ochi
,
S.
,
Imaizumi
,
Y.
,
Shimojo
,
H.
,
Miyachi
,
H.
and
Kageyama
,
R.
(
2020
).
Oscillatory expression of Hes1 regulates cell proliferation and neuronal differentiation in the embryonic brain
.
Development
147
,
dev182204
.
O'Reilly-Pol
,
T.
and
Johnson
,
S. L.
(
2008
).
Neocuproine ablates melanocytes in adult zebrafish
.
Zebrafish
5
,
257
-
264
.
Papatsenko
,
D.
,
Darr
,
H.
,
Kulakovskiy
,
I. V.
,
Waghray
,
A.
,
Makeev
,
V. J.
,
MacArthur
,
B. D.
and
Lemischka
,
I. R.
(
2015
).
Single-cell analyses of ESCs reveal alternative pluripotent cell states and molecular mechanisms that control self-renewal
.
Stem Cell Reports
5
,
207
-
220
.
Paratore
,
C.
,
Goerich
,
D. E.
,
Suter
,
U.
,
Wegner
,
M.
and
Sommer
,
L.
(
2001
).
Survival and glial fate acquisition of neural crest cells are regulated by an interplay between the transcription factor Sox10 and extrinsic combinatorial signaling
.
Development
128
,
3949
-
3961
.
Parichy
,
D. M.
and
Spiewak
,
J. E.
(
2015
).
Origins of adult pigmentation: diversity in pigment stem cell lineages and implications for pattern evolution
.
Pigment Cell Melanoma Res
28
,
31
-
50
.
Parichy
,
D. M.
,
Rawls
,
J. F.
,
Pratt
,
S. J.
,
Whitfield
,
T. T.
and
Johnson
,
S. L.
(
1999
).
Zebrafish sparse corresponds to an orthologue of c-kit and is required for the morphogenesis of a subpopulation of melanocytes, but is not essential for hematopoiesis or primordial germ cell development
.
Development
126
,
3425
-
3436
.
Parichy
,
D. M.
,
Turner
,
J. M.
and
Parker
,
N. B.
(
2003
).
Essential role for puma in development of postembryonic neural crest-derived cell lineages in zebrafish
.
Dev. Biol.
256
,
221
-
241
.
Peirano
,
R. I.
and
Wegner
,
M.
(
2000
).
The glial transcription factor Sox10 binds to DNA both as monomer and dimer with different functional consequences
.
Nucleic Acids Res.
28
,
3047
-
3055
.
Perez
,
S. E.
,
Rebelo
,
S.
and
Anderson
,
D. J.
(
1999
).
Early specification of sensory neuron fate revealed by expression and function of neurogenins in the chick embryo
.
Development
126
,
1715
-
1728
.
Petratou
,
K.
,
Spencer
,
S. A.
,
Kelsh
,
R. N.
and
Lister
,
J. A.
(
2021
).
The MITF paralog tfec is required in neural crest development for fate specification of the iridophore lineage from a multipotent pigment cell progenitor
.
PLoS ONE
16
,
e0244794
.
Petratou
,
K.
,
Subkhankulova
,
T.
,
Lister
,
J. A.
,
Rocco
,
A.
,
Schwetlick
,
H.
and
Kelsh
,
R. N.
(
2018
).
A systems biology approach uncovers the core gene regulatory network governing iridophore fate choice from the neural crest
.
PLoS Genet.
14
,
e1007402
.
Qiu
,
X. J.
,
Mao
,
Q.
,
Tang
,
Y.
,
Wang
,
L.
,
Chawla
,
R.
,
Pliner
,
H. A.
and
Trapnell
,
C.
(
2017
).
Reversed graph embedding resolves complex single-cell trajectories
.
Nat. Methods
14
,
979
-
982
.
Quigley
,
I. K.
,
Turner
,
J. M.
,
Nuckels
,
R. J.
,
Manuel
,
J. L.
,
Budi
,
E. H.
,
MacDonald
,
E. L.
and
Parichy
,
D. M.
(
2004
).
