avances en desarrollo craneofacial chai 2006

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

DEVELOPMENTAL DYNAMICS 235:2353–2375, 2006

SPECIAL ISSUE REVIEWS–A PEER REVIEWED FORUM

Recent Advances in Craniofacial


Morphogenesis
Yang Chai1* and Robert E. Maxson, Jr2

Craniofacial malformations are involved in three fourths of all congenital birth defects in humans, affecting
the development of head, face, or neck. Tremendous progress in the study of craniofacial development has
been made that places this field at the forefront of biomedical research. A concerted effort among
evolutionary and developmental biologists, human geneticists, and tissue engineers has revealed important
information on the molecular mechanisms that are crucial for the patterning and formation of craniofacial
structures. Here, we highlight recent advances in our understanding of evo– devo as it relates to
craniofacial morphogenesis, fate determination of cranial neural crest cells, and specific signaling
pathways in regulating tissue–tissue interactions during patterning of craniofacial apparatus and the
morphogenesis of tooth, mandible, and palate. Together, these findings will be beneficial for the
understanding, treatment, and prevention of human congenital malformations and establish the foundation
for craniofacial tissue regeneration. Developmental Dynamics 235:2353–2375, 2006. © 2006 Wiley-Liss, Inc.

Key words: cranial neural crest (CNC) cell; ectoderm; endoderm; evolution; mesoderm; mandible; palate; tooth
development

Accepted 30 March 2006

INTRODUCTION cranial neural crest (CNC) cells. The CNC cells and result in craniofacial
specification, emigration and migra- malformations. Significant progress
Development of the craniofacial re-
tion, proliferation, survival, and ulti- has been made in recent years toward
gion is a complex process with many
mate fate determination of the CNC the understanding of how this impor-
features that reflect strong evolution-
ary forces controlling morphology. The play an important role in regulating tant population of pluripotent cells is
vertebrate craniofacial region houses craniofacial development. Unlike the initially established in the early em-
and protects the brain and provides trunk neural crest, CNC cells give rise bryo and of the molecular mechanisms
the scaffold on which the sensory and to an array of cell types during embry- that mediate neural crest cell lineage
feeding organs are located. The ability onic development. For example, CNC segregation, differentiation, and final
to sense and devour prey is fundamen- cells form most of the hard tissues of contribution to a particular tissue
tal to animal survival. Variations in the head such as bone, cartilage, and type (Shah et al., 1996; LaBonne and
craniofacial anatomy and function teeth, whereas hard tissues in the rest Bronner-Fraser, 1999; Chai et al.,
provide the major driving force in evo- of the body are formed from mesoderm 2003; Le Douarin et al., 2004).
lutionary adaptation. cells. Genetic disorders, environmen- The tissues of the head are com-
One of the key features of craniofa- tal insults, or the combination of both posed of cells from all three germ layer
cial development is the formation of can alter the fate determination of origins: ectodermal, endodermal, and

1
Center for Craniofacial Molecular Biology School of Dentistry University of Southern California, Los Angeles, California
2
Department of Biochemistry and Molecular Biology, USC/Norris Comprehensive Cancer Center and Hospital, Keck School of Medicine,
University of Southern California, Los Angeles, California
Grant sponsor: National Institute of Dental and Craniofacial Research; Grant sponsor: NIH; Grant numbers: DE 014078; DE012711;
DE017007; DE12941; DE12450; Grant sponsor: March of Dimes Birth Defects Foundation.
*Correspondence to: Yang Chai, Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar
Street, CSA 103, Los Angeles, CA 90033. E-mail: ychai@usc.edu
DOI 10.1002/dvdy.20833
Published online 5 May 2006 in Wiley InterScience (www.interscience.wiley.com).

© 2006 Wiley-Liss, Inc.


