The neural crest (NC) is a multipotent cell population which can give rise to a vast array of derivatives including neurons and glia of the peripheral nervous system, cartilage, cardiac smooth muscle, melanocytes and sympathoadrenal cells. An attractive strategy to model human NC development and associated birth defects as well as produce clinically relevant cell populations for regenerative medicine applications involves the in vitro generation of NC from human pluripotent stem cells (hPSCs). However, in vivo, the potential of NC cells to generate distinct cell types is determined by their position along the anteroposterior (A–P) axis and, therefore the axial identity of hPSC-derived NC cells is an important aspect to consider. Recent advances in understanding the developmental origins of NC and the signalling pathways involved in its specification have aided the in vitro generation of human NC cells which are representative of various A–P positions. Here, we explore recent advances in methodologies of in vitro NC specification and axis patterning using hPSCs.

Human pluripotent stem cells (hPSCs) have become a powerful resource to study embryonic development, circumventing both the technical and ethical obstacles associated with the use of human embryos. By harnessing the correct developmental signalling pathways during hPSC differentiation, it has become increasingly feasible to generate a vast array of human progenitor cell types with distinct identities and differentiation potential. Combined with the use of patient-derived induced pluripotent stem cells (iPSCs) or CRISPR/Cas9 genome editing, it is possible to use these hPSC derived progenitors in disease modelling or for therapeutic applications such as cell replacement/regenerative medicine therapies. The position of the various cell types that make up the emerging nervous system along the embryonic anteroposterior (A–P) axis is a critical determinant of their functionality, developmental potential and disease vulnerability and an increasing body of evidence has pointed out that this is also the case for their hPSC-derived counterparts. Thus, shaping the desired A–P axial identity in vitro is a crucial aspect in the design of hPSC neural differentiation protocols. The A–P regionalisation of hPSC-derived central nervous system (CNS)-associated cell types has already been discussed in detail in some excellent recent reviews elsewhere [1,2]. Here, we focus on recent findings on the key cellular and signalling parameters directing axial identity acquisition during the transition of hPSCs toward NC cells, the precursors of the peripheral nervous system.

During mammalian embryonic development, the earliest cell lineage progenitors arise in the pluripotent post-implantation epiblast with the onset of gastrulation, which is marked by the emergence of a posterior structure known as the primitive streak (PS) under the influence of BMP, Wnt and Nodal signalling, followed by extensive morphogenetic movements leading to the continuous ingression of epiblast cells through the extending PS [3,4]. In turn, this leads to the rearrangement of the single epithelium epiblast into the three germ layers, which act as the building blocks of all embryonic tissues: ectoderm, mesoderm and endoderm. The central nervous system is derived from the ectoderm (neural plate) under the influence of an ‘organiser’ tissue possessing neural inductive properties, manifested though BMP, Wnt and Nodal signalling inhibition [5,6]. In amniote embryos, the principal neural-inductive organisers include the node (Hensen's node in the chick embryo), a region at the anterior tip of the PS, and axial mesoderm cells (its differentiated descendants); these have been shown in grafting experiments to induce a secondary body axis, including a nervous system, in pluripotent epiblast cells [7–11]. It is widely thought, based on early experiments in amphibians [12,13], that neural induction by an organiser, via BMP/Wnt/Nodal inhibition, leads by default to the generation of neural cells with anterior features (forebrain) (‘activation’). These anterior neural cells then become progressively ‘posteriorised’ under the influence of signals such as retinoic acid (RA) and WNT/FGF agonists to produce more posterior structures such as the hindbrain and the spinal cord (‘transformation’).

Fate-mapping/clonal analysis experiments in the mouse embryo have revealed that the ectodermal precursors of brain components and spinal cord are found in distinct, defined locations in the late-gastrula mouse epiblast. These reflect the A–P regionalisation of their descendants i.e. the forebrain is derived exclusively from cells in the anterior epiblast whereas the spinal cord originates from a more distal site around the node [14–16]. By the end of gastrulation, all head neural structures in mammalian embryos have been laid whereas the production of the spinal cord continues throughout embryonic axis elongation via the addition of tissue in a head-to-tail direction. This process of post-gastrulation spinal cord construction appears to be distinct from the generation of the anterior (brain) parts of the central nervous system, consistent with a mode of growth driven by a transient pool of axial progenitors exhibiting stem cell-like properties [17–19]. These are bipotent and are marked by the ability to generate both spinal cord and presomitic mesoderm cells and are thus referred as neuromesodermal progenitors (NMPs). NMPs are located in a ‘posterior growth zone’ around the PS/node, including the node-streak border (NSB) and the caudal lateral epiblast (CLE) during early somite stages, and later in the PS-derived tailbud in a region known as the chordoneural hinge (CNH) (reviewed in [20–22]). Typically, NMPs are characterised by the co-expression of the nascent mesoderm marker Brachyury (Bra/T or TBXT in humans) and the neural marker SOX2, together with other posterior markers such as CDX2 and NKX1–2. NMP specification, maintenance and differentiation are orchestrated primarily via the interplay between the Wnt, FGF and retinoic acid (RA) signalling pathways [20–22]. The final A–P coordinates of the cellular parts of the fully formed nervous system are defined and determined by the combinatorial expression of Hox genes, which are arranged as paralogous groups (PG) (1–13) in four chromosomal clusters (A–D) [23] (Figure 1).

HOX gene expression in neural crest along the anterior–posterior axis.

Figure 1.
HOX gene expression in neural crest along the anterior–posterior axis.

The A–P axis is defined by the combination of overlapping HOX(A–D) expression. Neural crest is divided into four main categories along the A–P axis based on HOX gene expression: cranial, vagal, trunk and sacral. Cranial neural crest (green) is divided into the anterior most HOX negative neural crest and the posterior cranial HOX positive neural crest, expressing HOX PG (1–3). Vagal neural crest (blue) express HOX PG (3–7) genes and are located parallel to the cervical region of the spinal cord. Trunk neural crest (purple) express HOX(6–9) genes are derived adjacent to the thoracic/lumbar region of the spinal cord. Sacral neural crest (orange), derived from the most posterior part of the A–P axis, express HOX (10–13 genes). The potential of NC cells to generate distinct cell types is determined by their position along the anteroposterior (A–P) axis and the major derivatives of each subpopulation is shown in the corresponding section.

Figure 1.
HOX gene expression in neural crest along the anterior–posterior axis.

The A–P axis is defined by the combination of overlapping HOX(A–D) expression. Neural crest is divided into four main categories along the A–P axis based on HOX gene expression: cranial, vagal, trunk and sacral. Cranial neural crest (green) is divided into the anterior most HOX negative neural crest and the posterior cranial HOX positive neural crest, expressing HOX PG (1–3). Vagal neural crest (blue) express HOX PG (3–7) genes and are located parallel to the cervical region of the spinal cord. Trunk neural crest (purple) express HOX(6–9) genes are derived adjacent to the thoracic/lumbar region of the spinal cord. Sacral neural crest (orange), derived from the most posterior part of the A–P axis, express HOX (10–13 genes). The potential of NC cells to generate distinct cell types is determined by their position along the anteroposterior (A–P) axis and the major derivatives of each subpopulation is shown in the corresponding section.

Close modal

The origin of NC is tightly linked with that of the central nervous system, specified at the neural plate border between the neural plate (future CNS) and the non-neural ectoderm (future epidermis). Initially, NC is induced by a combination of signals, primarily linked to the BMP, FGF and Wnt pathways [24,25] and these have been reviewed in depth elsewhere [26–31]. Combined signalling from these pathways induces the expression of key NC specifier transcription factors such as MSX1/2, PAX3/7 and ZIC genes which overlap at the future neural plate border [32–34]. These transcription factors, induce, in turn, definitive NC markers including SOX9/10, SNAI2 and FOXD3 forming a complex gene regulatory network which specifies and maintains NC cells [30,35,36].

The NC populates the neural tube along the entire A–P axis and can be subdivided into four main subregions along this axis: cranial, vagal, trunk and sacral. Their position along this axis determines their gene expression, migratory paths and subsequently their differentiation potential and contribution to the embryo. The cranial NC gives rise to the mesectoderm (e.g. cartilage and bone) in the head, melanocytes, sensory neurons and glia [37,38]. They are further divided into an anterior Hox negative and a more posterior Hox PG (1–3)-positive domain [39] (Figure 1). Parallel to the first seven somites of the neural tube, vagal NC express Hox PG (3–7) members and contribute to the enteric nervous system (ENS) and various heart structures [40–45]. Located between somites 8–24 (in mouse), the trunk NC contributes to the sympathetic nervous system, neuroendocrine cells, and melanocytes [46]. Trunk NC cells are characterised by the expression of more posterior Hox genes (Hox PG (6–9)) [43,47–49] (Figure 1). The region from somite 24 (in mouse) and below coincides with lumbosacral Hox PG 10–13 expression (Figure 1) and the presence of the sacral NC [50,51]. In amniotes, the sacral NC contributes to the most distal part of the enteric nervous system, unlike in zebrafish where the ENS is derived only from the vagal NC [52–56]. In addition to ENS, sacral NC contributes to the pelvic ganglia and therefore provides autonomic innovation of the lower urogenital tract [57].