Pigment pattern evolution by differential deployment of neural crest and post-embryonic melanophore lineages in Danio fishes
.
Development
131
,
6053
-
6069
.
Raible
,
D. W.
and
Eisen
,
J. S.
(
1994
).
Restriction of neural crest cell fate in the trunk of the embryonic zebrafish
.
Development
120
,
495
-
503
.
Real
,
C.
,
Glavieux-Pardanaud
,
C.
,
Le Douarin
,
N. M.
and
Dupin
,
E.
(
2006
).
Clonally cultured differentiated pigment cells can dedifferentiate and generate multipotent progenitors with self-renewing potential
.
Dev. Biol.
300
,
656
-
669
.
Reedy
,
M. V.
,
Faraco
,
C. D.
and
Erickson
,
C. A.
(
1998
).
The delayed entry of thoracic neural crest cells into the dorsolateral path is a consequence of the late emigration of melanogenic neural crest cells from the neural tube
.
Dev. Biol.
200
,
234
-
246
.
Sagner
,
A.
and
Briscoe
,
J.
(
2019
).
Establishing neuronal diversity in the spinal cord: a time and a place
.
Development
146
,
dev182154
.
Saunders
,
L. M.
,
Mishra
,
A. K.
,
Aman
,
A. J.
,
Lewis
,
V. M.
,
Toomey
,
M. B.
,
Packer
,
J. S.
,
Qiu
,
X.
,
McFaline-Figueroa
,
J. L.
,
Corbo
,
J. C.
,
Trapnell
,
C.
et al. 
(
2019
).
Thyroid hormone regulates distinct paths to maturation in pigment cell lineages
.
Elife
8
,
e45181
.
Schartl
,
M.
,
Larue
,
L.
,
Goda
,
M.
,
Bosenberg
,
M. W.
,
Hashimoto
,
H.
and
Kelsh
,
R. N.
(
2016
).
What is a vertebrate pigment cell?
Pigment Cell Melanoma Res
29
,
8
-
14
.
Schilling
,
T. F.
and
Kimmel
,
C. B.
(
1994
).
Segment and cell type lineage restrictions during pharyngeal arch development in the zebrafish embryo
.
Development
120
,
483
-
494
.
Serbedzija
,
G. N.
,
Bronner-Fraser
,
M.
and
Fraser
,
S. E.
(
1989
).
A vital dye analysis of the timing and pathways of avian trunk neural crest cell migration
.
Development
106
,
809
-
816
.
Shah
,
N. M.
,
Marchionni
,
M. A.
,
Isaacs
,
I.
,
Stroobant
,
P.
and
Anderson
,
D. J.
(
1994
).
Glial growth-factor restricts mammalian neural crest stem-cells to a glial fate
.
Cell
77
,
349
-
360
.
Shah
,
N. M.
,
Groves
,
A. K.
and
Anderson
,
D. J.
(
1996
).
Alternative neural crest cell fates are instructively promoted by TGFbeta superfamily members
.
Cell
85
,
331
-
343
.
Shen
,
C. N.
,
Slack
,
J. M.
and
Tosh
,
D.
(
2000
).
Molecular basis of transdifferentiation of pancreas to liver
.
Nat. Cell Biol.
2
,
879
-
887
.
Sieber-Blum
,
M.
(
1989
).
Commitment of neural crest cells to the sensory neuron lineage
.
Science
243
,
1608
-
1611
.
Sieber-Blum
,
M.
and
Cohen
,
A. M.
(
1980
).
Clonal analysis of quail neural crest cells: they are pluripotent and differentiate in vitro in the absence of noncrest cells
.
Dev. Biol.
80
,
96
-
106
.
Singh
,
A. P.
,
Dinwiddie
,
A.
,
Mahalwar
,
P.
