Anda di halaman 1dari 12

Developmental Biology 366 (2012) 2233

Contents lists available at SciVerse ScienceDirect

Developmental Biology
journal homepage: www.elsevier.com/developmentalbiology

Review

Neural crest induction at the neural plate border in vertebrates


Ccile Milet, Anne H. Monsoro-Burq
Institut Curie; INSERM U1021; CNRS, UMR 3347, F-91405 Orsay, France
Universit Paris Sud-11, F-91405 Orsay, France

a r t i c l e

i n f o

Article history:
Received for publication 13 January 2012
Accepted 13 January 2012
Available online 25 January 2012
Keywords:
Neural crest
Neural plate border
AP2a
Pax3
Zic1
Hairy2
Msx1
Xenopus embryo
Vertebrate gene regulatory network

a b s t r a c t
The neural crest is a transient and multipotent cell population arising at the edge of the neural plate in
vertebrates. Recent ndings highlight that neural crest patterning is initiated during gastrulation, i.e. earlier
than classically described, in a progenitor domain named the neural border. This chapter reviews the dynamic
and complex molecular interactions underlying neural border formation and neural crest emergence.
2012 Published by Elsevier Inc.

Introduction
The neural crest, a key feature of vertebrate embryos, is a population
of highly multipotent and migratory progenitors. Neural crest cells give
rise to the many types of neurons and glia of the enteric, sensory and autonomous peripheral nervous system. They also differentiate into a variety of pigment cells such as melanocytes or melanophores, iridophores
and xanthophores. Moreover, they form chromafn cells, participate in
heart outow tract formation in amniotes, and are major progenitors of
craniofacial mesenchyme and skeleton (Le Douarin and Kalcheim,
1999). Neurocristopathies are a large group of birth defects resulting
from alterations in the complex succession of events required for the
induction, proliferation, migration and nal differentiation of neural
crest derivatives.
The neural crest originates from the border between the non-neural
ectoderm and the neurectoderm
Neural crest (NC) cells have been dened morphologically, as they
emerge from the edge of the neural folds (His, 1868; Le Douarin and
Kalcheim, 1999). At trunk levels, neural crest cells exit from the closing
dorsal neural tube via a stereotypical epithelium-to-mesenchyme transition (see Theveneau and Mayor, 2012this issue) (Ahlstrom and Erickson,
2009; Cheung et al., 2005; Serbedzija et al., 1989). At head levels, the
Corresponding author at: Institut Curie, UMR 3347, Batiment 110, Centre Universitaire,
F-91405 Orsay Cedex, France.
E-mail address: anne-helene.monsoro-burq@curie.fr (A.H. Monsoro-Burq).
0012-1606/$ see front matter 2012 Published by Elsevier Inc.
doi:10.1016/j.ydbio.2012.01.013

cranial neural crest forms before neural folds fusion, resulting in two bilateral masses of premigratory neural crest cells lying on both sides of the
neural plate. Soon thereafter, cranial neural crest cells initiate migration
toward the craniofacial areas, either as individuals or via collective migration recently described by live imaging (Theveneau et al., 2007, 2010; for
further details on migration events, see Theveneau and Mayor, 2012this
issue).
At both head and trunk locations, premigratory neural crest progenitors can be dened prior to any morphological change, by their specic
expression of characteristic molecular markers that distinguish them
from the adjacent ectoderm and neurectoderm (also see Labonne and
Sauka-Spengler, this issue on neural crest specication). Many of
these molecules are involved in the cellular process underlying the
epithelial to mesenchymal transition. Among the earliest and most
specic neural crest markers, Snai1, Snai2 (previously called Snail
and Slug) and SoxE family members are common to all vertebrate species examined (see references in Table 1).
Using both morphological and molecular analyses to identify neural
crest derivatives, lineage tracing was performed at neural plate and
neural tube stages, to follow the progeny of single or small groups of
cells from neurulation until organogenesis. Most classical analyses
were performed in the closing trunk neural tube and demonstrate the
common lineage between dorsal neural tube and neural crest, in avians,
amphibians and mammals during neurulation (Ahlstrom and Erickson,
2009; Bronner-Fraser and Fraser, 1988; Collazo et al., 1993; Serbedzija
et al., 1994). However, when such labeling is performed on small groups
of cells at the end of neurulation in the trunk of avian embryos, i.e. after
neural tube closure and just prior to cell emigration, the neural crest

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

lineage seems segregated from the roof plate lineage (Krispin et al.,
2010). Earlier mapping around the neural plate during avian gastrulation provides evidence for an area posterior and lateral to the neural

23

plate where progenitor for epidermis, placodes, neural crest and dorsal
neural tube are still intermingled (Basch et al., 2006; Fernandez-Garre
et al., 2002; Streit, 2002). In the early frog gastrula, neural crest

Table 1
List of the main neural border and neural crest markers in various vertebrates.
Each ectoderm subdomain (i.e. the non-neural ectoderm, the placodes, the neural border and neural crest, the neural plate and neural tube) can be characterized by the co-expression of
a group of markers. Although these markers are mostly conserved in vertebrates, some species-specic changes have been recorded and indicate the need for a careful characterization of
each model organism. The white boxes indicate genes without a described homolog in a given species; or that have not been implicated in NC development in the given
species. Reference links: Aoki et al., 2003; Aruga et al., 2002; Avilion et al., 2003; Barrionuevo et al., 2008; Bell et al., 2000; Bellefroid et al., 1998; Bellmeyer et al., 2003; Bulfone et al., 1993;
Byrd and Meyers, 2005; Cheng et al., 2000; Cheung and Briscoe, 2003; Davidson and Hill, 1991; Dottori et al., 2001; Dy et al., 2008; Essex et al., 1993; Feledy et al., 1999; Hill et al., 1989;
Honore et al., 2003; Inoue et al., 2004; Jen et al., 1996; Jiang et al., 1998; Jostes et al., 1990; Kee and Bronner-Fraser, 2001a; Kee and Bronner-Fraser, 2001b; Kos et al., 2001; Kuhlbrodt et al.,
1998; Liu and Harland, 2003; Mansouri et al., 1996; Marin and Nieto, 2004; Martin and Harland, 2001; Martinsen and Bronner-Fraser, 1998; Matsunaga et al., 2001; Mayor et al., 1995;
Mitchell et al., 1991; Myat et al., 1996; Nagai et al., 1997; Nakata et al., 1997; Nakata et al., 1998; Nakata et al., 2000; Nieto et al., 1994; Niss and Leutz, 1998; O'Donnell et al., 2006; Pera and
Kessel, 1999; Perez-Alcala et al., 2004; Robinson and Mahon, 1994; Saint-Germain et al., 2004; Salzberg et al., 1999; Sanchez-Guardado et al., 2011; Sasai et al., 2001; Sauka-Spengler et al.,
2007; Sawai et al., 1990; Schepers et al., 2000; Shen et al., 1997 Smith et al., 1992; Su et al., 1991; Suzuki et al., 1991; Suzuki et al., 1997; Thomas et al., 2008; Tour et al., 2001;
Uwanogho et al., 1995; Warner et al., 2003; Williams et al., 1995; Yang et al., 1998. Abbreviations: NC: neural crest; pNC: premigratory neural crest; mNC: migrating neural crest;
NP: neural plate; NT: neural tube; NB: neural border; HH Hamburger and Hamilton stage. Xenopus stages refer to Nieuwkoop and Faber stage table.

A- NEURAL BORDER SPECIFIERS (AND SOME ADJACENT NEURAL AND ECTODERM MARKERS)
Xenopus

Chick

Mouse

Lamprey

Human

Ectoderm lateral and anterior


to the neural plate in the
subcephalic pocket at HH st. 8

Neural folds corresponding to


pNC location atE 8.5

Ectoderm of early gastrula


(E3), ectoderm and NP border
(E4)

mNC in rostral sections,


branchial arches and cranial
mesenchyme at CS12

Luo et al., 2003

Shen et al., 1997

Mitchell et al.,1991

Sauka-Spengler et al., 2007;


Nikitina et al., 2008

Thomas et al., 2008

Neural fold at st12.5

Hindbrain at HH st. 10

Neural plate/ectoderm

Delta A

Not described in human early


neural development

Glavic et al., 2004

Myat et al., 1996

Williams et al., 1995

Sauka-Spengler et al., 2007

pNC in early neurula

ap2

Delta/notch

dlx3

dlx5

Anterior region of the


prospective NP at st12

pNC at HH st. 10

Branchial arches

DlxA, B, and C: Ventro-lateral


ectoderm, excluded from NP
(E4)

Feledy et al., 1999

Pera and Kessel, 1999

Robinson and Mahon, 1994

Sauka-Spengler et al., 2007

Not described in chick early


neural development

Lateral edges of the neural


plate at E7,75

Central part of the epiblast,


excluding the primitive streak
and Hensen's node at HH st. 4

Gastrula (E6.5), neuroectoderm and underlying


mesoderm (E7.5-E8.5), mNC
(pharyngeal arches)

Niss and Leutz, 1998

Bulfone et al., 1993, Byrd and


Meyers, 2005

No described homologue in
chick

No described homologue in
mouse

Not described in chick early


neural development

Ectoderm at the end of


gastrulation (E7.5)

No chick homologue

Ectoderm except in prospective


NP at mid to late gastrula
Luo et al., 2001
NB and lateral ectoderm at st 12

gbx2
Tour et al., 2001; Li et al. 2009

Hairy2

Neural fold at st12.5-13

iro3

meis3
Salzberg et al., 1999

msx1

msx2

pax7

serrate

sox2

Not described in human early


neural development

Hairy1/2A and B

Not described in human early


neural development

Not described in lamprey early


neural development

Not described in human early


neural development

Not described in mouse early


neural development

MeisA and B

Not described in human early


neural development

Bellefroid et al., 1998

Sanchez-Guardado et al., 2011

Sauka-Spengler et al., 2007

confined dorsally to the lateral


edge of the neural plate at
st14

NC

NC at E9.5

MsxA: Ectoderm of gastrula


(E3.5), ectoderm and NP
border (E4)

Suzuki et al., 1997

Suzuki et al., 1991

Hill et al., 1989

Sauka -Spengler et al., 2007;


Nikitina et al., 2008

dorsal mesoderm in gastrula

not described in chick early


neural development

tip of the neural folds at E8,


cephalic NC

Su et al., 1991

pax3

No described homologue in
lamprey

Sauka-Spengler et al., 2007

Bellefroid et al., 1998


NP at early mid neurula

Not described in human early


neural development

Yang et al., 1998

Glavic et al., 2004


Prospective NP in gastrula

Not described in human early


neural development

NT emigrating cells in culture,


late C10 to late C12
Thomas et al., 2008
not described in human early
neural development

Davidson and Hill, 1991

Lateral domains of the


prospective NP at st11.5

NT at HH st. 10

Dorsal part of the


neuroepithelium at E8.5

Pax3/7: Ectoderm of early


gastrula (E3), NP border (E4)

NT emigrating cells in culture,


late C10 to late C12, pNC of
dorsal NT at C12

Bang et al., 1999

Matsunaga et al., 2001

Goulding et al., 1991

Sauka-Spengler et al., 2007;


Nikitina et al., 2008

Thomas et al., 2008, Betters


et al., 2010

Brain (st14), paraxial


mesoderm (st18)

Bilaterally symmetric oblique


bands lateral to Hensens node
in epiblast at HH st. 4+

Alar plate of the neural tube


(E8.5)

Maczkowiak et al., 2010

Basch et al., 2006

Neural fold at st12.5-13

Neural fold HH st. 10

Glavic et al., 2004

Myat et al., 1996

End of gastrulation :
prospective neurectoderm.