Fate mapping and lineage tracing experiments have indicated a close link between NMPs and posterior NC precursors [58–62]. For example, homotopic grafting experiments of SOX2+/T+ cells taken from the E8.5 mouse CLE were found to result in NC contribution [59]. Similarly, lineage tracing of cells expressing NMP markers such as Nkx1–2, Tbx6/Sox2, Bra/T and FoxB1 revealed that these contribute to neural, mesodermal and NC lineages [58,61,63–68]. More recent work, using single-cell transcriptomics and epigenomic profiling in zebrafish, has further demonstrated that posterior NC is derived from an NMP-derived pre-neural intermediate [69]. Taken together these data strongly suggest that, at least a fraction of trunk and sacral NC are directly derived from axial progenitors, which are competent to generate presomitic mesoderm.

Cranial NC

Early in vitro differentiation experiments focused on the induction of NC cells from a hPSC-derived neuroectodermal progenitor intermediate [70–72]. These initial protocols relied on recapitulating the signalling events which accompany neural induction in vivo, by blocking BMP and TGFB/Activin/Nodal signalling using corresponding small molecule inhibitors (termed dual SMAD inhibition) [70,73]. However, these protocols typically yielded a variable and low percentage of NC progenitors and as NC specification in vivo primarily occurs under the influence of intermediate BMP and Wnt signalling, subsequent studies found that a short pulse of dual SMAD-inhibition followed by the addition of BMP and Wnt signalling yielded a higher proportion of NC progenitors [74,75]. Further refinement and characterisation of these strategies revealed that hPSC-derived NC appears to emerge from a pre-neural border intermediate which relies on intermediate BMP and Wnt activity [76–78]. PAX6 expression (indicating neural progenitor specification) in these protocols occurred at the expense of the NC specifier SOX10 in line with in vivo findings indicating these cells may diverge at an earlier timepoint (reviewed further in [31]). Varying endogenous BMP signalling levels within the cells and/or BMP ligands in the media were found to affect overall BMP activity and lead to large variations in the efficiency in NC specification thus complicating the view of BMP requirement in vitro[79]. Furthermore, high BMP signalling can lead to specification of the cranial placode or non-neural ectoderm whereas low BMP can result in neuroectoderm specification [78]. As a result, more recent improved protocols which modulate BMP levels through top-down inhibition demonstrated that a precise level of intermediate BMP activity is required to specify NC efficiently and reproducibly from hPSCs in vitro [79,80]. A common feature of these approaches is that the NC cells they give rise to exhibit minimal HOX gene expression indicating a cranial axial identity. This is also supported by the demonstration that they can be further directed to differentiate toward peripheral neurons including (nociceptors, mechanoreceptors, and proprioceptors) and glia, melanocytes and mesenchymal lineages (smooth muscle, osteogenic and chondrogenic cells) [39,81].

Vagal NC

Early efforts in the posterior patterning of human NC cells in vitro have been heavily influenced by Nieuwkoop's ‘activation-transformation’ model focusing on the production of an anterior neuroectodermal precursor followed by addition of NC promoting signals in combination with posteriorising agents such as RA. Such approaches have been shown to yield predominantly NC cells marked by HOX PG (1–7) members denoting the acquisition of a posterior cranial/vagal axial identity [74,82–85]. Using RA to posteriorise HOX negative cells requires optimisation as both the timing of RA addition and RA concentration appear to impact NC specification in vitro [86]. As such, although increasing concentrations of RA results in increased expression of posterior HOX genes, this occurs at the expense of NC markers such as SOX10 [83,86]. Similar to their in vivo counterparts, vagal NC cells generated through these protocols have been shown to further differentiate, via culture in neurotrophic media, toward various components of the ENS including distinct enteric neuronal subtypes marked by expression of 5HT, GABA and NOS [82,83,85,86]. Interestingly, cranial NC when treated in the same conditions, differentiated into TH positive neurons which are positive for TRKB, indicating that they preferentially give rise to sympathetic neurons [82]. Similarly, when cranial and vagal NC cells were grafted into the muscle wall of the mouse cecum at 3-6 weeks, vagal NC migrated and repopulated the host colon to a greater extent than cranial NC [82]. This finding highlights the importance of generating cell progenitors of the correct regional identity for potential use in clinical applications from hPSCs and demonstrates that early axial specification impacts cell functionality in vivo. Crucially, transplantation experiments revealed that hPSC-derived vagal NC/ENS progenitors can colonise and differentiate efficiently within the gut of host mice and even rescue the lethal aganglionic phenotype exhibited by Ednrb mutant mice, a model of Hirschsprung disease thus serving as a promising population for the development of cell therapy approaches against conditions affecting ENS specification [82,86].

Recent advances in organoid-based differentiation approaches have opened new avenues toward the establishment of more sophisticated in vitro models for dissecting NC function and related diseased states. To effectively study the complex role of ENS within the gastrointestinal (GI) tract, several studies have combined hPSC-derived vagal NC cells with human intestinal organoids (HIO), thereby generating an hPSC-derived assembloid model of intestinal tissue with functional ENS [82,84,87]. When combined, vagal NC progenitors generated glia and neurons of the ENS which integrated into the smooth muscle layer of the HIO and were found to promote cell proliferation in intestinal crypts and gene expression indicative of intestinal cell development. More recent work has described the generation of assembloid cultures containing colonic intestinal tissue with ENS and blood vessels; a model perhaps more suited to diseases which impact the distal part of the GI tract, including Crohn's and Hirschsprung's disease [88].

Trunk NC

Although RA addition to cranial NC cells results in posteriorisation to a vagal axial identity, it does not lead to the induction of HOX genes posterior to HOX PG 7 [82,83,86] and early attempts to produce trunk NC derivatives from hPSCs have relied on the expression of PHOX2B or ASCL1, which also mark vagal NC derivatives in addition to trunk NC-derived sympathoadrenal progenitors [83,89]. A number of recent studies have demonstrated the efficient generation of NMP-like cells from both mouse and human PSCs following stimulation with Wnt (using the GSK-3 inhibitor CHIR99021 (CHIR) or Wnt3a) and FGF2/8 for 2–4 days [67,85,90–95]. Since then, extensive studies have optimised their generation and utilised these progenitors in vitro (reviewed in [20]). PSC-derived NMP-like cells have been shown to give rise to both neural spinal cord and paraxial mesoderm cells [1,93,96–100]. Interestingly, some studies have demonstrated that trunk NC can be generated via an NMP intermediate reflecting the developmental origin of these cells in vivo [85,91,101–105]. The earliest example of trunkNC induction and differentiation in vitro found that treating NMP-like cells with BMP resulted in up-regulation of NC markers and generated NC-like cells capable of generating peripheral nervous system (e.g. dorsal root ganglia and sympathetic ganglia) and Schwann cells and when transplanted to the mid/hindgut in quail migrated into the gut wall [102]. Subsequent studies, using a similar NMP cell induction method found that NC cells expressed trunk HOX genes and could be differentiated to sympathoadrenal progenitors and chromaffin cells [85,103,104,106]. Expanding these findings, Frith and colleagues demonstrated that a fraction of TBXT+ hPSC-derived NMPs expressed neural plate border genes and early neural crest markers (e.g. PAX3/7, MSX1/2 and ZIC1/3) [85]. Intermediate BMP and Wnt signalling then facilitated the production of high yields of SOX10+ NC-like cells from TBXT+ NMPs, which express HOX PG (6–9), the posterior spinal cord/NC marker CDX2 and can be differentiated to sympathoadrenal progenitors/sympathetic neurons [85,103]. Transplantations into the chick dorsal neural tube also showed the potential of these cells to integrate and migrate to the dorsal root ganglia recapitulating similar behaviour to their in vivo counterparts [85]. Interestingly, NMP-derived trunk NC cells have been reported to remain capable of generating adipocytes, chondrocytes and osteocytes [101,102]. This indicates either the co-emergence of minor cranial NC cell subpopulations or suggests a greater developmental potential of hPSC-derived trunk NC in line with data showing that in vitro cultured embryonic avian trunk NC exhibits the ability to generate some ectomesenchymal derivatives [107,108].