2354 CHAI AND MAXSON

mesenchymal. As seen in the develop- well as branchial arches. More re- face, skull, teeth, and jaw. How this
ment of many organs, craniofacial cently, many animal models with trajectory of inductive interactions oc-
morphogenesis depends upon continu- specific craniofacial malformations curs in molecular terms is a key ques-
ous and reciprocal tissue–tissue inter- have facilitated human genetic link- tion in developmental biology; how it
actions, with tooth, palate, and mandi- age analysis, in which a genetic de- has been modified during evolution to
ble development as classic examples. fect has been identified as the cause produce the stunning variety of
Research on the development of the of congenital malformation(s) (Thya- craniofacial structures in vertebrates
head requires a thorough understand- garajan et al., 2003; Murray and is of major interest in evolutionary bi-
ing of normal morphology, cell move- Schutte, 2004). Overall, genetically ology.
ment, cell signaling, gene/gene interac- mutated mouse models highlight the We begin our review with a brief
tion, and transcriptional regulation in enormous challenges of uncovering consideration of some current issues
time and space. complex genetic mechanisms under- concerning the evolution of the head
Investigation of craniofacial devel- lying craniofacial development and (for comprehensive treatments, see
opment uses different animal species malformations. Santagati and Rijli, 2003; Kuratani,
as models as with other areas of re- Progress and understanding of the 2004, 2005; Northcutt, 2005; Morriss-
search in developmental biology. key control processes of head develop- Kay and Wilkie, 2005; Depew et al.,
Studies in mice combine the power of ment have advanced to an extent 2005). We focus on recent findings
genetics and genome manipulation where developmental biologists are that pertain to two broad questions:
together with in vitro organ culture interacting with tissue engineers to First, how did the head evolve in the
techniques, leading to great progress devise cell-based approaches to treat chordate ancestor of vertebrates? Sec-
in recent years. Avian embryos clinical malformations in humans. ond, how, over shorter evolutionary
(chicken and quail) are easily acces- The prospect of harnessing develop- periods, did modifications of the head
sible and are used for grafting/ mental processes to repair or replace developmental program produce the
transplantation experiments. Ze- damaged or diseased craniofacial tis- evolutionary novelties that made pos-
brafish craniofacial developmental sues and organs is an exciting new sible the huge array of craniofacial
studies have emerged more recently, area that links basic animal research morphologies evident in vertebrates?
and mutant screens have led to the with human genetics and provides Although the details of how the
identification of new cell signaling great promise in our effort to reduce head first evolved are as elusive as
interactions (Trainor and Krumlauf, the pain and suffering associated with fossils of the early chordates that once
2000; Yelick and Schilling, 2002). craniofacial malformations. lived in pre-Cambrian seas, there is
Central to these models and tech- In this review, we highlight some broad agreement with a scenario put
niques lies morphology. The head is recent advances in our understanding forward by Northcutt and Gans (1983)
a very complex structure, and a de- of evo– devo (evolutionary– develop- and modified recently by Northcutt
tailed analysis and appreciation of ment) as it relates to craniofacial de- (2005). Their New Head hypothesis
morphology are essential to under- velopment, the fate determination of postulates that the ancestral verte-
standing the mechanism of craniofacial cranial neural crest cells, craniofacial brate was an animal similar to the
malformations. Ultimately, “molecular patterning and organogenesis. We modern day cephalochordate Am-
morphology,” the combination of classic also review new discoveries on the de- phioxus. Like Amphioxus, this animal
morphology and molecular biology, pro- velopment of craniofacial bones, sig- was a filter feeder that lacked pharyn-
vides the basis for understanding the naling interactions, and specificity in geal arch muscles to move water
evolutionary changes in head structure craniofacial morphogenesis. Finally, through the gills. Also lacking was a
and formation that in turn helps to clar- we will discuss how research advance- braincase and the structures charac-
ify key developmental principals as well ments will be beneficial for the under- teristic of the rostral head of verte-
as the mechanism of craniofacial mal- standing, treatment, and prevention brates, including olfactory bulbs and
formations. of human congenital malformations telencephalic vesicles.
Mouse models are extremely valu- and prospectus of tissue engineering. According to the New Head hypoth-
able in our effort to gain a better esis, this stem organism underwent
understanding of human craniofa- an evolutionary transition from filter
cial birth defects. The remarkable
EVOLUTION feeding to active predation (Fig. 1).
progress being made in the human Perhaps no other anatomical feature Underlying this transition were sev-
genome, in parallel with exquisite more closely epitomizes vertebrates eral key innovations, including the de-
functional genomic investigations in than the head. Comprising paired sen- velopment of muscularized jaws and
various mouse models, has resulted sory elements, a muscularized masti- gill arches, a nerve plexus that en-
in the discovery of morphoregulatory catory apparatus, and a cartilaginous abled the animal to detect and capture
genes that determine craniofacial or bony braincase, the head develops prey, and cartilaginous and skeletal
morphogenesis. For example, we from complex and massive movements elements that provided a fixed spatial
have uncovered genes that are criti- of mesenchymal cells derived from organization for this plexus. In princi-
cal for determining cranial– caudal mesoderm and neural crest. These ple, these new structures could be pro-
axis, dorsal–ventral patterning, cells interact with each other and with duced in two different ways: the ante-
left–right symmetry and segmenta- a variety of craniofacial epithelia to rior portion of the trunk could be
tion in early forming neurulation as produce the intricate structures of the restructured, or a section could be
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2355