Neural epithelium at HH st. 10

Mizuseki et al., 1998

Uwanogho et al., 1995

Jostes et al., 1990, Mansouri


et al., 1996
not described in mouse early
neural development

Dorsal NT at CS12
Thomas et al., 2008
No described homologue in
lamprey

Not described in human early


neural development

Anterior neurectderm at E77.5, neural tube and branchial


arches at E9.5

SoxB1: NP border (E4)

NT emigrating cells in culture,


late C10 to late C12

Avilion et al., 2003

Sauka-Spengler et al., 2007

Thomas et al., 2008

24

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

Table 1 (continued)

B- EARLY NEURAL CREST MARKERS


Xenopus

zic1

zic2

zic3

id4

snail1

snail2

sox5

sox 8

sox 9

sox 10

zic 5

Lamprey
ZicA: Ectoderm of early
gastrula (E3), NP (E4)

Mizuseki et al., 1998, Nakata


et al., 1998

Aruga et al., 2002, Warner et


al., 2003

Nagai et al., 1997

Sauka-Spengler et al., 2007;


Nikitina et al., 2008

Prospective NP in early
gastrula

NT HH st. 13

Embryonic mesoderm and


ectodermal cells in gastrula
(E7), neurectoderm (E7.25),
dorsal NT and dorsal
mesoderm derivatives (E9.5)

Nakata et al., 1998

Warner et al., 2003

Nagai et al., 1997

Prospective NP in gastrula

NT HH st. 13, dorsal


epithelium st23

Embryonic mesoderm and


ectodermal cells in gastrula
(E7), dorsal NT and dorsal
mesoderm derivatives (E9.5)

Nakata et al., 1997

Warner et al., 2003

Nagai et al., 1997

pNC at st14 (early neurula)

N-Myc: dorsal NT and NC


derivatives at E3.5

Not described in mouse NC

Bellmeyer et, al., 2003

Sawai et al., 1990

Presumptive NC region in late


gastrula

pNC and mNC at HH st. 6 to 10

Human
Not described in human early
neural development

Not described in human early


neural development

Not described in human early


neural development

N-myc: Ectoderm of early


gastrula (E3), ectoderm
including the NP (E4)

Not described in human NC

Sauka-Spengler et al., 2007;


Nikitina et al., 2008
pNC and mNC at E9.5-E10.5

FoxD-A: pNC, mNC at E6 (NT


formation)

NT emigrating cells in culture,


late C10 to late C12

Sasai et al., 2001

Kos et al., 2001

Dottori et al., 2001

Sauka-Spengler et al., 2007

Thomas et al., 2008

mNC

Cranial neural folds and mNC


in 5 somite stage embryo

mNC at E8.5

Id: Ectoderm of early gastrula


(E3), ectoderm and NP border
(E4)

Not described in human NC

Liu and Harland, 2003

Martinsen and Bronner-Fraser,


1998

Jen et al., 1996

Sauka-Spengler et al., 2007;


Nikitina et al., 2008

Migrating NC

Border of neural plate at HH


st. 5-. Dorsal NT at HH st 8.
Olfactory, lens and otic
placodes at stage 12

mNC at E8.5

Liu and Harland, 2003

Kee et al., 2001b

Jen et al., 1996

Ectoderm at the gastrula


stages

Epiblast ectoderm at HH st. 5.Dorsal NT at HH st. 10. mNC


at HH st. 12

Not described in mouse NC

Liu and Harland,2003

Kee et al., 2001a

Gastrula st 11,5

mNC at HH st. 21

Cephalic NC at E9.5

SnailA: Open NP and border at


E4

Essex et al., 1993

Marin and Nieto, 2004

Smith et al., 1992

Sauka-Spengler et al., 2007

Gastrula st 12,5

pNC, mesoderm (5 somite


stage)

Not in pNC, experssed in mNC

No described homologue in
lamprey

NT emigrating cells in culture,


late C10 to late C12

Not described in lamprey NC

Not described in human NC

Not described in human NC

id2

id3

Mouse

dorsal NT HH st. 10, NC HH


st. 23

c-myc

foxd3

Chick

Embryonic mesoderm in
gastrula (E7), neurectoderm
(E7.25), dorsal NT and dorsal
mesoderm derivatives (E9.5)

End of gastrulation :
prospective neurectoderm.
NP, NB

Not described in human NC

Not described in human NC

Mayor et al., 1995

Nieto et al., 1994

Jiang et al., 1998

migrating NC

Neural folds HH st. 7

NC

Martin and Harland, 2001

Perez-Alcala et al., 2004

Dy et al., 2008

Gastrula st 11,5

Dorsal NT at HH st. 11-13

mNC (branchial arches, dorsal


root ganglia) at E10

SoxE1, 2 and 3: pNC, mNC at


E6 (NT formation)

O'Donnell et al., 2006

Bell and Briscoe, 2000

Schepers et al., 2000

Sauka-Spengler et al., 2007

Gastrula st 12

Prospective NC at HH st. 10-12

Not described in human NC

Thomas et al., 2008

NT emigrating cells in culture


late C10 to late C12; pNC of
dorsal NT, branchial arches
and cranial mesenchyme,
mNC in rostral sections at CS12

NC at 5-6 somites stage

Saint-Germain et al., 2004

Cheung and Briscoe, 2003

Barrionuevo et al., 2008

Thomas et al., 2008, Betters


et al., 2010

Mid-neurula st 14

Prospective NC from HH st.7-8


to NC derivatives HH st. 26

pNC at E8.5

pNC of dorsal NT, mNC in


rostral sections at CS12

Aoki et al., 2003, Honore et


al., 2003

Cheng et al., 2000

Kuhlbrodt et al., 1998

pNC at late gastrula (stage


11.5-13)

No described homologue in
chick

Nakata et al., 2000

Betters et al., 2010

NP at E8, dorsal NT

ZicA: Ectoderm of early


gastrula (E3), NP (E4)

Inoue et al., 2004

Sauka-Spengler et al., 2007;


Nikitina et al., 2008

progenitors were mapped across an ectodermal area and the lateral part
of the neural plate, lying adjacent to the dorsallateral marginal zone
(Steventon et al., 2009). Similar fate maps were obtained for mouse
(Tam, 1989) and chick embryos (Ezin et al., 2009). This posterior and
lateral ectoderm area is the site of key patterning events that will dene
the neural crest lineage from these adjacent fates and constitutes the
domain named the neural border (or the neural plate border) (Fig. 1).

Not described in human NC

The neural border (NB) could thus be dened as the broad competence domain, established between neural and non-neural ectoderm,
within which subtle and sequential cell autonomous and non-cellautonomous molecular interactions will progressively dene the neural
crest fate from adjacent dorsal neural fates. This chapter will describe
the current state of knowledge regarding neural border patterning during the course of vertebrate gastrulation and neurulation, the process of

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

25

anterior
Ectoderm

high BMP

Preplacodal
field
low Wnt / low BMP
Otx2

Anterior NF
Neural border
Medial NP
Midline

Late gastrula

high Wnt / medium BMP


Gbx2
AP2 (border / ectoderm)
high Wnt / low BMP
Gbx, Cdx
posterior
anterior

Ectoderm
keratins

Preplacodal
field

eya1, six1

Anterior NF

anf1,
sox2

Neural border

pax3

Medial NP

pax3/snail2

Midline

Mid-neurula

posterior

Fig. 1. Schematic representation of the ectoderm at late gastrula and mid-neurula stages.The non-neural ectoderm (yellow) surrounds the neural border and the pre-placodal ectoderm (pink and green respectively), which themselves surround the neural plate (blue). At gastrulation stage, the main secreted inducers and key patterning transcription factors
expressed in these areas are indicated. At neurula stage, each domain can be dened by expression of a few characteristic markers (one is indicated, see text and Table 1 for additional information).

neural crest induction from neural border progenitors and link these
molecular events to the neural crest gene regulatory network.
A combination of secreted signals patterns the neural border
during gastrulation and neurulation
Molecular characterization of the neural border during gastrulation and
neurulation
The earliest detection of specic neural crest markers is reported
at mid-to-late gastrula stage in frog, and at 57 somites stage in
chick and mouse head neural folds stage (see references in Table 1).
This suggests that neural border patterning occurs prior to these
stages, namely during early- or mid-gastrulation. At similar stages,
neural patterning has also started (Puelles et al., 2005). In agreement
with its location at the prospective edge between neural and nonneural ectoderm, the neural border can be rst identied as the
overlap between two types of more broadly expressed markers:
genes also expressed in the lateral ectoderm (e.g. tfap2, hairy2,
msx1, msx2, dlx5) and genes also expressed in the more medial neural
plate (e.g. zic1, zic2, gbx2, meis3) (Fig. 1, see references in Table 1). More
specic neural border expression is described for pax3 and pax7 paralogs whose expression is initiated at early gastrula stage and can be
traced later to the dorsal neural fold and neural crest in both in frog
and chick embryos (Bang et al., 1999; Basch et al., 2006; de Croze
et al., 2011; Monsoro-Burq et al., 2005). Although mouse and human

pax3 and pax7 have been described in the dorsal neural tube, there is little information about their expression in the open neural plate (Betters
et al., 2010; Gerard et al., 1995; Goulding et al., 1991; Terzic and SaragaBabic, 1999). In summary, during gastrulation, the prospective neural
border encompasses a broad lateral domain (rather than a dened
line), comparable to the neural plate edge described at a similar stage
(Puelles et al., 2005; Rodriguez-Gallardo et al., 2005).
As the neural folds thicken and elevate, the neural border of vertebrate embryos is characterized by the enhanced co-expression of pax3/
7, msx1/2, zic1/2, tfap2a, iroquois1 (Xiro1) and hairy2, while it is outlined
by the expression of serrate and delta, as reported in chick, frog, zebrash,
lamprey, mouse and human (see references in Table 1). Simultaneously,
many of these NB genes are either down regulated or excluded from the
immediately adjacent ectoderm and the medial neural plate (e.g. msx1,
ap2a, hairy2, zic1, zic2). In contrast, the non-neural lateral ectoderm is
characterized by specic markers, such as dlx3 and ectodermal keratins
(Fuchs, 1995; Luo et al., 2001). Anteriorly, between the neural plate
(expressing pan-neural markers such as sox2 and sox3) (Iwafuchi-Doi
et al., 2011; Mizuseki et al., 1998; Rogers et al., 2008) and the nonneural ectoderm, the preplacodal markers (e.g. six1, eya1) mark the prospective placodal eld (Schlosser and Ahrens, 2004; Streit, 2002) (Fig. 1).
At mid-neurula stage, NC markers' expression is enhanced within the
neural border and overlaps with some NB markers such as pax3/7. In
contrast, other early NB markers are soon excluded from the prospective
neural crest domain (e.g. msx1) (Monsoro-Burq et al., 2005). In summary, the neural border can be dened molecularly as early as gastrulation,

26

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

by the overlap in expression of several neural border genes, a domain


within which early neural crest markers will appear.
Tissue interactions control early neural crest patterning
Explants recombination in vitro and grafting experiments in vivo have
explored how the neural crest is induced in the embryo. These
experiments have uncovered the critical role of ectodermneurectoderm
and mesodermectoderm interactions during neural crest formation.
Moreover, heterochronic recombination experiments have explored the
timing of neural crest patterning.
The ectoderm induces neural crest when apposed to a nonpatterned neural plate fragment (Liem et al., 1995). Similarly, an
early ventral neural plate implanted into lateral ectoderm in vivo,
leads to snai2 induction at the edge between the two tissues, with
both neural and ectoderm-derived cells expressing the NC markers
(Mancilla and Mayor, 1996; Selleck and Bronner-Fraser, 1995). The
effect of such experiments on neural border gene expression has
not been reported. Recombination of the non-neural ectoderm (or
early ectoderm taken from the blastocoele roof in amphibians, the
animal cap) and prospective paraxial/intermediate mesoderm (i.e.
the dorsal lateral marginal zone in amphibians) also lead to neural
crest induction followed by melanocyte differentiation in the ectoderm
(Bonstein et al., 1998; Marchant et al., 1998). Similar recombinations
have resulted in pax3 and msx1 induction using chick inducing tissues
and Xenopus responding ectoderm (Bang et al., 1999). In contrast, the
anterior neural fold or the notochord prevents either anterior or medial
snai2 expression in the neural plate (Carmona-Fontaine et al., 2007;
Rufns et al., 1998). However, notochord grafts performed after neural
tube closure do not prevent neural crest emigration (Artinger and
Bronner-Fraser, 1992), consistent with commitment to neural fold
fate after gastrulation.
In addition, explants cultured in vitro and heterochronic recombination experiments in vivo show that Xenopus neural border ectoderm is
committed to form snai2-positive cells by the end of gastrulation
(around Nieuwkoop and Faber stage 12) and that competence to respond to inducing cues is lost at the same stage (Mancilla and Mayor,
1996). This suggests that the neural border is induced and determined
prior to this stage. In chick embryos, explantation of the prospective
(pax3/7+) neural border ectoderm shows that these cells are also determined to form NC cells by gastrulation stage 4+ (Basch et al., 2006).
These experiments have opened the path for the identication of
the molecular signals responsible for neural border and neural crest
induction, as it has been assumed that similar secreted molecules
may act during both processes.