To achieve a homogeneous TBXT/SOX2-expressing NMP population and subsequent trunk NC formation, the intensity of Wnt stimulation requires titration to an intermediate to high level (CHIR; 3–12 µM) level [85,91,101,105]. The level required for NMP specification is higher than the low-intermediate level (<3 µM) activation which is required to generate cranial NC-like cells [101,105]. Increasing levels of Wnt stimulation also induce a higher expression of posterior HOX genes (e.g. HOXC9), most likely as a result of CDX up-regulation which drives posterior HOX gene expression and mediates NC induction [101,109–111]. Similarly to the fine tuning required to achieve correct BMP signalling, the specific level of CHIR required is dependent on each cell line, confluency of starting cell population and CHIR batch to batch variation, therefore, the unique balance of Wnt signalling requires optimisation in each differentiation protocol. Furthermore, the length as well as magnitude of Wnt signalling activation is also important in trunk NC specification as prolonged activation of Wnt signalling may drive mesoderm specification from NMPs [90], while combined Wnt/FGF signalling results in neural commitment [91,98,100].

FGF signalling is known to play a role in NMP specification and shaping axial identity, although some in vitro protocols omit FGF ligands indicating endogenous FGF signalling may be sufficient to induce an NMP identity [99,102,105,106]. Protocols which rely on CHIR alone or CHIR/FGF induced NMPs produce NC at a similar efficiency (Table 1). In axial progenitors, FGF signalling is required to pace the ‘HOX clock’ and mediates temporal HOX gene activation [92]. Similarly, inhibition of FGF during NC induction impairs HOX and CDX2 expression, although it does not impact SOX10 induction [101,105]. Conversely, high levels of FGF signalling may drive more sacral HOX expression (HOX PG (10–13)) in NC cultures at the expense of SOX10 expression [105]. Together, these data suggest that a certain level of FGF signalling activity is required to induce a trunk HOX identity and posterior markers such as CDX2 [101,105]. Transcriptomic analysis of NMPs and NMP-derived NC cells indicate that collinear HOX gene expression progresses over time in culture [85,91]. However, further work is required to define the signalling determinants of posterior axial identity/collinear HOX gene activation during NC specification from NMPs and more specifically whether this occurs in NMP/pre-neural progenitors, committed NC cells, or both.

A promising avenue in 3-dimensional (3D) cell culture models containing trunk NC derivatives, is the recent development of trunk neuromuscular organoids (NMOs) [112,113]. Using hPSC-derived NMPs aggregated to form 3D spheres, these bipotent progenitors are capable of generating mesodermal and ectodermal derivatives which self-organise into 3D organoids containing neuromuscular junctions (NMJ). As NMPs also contribute to trunk NC lineages, NMOs were found to contain Schwann cells which are essential for maturation and support of the neuromuscular junction in vivo and have been implicated in diseases such as amyotrophic lateral sclerosis [112,114]. Thus, this approach enables the analysis of the complex interaction of cells at the NMJ and the role of distinct cell types in disease onset and progression e.g. through the use of patient-derived induced pluripotent stem cell lines. More complex 3D models (known as gastruloids) reflecting early mammalian development have also been developed and these involve the induction of multiple interacting cell lineages [115–118]. These models have been shown to undergo transitions which recapitulate axis elongation and symmetry breaking events, somitogenesis, rudimentary neural tube formation and cardiogenesis [115–117,119,120]. Moreover, they have recently been shown to contain NC progenitors which differentiate to the peripheral nervous system and integrate with a primitive gut tube structure making them a useful model to study early human NC development [121].

Sacral NC

To date sacral neural crest has received little attention in in vitro studies and has limited characterisation in vivo. Recent work in vivo, revealed that the trunk to tail transition in mouse embryos, is driven by a gene regulatory network centred around GDF11, LIN28A and HOX13 genes [122–124]. GDF11, a member of the TGFβ signalling pathway, regulates HOX gene expression at the sacral-most level of the axis. In a negative feedback loop, acting to terminate axis elongation, up-regulation of HOX13 genes inhibits LIN28A expression which results in cell cycle exit. In line with this, in vitro studies have demonstrated that GDF11 drives sacral HOX expression in derivatives of NMP-like cells in vitro [91–93]. Similarly, a rise in endogenous GDF11 signalling in mixed pre-neural progenitors/neural crest cultures was shown to induce sacral HOX expression thus facilitating the generation of neural crest with a sacral identity [91]. Further work to characterise these cells and their differentiation potential may yield an exciting sacral NC model with potential for therapeutic application and disease modelling for both the ENS and lower urogenital tract.

Over the last decade much progress has been made in developing human in vitro models of neural crest formation, aiding our understanding of the precise signalling pathways required to specify NC cells corresponding to all axial identities and consequently human neural crest biology (Table 1 and Figure 2). The success and advances in generating specific neural crest derivatives has also opened new opportunities to generate complex multilineage 3D models which mimic early development and exhibit complex multi-tissue interactions. Specifically, cranial NC cultures have demonstrated a potential application in tendon healing whereas vagal NC hold promise for treatment of ENS defects such as in Hirschsprung's disease [82,86,125]. However, further work is required to produce purified NC populations of defined regional identities, to understand the heterogeneity and potential of distinct NC populations and rigorously test their differentiation potential in vitro and in vivo. The increasing availability of single-cell transcriptomic datasets and 3D and 2D culture systems combined with improved fully defined differentiation protocols will significantly aid this goal.

Anterior–posterior patterning in in vitro derived neural crest.

Figure 2.
Anterior–posterior patterning in in vitro derived neural crest.

Schematic diagram summarising the generation of the four subcategories of NC from human pluripotent stem cells (hPSCs). The major signalling pathways known to regulate differentiation are displayed for each subcategory. Cranial and vagal NC differentiation methods use the same initial signalling pathways but vagal NC are posteriorised using the addition of retinoic acid (RA) signalling. Trunk and sacral NC are derived via a common neuromesodermal progenitor (NMP) population. When sacral NC obtain a sacral identity remains unclear, but two potential routes and signalling pathways are displayed in grey. In the first route, trunk NC are posteriorized by GDF signalling, whereas in the second NMPs or pre-neural progenitors (PNP) are posteriorized by GDF prior to NC specification.

Figure 2.
Anterior–posterior patterning in in vitro derived neural crest.

Schematic diagram summarising the generation of the four subcategories of NC from human pluripotent stem cells (hPSCs). The major signalling pathways known to regulate differentiation are displayed for each subcategory. Cranial and vagal NC differentiation methods use the same initial signalling pathways but vagal NC are posteriorised using the addition of retinoic acid (RA) signalling. Trunk and sacral NC are derived via a common neuromesodermal progenitor (NMP) population. When sacral NC obtain a sacral identity remains unclear, but two potential routes and signalling pathways are displayed in grey. In the first route, trunk NC are posteriorized by GDF signalling, whereas in the second NMPs or pre-neural progenitors (PNP) are posteriorized by GDF prior to NC specification.