added onto the trunk de novo. The early vertebrate evolution took on a upper and lower lips with distinct
latter possibility—that the head is a function in the early neural crest. morphologies. These authors exam-
neomorph—is a central proposal of On the other hand, recent work ined the expression of Dlx and Msx
the New Head hypothesis. Also key to from Jeffery’s group has shown that a homeobox genes, which, in amniotes,
this hypothesis is the idea that the cell urochordate (ascidian) possesses cells are expressed in neural crest-derived
type most critical for the evolution of that exhibit neural crest-like behavior mesenchymal cells along the future
the new head was the neural crest. (Jeffery et al., 2004). These cells mi- proximal– distal axis of the mandibu-
The neural crest is induced by an in- grate from the neural tube and ex- lar arch. Additional markers with re-
teraction between cells of the neural press the neural crest markers hnk gion-specific expression included Fgf8
plate and adjacent surface ectoderm and Zic1. Ultimately they give rise to and Bmp4.
(reviewed by Meulemans and Bronner- pigment cells, leading Jeffery and co- The expression patterns of these
Fraser, 2004). Nascent neural crest workers to propose that the neural genes in lamprey embryos appeared
cells delaminate from the developing crest first arose as pigment cell pre- at first to support the view that lam-
neural tube, take on a mesenchymal cursors in a common ancestor of ver- prey larval lips and gnathostome jaws
character and migrate to distant sites tebrates and ascidians, and later ac- are homologous. However, DiI label-
in the developing embryo. In the cranio- quired additional fates. ing of neural crest populations sug-
facial region, they give rise to a wide Whether such neural crest-like cells gested a more complex picture. Neural
variety of cell types and tissues, includ- exist in other nonvertebrate chordate crest cells contributing to the upper
ing intramembranous bone, cartilage, groups remains to be determined. Re- and lower lips are derived from fore-
muscle, and nerves (Creuzet et al., sults to date show that, although cells brain and midbrain crest populations,
2005; Noden and Trainor, 2005). North- with neural crest-like properties are unlike the situation in gnathostomes
cutt and Gans (1983) proposed that the present in some nonvertebrates, cells in which mandibular arch crest de-
neural crest emerged in an early verte- possessing the full spectrum of neural rives from more posterior populations.
brate ancestor and served as a key crest behavior are unique to verte- The lamprey Dlx1 homologue is ex-
source of evolutionary novelty that brates. Thus, the idea that the neural pressed in different regions of the neu-
made the New Head possible. They crest is a major source of New Head ral crest compared with gnathos-
based this view on the apparent unique- structures remains intact (Northcutt, tomes. This shift in expression is
ness of neural crest to vertebrates and 2005). correlated with a shift in expression of
on the ability of neural crest both to Fgf8, an upstream regulator of Dlx1 in
contribute to structures that form the Evolutionary Novelty in the both lamprey and gnathostomes, in