During gastrulation in mouse, Otx2 is required for expression of the


canonical Wnt antagonist dickkopf-1 (dkk1) in the anterior visceral
endoderm, the mouse head organizer (Zakin et al., 2000). Dkk1 is essential for head formation in vertebrates; it allows placode formation and
represses posterior (i.e. neural crest) fates at the anterior neural border
(Carmona-Fontaine et al., 2007; Fossat et al., 2011; Niehrs et al., 2001).
The negative feedback loop established between the anterior otx2 positive domain and the posterior gbx1/2-positive domain establishes a
sharp MHB as early as E7.5 in mouse embryos (Millet et al., 1999;
Sunmonu et al., 2011). In the posterior domain, Gbx2 represses isthmus
markers (engrailed2, wnt1, fgf8) and activates posterior neural platespecic genes, both in the medial (cad2) and lateral (pax3) domains
(Li et al., 2005, 2009; Tour et al., 2002) (Fig. 1).
Mediolateral patterning involves a combination of secreted signals at the
neural border
Within the posterior gbx2-positive domain, another complex
combination of secreted molecules establishes neural border patterning,
as gastrulation movements place the prospective paraxial/intermediate
mesoderm under the lateral neural plate (Schroeder, 1970; Steventon
et al., 2009). Neural induction, primed by FGF signals at the end of blastula stage, is controlled by simultaneous inhibition of Smad1 and Smad2
signaling (Chang and Harland, 2007; Delaune et al., 2005; Londin et al.,
2005; Wilson et al., 2000). A medial (low) to ventral (high) BMP signaling gradient, visualized by phospho-Smad1-5-8 staining, indicates the
establishment of a zone of intermediate BMP activity at the level of the
prospective neural border at the onset of gastrulation in chick, sh and
frog embryos in vivo (Faure et al., 2000, 2002; Nguyen et al., 1998).
Later, as the neural folds elevate, increased BMP4/7 mRNA expression and high BMP signaling mark the neural folds in chick, mouse,
frog and sh (Barth et al., 1999; Faure et al., 2002; Monsoro-Burq
and Le Douarin, 2001; Sakai et al., 2005; Wu et al., 2011). At
gastrula-stages, BMP4 appears to act as a morphogen in a graded
fashion by providing initial positional information in the eld of
the ectoderm, which denes main ectodermal fates: neural (low
activity); neural crest/placodes (intermediate activity); and ectoderm/cement gland (high activity).
However, the experimental modulation of BMP signaling in vivo
or in ectoderm isolated in vitro, is an inefcient neural crest inducer

Neural plate patterning cues position the neural crest-forming neural


border along anteriorposterior and mediallateral axes
Early anteriorposterior patterning denes the boundary between neural
crest-forming and neural crest-free neural border
Anterior-to-posterior neural patterning denes the extent of the
neural crest forming neural border along the body axis. Classical fatemapping experiments have shown that the neural crest originates
posterior to the mid-diencephalon (reviewed in Le Douarin, 2004).
During gastrulation, anterior and posterior brain domains are established by the mutual antagonism between otx2 and gbx1/2 transcription
factors, dening the prospective midbrainhindbrain boundary (MHB)
(Heimbucher et al., 2007; Martinez-Barbera et al., 2001; Tour et al.,
2002). This results from the combined action of Wnts, FGFs, and retinoic
acid, that impose posterior fates to the forming neural plate (Kudoh et
al., 2002; Ribes et al., 2009; Takemoto et al., 2006). Studies in zebrash,
mouse and frog embryos show that retinoic acid and FGF8 regulate the
early neural expression of gbx2; and that Wnt8 signals, diffusing from
the lateral mesendoderm, directly activate gbx1/2 (Li et al., 2009; Liu
et al., 1999; Rhinn et al., 2003, 2005, 2009; von Bubnoff et al., 1996).

Fig. 2. Neural border and neural crest patterning is a highly dynamic process which involves reiterated action of secreted signals. Several signaling pathways (BMP, WNT,
FGF, Notch) cooperate and dene the posterior neural border and the neural crest. Recently described novel modulators of these signals are activated during gastrulation or
neurulation. Collectively, their action is critical to achieve correct spatial and temporal
patterning.

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

(LaBonne and Bronner-Fraser, 1998). Wnts and FGFs potentiate NC


induction and its further development, both in vivo and in neuralized
ectoderm, leading to the two-signal model of neural crest induction
(Chang and Hemmati-Brivanlou, 1998; LaBonne and Bronner-Fraser,
1998; Saint-Jeannet et al., 1997; Villanueva et al., 2002). Similar
rules control neural border formation (de Croze et al., 2011; Hong
et al., 2008; Luo et al., 2003; Monsoro-Burq et al., 2005; Nichane et
al., 2008b; Sato et al., 2005; Tribulo et al., 2003). Multiple Wnts are
expressed in the ectoderm; some being restricted to the non-neural
ectoderm from where they diffuse towards the neural border (wnt6
in chick and wnt10a in Xenopus) (Garcia-Castro et al., 2002;
Garriock et al., 2007). Several FGFs are expressed in the paraxial/
intermediate mesoderm (Kimelman et al., 1992; Monsoro-Burq et
al., 2003; Moon, 1993) (Fig. 2). Thus far, the respective role of Wnts
and FGFs secreted from the mesoderm has been a matter of debate.
The requirement for Wnt signals at multiple steps of NB/NC
development makes it particularly difcult to evaluate the action of
other signals independently of Wnt activity (de Croze et al., 2011;
Lewis et al., 2004; Monsoro-Burq et al., 2005; Sato et al., 2005). In
addition, there are indications that wnt8 is activated by FGFs in the
mesoderm and reciprocally that Wnts activate fgf3/fgf8 in the
ectoderm (Hong et al., 2008; Zhao et al., 2008) (Fig. 2).
There are several lines of evidence suggesting that FGFs participate in NC induction. First, although FGFs seem to be weaker NC inducers than canonical Wnt signals, when applied after neural and
mesoderm induction, they display clear neural crest inducing activity
on isolated ectoderm. Basic FGF (bFGF/FGF2) also induces melanocytes in mid-gastrula ectoderm cells grown in vitro, while an earlier
treatment, at late blastula stage, activates neural fates (Kengaku and
Okamoto, 1993). Secondly, the FGF8 isoform-a (FGF8a), which does
not induce mesoderm but efciently promotes posterior neural
fates, is sufcient to induce some NC markers (foxd3, sox9 but not
snail2) in isolated blastula ectoderm (Fletcher et al., 2006; MonsoroBurq et al., 2003). The mechanism of neural crest induction by FGFs
seems to involve Wnt signals since Wnt8 is a target of FGF8 in the mesoderm, and FGFs induce neural crest in a Wnt-dependent manner in
neuralized ectoderm (Hong et al., 2008; LaBonne and Bronner-Fraser,
1998). These observations suggest that FGFs would activate Wnt signals within the mesoderm, and that in turn, Wnts would induce neural border and neural crest in the overlying ectoderm (Hong et al.,
2008).
There are, however, several strong indications of a parallel Wntindependent activation of neural border/neural crest by FGF signals.
First, in the explants recombination assay between dorsallateral
mesoderm and blastula ectoderm, blocking response to Wnts in the
ectoderm still allows efcient NC induction (Monsoro-Burq et al.,
2003). It is only when extremely high doses of Wnt inhibitors are
used that neural crest induction is lost, suggesting a non-specic
effect (Steventon et al., 2009). Secondly, FGF receptors are expressed
at the neural border and blocking FGFR signaling in the ectoderm
prevents NC induction in the mesoderm/ectoderm recombinant
assay. Furthermore, FGF-dependent Stat3 activity is essential in the
prospective NC cells (Hongo et al., 1999; Monsoro-Burq et al., 2003;
Nichane et al., 2010; Zhao et al., 2008). Finally, while Wnt and FGF
pathways may have common modulators such as the transmembrane
protein Lrig3, Wnt/Lrig3 and FGF/Lrig3 interactions display different
roles in neural crest induction (Zhao et al., 2008). Resolution of
these differences will likely occur within the next few years, thanks
to the availability of new tools and markers for analyzing discrete
steps of neural border and neural crest formation.
Fine-tuned modulation of the secreted signals allows further neural crest
induction
The precise timing of signal activity is a key parameter for neural
border and neural crest induction, highlighted by recent studies of
novel signaling modulators. In relationship to the two phases of

27

BMP signaling mentioned above, initial BMP inhibition is required


for the rst phase of NB/NC induction during gastrulation, while
further NC specication requires simultaneously increased BMP and
Wnts (Patthey et al., 2009; Steventon et al., 2009).
The timing of BMP activity also varies along the anteriorposterior
axis (Tucker et al., 2008). This dynamic activity is tightly regulated, in
part by newly identied molecules, which shape the initial BMP
gradient and rene neural border positioning (Fig. 2). For example,
a dynamic negative BMP regulation has been demonstrated for the
novel secreted antagonist Tsukushi (Ohta et al., 2004). At gastrula
stage, the BMP gradient is shaped by BMP antagonists such as Noggin,
Chordin, Follistatin, Dan, Cerberus and Tsukushi, secreted from the
node/organizer (Harland, 2000; Ohta et al., 2004). At early neurula
stage, tsukushi is secondarily activated by BMP signals around the
anterior neural border and participates in modulating BMP signaling,
by a complex, dose-dependent effect on Notch signaling, which in
turn, affects bmp4 expression at the neural border (Glavic et al.,
2004; Kuriyama et al., 2006). By this activity, Tsukushi synergizes
with Wnt signals and allows mid-neurula stage hairy2, msx1,
snail2 and sox9 expression at the neural border (Kuriyama et al.,
2006).
Simultaneously, positive BMP regulators are also needed for
neural border patterning. For example, Snw1 (also known as skiinteracting protein SKIP), which encodes a highly conserved putative
nuclear protein, was isolated in a large-scale functional screening
conducted to identify novel modulators of the neural crest fate in
Xenopus embryos (Wu et al., 2011). Snw1/SKIP is expressed dorsally
during gastrulation and enriched in the neural and neural crest tissue
at neurulation. The neural border does not form in Snw1/SKIP
morphants (lack of msx1 expression) while ectoderm and neural
plate markers are expressed in adjacent ectoderm territories. Snw1
morphants exhibit decreased phospho-Smad signals after (but not
during) gastrulation, indicating that Snw1/SKIP is essential to
promote BMP signaling and neural plate border sharpening during
neurulation (Wu et al., 2011). Another novel regulator of BMP signals, Tumor Necrosis Factor-Receptor associated Factor-4 (TRAF4),
isolated as a target of the ubiquitin ligase Smurf1, is strongly
expressed in mid-neurula stage premigratory neural crest, and potentiates both BMP and Nodal signals at the neural border (Kalkan
et al., 2009).
Similarly, several modulators of Wnt and FGF pathways have been
implicated in NC and NB formation. In addition to its action upstream
of BMP receptors, Snw1/SKIP is also an essential partner of the
canonical Wnt pathway by interacting with the transcription factor
LEF1 and repressing Wnt targets (Wang et al., 2010). In the anterior
neural plate, attenuation of Wnt signaling has also been reported for
the secreted Wnt antagonist Dkk1, and for Kremen when bound to
Dkk1 and the Wnt co-receptor LRP6 (Davidson et al., 2002; Fossat
et al., 2011; Glinka et al., 1998). Similarly, a tight control of Wnt
signaling around the neural plate or in trunk neural tube patterning,
involves various Wnt negative regulators, such as SFRPs, Kcdt15, or
ephrins (Dutta and Dawid, 2010; Misra and Matise, 2010). In contrast,
in the posterior neural border, activation of Wnt signaling is achieved
by Kremen, by Dkk1-independent LRP6 signaling activation (Hassler
et al., 2007). Similarly, the scaffolding transmembrane protein
TEM198 stimulates LRP6 signaling and is essential for Wnt signaling
in neural crest formation, while the metalloprotease ADAM13
stimulates Wnt signaling via cleavage of ephrinb2 (Liang et al.,
2011; Wei et al., 2010). Finally, the transmembrane protein Lrig3,
acts downstream of neural border formation and differentially
modulates Wnt and FGF/FGFR signaling in a context-dependent manner (Zhao et al., 2008).
These results highlight that ne-tuned signaling activities are
essential in controlling neural border positioning, and that these signals are highly dynamic both in space and time. These observations
underline the importance of a careful stage-specic analysis, especially