Close modal
Table 1
Published studies reporting the generation and axial characterisation, determined by HOX gene expression, of in vitro derived neural crest
StudyAxial identity based on HOX gene expressionBasal mediaDifferentiation protocolSOX10 expression (%)Differentiation potential (in vitro)
[74Cranial KSR/N2 (DMEM:F12) LDN (D0–3); SB43 (D0–4); 3 µM CHIR (D2–11) 53% Melanocytes 
[82Cranial KSR/N2 (DMEM:F12) SB43, LDN (D0–11); 3 µM CHIR (D2–11) 65% Enteric neurons/glia 
[101,126Cranial B27 (DMEM:F12) + 0.5% BSA 2.5–3 µM CHIR, ROCKi (D0–2); basal media (D0–5) 60% Osteocytes, chondrocytes, smooth muscle, adipocytes, peripheral neurons, glia and melanocytes 
[76Cranial B27 (DMEM:F12) + 0.5% BSA 3 µM CHIR (D0–5); ROCKi (D0–2) 63% Chondrocytes, osteoblasts, peripheral neurons, glia and melanoblasts 
[86Vagal N2 (DMEM:F12) SB43, DMH1, BMP4, 1 μM CHIR, (D0-6); ROCKi (D0-2); RA (D4-6) 40% Enteric neurons/glia 
[82Vagal KSR/N2 (DMEM:F12) SB43, LDN (D0-11); 3 μM CHIR (D2-11); RA (D6-11) 60% Enteric neurons/glia 
[74Vagal KSR/N2 (DMEM:F12) LDN (D0–3); SB43 (D0–4); 3 µM CHIR (D2–11); FGF or RA (various time points) 10–60% Melanocytes 
[83Vagal KSR/N2 (DMEM:F12) SB43 (D0–11); 3 µM CHIR (D0–11) n.d. (20% SOX10E1+ or 20% PHOX2B+Sympathoadrenal cells and melanoblasts 
[84Vagal N2B27 (Neurobasal)/DMEM:F12 + insulin FGF2, EGF (D0–6); RA (D4–6) n.d. (99.5% p75NTR+/HNK-1+Enteric neurons/glia 
[105Trunk N2 (DMEM:F12) 3 µM CHIR, ROCKi (D0–2); BMP4 and DMH1 (D2–5) 80–90% Melanocytes, peripheral neurons, sympathoadrenal, glia, osteocytes and smooth muscle 
[101Trunk B27 (DMEM:F12) + 0.5% BSA 7–12 µM CHIR, ROCKi (D0–2); basal media (D0–5) 75% Osteoblasts, smooth muscle, peripheral neurons, glia, melanocytes and sympathoadrenal cells 
[85Trunk N2B27 (DMEM:F12, D0–3); N2 (DMEM:F12, D3–9) FGF2 and 3–4 µM CHIR (D0–D3); SB42, DMH1, BMP4, 1 µM CHIR (D3–9) 60% Sympathoadrenal progenitors/sympathetic neurons 
[104Trunk N2B27 (D1–5); 1NB (D5–D11) 3 µM CHIR, SB43 (D0–5); BMP2, FGF2 (D5–D11) n.d. (98% p75NTR+/HNK-1+) Sympathoadrenal cells/adrenomedullary chromaffin cells 
[106Trunk E6 2 µM CHIR, SB (D0–3); FGF2, BMP, RA (D3–10) 40–50% Sympathetic neurons/glia 
[91Trunk and Sacral N2B27 -vitamin A (DMEM:F12) FGF2, 3–5 µM CHIR, AGN, ROCKi (D0-36h); FGF2, 3–5 µM CHIR, AGN, ROCKi (36h — P10) 40% Smooth muscle 
StudyAxial identity based on HOX gene expressionBasal mediaDifferentiation protocolSOX10 expression (%)Differentiation potential (in vitro)
[74Cranial KSR/N2 (DMEM:F12) LDN (D0–3); SB43 (D0–4); 3 µM CHIR (D2–11) 53% Melanocytes 
[82Cranial KSR/N2 (DMEM:F12) SB43, LDN (D0–11); 3 µM CHIR (D2–11) 65% Enteric neurons/glia 
[101,126Cranial B27 (DMEM:F12) + 0.5% BSA 2.5–3 µM CHIR, ROCKi (D0–2); basal media (D0–5) 60% Osteocytes, chondrocytes, smooth muscle, adipocytes, peripheral neurons, glia and melanocytes 
[76Cranial B27 (DMEM:F12) + 0.5% BSA 3 µM CHIR (D0–5); ROCKi (D0–2) 63% Chondrocytes, osteoblasts, peripheral neurons, glia and melanoblasts 
[86Vagal N2 (DMEM:F12) SB43, DMH1, BMP4, 1 μM CHIR, (D0-6); ROCKi (D0-2); RA (D4-6) 40% Enteric neurons/glia 
[82Vagal KSR/N2 (DMEM:F12) SB43, LDN (D0-11); 3 μM CHIR (D2-11); RA (D6-11) 60% Enteric neurons/glia 
[74Vagal KSR/N2 (DMEM:F12) LDN (D0–3); SB43 (D0–4); 3 µM CHIR (D2–11); FGF or RA (various time points) 10–60% Melanocytes 
[83Vagal KSR/N2 (DMEM:F12) SB43 (D0–11); 3 µM CHIR (D0–11) n.d. (20% SOX10E1+ or 20% PHOX2B+Sympathoadrenal cells and melanoblasts 
[84Vagal N2B27 (Neurobasal)/DMEM:F12 + insulin FGF2, EGF (D0–6); RA (D4–6) n.d. (99.5% p75NTR+/HNK-1+Enteric neurons/glia 
[105Trunk N2 (DMEM:F12) 3 µM CHIR, ROCKi (D0–2); BMP4 and DMH1 (D2–5) 80–90% Melanocytes, peripheral neurons, sympathoadrenal, glia, osteocytes and smooth muscle 
[101Trunk B27 (DMEM:F12) + 0.5% BSA 7–12 µM CHIR, ROCKi (D0–2); basal media (D0–5) 75% Osteoblasts, smooth muscle, peripheral neurons, glia, melanocytes and sympathoadrenal cells 
[85Trunk N2B27 (DMEM:F12, D0–3); N2 (DMEM:F12, D3–9) FGF2 and 3–4 µM CHIR (D0–D3); SB42, DMH1, BMP4, 1 µM CHIR (D3–9) 60% Sympathoadrenal progenitors/sympathetic neurons 
[104Trunk N2B27 (D1–5); 1NB (D5–D11) 3 µM CHIR, SB43 (D0–5); BMP2, FGF2 (D5–D11) n.d. (98% p75NTR+/HNK-1+) Sympathoadrenal cells/adrenomedullary chromaffin cells 
[106Trunk E6 2 µM CHIR, SB (D0–3); FGF2, BMP, RA (D3–10) 40–50% Sympathetic neurons/glia 
[91Trunk and Sacral N2B27 -vitamin A (DMEM:F12) FGF2, 3–5 µM CHIR, AGN, ROCKi (D0-36h); FGF2, 3–5 µM CHIR, AGN, ROCKi (36h — P10) 40% Smooth muscle 

The concentration of CHIR used in each experiment has been noted due to the role of Wnt signalling in mediated the cranial-trunk switch [101,105,127]. Only studies which have determined axial identity through HOX gene analysis have been included in this table [74,76,82–86,91,101,104–106,126].

1

NB, neurobasal [102]; CHIR, CHIR99021; D, day; P, passage; SB43, SB431542; ROCKi, ROCK inhibition; LDN, LDN193189; AGN, AGN193109; RA, retinoic acid.

  • NC can contribute to up to 47 cell types in humans [128]. NC differentiation potential is shaped by its A–P position, therefore defining and modulating A–P axial identity in vitro is a crucial aspect in the design of hPSC neural differentiation protocols.

  • Cranial NC are specified from hPSCs through a pre-neural/neural plate border intermediate and under the influence of intermediate BMP activity and low Wnt signalling. RA signalling posteriorises cranial neural crest to a vagal identity but does not efficiently induce trunk NC. Instead, trunk NC specification relies on the generation of an NMP intermediate and subsequent intermediate BMP signalling. Generating sacral neural crest requires further work although GDF11 signalling may be critical in inducing a lumbosacral axial identity in hPSC neural derivatives.

  • The therapeutic potential of each of these subgroups remains an exciting avenue to explore and calls for in depth understanding of heterogeneity of progenitor pool and their in vivo functionality.

The authors declare that there are no competing interests associated with the manuscript.

Open access for this article was enabled by the participation of University of Sheffield in an all-inclusive Read & Publish pilot with Portland Press and the Biochemical Society under a transformative agreement with JISC.

F.C. and A.T. wrote and edited the manuscript

This work is supported by the Biological Sciences Research Council (New Investigator Research Grant BB/P000444/1), H2020 European Research Council (824070), Children's Cancer and Leukaemia Group/Neuroblastoma U.K. (CCLGA 2019 28) and the Medical Research Council (MR/V002163/1).