new head as well as to serve as a source the epidermis. This shift is seen as the
of patterning information (Noden, 1978;
Craniofacial Complex cause of a corresponding shift in the
Couly et al., 1993; Jiang et al., 2002; The first vertebrates exhibiting a fate of subpopulations of neural crest.
Schneider and Helms, 2003). “New Head” were probably similar to An evolutionary change such as this
That the fate of the neural crest modern day jawless fishes (ag- one in a jawless gnathostome ancestor
includes elements of the rostral most nathans; Fig. 1). These organisms, could have led to the emergence of the
portion of the new head and that the which include lampreys and hagfish, jaw. If this scenario is correct, then
neural crest has a key role in pat- are similar to higher vertebrates in the gnathostome jaw is an evolution-
terning these tissues are clear their embryology, but lack jaws, pos- ary innovation resulting from a
(Couly et al., 1993; Jiang et al., 2002; sessing instead a filter-feeding appa- change in the topographic location of
Gross and Hanken, 2005; Evans and ratus (Kuratani et al., 2001). A major an epithelial–mesenchymal interac-
Noden, 2006). What is less clear is question is what were the sources of tion. This change resulted in a shift in
whether the neural crest is unique to evolutionary novelty that made possi- fate of neural crest cells and, thus, to
vertebrates. Efforts to identify neu- ble changes that led to more complex the innovation that became the verte-
ral crest-like cell populations in ex- craniofacial morphologies of later ver- brate jaw.
tant nonvertebrate chordates have tebrate groups? One approach to this Recent work from the Tabin and
produced mixed results. Yu et al. problem has been to carry out compar- Chuong labs has identified potential
(2002) found that amphioxus has ative embryological studies on the sources of evolutionary novelty in
cells that express an amphioxus ho- lamprey. the epithelial–mesenchymal interac-
mologue of the crest marker FoxD3. Kuratani and colleagues have ex- tions that pattern the avian beak
AmphiFoxD is expressed in the an- amined marker gene expression in the (Abzhanov et al., 2004; Wu et al.,
terior neural plate but not in cells Japanese lamprey, comparing expres- 2004). This work suggests that evo-
bordering the neural plate, as is the sion patterns with those seen in gna- lutionary changes in beak morphol-
case for FoxD3 in vertebrates. Am- thostomes (Shigetani et al., 2002). A ogy in birds may be caused by simi-
phiFoxD is also expressed in axial generally accepted view of jaw evolu- lar shifts in epithelial–mesenchymal
and paraxial mesoderm. Yu and co- tion is that the mandibular arch, the signaling. Darwin’s finches, in the
workers speculate that an amphi- most rostral of the pharyngeal arches, course of their radiation into various
FoxD congener in a common ances- was modified to produce the mandible ecological niches in the Galapagos,
tor of vertebrates and amphioxus and maxilla. In ancient vertebrates, evolved diverse beak morphologies.
had a role in the development of the the arches were morphologically sim- By examining the expression of var-
mesoderm, and subsequently, during ilar. The lamprey lacks a jaw but has ious growth factors among Galapa-
2356 CHAI AND MAXSON