28

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

when these regulations are compared between different vertebrate


species.
Neural border speciers crosstalk to induce neural crest
In 2004, Meulemans and Bronner proposed a model primarily based
on expression patterns, grouping actors of neural crest development
according to their putative timing of activity at the neural border.
Hence, secreted signaling molecules act upstream of neural border
formation while stabilizing the neural border which expresses genes
that activate downstream NC specic genes. In turn, the latter genes
control EMT and migration (Meulemans and Bronner-Fraser, 2004).
This hierarchical view has since proven a very useful framework for
validation of the proposed interactions (Betancur et al., 2010). It has
dened the notion of neural border and neural crest speciers
compared to neural border/crest markers. While a marker (better a
combination of markers) is useful to identify a given tissue, a specier
has been shown to be expressed in this tissue, and to play an essential
function in its formation. Perturbed expression of a NB specier will
affect both neural border formation and downstream neural crest
development. Altered NC speciers expression does not affect neural
border formation or marker expression, but rather impacts the
premigratory neural crest and its later development.
Cross-regulation between the NB speciers has been explored
mainly in sh, lamprey and Xenopus embryos, as these vertebrate
models allow numerous types of experimental manipulations at
blastula and gastrula stages, using morpholino knock-down or
analysis of mutants. These studies have identied three sequential
steps upstream of neural crest formation: neural border induction
and maintenance, and NC speciers' induction.
Neural border speciers cooperate and stabilize the neural border
Neural border gene expression is initiated during early gastrulation,
in the posterior half of the developing neural plate (de Croze et al.,
2011; Nikitina et al., 2008). Morpholino-mediated knockdown
experiments have explored which factors initiate neural border
induction. When AP2 is depleted, neither pax3, hairy2 nor msx1
expression occurs at the edge of the neural plate of gastrula embryos,
while ap2 expression remains normal when all these other NB speciers
are depleted (de Croze et al., 2011). This indicates that AP2 is essential
in initiating the neural border. In addition, Wnt signals activate ap2,
pax3 and msx1 expression as immediate-early targets. However, this
induction depends on the presence of AP2, which directly binds to
pax3 regulatory elements (de Croze et al., 2011). A parallel observation
shows that Wnt signals directly activate gbx2; in turn, Gbx2 is essential
for early onset of the neural border by Wnts (Li et al., 2009). Additional
direct regulations involve Pou-domain factors Brn1/2 and Pbx/Hox
genes on pax3 promoter to dene neural expression and its most
anterior level of expression, respectively (Pruitt et al., 2004). Thus,
within the posterior neural domain, cooperation between Wnt signals,
Gbx2 and AP2 is responsible for neural border initiation; these factors
cooperate with additional transcription factors to directly control pax3
induction.
In a second step, as soon as neurulation starts, NB speciers
cooperate with each other, resulting in the maintenance of a proper
neural border development. Hence, AP2 depletion also prevents
later expression of the NB specier, indicating that neural border
development is not just delayed but blocked, in favor of neural formation (zic1 and sox2 are expanded) (de Croze et al., 2011; Li and
Cornell, 2007; Nikitina et al., 2008). In addition, in Xenopus embryos,
when either Msx1, Zic1, Pax3 or Hairy2 are depleted, hairy2, ap2, or
msx1 are lost, while pax3 and/or zic1 expression are increased (de
Croze et al., 2011; Monsoro-Burq et al., 2005; Nichane et al., 2008a;
Nikitina et al., 2008; Sato et al., 2005). Some species-specic
differences are observed in these regulatory relationships, since pax3

or zic1 can be either increased of diminished in lamprey embryos subjected to similar knockdown experiments (Nikitina et al., 2008).
Species-specic differences can also exist in the differential deployment
of paralog genes (pax3 or pax7) as well as in redundancy between duplicated genes (ap2a and ap2c in sh) (Basch et al., 2006; Li and Cornell,
2007; Maczkowiak et al., 2010).
As a whole, these results indicate that the neural border is not
patterned and maintained normally after perturbation of any of the
NB speciers. Consequently all these manipulations result in the
lack of neural crest induction.
Neural border speciers cooperate to induce premigratory neural crest
NB speciers' activity is tightly regulated, both positively and
negatively to activate NC speciers. For example, Nichane and
colleagues have provided a detailed view of the complex interactions
taking place between the NB speciers Hairy2, Id3 and Stat3 in neural
crest induction. By loss and gain of function experiments followed by
NC markers detection (snail2, foxd3, sox10), they showed that Id3 is
required for Hairy2 to expand the neural crest, and that Hairy2 and
Stat3 act synergistically in NC induction (Nichane et al., 2008a,
2010). In particular, Hairy2 plays a dual role in NC specication:
through cell-autonomous mechanisms involving Hairy2 DNAbinding activity, Hairy2 promotes undifferentiated NC progenitor
survival and maintenance, while a non-cell-autonomous mechanism
involves Hairy2 binding to FGFR4/Stat3 complex leading to id3
activation. In turn, Id3 promotes neural crest proliferation and
differentiation (Kee and Bronner-Fraser, 2005; Light et al., 2005).
Stat3 plays a central role in regulating this balance. At low levels,
Stat3 activates hairy2, which will prevent id3 activation, via bmp4
inhibition (Nichane et al., 2008a, 2010). Conversely, at high activity
levels, Stat3 induces id3, which in turn down regulates hairy2 and
Stat3 signaling by disrupting the Stat3-Hairy2-FGFR4 ternary
complex (Nichane et al., 2010). Thus, low Stat3 activity maintains
prospective NC cells undifferentiated; high Stat3 activity induces NC
specication; respectively, Hairy2 and Id3 function as positive and
negative feedback regulators of Stat3 activation, allowing ne-tuned
control of the system.
Other NB speciers have been shown to cooperate in NC
induction. In particular, Pax3 and Zic1 play a key role in premigratory
NC induction. Pax3 and Zic1 co-expression induces ectopic ventral
snai2 expression in whole embryos, as well as snai2 and foxd3 expression in animal caps (Monsoro-Burq et al., 2005; Sato et al., 2005).
Over expressing Zic1 (or Pax3) cannot compensate for Pax3 (or Zic1
respectively) depletion (Sato et al., 2005). Balanced Pax3 and Zic1
levels are critical for allowing neural crest fate choice, at the expense
of other ectoderm fates (placodes or hatching gland in Xenopus embryos) (Hong and Saint-Jeannet, 2007). In parallel, Msx1 activates
pax3 cell-autonomously, while Pax3 activity is necessary for Msx1
to induce snai2, and Pax3 gain-of-function compensates for Msx1
knockdown (Monsoro-Burq et al., 2005). Finally, downstream of
Pax3 and Zic1 cooperation, AP2 plays another role: it is both required
to allow NC marker induction by Pax3/Zic1 in explants, and sufcient
to compensate for Pax3 or Zic1 knockdown in snai2 and foxd3
induction in vivo (de Croze et al., 2011). Together, these results
indicate hierarchical (i.e. epistatic) relationships between the NB
speciers. Future work will further validate these as direct or indirect interactions (Betancur et al., 2010).
NB speciers are essential mediators of secreted factors in NC induction
By their cooperative response, the NB speciers integrate the
multiple developmental cues provided by the tissues surrounding
the neural plate. Hairy2 and Msx1 act downstream of FGF and BMP
signaling to induce NB (pax3) and NC markers' expression (snai2,
foxd3) while Wnt signaling induces NC in Hairy2 and Msx1 morphant

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

embryos (Nichane et al., 2008a) (Monsoro-Burq et al., 2005). In


parallel, AP2, Pax3 and Zic1 are essential for Wnt, FGF and BMP
signaling to induce NC (de Croze et al., 2011; Monsoro-Burq et al.,
2005; Sato et al., 2005). In addition, several combinations between a
given NB specier and a secreted signal have been validated for
neural border and neural crest induction in animal caps (e.g. Msx1
and Noggin, AP2 and Chordin/Wnt and AP2 and Noggin; Pax3 and
Wnt or BMP, Zic1 and Wnt or BMP etc.) (de Croze et al., 2011; Luo
et al., 2003; Monsoro-Burq et al., 2005; Sato et al., 2005). These
experiments indicate that, BMP levels must be tightly balanced, and
that such assays result into the activation of the whole complement
of NB speciers followed by neural crest induction.

29

affects the expression of the NC speciers sox9, foxd3 and snai2 but
not dorsal neural tube markers. Changes in H3K9me3, H3K36me3
and JMJD2A occupancy on NC specier genes (sox10, snail2) were
observed during chick development coincident with NC specication.
JmjD2A is responsible for derepressing sox10 by H3K9me3 demethylation (Strobl-Mazzulla et al., 2010).
Chromatin structure regulation is known to provide spatiotemporal control of stem cell maintenance, specication and differentiation. The discovery of a developmental role for epigenetic regulators in
controlling neural crest specication opens exciting avenues for novel
research.
Conserved evolutionary function of the neural border speciers

Downstream regulators of NC fate and secondary use of signaling


pathways
Downstream of Pax3 and Zic1, several novel effectors and a reiterated
activation of the Wnt, FGF, BMP and Notch pathway are necessary to
allow further NC development. The TALE homeodomain Meis proteins
are important candidates for coordinating hindbrain neural and neural
crest patterning. Meis3 is a co-factor of Hox proteins (Nakamura et al.,
1996) expressed in the neural plate and regulated by mesodermal
Wnt3a (Elkouby et al., 2010; Gutkovich et al., 2010). Loss of function
experiments have established that Meis3 acts downstream of Pax3/
Zic1 and is essential for the expression of a subset of NC genes and NC
formation (Gutkovich et al., 2010; Maeda et al., 2002). In addition,
Meis3 directly activates FGF3 and FGF8 and interacts with retinoic acid
signaling in the neurectoderm, promoting further secondary signaling
necessary for NC induction (Aamar and Frank, 2004; Dibner et al.,
2004; Elkouby et al., 2010). In parallel, Lrig3 also acts downstream of
Pax3 and Zic1 in neural crest specication (Zhao et al., 2008).
Furthermore, dissociated AC experiments showed that after initial
induction of Msx1 and Pax3 by Wnt, Pax3 and Zic1 cooperate with the
canonical Wnt pathway to induce snail2 and foxd3 expression in a
non-cell-autonomous manner (Monsoro-Burq et al., 2005; Sato
et al., 2005). Downstream of Hairy2/Stat3 interactions, high Stat3
signaling activates Delta/Notch signaling which results in bmp4/id3
expression modulation and NC specication (Nichane et al., 2008a,
2008c, 2010). As described previously, modulation of several other
signaling pathways is involved in NC formation at the neurula stage.
These include the modulation of BMP and Wnt pathways by Tsukushi,
requirement of Snw1/SKIP for BMP signaling, Nodal and BMP signals
potentiated by TRAF4, modulation of Wnt signaling by positive and
negative regulators. In conclusion, neural border patterning sets the
scene for NC specication, by the induction and maintenance of
expression of essential transcription factors and signaling molecules
that will, in turn, activate NC speciers' gene expression.
Epigenetic regulation: jumonji and CDH7 control timing and access to
neural crest speciers' promoters
Recently, several epigenetic regulators have been found to play a
crucial role in NC induction, in Xenopus, chick and human. CHD7, an
ATP-dependent chromatin remodeler, is essential for multipotent
migratory neural crest formation, both in human embryonic stem
cells (hESCs) in vitro and in Xenopus embryos. CHD7, together with
BRG1 (a SWI/SNF family member), occupies distal regulatory
elements controlling the expression of critical NC speciers (SOX9
and TWIST) in human cells. Furthermore, they act synergistically in
vivo in the Xenopus embryo to regulate NC gene expression and migration (but not neural border formation) (Bajpai et al., 2010).
Another chromatin regulator, JumonjiD2A, regulates NC formation
in chick embryos. JMJD2A, a Histone Lysine Demethylase, is a subunit
of the PRC2 complex. Fine-tuned, dynamic regulation of H3K27me3
by JMJD2A and PRC2 balances stem cell and differentiation status in
mouse ESCs (Shen et al., 2009). In chick, JmjD2A loss of function