A–P

Anterior–posterior

CHIR

CHIR99021

ENS

enteric nervous system

hPSC

human pluripotent stem cells

NC

neural crest

NMPs

Neuromesodermal progenitors

PG

paralog groups

1
Nedelec
,
S.
and
Martinez-Arias
,
A.
(
2021
)
In vitro models of spinal motor circuit's development in mammals: achievements and challenges
.
Curr. Opin. Neurobiol.
66
,
240
249
2
Sances
,
S.
,
Bruijn
,
L.I.
,
Chandran
,
S.
,
Eggan
,
K.
,
Ho
,
R.
,
Klim
,
J.R.
et al (
2016
)
Modeling ALS with motor neurons derived from human induced pluripotent stem cells
.
Nat. Neurosci.
19
,
542
553
3
Arnold
,
S.J.
and
Robertson
,
E.J.
(
2009
)
Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo
.
Nat. Rev. Mol. Cell Biol.
10
,
91
103
4
Tam
,
P.P.
and
Loebel
,
D.A.
(
2007
)
Gene function in mouse embryogenesis: get set for gastrulation
.
Nat. Rev. Genet.
8
,
368
381
5
Anderson
,
C.
and
Stern
,
C.D.
(
2016
)
Organizers in development
.
Curr. Top. Dev. Biol.
117
,
435
454
6
Martinez Arias
,
A.
and
Steventon
,
B.
(
2018
)
On the nature and function of organizers
.
Development
145
,
dev159525
7
Waddington
,
C.H.
(
1933
)
Induction by the primitive streak and its derivatives in the chick
.
J. Exp. Biol.
10
,
38
46
8
Waddington
,
C.H.
and
Cohen
,
A.
(
1936
)
Experiments on the development of the head of the chick embryo
.
J. Exp. Biol.
13
,
219
236
9
Beddington
,
R.S.
(
1994
)
Induction of a second neural axis by the mouse node
.
Development
120
,
613
620
10
Waddington
,
C.H.
and
Gray
,
J.
(
1932
)
III. Experiments on the development of chick and duck embryos, cultivated in vitro
.
Philos. Trans. R. Soc. Lond. B
221
,
179
230
11
Tam
,
P.P.
and
Steiner
,
K.A.
(
1999
)
Anterior patterning by synergistic activity of the early gastrula organizer and the anterior germ layer tissues of the mouse embryo
.
Development
126
,
5171
5179
12
Nieuwkoop
,
P.D.
(
1952
)
Activation and organization of the central nervous system in amphibians. Part III. Synthesis of a new working hypothesis
.
J. Exp. Zool.
120
,
83
108
13
Nieuwkoop
,
P.D.
and
Nigtevecht
,
G.
(
1954
)
Neural activation and transformation in explants of competent ectoderm under the influence of fragments of anterior notochord in urodeles
.
Development
2
,
175
193
14
Cajal
,
M.
,
Lawson
,
K.A.
,
Hill
,
B.
,
Moreau
,
A.
,
Rao
,
J.
,
Ross
,
A.
et al (
2012
)
Clonal and molecular analysis of the prospective anterior neural boundary in the mouse embryo
.
Development
139
,
423
436
15
Forlani
,
S.
,
Lawson
,
K.A.
and
Deschamps
,
J.
(
2003
)
Acquisition of Hox codes during gastrulation and axial elongation in the mouse embryo
.
Development
130
,
3807
3819
16
Tam
,
P.P.
(
1989
)
Regionalisation of the mouse embryonic ectoderm: allocation of prospective ectodermal tissues during gastrulation
.
Development
107
,
55
67
17
Mathis
,
L.
and
Nicolas
,
J.F.
(
2000
)
Different clonal dispersion in the rostral and caudal mouse central nervous system
.
Development
127
,
1277
18
Tzouanacou
,
E.
,
Wegener
,
A.
,
Wymeersch
,
F.J.
,
Wilson
,
V.
and
Nicolas
,
J.F.
(
2009
)
Redefining the progression of lineage segregations during mammalian embryogenesis by clonal analysis
.
Dev. Cell
17
,
365
376
19
Roszko
,
I.
,
Faure
,
P.
and
Mathis
,
L.
(
2007
)
Stem cell growth becomes predominant while neural plate progenitor pool decreases during spinal cord elongation
.
Dev Biol.
304
,
232
245
20
Wymeersch
,
F.J.
,
Wilson
,
V.
and
Tsakiridis
,
A.
(
2021
)
Understanding axial progenitor biology in vivo and in vitro
.
Development
148
,
dev18062
21
Henrique
,
D.
,
Abranches
,
E.
,
Verrier
,
L.
and
Storey
,
K.G.
(
2015
)
Neuromesodermal progenitors and the making of the spinal cord
.
Development
142
,
2864
2875
22
Sambasivan
,
R.
and
Steventon
,
B.
(
2021
)
Neuromesodermal progenitors: a basis for robust axial patterning in development and evolution
.
Front. Cell Dev. Biol.
8
,
607516
23
Deschamps
,
J.
and
van Nes
,
J.
(
2005
)
Developmental regulation of the Hox genes during axial morphogenesis in the mouse
.
Development
132
,
2931
2942
24
LaBonne
,
C.
and
Bronner-Fraser
,
M.
(
1998
)
Neural crest induction in Xenopus: evidence for a two-signal model
.
Development
125
,
2403
2414
25
Streit
,
A.
and
Stern
,
C.D.
(
1999
)
Establishment and maintenance of the border of the neural plate in the chick: involvement of FGF and BMP activity
.
Mech. Dev.
82
,
51
66
26
Steventon
,
B.
,
Carmona-Fontaine
,
C.
and
Mayor
,
R.
(
2005
)
Genetic network during neural crest induction: from cell specification to cell survival
.
Semin. Cell Dev. Biol.
16
,
647
654
27
Pla
,
P.
and
Monsoro-Burq
,
A.H.
(
2018
)
The neural border: induction, specification and maturation of the territory that generates neural crest cells
.
Dev Biol.
444
,
S36
S46
28
Stuhlmiller
,
T.J.
and
Garcia-Castro
,
M.I.
(
2012
)
Current perspectives of the signaling pathways directing neural crest induction
.
Cell. Mol. Life Sci.
69
,
3715
3737
29
Milet
,
C.
and
Monsoro-Burq
,
A.H.
(
2012
)
Neural crest induction at the neural plate border in vertebrates
.
Dev. Biol.
366
,
22
33
30
Prasad
,
M.S.
,
Sauka-Spengler
,
T.
and
LaBonne
,
C.
(
2012
)
Induction of the neural crest state: control of stem cell attributes by gene regulatory, post-transcriptional and epigenetic interactions
.
Dev. Biol.
366
,
10
21
31
Prasad
,
M.S.
,
Charney
,
R.M.
and
Garcia-Castro
,
M.I.
(
2019
)
Specification and formation of the neural crest: perspectives on lineage segregation
.
Genesis
57
,
e23276
32
Bang
,
A.G.
,
Papalopulu
,
N.
,
Goulding
,
M.D.
and
Kintner
,
C.
(
1999
)
Expression of Pax-3 in the lateral neural plate is dependent on a Wnt-mediated signal from posterior nonaxial mesoderm
.
Dev. Biol.
212
,
366
380
33
Monsoro-Burq
,
A.H.
,
Wang
,
E.
and
Harland
,
R.
(
2005
)
Msx1 and Pax3 cooperate to mediate FGF8 and WNT signals during Xenopus neural crest induction
.
Dev. Cell
8
,
167
178
34
Garnett
,
A.T.
,
Square
,
T.A.
and
Medeiros
,
D.M.
(
2012
)
BMP, Wnt and FGF signals are integrated through evolutionarily conserved enhancers to achieve robust expression of Pax3 and Zic genes at the zebrafish neural plate border
.
Development
139
,
4220
4231
35
Martik
,
M.L.
and
Bronner
,
M.E.
(
2017
)
Regulatory logic underlying diversification of the neural crest
.
Trends Genet.
33
,
715
727
36
Simoes-Costa
,
M.
and
Bronner
,
M.E.
(
2015
)
Establishing neural crest identity: a gene regulatory recipe
.
Development
142
,
242
257
37
Rothstein
,
M.
,
Bhattacharya
,
D.
and
Simoes-Costa
,
M.
(
2018
)
The molecular basis of neural crest axial identity
.
Dev. Biol.
444
,
S170
SS80
38
Le Douarin
,
N.M.
,
Creuzet
,
S.
,
Gr
,
C.
and
Dupin
,
E.
(
2004
)
Neural crest cell plasticity and its limits
.
Development
131
,
4637
4650
39
Trainor
,
P.A.