,
Schach
,
U.
,
Linker
,
C.
,
Irion
,
U.
and
Nusslein-Volhard
,
C.
(
2016
).
Pigment cell progenitors in Zebrafish remain multipotent through metamorphosis
.
Dev. Cell
38
,
316
-
330
.
Soldatov
,
R.
,
Kaucka
,
M.
,
Kastriti
,
M. E.
,
Petersen
,
J.
,
Chontorotzea
,
T.
,
Englmaier
,
L.
,
Akkuratova
,
N.
,
Yang
,
Y.
,
Haring
,
M.
,
Dyachuk
,
V.
et al. 
(
2019
).
Spatiotemporal structure of cell fate decisions in murine neural crest
.
Science
364
,
eaas9536
.
Sonnenberg-Riethmacher
,
E.
,
Miehe
,
M.
,
Stolt
,
C. C.
,
Goerich
,
D. E.
,
Wegner
,
M.
and
Riethmacher
,
D.
(
2001
).
Development and degeneration of dorsal root ganglia in the absence of the HMG-domain transcription factor Sox10
.
Mech. Dev.
109
,
253
-
265
.
Steingrimsson
,
E.
,
Moore
,
K. J.
,
Lamoreux
,
M. L.
,
Ferre-D'Amare
,
A. R.
,
Burley
,
S. K.
,
Zimring
,
D. C.
,
Skow
,
L. C.
,
Hodgkinson
,
C. A.
,
Arnheiter
,
H.
,
Copeland
,
N. G.
et al. 
(
1994
).
Molecular basis of mouse microphthalmia (mi) mutations helps explain their developmental and phenotypic consequences
.
Nat. Genet.
8
,
256
-
263
.
Stemple
,
D. L.
and
Anderson
,
D. J.
(
1992
).
Isolation of a stem cell for neurons and glia from the mammalian neural crest
.
Cell
71
,
973
-
985
.
Tassabehji
,
M.
,
Newton
,
V. E.
and
Read
,
A. P.
(
1994
).
Waardenburg syndrome type 2 caused by mutations in the human microphthalmia (MITF) gene [see comments]
.
Nat. Genet.
8
,
251
-
255
.
Tessarollo
,
L.
,
Tsoulfas
,
P.
,
Martin-Zanca
,
D.
,
Gilbert
,
D. J.
,
Jenkins
,
N. A.
,
Copeland
,
N. G.
and
Parada
,
L. F.
(
1993
).
trkC, a receptor for neurotrophin-3, is widely expressed in the developing nervous system and in non-neuronal tissues
.
Development
118
,
463
-
475
.
Than-Trong
,
E.
and
Bally-Cuif
,
L.
(
2015
).
Radial glia and neural progenitors in the adult zebrafish central nervous system
.
Glia
63
,
1406
-
1428
.
Trentin
,
A.
,
Glavieux-Pardanaud
,
C.
,
Le Douarin
,
N. M.
and
Dupin
,
E.
(
2004
).
Self-renewal capacity is a widespread property of various types of neural crest precursor cells
.
Proc. Natl. Acad. Sci. U. S. A.
101
,
4495
-
4500
.
Tryon
,
R. C.
,
Higdon
,
C. W.
and
Johnson
,
S. L.
(
2011
).
Lineage relationship of direct-developing melanocytes and melanocyte stem cells in the zebrafish
.
PLoS ONE
6
,
e21010
.
Vogel
,
K.
and
Weston
,
J. A.
(
1988
).
A subpopulation of cultured avian neural crest cells has transient neurogenic potential
.
Neuron
1
,
569
-
577
.
Waddington
,
C. H.
(
1940
).
Organisers and Genes
.
Cambridge
:
Cambridge University press
.
Walderich
,
B.
,
Singh
,
A. P.
,
Mahalwar
,
P.
and
Nusslein-Volhard
,
C.
(
2016
).