Although the function of NB speciers on NC formation during


human gastrulation cannot be directly studied, some NB speciers
are found mutated in neurocristopathies. Interestingly, human Zic3
can promote NC formation in Xenopus embryos (Chin et al., 2007)
and one of the mutated human alleles tested in this study has lost
its NC-inducing properties when over expressed. Pax3 is mutated in
the Waardenburg syndrome (WS type I and III), is responsible for
the mouse Splotch phenotype (Epstein et al., 1991; Tassabehji et al.,
1992), and is expressed in human premigratory NC (Betters et al.,
2010). However, no functional analysis of Pax3 on NC formation in
humans has been performed thus far.
Thanks to the development of protocols allowing hESCs manipulation,
it is now possible to induce the in vitro differentiation of hESCs into NC
cells (for a review see Chimge and Bayarsaihan, 2010; and Milet and
Monsoro-Burq, 2012this issue). Several genes involved in early NC specication are expressed in neuralized hESCs and in the derived NC-like
cells: ap2, pax3 and msx1 (Pomp et al., 2005) (Curchoe et al., 2010). This
model is of great interest to test the role of known NB specier genes
on the onset of human NC specication, or the role of genes that are
found mutated in neurocristopathies. For example, CHD7, important for
sox9 and twist induction, is mutated in the CHARGE syndrome (Bajpai
et al., 2010).
Conclusion and perspectives
In the last few years, studies of the earliest steps of neural crest
formation have revealed the complexity of the signaling and
transcriptional events taking place at the edge of the neural plate, as
early as gastrulation starts. This region, now dened as the neural
border in the young gastrula, is mapped in several vertebrate embryos,
and is marked by early molecular markers, the most specic being the
transcription factor pax3/7. The neural border includes progenitors for
the dorsal neural tube, the neural crest, placodes and ectoderm. The
neural border is rst induced by synergy between various secreted signals. It is then stabilized by interactions between several transcription
factors co-expressed in this region, the neural border speciers. Finally,
in coordination with additional secreted signaling pathways, neural
border speciers promote uncommitted neural crest progenitor proliferation on one hand, and induce the neural crest-specic regulators
on the other hand. These studies have provided a novel framework
that will allow building the detailed neural border gene regulatory network in the near future.
Acknowledgments
The authors are grateful to the members of the Monsoro-Burq
laboratory for their critical comments on the manuscript. Our research
is supported by grants from Ligue Contre le Cancer, Association pour
la Recherche contre le Cancer (SFI20101201882), CNRS, Universit
Paris Sud-11 (Attractivit), and Agence Nationale pour la Recherche to
A. M-B. C.M. is supported by fellowships from DIM Stem-Pole/Rgion
Ile-de-France and Universit Paris-Sud-11.

30

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

References
Aamar, E., Frank, D., 2004. Xenopus Meis3 protein forms a hindbrain-inducing center
by activating FGF/MAP kinase and PCP pathways. Development 131, 153163.
Ahlstrom, J.D., Erickson, C.A., 2009. The neural crest epithelial-mesenchymal transition
in 4D: a tail of multiple non-obligatory cellular mechanisms. Development 136,
18011812.
Aoki, Y., Saint-Germain, N., Gyda, M., Magner-Fink, E., Lee, Y.H., Credidio, C., Saint-Jeannet,
J.P., 2003. Sox10 regulates the development of neural crest-derived melanocytes in
Xenopus. Dev. Biol. 259, 1933.
Artinger, K.B., Bronner-Fraser, M., 1992. Notochord grafts do not suppress formation of
neural crest cells or commissural neurons. Development 116, 877886.
Aruga, J., Tohmonda, T., Homma, S., Mikoshiba, K., 2002. Zic1 promotes the expansion
of dorsal neural progenitors in spinal cord by inhibiting neuronal differentiation.
Dev. Biol. 244, 329341.
Avilion, A.A., Nicolis, S.K., Pevny, L.H., Perez, L., Vivian, N., Lovell-Badge, R., 2003. Multipotent
cell lineages in early mouse development depend on SOX2 function. Genes Dev. 17,
126140.
Bajpai, R., Chen, D.A., Rada-Iglesias, A., Zhang, J., Xiong, Y., Helms, J., Chang, C.P., Zhao,
Y., Swigut, T., Wysocka, J., 2010. CHD7 cooperates with PBAF to control multipotent
neural crest formation. Nature 463, 958962.
Bang, A.G., Papalopulu, N., Goulding, M.D., 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, 366380.
Barrionuevo, F., Naumann, A., Bagheri-Fam, S., Speth, V., Taketo, M.M., Scherer, G.,
Neubuser, A., 2008. Sox9 is required for invagination of the otic placode in mice.
Dev. Biol. 317, 213224.
Barth, K.A., Kishimoto, Y., Rohr, K.B., Seydler, C., Schulte-Merker, S., Wilson, S.W., 1999.
Bmp activity establishes a gradient of positional information throughout the entire
neural plate. Development 126, 49774987.
Basch, M.L., Bronner-Fraser, M., Garcia-Castro, M.I., 2006. Specication of the neural
crest occurs during gastrulation and requires Pax7. Nature 441, 218222.
Bell, K.M., Western, P.S., Sinclair, A.H., 2000. SOX8 expression during chick embryogenesis.
Mech. Dev. 94, 257260.
Bellefroid, E.J., Kobbe, A., Gruss, P., Pieler, T., Gurdon, J.B., Papalopulu, N., 1998. Xiro3
encodes a Xenopus homolog of the Drosophila Iroquois genes and functions in neural
specication. EMBO J. 17, 191203.
Bellmeyer, A., Krase, J., Lindgren, J., LaBonne, C., 2003. The protooncogene c-myc is
an essential regulator of neural crest formation in xenopus. Dev. Cell 4,
827839.
Betancur, P., Bronner-Fraser, M., Sauka-Spengler, T., 2010. Assembling neural crest
regulatory circuits into a gene regulatory network. Annu. Rev. Cell Dev. Biol. 26,
581603.
Betters, E., Liu, Y., Kjaeldgaard, A., Sundstrom, E., Garcia-Castro, M.I., 2010. Analysis of
early human neural crest development. Dev. Biol. 344, 578592.
Bonstein, L., Elias, S., Frank, D., 1998. Paraxial-fated mesoderm is required for neural
crest induction in Xenopus embryos. Dev. Biol. 193, 156168.
Bronner-Fraser, M., Fraser, S.E., 1988. Cell lineage analysis reveals multipotency of
some avian neural crest cells. Nature 335, 161164.
Bulfone, A., Puelles, L., Porteus, M.H., Frohman, M.A., Martin, G.R., Rubenstein, J.L., 1993.
Spatially restricted expression of Dlx-1, Dlx-2 (Tes-1), Gbx-2, and Wnt-3 in the
embryonic day 12.5 mouse forebrain denes potential transverse and longitudinal
segmental boundaries. J. Neurosci. 13, 31553172.
Byrd, N.A., Meyers, E.N., 2005. Loss of Gbx2 results in neural crest cell patterning and
pharyngeal arch artery defects in the mouse embryo. Dev. Biol. 284, 233245.
Carmona-Fontaine, C., Acuna, G., Ellwanger, K., Niehrs, C., Mayor, R., 2007. Neural crests
are actively precluded from the anterior neural fold by a novel inhibitory mechanism
dependent on Dickkopf1 secreted by the prechordal mesoderm. Dev. Biol. 309,
208221.
Chang, C., Harland, R.M., 2007. Neural induction requires continued suppression of
both Smad1 and Smad2 signals during gastrulation. Development 134, 38613872.
Chang, C., Hemmati-Brivanlou, A., 1998. Neural crest induction by Xwnt7B in Xenopus.
Dev. Biol. 194, 129134.
Cheng, Y., Cheung, M., Abu-Elmagd, M.M., Orme, A., Scotting, P.J., 2000. Chick sox10, a
transcription factor expressed in both early neural crest cells and central nervous
system. Brain Res. Dev. Brain Res. 121, 233241.
Cheung, M., Briscoe, J., 2003. Neural crest development is regulated by the transcription
factor Sox9. Development 130, 56815693.
Cheung, M., Chaboissier, M.C., Mynett, A., Hirst, E., Schedl, A., Briscoe, J., 2005. The
transcriptional control of trunk neural crest induction, survival, and delamination.
Dev. Cell 8, 179192.
Chhin, B., Hatayama, M., Bozon, D., Ogawa, M., Schon, P., Tohmonda, T., Sassolas, F.,
Aruga, J., Valard, A.G., Chen, S.C., et al., 2007. Elucidation of penetrance variability of a ZIC3 mutation in a family with complex heart defects and functional
analysis of ZIC3 mutations in the rst zinc nger domain. Hum. Mutat. 28,
563570.
Chimge, N.O., Bayarsaihan, D., 2010. Generation of neural crest progenitors from
human embryonic stem cells. J. Exp. Zool. B Mol. Dev. Evol. 314, 95103.
Collazo, A., Bronner-Fraser, M., Fraser, S.E., 1993. Vital dye labelling of Xenopus laevis trunk
neural crest reveals multipotency and novel pathways of migration. Development
118, 363376.
Curchoe, C.L., Maurer, J., McKeown, S.J., Cattarossi, G., Cimadamore, F., Nilbratt, M., Snyder,
E.Y., Bronner-Fraser, M., Terskikh, A.V., 2010. Early acquisition of neural crest
competence during hESCs neuralization. PLoS One 5, e13890.
Davidson, D.R., Hill, R.E., 1991. Msh-like Genes: A Family of Homeobox Genes with
Wide-ranging Expression during Vertebrate Development.