(
2003
)
Making headway: the roles of Hox genes and neural crest cells in craniofacial development
.
ScientificWorldJournal
3
,
240
264
40
Diman
,
N.Y.S.G.
,
Remacle
,
S.
,
Bertrand
,
N.
,
Picard
,
J.J.
,
Zaffran
,
S.
and
Rezsohazy
,
R.
(
2011
)
A retinoic acid responsive Hoxa3 transgene expressed in embryonic pharyngeal endoderm, cardiac neural crest and a subdomain of the second heart field
.
PLoS ONE
6
,
e27624
41
Fu
,
M.
,
Lui
,
V.C.
,
Sham
,
M.H.
,
Cheung
,
A.N.
and
Tam
,
P.K.
(
2003
)
HOXB5 expression is spatially and temporarily regulated in human embryonic gut during neural crest cell colonization and differentiation of enteric neuroblasts
.
Dev. Dyn.
228
,
1
10
42
Pitera
,
J.E.
,
Smith
,
V.V.
,
Thorogood
,
P.
and
Milla
,
P.J.
(
1999
)
Coordinated expression of 3’ hox genes during murine embryonal gut development: an enteric Hox code
.
Gastroenterology
117
,
1339
1351
43
Kam
,
M.K.M.
and
Lui
,
V.C.H.
(
2015
)
Roles of Hoxb5 in the development of vagal and trunk neural crest cells
.
Dev. Growth Differ.
57
,
158
168
44
Chan
,
K.K.
,
Chen
,
Y.S.
,
Yau
,
T.O.
,
Fu
,
M.
,
Lui
,
V.C.
,
Tam
,
P.K.
et al (
2005
)
Hoxb3 vagal neural crest-specific enhancer element for controlling enteric nervous system development
.
Dev. Dyn.
233
,
473
483
45
George
,
R.M.
,
Maldonado-Velez
,
G.
and
Firulli
,
A.B.
(
2020
)
The heart of the neural crest: cardiac neural crest cells in development and regeneration
.
Development
147
,
dev188706
46
Vega-Lopez
,
G.A.
,
Cerrizuela
,
S.
and
Aybar
,
M.J.
(
2017
)
Trunk neural crest cells: formation, migration and beyond
.
Int. J. Dev. Biol.
61
,
5
15
47
Nguyen
,
M.T.
,
Zhu
,
J.
,
Nakamura
,
E.
,
Bao
,
X.
and
Mackem
,
S.
(
2009
)
Tamoxifen-dependent, inducible Hoxb6CreERT recombinase function in lateral plate and limb mesoderm, CNS isthmic organizer, posterior trunk neural crest, hindgut, and tailbud
.
Dev. Dyn.
238
,
467
474
48
Ishikawa
,
S.
and
Ito
,
K.
(
2009
)
Plasticity and regulatory mechanisms of Hox gene expression in mouse neural crest cells
.
Cell Tissue Res.
337
,
381
391
49
Huber
,
L.
,
Ferdin
,
M.
,
Holzmann
,
J.
,
Stubbusch
,
J.
and
Rohrer
,
H.
(
2012
)
Hoxb8 in noradrenergic specification and differentiation of the autonomic nervous system
.
Dev. Biol.
363
,
219
233
50
Philippidou
,
P.
and
Dasen Jeremy
,
S.
(
2013
)
Hox genes: choreographers in neural development, architects of circuit organization
.
Neuron
80
,
12
34
51
Serbedzija
,
G.N.
,
Burgan
,
S.
,
Fraser
,
S.E.
and
Bronner-Fraser
,
M.
(
1991
)
Vital dye labelling demonstrates a sacral neural crest contribution to the enteric nervous system of chick and mouse embryos
.
Development
111
,
857
866
52
Heanue
,
T.A.
,
Shepherd
,
I.T.
and
Burns
,
A.J.
(
2016
)
Enteric nervous system development in avian and zebrafish models
.
Dev. Biol.
417
,
129
138
53
Le Douarin
,
N.M.
and
Teillet
,
M.A.
(
1973
)
The migration of neural crest cells to the wall of the digestive tract in avian embryo
.
J. Embryol. Exp. Morphol.
30
,
31
48
54
Kapur
,
R.P.
(
2000
)
Colonization of the murine hindgut by sacral crest-derived neural precursors: experimental support for an evolutionarily conserved model
.
Dev. Biol.
227
,
146
155
55
Anderson
,
R.
,
Stewart
,
A.
and
Young
,
H.
(
2006
)
Phenotypes of neural-crest-derived cells in vagal and sacral pathways
.
Cell Tissue Res.
323
,
11
25
56
Wang
,
X.
,
Chan
,
A.K.
,
Sham
,
M.H.
,
Burns
,
A.J.
and
Chan
,
W.Y.
(
2011
)
Analysis of the sacral neural crest cell contribution to the hindgut enteric nervous system in the mouse embryo
.
Gastroenterology
141
,
992
1002.e6
57
Wiese
,
C.B.
,
Deal
,
K.K.
,
Ireland
,
S.J.
,
Cantrell
,
V.A.
and
Southard-Smith
,
E.M.
(
2017
)
Migration pathways of sacral neural crest during development of lower urogenital tract innervation
.
Dev. Biol.
429
,
356
369
58
Javali
,
A.
,
Misra
,
A.
,
Leonavicius
,
K.
,
Acharyya
,
D.
,
Vyas
,
B.
and
Sambasivan
,
R.
(
2017
)
Co-expression of Tbx6 and Sox2 identifies a novel transient neuromesoderm progenitor cell state
.
Development
144
,
4522
4529
59
Wymeersch
,
F.J.
,
Huang
,
Y.
,
Blin
,
G.
,
Cambray
,
N.
,
Wilkie
,
R.
,
Wong
,
F.C.
et al (
2016
)
Position-dependent plasticity of distinct progenitor types in the primitive streak
.
eLife
5
,
e10042
60
McGrew
,
M.J.
,
Sherman
,
A.
,
Lillico
,
S.G.
,
Ellard
,
F.M.
,
Radcliffe
,
P.A.
,
Gilhooley
,
H.J.
et al (
2008
)
Localised axial progenitor cell populations in the avian tail bud are not committed to a posterior Hox identity
.
Development
135
,
2289
2299
61
Rodrigo Albors
,
A.
,
Halley
,
P.A.
and
Storey
,
K.G.
(
2018
)
Lineage tracing of axial progenitors using Nkx1-2CreER(T2) mice defines their trunk and tail contributions
.
Development
145
,
dev164319
62
Catala
,
M.
,
Teillet
,
M.-A.
and
Le Douarin
,
N.M.
(
1995
)
Organization and development of the tail bud analyzed with the quail-chick chimaera system
.
Mech. Dev.
51
,
51
65
63
Anderson
,
M.J.
,
Naiche
,
L.A.
,
Wilson
,
C.P.
,
Elder
,
C.
,
Swing
,
D.A.
and
Lewandoski
,
M.
(
2013
)
TCreERT2, a transgenic mouse line for temporal control of Cre-mediated recombination in lineages emerging from the primitive streak or tail bud
.
PLoS ONE
8
,
e62479
64
Feller
,
J.
,
Schneider
,
A.
,
Schuster-Gossler
,
K.
and
Gossler
,
A.
(
2008
)
Noncyclic notch activity in the presomitic mesoderm demonstrates uncoupling of somite compartmentalization and boundary formation
.
Genes Dev.
22
,
2166
2171
65
Garriock
,
R.J.
,
Chalamalasetty
,
R.B.
,
Kennedy
,
M.W.
,
Canizales
,
L.C.
,
Lewandoski
,
M.
and
Yamaguchi
,
T.P.
(
2015
)
Lineage tracing of neuromesodermal progenitors reveals novel Wnt-dependent roles in trunk progenitor cell maintenance and differentiation
.
Development
142
,
1628
1638
66
Perantoni
,
A.O.
,
Timofeeva
,
O.
,
Naillat
,
F.
,
Richman
,
C.
,
Pajni-Underwood
,
S.
,
Wilson
,
C.
et al (
2005
)
Inactivation of FGF8 in early mesoderm reveals an essential role in kidney development
.
Development
132
,
3859
3871
67
Turner
,
D.A.
,
Hayward
,
P.C.
,
Baillie-Johnson
,
P.
,
Rue
,
P.
,
Broome
,
R.
,
Faunes
,
F.
et al (
2014
)
Wnt/beta-catenin and FGF signalling direct the specification and maintenance of a neuromesodermal axial progenitor in ensembles of mouse embryonic stem cells
.
Development
141
,
4243
4253
68
Zhao
,
T.
,
Zhou
,
X.
,
Szabó
,
N.
,
Leitges
,
M.
and
Alvarez-Bolado
,
G.
(
2007
)
Foxb1-driven Cre expression in somites and the neuroepithelium of diencephalon, brainstem, and spinal cord
.
Genesis
45
,
781
787
69
Lukoseviciute
,
M.
,
Mayes
,
S.
and
Sauka-Spengler
,
T.
(
2021
)
Neuromesodermal progenitor origin of trunk neural crest in vivo
.
bioRxiv
70
Chambers
,
S.M.
,
Fasano
,
C.A.
,
Papapetrou
,
E.P.
,
Tomishima
,
M.
,
Sadelain
,
M.
and
Studer