Homotypic cell competition regulates proliferation and tiling of zebrafish pigment cells during colour pattern formation
.
Nat. Commun.
7
,
11462
.
Watanabe
,
N.
,
Motohashi
,
T.
,
Nishioka
,
M.
,
Kawamura
,
N.
,
Hirobe
,
T.
and
Kunisada
,
T.
(
2016
).
Multipotency of melanoblasts isolated from murine skin depends on the Notch signal
.
Dev. Dyn.
245
,
460
-
471
.
Wehrle-Haller
,
B.
and
Weston
,
J. A.
(
1995
).
Soluble and cell-bound forms of steel factor activity play distinct roles in melanocyte precursor dispersal and survival on the lateral neural crest migration pathway
.
Development
121
,
731
-
742
.
Weston
,
J. A.
(
1982
).
Neural crest cell development
.
Prog. Clin. Biol. Res.
85
,
359
-
379
.
Weston
,
J. A.
(
1983
).
Regulation of neural crest cell migration and differentiation
. In
Cell Interactions and Development: Molecular Mechanisms
(ed.
K. M.
Yamada
), pp.
153
-
184
:
John Wiley and Sons, Inc
.
Weston
,
J. A.
(
1991
).
Sequential segregation and fate of developmentally restricted intermediate cell populations in the neural crest lineage
.
Curr. Topics Dev. Biol.
25
,
133
-
153
.
Weston
,
J. A.
and
Thiery
,
J. P.
(
2015
).
Pentimento: neural crest and the origin of mesectoderm
.
Dev. Biol.
401
,
37
-
61
.
White
,
P. M.
,
Morrison
,
S. J.
,
Orimoto
,
K.
,
Kubu
,
C. J.
,
Verdi
,
J. M.
and
Anderson
,
D. J.
(
2001
).
Neural crest stem cells undergo cell-intrinsic developmental changes in sensitivity to instructive differentiation signals
.
Neuron
29
,
57
-
71
.
Wolf
,
F. A.
,
Hamey
,
F. K.
,
Plass
,
M.
,
Solana
,
J.
,
Dahlin
,
J. S.
,
Gottgens
,
B.
,
Rajewsky
,
N.
,
Simon
,
L.
and
Theis
,
F. J.
(
2019
).
PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells
.
Genome Biol.
20
,
59
.
Yang
,
C. T.
and
Johnson
,
S. L.
(
2006
).
Small molecule-induced ablation and subsequent regeneration of larval zebrafish melanocytes
.
Development
133
,
3563
-
3573
.
Yang
,
C. T.
,
Sengelmann
,
R. D.
and
Johnson
,
S. L.
(
2004
).
Larval melanocyte regeneration following laser ablation in zebrafish
.
J. Invest. Dermatol.
123
,
924
-
929
.
Ziller
,
C.
,
Dupin
,
E.
,
Brazeau
,
P.
,
Paulin
,
D.
and
Le Douarin
,
N. M.
(
1983
).
Early segregation of a neuronal precursor cell line in the neural crest as revealed by culture in a chemically defined medium
.
Cell
32
,
627
-
638
.
Ziller
,
C.
,
Fauquet
,
M.
,
Kalcheim
,
C.
,
Smith
,
J.
and
Le Douarin
,
N. M.
(
1987
).
Cell lineages in peripheral nervous system ontogeny: medium-induced modulation of neuronal phenotypic expression in neural crest cell cultures
.
Dev. Biol.
120
,
101
-
111
.
Zirlinger
,
M.
,
Lo
,
L.
,
McMahon
,
J.
,
McMahon
,
A. P.
and
Anderson
,
D. J.
(
2002
).
Transient expression of the bHLH factor neurogenin-2 marks a subpopulation of neural crest cells biased for a sensory but not a neuronal fate
.
Proc. Natl. Acad. Sci. U. S. A.
99
,
8084
-
8089
.

Competing interests

The authors declare no competing or financial interests.