Davidson, G., Mao, B., del Barco Barrantes, I., Niehrs, C., 2002. Kremen proteins interact
with Dickkopf1 to regulate anteroposterior CNS patterning. Development 129,
55875596.
de Croze, N., Maczkowiak, F., Monsoro-Burq, A.H., 2011. Reiterative AP2a activity
controls sequential steps in the neural crest gene regulatory network. Proc. Natl.
Acad. Sci. U. S. A. 108, 155160.
Delaune, E., Lemaire, P., Kodjabachian, L., 2005. Neural induction in Xenopus requires
early FGF signalling in addition to BMP inhibition. Development 132, 299310.
Dibner, C., Elias, S., Or, R., Souopgui, J., Kolm, P.J., Sive, H., Pieler, T., Frank, D., 2004. The
Meis3 protein and retinoid signaling interact to pattern the Xenopus hindbrain.
Dev. Biol. 271, 7586.
Dottori, M., Gross, M.K., Labosky, P., Goulding, M., 2001. The winged-helix transcription
factor Foxd3 suppresses interneuron differentiation and promotes neural crest cell
fate. Development 128, 41274138.
Dutta, S., Dawid, I.B., 2010. Kctd15 inhibits neural crest formation by attenuating Wnt/
beta-catenin signaling output. Development 137, 30133018.
Dy, P., Han, Y., Lefebvre, V., 2008. Generation of mice harboring a Sox5 conditional null
allele. Genesis 46, 294299.
Elkouby, Y.M., Elias, S., Casey, E.S., Blythe, S.A., Tsabar, N., Klein, P.S., Root, H., Liu, K.J.,
Frank, D., 2010. Mesodermal Wnt signaling organizes the neural plate via Meis3.
Development 137, 15311541.
Epstein, D.J., Vekemans, M., Gros, P., 1991. Splotch (Sp2H), a mutation affecting development
of the mouse neural tube, shows a deletion within the paired homeodomain of Pax-3.
Cell 67, 767774.
Essex, L.J., Mayor, R., Sargent, M.G., 1993. Expression of Xenopus snail in mesoderm and
prospective neural fold ectoderm. Dev. Dyn. 198, 108122.
Ezin, A.M., Fraser, S.E., Bronner-Fraser, M., 2009. Fate map and morphogenesis of
presumptive neural crest and dorsal neural tube. Dev. Biol. 330, 221236.
Faure, S., Lee, M.A., Keller, T., ten Dijke, P., Whitman, M., 2000. Endogenous patterns of
TGFbeta superfamily signaling during early Xenopus development. Development
127, 29172931.
Faure, S., de Santa Barbara, P., Roberts, D.J., Whitman, M., 2002. Endogenous patterns of
BMP signaling during early chick development. Dev. Biol. 244, 4465.
Feledy, J.A., Beanan, M.J., Sandoval, J.J., Goodrich, J.S., Lim, J.H., Matsuo-Takasaki,
M., Sato, S.M., Sargent, T.D., 1999. Inhibitory patterning of the anterior neural
plate in Xenopus by homeodomain factors Dlx3 and Msx1. Dev. Biol. 212,
455464.
Fernandez-Garre, P., Rodriguez-Gallardo, L., Gallego-Diaz, V., Alvarez, I.S., Puelles, L.,
2002. Fate map of the chicken neural plate at stage 4. Development 129,
28072822.
Fletcher, R.B., Baker, J.C., Harland, R.M., 2006. FGF8 spliceforms mediate early mesoderm
and posterior neural tissue formation in Xenopus. Development 133, 17031714.
Fossat, N., Jones, V., Khoo, P.L., Bogani, D., Hardy, A., Steiner, K., Mukhopadhyay, M.,
Westphal, H., Nolan, P.M., Arkell, R., et al., 2011. Stringent requirement of a proper
level of canonical WNT signalling activity for head formation in mouse embryo.
Development 138, 667676.
Fuchs, E., 1995. Keratins and the skin. Annu. Rev. Cell Dev. Biol. 11, 123153.
Garcia-Castro, M.I., Marcelle, C., Bronner-Fraser, M., 2002. Ectodermal Wnt function as
a neural crest inducer. Science 297, 848851.
Garriock, R.J., Warkman, A.S., Meadows, S.M., D'Agostino, S., Krieg, P.A., 2007. Census of
vertebrate Wnt genes: isolation and developmental expression of Xenopus Wnt2,
Wnt3, Wnt9a, Wnt9b, Wnt10a, and Wnt16. Dev. Dyn. 236, 12491258.
Gerard, M., Abitbol, M., Delezoide, A.L., Duer, J.L., Mallet, J., Vekemans, M., 1995. PAXgenes expression during human embryonic development, a preliminary report. C.
R. Acad. Sci. III 318, 5766.
Glavic, A., Silva, F., Aybar, M.J., Bastidas, F., Mayor, R., 2004. Interplay between Notch
signaling and the homeoprotein Xiro1 is required for neural crest induction in
Xenopus embryos. Development 131, 347359 (Epub 2003 Dec 2017).
Glinka, A., Wu, W., Delius, H., Monaghan, A.P., Blumenstock, C., Niehrs, C., 1998. Dickkopf-1
is a member of a new family of secreted proteins and functions in head induction.
Nature 391, 357362.
Goulding, M.D., Chalepakis, G., Deutsch, U., Erselius, J.R., Gruss, P., 1991. Pax-3, a novel murine
DNA binding protein expressed during early neurogenesis. EMBO J. 10, 11351147.
Gutkovich, Y.E., Or, R., Elkouby, Y.M., Dibner, C., Gefen, A., Elias, S., Frank, D., 2010.
Xenopus Meis3 protein lies at a nexus downstream to Zic1 and Pax3 proteins,
regulating multiple cell-fates during early nervous system development. Dev.
Biol. 338, 5062.
Harland, R., 2000. Neural induction. Curr. Opin. Genet. Dev. 10, 357362.
Hassler, C., Cruciat, C.M., Huang, Y.L., Kuriyama, S., Mayor, R., Niehrs, C., 2007. Kremen
is required for neural crest induction in Xenopus and promotes LRP6-mediated
Wnt signaling. Development 134, 42554263.
Heimbucher, T., Murko, C., Bajoghli, B., Aghaallaei, N., Huber, A., Stebegg, R., Eberhard,
D., Fink, M., Simeone, A., Czerny, T., 2007. Gbx2 and Otx2 interact with the WD40
domain of Groucho/Tle corepressors. Mol. Cell. Biol. 27, 340351.
Hill, R.E., Jones, P.F., Rees, A.R., Sime, C.M., Justice, M.J., Copeland, N.G., Jenkins, N.A.,
Graham, E., Davidson, D.R., 1989. A new family of mouse homeo box-containing
genes: molecular structure, chromosomal location, and developmental expression
of Hox-7.1. Genes Dev. 3, 2637.
His, W., 1868. Untersuchungen ber die erste Anlage des Wirbeltierleibes: die erste
Entwickelung des Hhnchens im Ei. Vogel FCW, Leipzig, p. 237.
Hong, C.S., Saint-Jeannet, J.P., 2007. The activity of Pax3 and Zic1 regulates three distinct
cell fates at the neural plate border. Mol. Biol. Cell 18, 21922202.
Hong, C.S., Park, B.Y., Saint-Jeannet, J.P., 2008. Fgf8a induces neural crest indirectly through
the activation of Wnt8 in the paraxial mesoderm. Development 135, 39033910.
Hongo, I., Kengaku, M., Okamoto, H., 1999. FGF signaling and the anterior neural induction
in Xenopus. Dev. Biol. 216, 561581.

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233


Honore, S.M., Aybar, M.J., Mayor, R., 2003. Sox10 is required for the early development
of the prospective neural crest in Xenopus embryos. Dev. Biol. 260, 7996.
Inoue, T., Hatayama, M., Tohmonda, T., Itohara, S., Aruga, J., Mikoshiba, K., 2004. Mouse
Zic5 deciency results in neural tube defects and hypoplasia of cephalic neural
crest derivatives. Dev. Biol. 270, 146162.
Iwafuchi-Doi, M., Yoshida, Y., Onichtchouk, D., Leichsenring, M., Driever, W., Takemoto,
T., Uchikawa, M., Kamachi, Y., Kondoh, H., 2011. The Pou5f1/Pou3f-dependent but
SoxB-independent regulation of conserved enhancer N2 initiates Sox2 expression during epiblast to neural plate stages in vertebrates. Dev. Biol. 352,
354366.
Jen, Y., Manova, K., Benezra, R., 1996. Expression patterns of Id1, Id2, and Id3 are highly
related but distinct from that of Id4 during mouse embryogenesis. Dev. Dyn. 207,
235252.
Jiang, R., Lan, Y., Norton, C.R., Sundberg, J.P., Gridley, T., 1998. The Slug gene is not essential
for mesoderm or neural crest development in mice. Dev. Biol. 198, 277285.
Jostes, B., Walther, C., Gruss, P., 1990. The murine paired box gene, Pax7, is expressed
specically during the development of the nervous and muscular system. Mech.
Dev. 33, 2737.
Kalkan, T., Iwasaki, Y., Park, C.Y., Thomsen, G.H., 2009. Tumor necrosis factor-receptorassociated factor-4 is a positive regulator of transforming growth factor-beta
signaling that affects neural crest formation. Mol. Biol. Cell 20, 34363450.
Kee, Y., Bronner-Fraser, M., 2001a. Id4 expression and its relationship to other Id genes
during avian embryonic development. Mech. Dev. 109, 341345.
Kee, Y., Bronner-Fraser, M., 2001b. The transcriptional regulator Id3 is expressed in cranial
sensory placodes during early avian embryonic development. Mech. Dev. 109,
337340.
Kee, Y., Bronner-Fraser, M., 2005. To proliferate or to die: role of Id3 in cell cycle
progression and survival of neural crest progenitors. Genes Dev. 19, 744755.
Kengaku, M., Okamoto, H., 1993. Basic broblast growth factor induces differentiation
of neural tube and neural crest lineages of cultured ectoderm cells from Xenopus
gastrula. Development 119, 10671078.
Kimelman, D., Christian, J.L., Moon, R.T., 1992. Synergistic principles of development: overlapping patterning systems in Xenopus mesoderm induction. Development 116, 19.
Kos, R., Reedy, M.V., Johnson, R.L., Erickson, C.A., 2001. The winged-helix transcription
factor FoxD3 is important for establishing the neural crest lineage and repressing
melanogenesis in avian embryos. Development 128, 14671479.
Krispin, S., Nitzan, E., Kalcheim, C., 2010. The dorsal neural tube: a dynamic setting for
cell fate decisions. Dev. Neurobiol. 70, 796812.
Kudoh, T., Wilson, S.W., Dawid, I.B., 2002. Distinct roles for Fgf, Wnt and retinoic acid in
posteriorizing the neural ectoderm. Development 129, 43354346.
Kuhlbrodt, K., Herbarth, B., Sock, E., Hermans-Borgmeyer, I., Wegner, M., 1998. Sox10, a
novel transcriptional modulator in glial cells. J. Neurosci. 18, 237250.
Kuriyama, S., Lupo, G., Ohta, K., Ohnuma, S., Harris, W.A., Tanaka, H., 2006. Tsukushi
controls ectodermal patterning and neural crest specication in Xenopus by direct
regulation of BMP4 and X-delta-1 activity. Development 133, 7588.
LaBonne, C., Bronner-Fraser, M., 1998. Neural crest induction in Xenopus: evidence for
a two-signal model. Development 125, 24032414.
Le Douarin, N.M., 2004. The avian embryo as a model to study the development of the
neural crest: a long and still ongoing story. Mech. Dev. 121, 10891102.
Le Douarin, M.L., Kalcheim, C., 1999. The Neural Crest. Cambridge university press.
Lewis, J.L., Bonner, J., Modrell, M., Ragland, J.W., Moon, R.T., Dorsky, R.I., Raible, D.W., 2004.
Reiterated Wnt signaling during zebrash neural crest development. Development
131, 12991308 (Epub 2004 Feb 1218).
Li, W., Cornell, R.A., 2007. Redundant activities of Tfap2a and Tfap2c are required for
neural crest induction and development of other non-neural ectoderm derivatives
in zebrash embryos. Dev. Biol. 304, 338354.
Li, J.Y., Lao, Z., Joyner, A.L., 2005. New regulatory interactions and cellular responses in
the isthmic organizer region revealed by altering Gbx2 expression. Development
132, 19711981.
Li, B., Kuriyama, S., Moreno, M., Mayor, R., 2009. The posteriorizing gene Gbx2 is a
direct target of Wnt signalling and the earliest factor in neural crest induction.
Development 136, 32673278.
Liang, J., Fu, Y., Cruciat, C.M., Jia, S., Wang, Y., Tong, Z., Tao, Q., Ingelnger, D., Boutros,
M., Meng, A., Niehrs, C., Wu, W., 2011. Transmembrane protein 198 promotes
LRP6 phosphorylation and Wnt signaling activation. Mol. Cell. Biol. 31 (13),
25772590.
Liem Jr., K.F., Tremml, G., Roelink, H., Jessell, T.M., 1995. Dorsal differentiation of neural
plate cells induced by BMP-mediated signals from epidermal ectoderm. Cell 82,
969979.
Light, W., Vernon, A.E., Lasorella, A., Iavarone, A., LaBonne, C., 2005. Xenopus Id3 is
required downstream of Myc for the formation of multipotent neural crest progenitor
cells. Development 132, 18311841.
Liu, K.J., Harland, R.M., 2003. Cloning and characterization of Xenopus Id4 reveals
differing roles for Id genes. Dev. Biol. 264, 339351.
Liu, A., Losos, K., Joyner, A.L., 1999. FGF8 can activate Gbx2 and transform regions of the
rostral mouse brain into a hindbrain fate. Development 126, 48274838.
Londin, E.R., Niemiec, J., Sirotkin, H.I., 2005. Chordin, FGF signaling, and mesodermal
factors cooperate in zebrash neural induction. Dev. Biol. 279, 119.
Luo, T., Matsuo-Takasaki, M., Lim, J.H., Sargent, T.D., 2001. Differential regulation of Dlx
gene expression by a BMP morphogenetic gradient. Int. J. Dev. Biol. 45, 681684.
Luo, T., Lee, Y.H., Saint-Jeannet, J.P., Sargent, T.D., 2003. Induction of neural crest in
Xenopus by transcription factor AP2alpha. Proc. Natl. Acad. Sci. U. S. A. 100,
532537.
Maczkowiak, F., Matos, S., Wang, E., Roche, D., Harland, R., Monsoro-Burq, A.H., 2010. The
Pax3 and Pax7 paralogs cooperate in neural and neural crest patterning using distinct
molecular mechanisms, in Xenopus laevis embryos. Dev. Biol. 340 (2), 381396.