,
L.
(
2009
)
Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling
.
Nat. Biotechnol.
27
,
275
280
71
Lee
,
G.
,
Chambers
,
S.M.
,
Tomishima
,
M.J.
and
Studer
,
L.
(
2010
)
Derivation of neural crest cells from human pluripotent stem cells
.
Nat. Protoc.
5
,
688
701
72
Lee
,
G.
,
Kim
,
H.
,
Elkabetz
,
Y.
,
Al Shamy
,
G.
,
Panagiotakos
,
G.
,
Barberi
,
T.
et al (
2007
)
Isolation and directed differentiation of neural crest stem cells derived from human embryonic stem cells
.
Nat. Biotechnol.
25
,
1468
1475
73
Chambers
,
S.M.
,
Qi
,
Y.
,
Mica
,
Y.
,
Lee
,
G.
,
Zhang
,
X.J.
,
Niu
,
L.
et al (
2012
)
Combined small-molecule inhibition accelerates developmental timing and converts human pluripotent stem cells into nociceptors
.
Nat. Biotechnol.
30
,
715
720
74
Mica
,
Y.
,
Lee
,
G.
,
Chambers
,
S.M.
,
Tomishima
,
M.J.
and
Studer
,
L.
(
2013
)
Modeling neural crest induction, melanocyte specification, and disease-related pigmentation defects in hESCs and patient-specific iPSCs
.
Cell Rep.
3
,
1140
1152
75
Chambers
,
S.M.
,
Mica
,
Y.
,
Lee
,
G.
,
Studer
,
L.
and
Tomishima
,
M.J.
(
2016
)
Dual-SMAD inhibition/WNT activation-based methods to induce neural crest and derivatives from human pluripotent stem cells
.
Methods Mol. Biol.
1307
,
329
343
76
Leung
,
A.W.
,
Murdoch
,
B.
,
Salem
,
A.F.
,
Prasad
,
M.S.
,
Gomez
,
G.A.
and
Garcia-Castro
,
M.I.
(
2016
)
WNT/beta-catenin signaling mediates human neural crest induction via a pre-neural border intermediate
.
Development
143
,
398
410
77
Menendez
,
L.
,
Yatskievych
,
T.A.
,
Antin
,
P.B.
and
Dalton
,
S.
(
2011
)
Wnt signaling and a smad pathway blockade direct the differentiation of human pluripotent stem cells to multipotent neural crest cells
.
Proc. Natl Acad. Sci. U.S.A.
108
,
19240
5
78
Tchieu
,
J.
,
Zimmer
,
B.
,
Fattahi
,
F.
,
Amin
,
S.
,
Zeltner
,
N.
,
Chen
,
S.
et al (
2017
)
A modular platform for differentiation of human PSCs into All major ectodermal lineages
.
Cell Stem Cell
21
,
399
410.e7
79
Hackland
,
J.O.S.
,
Frith
,
T.J.R.
,
Thompson
,
O.
,
Marin Navarro
,
A.
,
Garcia-Castro
,
M.I.
,
Unger
,
C.
et al (
2017
)
Top-Down inhibition of BMP signaling enables robust induction of hPSCs into neural crest in fully defined, xeno-free conditions
.
Stem Cell Rep.
9
,
1043
1052
80
Hackland
,
J.
(
2019
)
Top-Down inhibition (TDi) and baseline activation (BLa): controlling signal transduction when endogenous cytokines are ruining your differentiation
.
Curr. Protoc. Stem Cell Biol.
51
,
e98
81
Saito-Diaz
,
K.
,
Street
,
J.R.
,
Ulrichs
,
H.
and
Zeltner
,
N.
(
2021
)
Derivation of peripheral nociceptive, mechanoreceptive, and proprioceptive sensory neurons from the same culture of human pluripotent stem cells
.
Stem Cell Rep.
16
,
446
457
82
Fattahi
,
F.
,
Steinbeck
,
J.A.
,
Kriks
,
S.
,
Tchieu
,
J.
,
Zimmer
,
B.
,
Kishinevsky
,
S.
et al (
2016
)
Deriving human ENS lineages for cell therapy and drug discovery in hirschsprung disease
.
Nature
531
,
105
109
83
Huang
,
M.
,
Miller
,
M.L.
,
McHenry
,
L.K.
,
Zheng
,
T.
,
Zhen
,
Q.
,
Ilkhanizadeh
,
S.
et al (
2016
)
Generating trunk neural crest from human pluripotent stem cells
.
Sci. Rep.
6
,
19727
84
Workman
,
M.J.
,
Mahe
,
M.M.
,
Trisno
,
S.
,
Poling
,
H.M.
,
Watson
,
C.L.
,
Sundaram
,
N.
et al (
2017
)
Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system
.
Nat. Med.
23
,
49
59
85
Frith
,
T.J.
,
Granata
,
I.
,
Wind
,
M.
,
Stout
,
E.
,
Thompson
,
O.
,
Neumann
,
K.
et al (
2018
)
Human axial progenitors generate trunk neural crest cells in vitro
.
eLife
7
,
e35786
86
Frith
,
T.J.R.
,
Gogolou
,
A.
,
Hackland
,
J.O.S.
,
Hewitt
,
Z.A.
,
Moore
,
H.D.
,
Barbaric
,
I.
et al (
2020
)
Retinoic acid accelerates the specification of enteric neural progenitors from In-Vitro-Derived neural crest
.
Stem Cell Rep.
15
,
557
565
87
Schlieve
,
C.R.
,
Fowler
,
K.L.
,
Thornton
,
M.
,
Huang
,
S.
,
Hajjali
,
I.
,
Hou
,
X.
et al (
2017
)
Neural crest cell implantation restores enteric nervous system function and alters the gastrointestinal transcriptome in human tissue-engineered small intestine
.
Stem Cell Rep.
9
,
883
896
88
Park
,
C.S.
,
Nguyen
,
L.P.
and
Yong
,
D.
(
2020
)
Development of colonic organoids containing enteric nerves or blood vessels from human embryonic stem cells
.
Cells
9
,
2209
89
Oh
,
Y.
,
Cho
,
G.-S.
,
Li
,
Z.
,
Hong
,
I.
,
Zhu
,
R.
,
Kim
,
M.-J.
et al (
2016
)
Functional coupling with cardiac muscle promotes maturation of hPSC-derived sympathetic neurons
.
Cell Stem Cell
19
,
95
106
90
Gouti
,
M.
,
Tsakiridis
,
A.
,
Wymeersch
,
F.J.
,
Huang
,
Y.
,
Kleinjung
,
J.
,
Wilson
,
V.
et al (
2014
)
In vitro generation of neuromesodermal progenitors reveals distinct roles for wnt signalling in the specification of spinal cord and paraxial mesoderm identity
.
PLoS Biol.
12
,
e1001937
91
Cooper
,
F.
,
Gentsch
,
G.E.
,
Mitter
,
R.
,
Bouissou
,
C.
,
Healy
,
L.
,
Rodriguez
,
A.H.
et al (
2021
)
Rostrocaudal patterning and neural crest differentiation of human pre-neural spinal cord progenitors in vitro
.
bioRxiv.
92
Mouilleau
,
V.
,
Vaslin
,
C.
,
Robert
,
R.
,
Gribaudo
,
S.
,
Nicolas
,
N.
,
Jarrige
,
M.
et al (
2021
)
Dynamic extrinsic pacing of the HOX clock in human axial progenitors controls motor neuron subtype specification
.
Development
148
,
dev194514
93
Lippmann
,
E.S.
,
Williams
,
C.E.
,
Ruhl
,
D.A.
,
Estevez-Silva
,
M.C.
,
Chapman
,
E.R.
,
Coon
,
J.J.
et al (
2015
)
Deterministic HOX patterning in human pluripotent stem cell-derived neuroectoderm
.
Stem Cell Rep.
4
,
632
644
94
Cunningham
,
T.J.
,
Colas
,
A.
and
Duester
,
G.
(
2016
)
Early molecular events during retinoic acid induced differentiation of neuromesodermal progenitors
.
Biol. Open
5
,
1821
1833
95
Tsakiridis
,
A.
,
Huang
,
Y.
,
Blin
,
G.
,
Skylaki
,
S.
,
Wymeersch
,
F.
,
Osorno
,
R.
et al (
2014
)
Distinct Wnt-driven primitive streak-like populations reflect in vivo lineage precursors
.
Development
141
,
1209
1221
96
Gouti
,
M.
,
Delile
,
J.
,
Stamataki
,
D.
,
Wymeersch
,
F.J.
,
Huang
,
Y.
,
Kleinjung
,
J.
et al (
2017
)
A gene regulatory network balances neural and mesoderm specification during vertebrate trunk development
.
Dev. Cell
41
,
243
61 e7
97
Verrier
,
L.
,
Davidson
,
L.
,
Gierlinski
,
M.
,
Dady
,
A.
and
Storey
,
K.G.
(
2018
)
Neural differentiation, selection and transcriptomic profiling of human neuromesodermal progenitor-like cells in vitro
.
Development
145
,
dev166215
98
Edri
,
S.
,
Hayward
,
P.
,
Baillie-Johnson
,
P.
,
Steventon
,
B.J.
and
Martinez Arias
,
A.
(
2019
)
An epiblast stem cell-derived multipotent progenitor population for axial extension
.
Development
146
,
dev168187
99
Diaz-Cuadros
,
M.
,
Wagner
,
D.E.
,
Budjan
,
C.
,
Hubaud
,