Fig. 1. The New Head and the evolution of the jaw. Schematics of the oral region of Amphioxus, a
lamprey (jawless vertebrate), and a gnathostome (jawed vertebrate) are shown with their phylogenetic
relationships (redrawn from Shigetani et al., 2002, and Holland et al., 2004). As proposed by Shigetani
et al. (2002), the neural crest-derived mesenchyme of lampreys and gnathostomes is subdivided into
postoptic (po) and mandibular arch (ma) subdomains. Growth factors are secreted by the epidermis (red
line). In the lamprey, these induce homeobox genes (brown and blue) over the entire length of the neural
crest-derived mesenchyme, whereas in the gnathostome, such signals are effective only in the man-
dibular component. Thus, it is proposed that a shift in epithelial–mesenchymal interactions results in the
evolutionary emergence of the jaw. Ulp, upper lip; llp, lower lip; prc, prechordal cranium.

Fig. 2. Neural crest boundary relationships in


skull vaults of mouse, chicken, and frog. Skulls
are shown in the dorsal aspect. In the mouse,
the coronal suture (CS) marks a neural crest–
mesoderm boundary (blue line; neural crest an-
terior) (Jiang et al., 2002). Note also that the
frontal (F) and interparietal (IP) bones are neural
crest-derived (blue). In the chicken, the neural
crest–mesoderm boundary lies within the fron-
tal bone (F) and does not coincide with a suture
(Noden, 1975; Le Lievre, 1978; Noden and
Trainor, 2005; Evans and Noden, 2006). The
solid blue frontoparietal (FP) bone of the frog,
Xenopus, indicates that neural crest contributes
over its entire length (Gross and Hanken, 2005).
The extent to which mesoderm contributes to
this bone is not known. See Figure 3 for a
detailed depiction of neural crest- and meso-
derm-derived bones of the skull vault. P, pari-
etal bone.

gos finches, Tabin’s group found that


Fig. 3. Contribution of ectoderm, mesoderm, and endoderm during craniofacial development. the expression of Bmp4 in the mes-
A: Neural crest cells are formed at the junction of neural and surface ectoderm. These cells undergo
enchyme of the upper beak was
epithelial–mesenchymal transformation, become ectomesenchyme, and travel into multiple destina-
tions. B: Side view of an E9.5 mouse embryo shows unsegmented paraxial mesoderm in the head and closely associated with a particular
mesoderm-derived somites in the trunk. OP, optic vesicle; OV, otic vesicle. C: Transverse section of the morphology, a broad shape (Abzh-
developing first branchial arch that is covered by surface ectoderm. The core of the first arch contains anov et al., 2004). By misexpressing
cranial neural crest (CNC) -derived (blue) and paraxial mesoderm-derived (pink) cells. The pharyngeal bone morphogenetic protein-4
endoderm (yellow) lines the inner aspect of the branchial arch. D: Schematic drawing of an adult mouse
skull shows both the CNC- and paraxial mesoderm-derived elements (modified from Noden and
Trainor, 2005). Mesoderm-derived cells are in pink, and CNC-derived cells are in blue.
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2357