31

Maeda, R., Ishimura, A., Mood, K., Park, E.K., Buchberg, A.M., Daar, I.O., 2002. Xpbx1b and
Xmeis1b play a collaborative role in hindbrain and neural crest gene expression in
Xenopus embryos. Proc. Natl. Acad. Sci. U. S. A. 99, 54485453.
Mancilla, A., Mayor, R., 1996. Neural crest formation in Xenopus laevis: mechanisms of
Xslug induction. Dev. Biol. 177, 580589.
Mansouri, A., Stoykova, A., Torres, M., Gruss, P., 1996. Dysgenesis of cephalic neural
crest derivatives in Pax7/ mutant mice. Development 122, 831838.
Marchant, L., Linker, C., Ruiz, P., Guerrero, N., Mayor, R., 1998. The inductive properties
of mesoderm suggest that the neural crest cells are specied by a BMP gradient.
Dev. Biol. 198, 319329.
Marin, F., Nieto, M.A., 2004. Expression of chicken slug and snail in mesenchymal
components of the developing central nervous system. Dev. Dyn. 230, 144148.
Martin, B.L., Harland, R.M., 2001. Hypaxial muscle migration during primary myogenesis
in Xenopus laevis. Dev. Biol. 239, 270280.
Martinez-Barbera, J.P., Signore, M., Boyl, P.P., Puelles, E., Acampora, D., Gogoi, R., Schubert,
F., Lumsden, A., Simeone, A., 2001. Regionalisation of anterior neuroectoderm and its
competence in responding to forebrain and midbrain inducing activities depend on
mutual antagonism between OTX2 and GBX2. Development 128, 47894800.
Martinsen, B.J., Bronner-Fraser, M., 1998. Neural crest specication regulated by the
helix-loop-helix repressor Id2. Science 281, 988991.
Matsunaga, E., Araki, I., Nakamura, H., 2001. Role of Pax3/7 in the tectum regionalization.
Development 128, 40694077.
Mayor, R., Morgan, R., Sargent, M.G., 1995. Induction of the prospective neural crest of
Xenopus. Development 121, 767777.
Meulemans, D., Bronner-Fraser, M., 2004. Gene-regulatory interactions in neural crest
evolution and development. Dev. Cell 7, 291299.
Milet, Monsoro-Burq, 2012. Embryonic stem cell strategies to explore neural crest development in human embryos. Dev. Biol. 366, 9699 (this issue).
Millet, S., Campbell, K., Epstein, D.J., Losos, K., Harris, E., Joyner, A.L., 1999. A role for
Gbx2 in repression of Otx2 and positioning the mid/hindbrain organizer. Nature
401, 161164.
Misra, K., Matise, M.P., 2010. A critical role for sFRP proteins in maintaining caudal neural
tube closure in mice via inhibition of BMP signaling. Dev. Biol. 337, 7483.
Mitchell, P.J., Timmons, P.M., Hebert, J.M., Rigby, P.W., Tjian, R., 1991. Transcription factor
AP-2 is expressed in neural crest cell lineages during mouse embryogenesis. Genes
Dev. 5, 105119.
Mizuseki, K., Kishi, M., Matsui, M., Nakanishi, S., Sasai, Y., 1998. Xenopus Zic-related-1
and Sox-2, two factors induced by chordin, have distinct activities in the initiation
of neural induction. Development 125, 579587.
Monsoro-Burq, A., Le Douarin, N.M., 2001. BMP4 plays a key role in leftright patterning in chick embryos by maintaining Sonic Hedgehog asymmetry. Mol. Cell 7,
789799.
Monsoro-Burq, A.H., Fletcher, R.B., Harland, R.M., 2003. Neural crest induction by
paraxial mesoderm in Xenopus embryos requires FGF signals. Development 130,
31113124.
Monsoro-Burq, A.H., Wang, E., Harland, R., 2005. Msx1 and Pax3 cooperate to mediate
FGF8 and WNT signals during Xenopus neural crest induction. Dev. Cell 8,
167178.
Moon, R.T., 1993. In pursuit of the functions of the Wnt family of developmental
regulators: insights from Xenopus laevis. BioEssays 15, 9197.
Myat, A., Henrique, D., Ish-Horowicz, D., Lewis, J., 1996. A chick homologue of Serrate and
its relationship with Notch and Delta homologues during central neurogenesis. Dev.
Biol. 174, 233247.
Nagai, T., Aruga, J., Takada, S., Gunther, T., Sporle, R., Schughart, K., Mikoshiba, K., 1997.
The expression of the mouse Zic1, Zic2, and Zic3 gene suggests an essential role for
Zic genes in body pattern formation. Dev. Biol. 182, 299313.
Nakamura, T., Jenkins, N.A., Copeland, N.G., 1996. Identication of a new family of Pbxrelated homeobox genes. Oncogene 13, 22352242.
Nakata, K., Nagai, T., Aruga, J., Mikoshiba, K., 1997. Xenopus Zic3, a primary regulator
both in neural and neural crest development. Proc. Natl. Acad. Sci. U. S. A. 94,
1198011985.
Nakata, K., Nagai, T., Aruga, J., Mikoshiba, K., 1998. Xenopus Zic family and its role in
neural and neural crest development. Mech. Dev. 75, 4351.
Nakata, K., Koyabu, Y., Aruga, J., Mikoshiba, K., 2000. A novel member of the Xenopus
Zic family, Zic5, mediates neural crest development. Mech. Dev. 99, 8391.
Nguyen, V.H., Schmid, B., Trout, J., Connors, S.A., Ekker, M., Mullins, M.C., 1998. Ventral
and lateral regions of the zebrash gastrula, including the neural crest progenitors,
are established by a bmp2b/swirl pathway of genes. Dev. Biol. 199, 93110.
Nichane, M., de Croze, N., Ren, X., Souopgui, J., Monsoro-Burq, A.H., Bellefroid, E.J.,
2008a. Hairy2Id3 interactions play an essential role in Xenopus neural crest
progenitor specication. Dev. Biol. 322, 355367.
Nichane, M., de Croz, N., Ren, X., Souopgui, J., Monsoro-Burq, A.H., Bellefroid, E.J.,
2008b. Hairy2-Id3 interactions play an essential role in Xenopus neural crest
progenitor specication. Dev. Biol. 322, 355367.
Nichane, M., Ren, X., Souopgui, J., Bellefroid, E.J., 2008c. Hairy2 functions through both
DNA-binding and non DNA-binding mechanisms at the neural plate border in
Xenopus. Dev. Biol. 322, 368380.
Nichane, M., Ren, X., Bellefroid, E.J., 2010. Self-regulation of Stat3 activity coordinates
cell-cycle progression and neural crest specication. EMBO J. 29, 5567.
Niehrs, C., Kazanskaya, O., Wu, W., Glinka, A., 2001. Dickkopf1 and the SpemannMangold
head organizer. Int. J. Dev. Biol. 45, 237240.
Nieto, M.A., Sargent, M.G., Wilkinson, D.G., Cooke, J., 1994. Control of cell behavior during
vertebrate development by Slug, a zinc nger gene. Science 264, 835839.
Nikitina, N., Sauka-Spengler, T., Bronner-Fraser, M., 2008. Dissecting early regulatory
relationships in the lamprey neural crest gene network. Proc. Natl. Acad. Sci. U. S.
A. 105, 2008320088.