A.
,
Tarazona
,
O.A.
,
Donelly
,
S.
et al (
2020
)
In vitro characterization of the human segmentation clock
.
Nature
580
,
113
118
100
Wind
,
M.
,
Gogolou
,
A.
,
Manipur
,
I.
,
Granata
,
I.
,
Butler
,
L.
,
Andrews
,
P.W.
et al (
2021
)
Defining the signalling determinants of a posterior ventral spinal cord identity in human neuromesodermal progenitor derivatives
.
Development
148
,
dev194415
101
Gomez
,
G.A.
,
Prasad
,
M.S.
,
Wong
,
M.
,
Charney
,
R.M.
,
Shelar
,
P.B.
,
Sandhu
,
N.
et al (
2019
)
WNT/beta-catenin modulates the axial identity of embryonic stem cell-derived human neural crest
.
Development
146
,
dev175604
102
Denham
,
M.
,
Hasegawa
,
K.
,
Menheniott
,
T.
,
Rollo
,
B.
,
Zhang
,
D.
,
Hough
,
S.
et al (
2015
)
Multipotent caudal neural progenitors derived from human pluripotent stem cells that give rise to lineages of the central and peripheral nervous system
.
STEM CELLS
33
,
1759
1770
103
Frith
,
T.J.R.
and
Tsakiridis
,
A.
(
2019
)
Efficient generation of trunk neural crest and sympathetic neurons from human pluripotent stem cells Via a neuromesodermal axial progenitor intermediate
.
Curr. Protoc. Stem Cell Biol.
49
,
e81
104
Abu-Bonsrah
,
K.D.
,
Zhang
,
D.
,
Bjorksten
,
A.R.
,
Dottori
,
M.
and
Newgreen
,
D.F.
(
2018
)
Generation of adrenal chromaffin-like cells from human pluripotent stem cells
.
Stem Cell Rep.
10
,
134
150
105
Hackland
,
J.O.S.
,
Shelar
,
P.B.
,
Sandhu
,
N.
,
Prasad
,
M.S.
,
Charney
,
R.M.
,
Gomez
,
G.A.
et al (
2019
)
FGF modulates the axial identity of trunk hPSC-derived neural crest but Not the cranial-Trunk decision
.
Stem Cell Rep.
12
,
920
933
106
Kirino
,
K.
,
Nakahata
,
T.
,
Taguchi
,
T.
and
Saito
,
M.K.
(
2018
)
Efficient derivation of sympathetic neurons from human pluripotent stem cells with a defined condition
.
Sci. Rep.
8
,
12865
107
McGonnell
,
I.M.
and
Graham
,
A.
(
2002
)
Trunk neural crest Has skeletogenic potential
.
Curr. Biol.
12
,
767
771
108
Coelho-Aguiar
,
J.M.
,
Le Douarin
,
N.M.
and
Dupin
,
E.
(
2013
)
Environmental factors unveil dormant developmental capacities in multipotent progenitors of the trunk neural crest
.
Dev. Biol.
384
,
13
25
109
Sanchez-Ferras
,
O.
,
Bernas
,
G.
,
Farnos
,
O.
,
Touré
,
A.M.
,
Souchkova
,
O.
and
Pilon
,
N.
(
2016
)
A direct role for murine Cdx proteins in the trunk neural crest gene regulatory network
.
Development
143
,
1363
1374
110
Sanchez-Ferras
,
O.
,
Coutaud
,
B.
,
Djavanbakht Samani
,
T.
,
Tremblay
,
I.
,
Souchkova
,
O.
and
Pilon
,
N.
(
2012
)
Caudal-related homeobox (Cdx) protein-dependent integration of canonical Wnt signaling on paired-box 3 (Pax3) neural crest enhancer
.
J. Biol. Chem.
287
,
16623
16635
111
Neijts
,
R.
,
Amin
,
S.
,
van Rooijen
,
C.
and
Deschamps
,
J.
(
2017
)
Cdx is crucial for the timing mechanism driving colinear Hox activation and defines a trunk segment in the Hox cluster topology
.
Dev. Biol.
422
,
146
154
112
Faustino Martins
,
J.M.
,
Fischer
,
C.
,
Urzi
,
A.
,
Vidal
,
R.
,
Kunz
,
S.
,
Ruffault
,
P.L.
et al (
2020
)
Self-Organizing 3D human trunk neuromuscular organoids
.
Cell Stem Cell
26
,
172
86 e6
113
Pereira
,
J.D.
,
DuBreuil
,
D.M.
,
Devlin
,
A.-C.
,
Held
,
A.
,
Sapir
,
Y.
,
Berezovski
,
E.
et al (
2021
)
Human sensorimotor organoids derived from healthy and amyotrophic lateral sclerosis stem cells form neuromuscular junctions
.
Nat. Commun.
12
,
4744
114
Arbour
,
D.
,
Vande Velde
,
C.
and
Robitaille
,
R.
(
2017
)
New perspectives on amyotrophic lateral sclerosis: the role of glial cells at the neuromuscular junction
.
J. Physiol.
595
,
647
661
115
Beccari
,
L.
,
Moris
,
N.
,
Girgin
,
M.
,
Turner
,
D.A.
,
Baillie-Johnson
,
P.
,
Cossy
,
A.C.
et al (
2018
)
Multi-axial self-organization properties of mouse embryonic stem cells into gastruloids
.
Nature
562
,
272
276
116
van den Brink
,
S.C.
,
Alemany
,
A.
,
van Batenburg
,
V.
,
Moris
,
N.
,
Blotenburg
,
M.
,
Vivié
,
J.
et al (
2020
)
Single-cell and spatial transcriptomics reveal somitogenesis in gastruloids
.
Nature
582
,
405
409
117
van den Brink
,
S.C.
,
Baillie-Johnson
,
P.
,
Balayo
,
T.
,
Hadjantonakis
,
A.K.
,
Nowotschin
,
S.
,
Turner
,
D.A.
et al (
2014
)
Symmetry breaking, germ layer specification and axial organisation in aggregates of mouse embryonic stem cells
.
Development
141
,
4231
4242
118
Moris
,
N.
,
Alev
,
C.
,
Pera
,
M.
and
Martinez Arias
,
A.
(
2021
)
Biomedical and societal impacts of in vitro embryo models of mammalian development
.
Stem Cell Rep.
16
,
1021
1030
119
Veenvliet
,
J.V.
,
Bolondi
,
A.
,
Kretzmer
,
H.
,
Haut
,
L.
,
Scholze-Wittler
,
M.
,
Schifferl
,
D.
et al (
2020
)
Mouse embryonic stem cells self-organize into trunk-like structures with neural tube and somites
.
Science
370
,
eaba4937
120
Rossi
,
G.
,
Broguiere
,
N.
,
Miyamoto
,
M.
,
Boni
,
A.
,
Guiet
,
R.
,
Girgin
,
M.
et al (
2021
)
Capturing cardiogenesis in gastruloids
.
Cell Stem Cell
28
,
230
40.e6
121
Olmsted
,
Z.T.
and
Paluh
,
J.L.
(
2021
)
Co-development of central and peripheral neurons with trunk mesendoderm in human elongating multi-lineage organized gastruloids
.
Nat. Commun.
12
,
3020
122
Aires
,
R.
,
de Lemos
,
L.
,
Novoa
,
A.
,
Jurberg
,
A.D.
,
Mascrez
,
B.
,
Duboule
,
D.
et al (
2019
)
Tail Bud progenitor activity relies on a network comprising Gdf11, Lin28, and Hox13 genes
.
Dev. Cell
48
,
383
95 e8
123
Robinton
,
D.A.
,
Chal
,
J.
,
Lummertz da Rocha
,
E.
,
Han
,
A.
,
Yermalovich
,
A.V.
,
Oginuma
,
M.
et al (
2019
)
The Lin28/let-7 pathway regulates the mammalian caudal body axis elongation program
.
Dev. Cell
48
,
396
405.e3
124
Jurberg
,
A.D.
,
Aires
,
R.
,
Varela-Lasheras
,
I.
,
Novoa
,
A.
and
Mallo
,
M.
(
2013
)
Switching axial progenitors from producing trunk to tail tissues in vertebrate embryos
.
Dev. Cell
25
,
451
462
125
Xu
,
W.
,
Wang
,
Y.
,
Liu
,
E.
,
Sun
,
Y.
,
Luo
,
Z.
,
Xu
,
Z.
et al (
2013
)
Human iPSC-derived neural crest stem cells promote tendon repair in a rat patellar tendon window defect model
.
Tissue Eng. Part A
19
,
2439
2451
126
Gomez
,
G.A.
,
Prasad
,
M.S.
,
Sandhu
,
N.
,
Shelar
,
P.B.
,
Leung
,
A.W.
and
García-Castro
,
M.I.
(
2019
)
Human neural crest induction by temporal modulation of WNT activation
.
Dev. Biol.
449
,
99
106
127
Gogolou
,
A.
,
Souilhol
,
C.
,
Granata
,
I.
,
Wymeersch
,
F.J.
,
Manipur
,
I.
,
Wind
,
M.
et al (
2021
)
Early anteroposterior regionalisation of human neural crest is shaped by a pro-mesodermal factor
.
bioRxiv
128
Vickaryous
,
M.K.
and
Hall
,
B.K.
(
2006
)
Human cell type diversity, evolution, development, and classification with special reference to cells derived from the neural crest
.
Biol. Rev. Camb. Philos. Soc.
81
,
425
455
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).