(Bmp4) in chick embryos, a similar Tissue Boundaries in (2005), the situation may not be sub-
broad shape could be produced. Cranial Evolution stantially different from the mouse.
The Chuong group (Wu et al., On the other hand, if, in the chicken,
2004) addressed a related question The finding that the neural crest can crest indeed contributes to the entire
by examining the basis of morpho- provide patterning information sug- skull vault, as it apparently does in
gests that evolutionary shifts in neu- Xenopus, then there may have been an
logical differences between the
ral crest–non-neural crest boundaries evolutionary shift in the neural crest–
shapes of chicken and duck beaks.
could be an additional source of nov- mesoderm boundary in the ancestor of
Ducks have two growth zones in the
elty (Fig. 2). Recent studies have ad- birds and mammals (Gross and Han-
frontonasal mass, whereas chickens
dressed the significance of boundaries ken, 2005).
have only one. This difference ac-
in the development and evolution of It is also possible that, as pointed
counts for the difference in beak
the skull vault (Jiang et al., 2002; out by Gross and Hanken (2005), am-
morphology between these two birds. Merrill et al., 2006; Evans and Noden, phibians, birds, and mammals may
Bmp4 is expressed in the growth 2006). each exhibit distinct contributions of
zones. By manipulating the location As discussed above, the skull vault neural crest to the skull vault. We
of Bmp4 expression, Chuong’s group develops from populations of mesen- note as well that there remains some
was able to modulate the shape of chymal cells with distinct embryolog- uncertainty about the homology rela-
the beak. Together, these studies on ical origins, neural crest, and head tionships between the frontal, pari-
beak development demonstrate that mesoderm. The extent of the contribu- etal, and frontoparietal bones of these
subtle changes in the topography of tion of neural crest to calvarial bones three taxa—an uncertainty that clearly
Bmp signaling within the developing of birds has been controversial. Sev- complicates the interpretation of neural
beak can account for dramatic evolu- eral studies using quail– chick chime- crest labeling studies.
tionary shifts in beak morphology. ras or retroviral infection to produce In the mouse, the coronal suture is
Schneider and Helms (2003) ad- fate maps (Noden, 1975; Le Lievre, located at the neural crest–mesoderm
dressed the role of the neural crest in 1978; Evans and Noden, 2006) sug- interface and is a major growth center
beak patterning and the evolution of gest that the neural crest–non-neural of the skull vault. Boundaries between
beak shape. These authors exchanged crest boundary lies within the frontal dissimilar cell populations often serve
neural crest cells fated to contributed bone (reviewed in Noden and Trainor, as important signaling centers that
to the beak between quail and chicks, 2005). The frontal bones develop after function in growth control (reviewed
which exhibit distinct beak morpholo- the fusion of two intramembranous in Dahmann and Basler, 1999). The
gies. They sought to test the tripartite centers (Jollie, 1981; discussed in No- location of the tissue boundary at the
hypothesis that neural crest cells con- den and Trainor, 2005). The more ros- coronal suture in mammals might rep-
tain patterning information for beak tral of these is of neural crest origin, resent an evolutionary innovation
morphology, that such cells possess an the caudal of mesoderm origin. Thus, that makes possible, for example, new
autonomous program by which they the boundary lies at the interface of modes of mastication, locomotion, or
express this information, and that these two ossification centers. Later cranial growth. Further analysis of
such cells influence the fates of non- studies, however, have suggested that contributions of neural crest to the
neural crest cells. the entire chick cranial vault, includ- skull vault in extant vertebrates will
Reciprocal transplantation experi- ing the frontal and parietal bones, is of be required to illuminate this issue.
neural crest origin (Couly et al., 1993).
ments confirmed each of these points.
Similarly, in Xenopus, dye-marking FATE DETERMINATION OF
Thus, for example, quail neural crest
experiments have shown that crest
cells, when transplanted into duck CRANIAL NEURAL CREST
contributes to the full length of the
embryos gave rise to beaks like those CELLS
frontoparietal bone, the major bone of
found in quail. That donor neural
the skull vault in anuran amphibians The vertebrate neural crest is a pluri-
crest cells pattern host structures was
(Gross and Hanken, 2005). In mouse, potent cell population derived from
shown by an examination of egg tooth
in contrast, the frontal bone appears the lateral ridges of the neural plate
morphology. Duck and quail egg teeth, to be derived entirely from neural during early stages of embryogenesis
which are derived from epidermis, not crest, whereas the parietal bone is de- (Fig. 3). The functions of neural crest
neural crest, exhibit distinct morphol- rived from mesoderm (Jiang et al., cells include coordination of various
ogies. The results showed that the mor- 2002; Ishii et al., 2003). Thus, in the visceral activities, such as in the pe-
phology was characteristic of the trans- mouse, the interface between the fron- ripheral nervous system; protection of
planted tissue. Transplanted neural tal and parietal bones—the coronal the body from external conditions,
crest cells express molecular markers suture—is a boundary between neural such as by melanocytes; and partici-
characteristic of the donor species, con- crest and mesoderm (Merrill et al., pation in craniofacial skeletal devel-
firming the idea that such cells express 2006). opment (Le Douarin et al., 2004).
an autonomous molecular program. It If we make the assumption that the The formation of the neural crest is
is not hard to envisage that evolution- boundary in the chick is within the a classic example of embryonic induc-
ary shifts in neural crest populations frontal bone (Noden, 1975; Le Lievre, tion, in which tissue–tissue interac-
could produce profound morphological 1978; Evans and Noden, 2006), then, tions and the concerted action of sig-
changes. as pointed out by Noden and Trainor naling pathways are critical for the
2358 CHAI AND MAXSON