32

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233

Niss, K., Leutz, A., 1998. Expression of the homeobox gene GBX2 during chicken development.
Mech. Dev. 76, 151155.
O'Donnell, M., Hong, C.S., Huang, X., Delnicki, R.J., Saint-Jeannet, J.P., 2006. Functional
analysis of Sox8 during neural crest development in Xenopus. Development 133,
38173826.
Ohta, K., Lupo, G., Kuriyama, S., Keynes, R., Holt, C.E., Harris, W.A., Tanaka, H., Ohnuma,
S., 2004. Tsukushi functions as an organizer inducer by inhibition of BMP activity in
cooperation with chordin. Dev. Cell 7, 347358.
Patthey, C., Edlund, T., Gunhaga, L., 2009. Wnt-regulated temporal control of BMP
exposure directs the choice between neural plate border and epidermal fate.
Development 136, 7383.
Pera, E., Kessel, M., 1999. Expression of DLX3 in chick embryos. Mech. Dev. 89,
189193.
Perez-Alcala, S., Nieto, M.A., Barbas, J.A., 2004. LSox5 regulates RhoB expression in the
neural tube and promotes generation of the neural crest. Development 131,
44554465.
Pomp, O., Brokhman, I., Ben-Dor, I., Reubinoff, B., Goldstein, R.S., 2005. Generation of
peripheral sensory and sympathetic neurons and neural crest cells from human
embryonic stem cells. Stem Cells 23, 923930.
Pruitt, S.C., Bussman, A., Maslov, A.Y., Natoli, T.A., Heinaman, R., 2004. Hox/Pbx and Brn
binding sites mediate Pax3 expression in vitro and in vivo. Gene Expr. Patterns 4,
671685.
Puelles, L., Fernandez-Garre, P., Sanchez-Arrones, L., Garcia-Calero, E., RodriguezGallardo, L., 2005. Correlation of a chicken stage 4 neural plate fate map with
early gene expression patterns. Brain Res. Brain Res. Rev. 49, 167178.
Rhinn, M., Lun, K., Amores, A., Yan, Y.L., Postlethwait, J.H., Brand, M., 2003. Cloning,
expression and relationship of zebrash gbx1 and gbx2 genes to Fgf signaling.
Mech. Dev. 120, 919936.
Rhinn, M., Lun, K., Luz, M., Werner, M., Brand, M., 2005. Positioning of the midbrainhindbrain boundary organizer through global posteriorization of the neuroectoderm
mediated by Wnt8 signaling. Development 132, 12611272.
Rhinn, M., Lun, K., Ahrendt, R., Geffarth, M., Brand, M., 2009. Zebrash gbx1 renes the
midbrainhindbrain boundary border and mediates the Wnt8 posteriorization
signal. Neural. Dev 4, 12.
Ribes, V., Le Roux, I., Rhinn, M., Schuhbaur, B., Dolle, P., 2009. Early mouse caudal
development relies on crosstalk between retinoic acid, Shh and Fgf signalling pathways.
Development 136, 665676.
Robinson, G.W., Mahon, K.A., 1994. Differential and overlapping expression domains of
Dlx-2 and Dlx-3 suggest distinct roles for Distal-less homeobox genes in craniofacial
development. Mech. Dev. 48, 199215.
Rodriguez-Gallardo, L., Sanchez-Arrones, L., Fernandez-Garre, P., Puelles, L., 2005.
Agreement and disagreement among fate maps of the chick neural plate. Brain
Res. Brain Res. Rev. 49, 191201.
Rogers, C.D., Archer, T.C., Cunningham, D.D., Grammer, T.C., Casey, E.M., 2008. Sox3
expression is maintained by FGF signaling and restricted to the neural plate by
Vent proteins in the Xenopus embryo. Dev. Biol. 313, 307319.
Rufns, S., Artinger, K.B., Bronner-Fraser, M., 1998. Early migrating neural crest cells
can form ventral neural tube derivatives when challenged by transplantation.
Dev. Biol. 203, 295304.
Saint-Germain, N., Lee, Y.H., Zhang, Y., Sargent, T.D., Saint-Jeannet, J.P., 2004. Specication
of the otic placode depends on Sox9 function in Xenopus. Development 131,
17551763.
Saint-Jeannet, J.P., He, X., Varmus, H.E., Dawid, I.B., 1997. Regulation of dorsal fate in the
neuraxis by Wnt-1 and Wnt-3a. Proc. Natl. Acad. Sci. U. S. A. 94, 1371313718.
Sakai, D., Tanaka, Y., Endo, Y., Osumi, N., Okamoto, H., Wakamatsu, Y., 2005. Regulation
of Slug transcription in embryonic ectoderm by beta-catenin-Lef/Tcf and BMPSmad signaling. Dev. Growth Differ. 47, 471482.
Salzberg, A., Elias, S., Nachaliel, N., Bonstein, L., Henig, C., Frank, D., 1999. A Meis family
protein caudalizes neural cell fates in Xenopus. Mech. Dev. 80, 313.
Sanchez-Guardado, L.O., Irimia, M., Sanchez-Arrones, L., Burguera, D., Rodriguez-Gallardo,
L., Garcia-Fernandez, J., Puelles, L., Ferran, J.L., Hidalgo-Sanchez, M., 2011. Distinct and
redundant expression and transcriptional diversity of MEIS gene paralogs during
chicken development. Dev. Dyn. 240, 14751492.
Sasai, N., Mizuseki, K., Sasai, Y., 2001. Requirement of FoxD3-class signaling for neural
crest determination in Xenopus. Development 128, 25252536.
Sato, T., Sasai, N., Sasai, Y., 2005. Neural crest determination by co-activation of Pax3
and Zic1 genes in Xenopus ectoderm. Development 132, 23552363.
Sauka-Spengler, T., Meulemans, D., Jones, M., Bronner-Fraser, M., 2007. Ancient evolutionary origin of the neural crest gene regulatory network. Dev. Cell 13,
405420.
Sawai, S., Kato, K., Wakamatsu, Y., Kondoh, H., 1990. Organization and expression of the
chicken N-myc gene. Mol. Cell. Biol. 10, 20172026.
Schepers, G.E., Bullejos, M., Hosking, B.M., Koopman, P., 2000. Cloning and characterisation of the Sry-related transcription factor gene Sox8. Nucleic Acids Res. 28,
14731480.
Schlosser, G., Ahrens, K., 2004. Molecular anatomy of placode development in Xenopus
laevis. Dev. Biol. 271, 439466.
Schroeder, T.E., 1970. Neurulation in Xenopus laevis. An analysis and model based upon
light and electron microscopy. J. Embryol. Exp. Morphol. 23, 427462.
Selleck, M.A., Bronner-Fraser, M., 1995. Origins of the avian neural crest: the role of
neural plateepidermal interactions. Development 121, 525538.
Serbedzija, G.N., Bronner-Fraser, M., Fraser, S.E., 1989. A vital dye analysis of the timing
and pathways of avian trunk neural crest cell migration. Development 106,
809816.
Serbedzija, G.N., Bronner-Fraser, M., Fraser, S.E., 1994. Developmental potential of
trunk neural crest cells in the mouse. Development 120, 17091718.

Shen, H., Wilke, T., Ashique, A.M., Narvey, M., Zerucha, T., Savino, E., Williams, T., Richman,
J.M., 1997. Chicken transcription factor AP-2: cloning, expression and its role in outgrowth of facial prominences and limb buds. Dev. Biol. 188 (2), 248266.
Shen, X., Kim, W., Fujiwara, Y., Simon, M.D., Liu, Y., Mysliwiec, M.R., Yuan, G.C., Lee, Y.,
Orkin, S.H., 2009. Jumonji modulates polycomb activity and self-renewal versus
differentiation of stem cells. Cell 139, 13031314.
Smith, D.E., Franco del Amo, F., Gridley, T., 1992. Isolation of Sna, a mouse gene homologous to the Drosophila genes snail and escargot: its expression pattern suggests
multiple roles during postimplantation development. Development 116,
10331039.
Steventon, B., Araya, C., Linker, C., Kuriyama, S., Mayor, R., 2009. Differential requirements
of BMP and Wnt signalling during gastrulation and neurulation dene two steps in
neural crest induction. Development 136, 771779.
Streit, A., 2002. Extensive cell movements accompany formation of the otic placode.
Dev. Biol. 249, 237254.
Strobl-Mazzulla, P.H., Sauka-Spengler, T., Bronner-Fraser, M., 2010. Histone demethylase
JmjD2A regulates neural crest specication. Dev. Cell 19, 460468.
Su, M.W., Suzuki, H.R., Solursh, M., Ramirez, F., 1991. Progressively restricted expression of a
new homeobox-containing gene during Xenopus laevis embryogenesis. Development
111, 11791187.
Sunmonu, N.A., Li, K., Guo, Q., Li, J.Y., 2011. Gbx2 and Fgf8 are sequentially required for
formation of the midbrainhindbrain compartment boundary. Development 138,
725734.
Suzuki, H.R., Padanilam, B.J., Vitale, E., Ramirez, F., Solursh, M., 1991. Repeating developmental expression of G-Hox 7, a novel homeobox-containing gene in the chicken.
Dev. Biol. 148, 375388.
Suzuki, A., Ueno, N., Hemmati-Brivanlou, A., 1997. Xenopus msx1 mediates epidermal induction and neural inhibition by BMP4. Development 124,
30373044.
Takemoto, T., Uchikawa, M., Kamachi, Y., Kondoh, H., 2006. Convergence of Wnt and
FGF signals in the genesis of posterior neural plate through activation of the Sox2
enhancer N-1. Development 133, 297306.
Tam, P.P., 1989. Regionalisation of the mouse embryonic ectoderm: allocation of prospective
ectodermal tissues during gastrulation. Development 107, 5567.
Tassabehji, M., Read, A.P., Newton, V.E., Harris, R., Balling, R., Gruss, P., Strachan, T.,
1992. Waardenburg's syndrome patients have mutations in the human homologue
of the Pax-3 paired box gene. Nature 355, 635636.
Terzic, J., Saraga-Babic, M., 1999. Expression pattern of PAX3 and PAX6 genes during
human embryogenesis. Int. J. Dev. Biol. 43, 501508.
Theveneau, E., Mayor, R., 2012. Neural crest delamination and migration: from epitheliumto-mesenchyme transition to collective cell migration. Dev. Biol. 366, 3454 (this issue).
Theveneau, E., Duband, J.L., Altabef, M., 2007. Ets-1 confers cranial features on neural
crest delamination. PLoS One 2, e1142.
Theveneau, E., Marchant, L., Kuriyama, S., Gull, M., Moepps, B., Parsons, M., Mayor, R.,
2010. Collective chemotaxis requires contact-dependent cell polarity. Dev. Cell
19, 3953.
Thomas, S., Thomas, M., Wincker, P., Babarit, C., Xu, P., Speer, M.C., Munnich, A.,
Lyonnet, S., Vekemans, M., Etchevers, H.C., 2008. Human neural crest cells display molecular and phenotypic hallmarks of stem cells. Hum. Mol. Genet. 17,
34113425.
Tour, E., Pillemer, G., Gruenbaum, Y., Fainsod, A., 2001. The two Xenopus Gbx2 genes
exhibit similar, but not identical expression patterns and can affect head formation.
FEBS Lett. 507, 205209.
Tour, E., Pillemer, G., Gruenbaum, Y., Fainsod, A., 2002. Gbx2 interacts with Otx2 and
patterns the anterior-posterior axis during gastrulation in Xenopus. Mech. Dev.
112, 141151.
Tribulo, C., Aybar, M.J., Nguyen, V.H., Mullins, M.C., Mayor, R., 2003. Regulation of Msx
genes by a Bmp gradient is essential for neural crest specication. Development
130, 64416452.
Tucker, J.A., Mintzer, K.A., Mullins, M.C., 2008. The BMP signaling gradient patterns
dorsoventral tissues in a temporally progressive manner along the anteroposterior
axis. Dev. Cell 14, 108119.
Uwanogho, D., Rex, M., Cartwright, E.J., Pearl, G., Healy, C., Scotting, P.J., Sharpe, P.T.,
1995. Embryonic expression of the chicken Sox2, Sox3 and Sox11 genes suggests
an interactive role in neuronal development. Mech. Dev. 49, 2336.
Villanueva, S., Glavic, A., Ruiz, P., Mayor, R., 2002. Posteriorization by FGF, Wnt,
and retinoic acid is required for neural crest induction. Dev. Biol. 241,
289301.
von Bubnoff, A., Schmidt, J.E., Kimelman, D., 1996. The Xenopus laevis homeobox gene
Xgbx-2 is an early marker of anteroposterior patterning in the ectoderm. Mech.
Dev. 54, 149160.
Wang, Y., Fu, Y., Gao, L., Zhu, G., Liang, J., Gao, C., Huang, B., Fenger, U., Niehrs, C., Chen,
Y.G., et al., 2010. Xenopus skip modulates Wnt/beta-catenin signaling and functions
in neural crest induction. J. Biol. Chem. 285, 1089010901.
Warner, S.J., Hutson, M.R., Oh, S.H., Gerlach-Bank, L.M., Lomax, M.I., Barald, K.F., 2003.
Expression of ZIC genes in the development of the chick inner ear and nervous
system. Dev. Dyn. 226, 702712.
Wei, S., Xu, G., Bridges, L.C., Williams, P., White, J.M., DeSimone, D.W., 2010. ADAM13
induces cranial neural crest by cleaving class B Ephrins and regulating Wnt signaling.
Dev. Cell 19, 345352.
Williams, R., Lendahl, U., Lardelli, M., 1995. Complementary and combinatorial patterns of
Notch gene family expression during early mouse development. Mech. Dev. 53,
357368.
Wilson, S.I., Graziano, E., Harland, R., Jessell, T.M., Edlund, T., 2000. An early requirement
for FGF signalling in the acquisition of neural cell fate in the chick embryo. Curr.
Biol. 10, 421429.

C. Milet, A.H. Monsoro-Burq / Developmental Biology 366 (2012) 2233


Wu, M.Y., Ramel, M.C., Howell, M., Hill, C.S., 2011. SNW1 is a critical regulator of spatial
BMP activity, neural plate border formation, and neural crest specication in
vertebrate embryos. PLoS Biol. 9, e1000593.
Yang, L., Zhang, H., Hu, G., Wang, H., Abate-Shen, C., Shen, M.M., 1998. An early phase of
embryonic Dlx5 expression denes the rostral boundary of the neural plate. J.
Neurosci. 18, 83228330.

33

Zakin, L., Reversade, B., Virlon, B., Rusniok, C., Glaser, P., Elalouf, J.M., Brulet, P., 2000. Gene
expression proles in normal and Otx2/ early gastrulating mouse embryos. Proc.
Natl. Acad. Sci. U. S. A. 97, 1438814393.
Zhao, H., Tanegashima, K., Ro, H., Dawid, I.B., 2008. Lrig3 regulates neural crest
formation in Xenopus by modulating Fgf and Wnt signaling pathways. Development 135, 12831293.

Anda mungkin juga menyukai