induction of neural crest precursor genitor cells must be instructed by sig- has a critical role in regulating the
cells. After induction, neural crest nals from other tissues to generate skel- fate of CNC. Discovery of the critical
cells disperse from the dorsal surface etal elements of appropriate shape and signaling pathway involved in this
of the neural tube, undergo epithelial– size in the craniofacial region. Tissues regulatory process awaits the develop-
mesenchymal transformation, and mi- that provide the instructive signaling ment of mouse pharyngeal endoderm-
grate extensively through the embryo, for CNC fate specification include, but specific gene inactivation models.
giving rise to a wide variety of differ- are not limited to, the pharyngeal
entiated cell types. The migration, endoderm, the branchial arch ectoderm,
proliferation, and differentiation of and the isthmic organizer at the mid- Orofacial Ectoderm
neural crest cells along multiple dis- brain– hindbrain boundary (Baker and
tinctive pathways have been studied Bronner-Fraser, 2001; Trainor et al., Early patterning of the oral ectoderm is
extensively in various animal models 2002; Couly et al., 2002; Le Douarin et independent of the neural crest (Veitch
(Bronner-Fraser, 1993; LaBonne and al., 2004). et al., 1999). In the mouse model, the
Bronner-Fraser, 1999; Le Douarin et branchial arch epithelium is correctly
al., 2004; Basch et al., 2004). Recent patterned, despite the CNC migration
studies suggest that fate determina- The Pharyngeal Endoderm
defect (Gavalas et al., 2001). After the
tion of the neural crest is strongly in- Most of our knowledge about the interaction with pharyngeal endoderm,
fluenced by environmental cues. CNC critical function of pharyngeal the orofacial ectoderm is roughly di-
cells interact with and are consequently endoderm in regulating the fate of vided into proximal and distal domains.
instructed by pharyngeal endoderm, ec- CNC cells and the patterning of mid- The establishment of the ectodermal
toderm, and mesoderm before giving dle and lower face derives from stud- domain greatly influences the fate de-
rise to various types of tissues in the ies using chick, quail, or zebrafish termination of migrating CNC cells as
craniofacial region. embryos as models. These embryos they populate the branchial arch. At
During craniofacial development, allow relatively easy manipulation later stages of embryonic develop-
neural crest cells migrate ventrolater- compared with the mouse model. ment, CNC cells interact with and
ally as they populate the craniofacial Surgical removal of pharyngeal provide instructive signals to the over-
region. The proliferative activity of endoderm during early stages of lying epithelium and have an effect on
these crest cells produces the fronto- chick embryogenesis resulted in de- epithelial patterning (Miletich and
nasal process and the discrete swell- fects in facial bone and cartilage devel- Sharpe, 2004). Tooth development is a
ings that demarcate each branchial opment (Couly et al., 2002). During the clear example of the consistent shift of
arch. As these ectodermally derived development of the first branchial arch, instructive signal between orofacial
cells migrate, they contribute exten- pharyngeal endoderm is thought to ectoderm and the CNC-derived mes-
sively to the formation of mesenchy- prepattern the orofacial epithelium, enchyme (see below for detailed dis-
mal structures in the head and neck. which in turn will provide instructive cussion).
Cell labeling studies have demon- signals to pattern the CNC-derived
strated that neural crest cells arising mesenchyme (Haworth et al., 2004).
from rhombomeres 1–3 (r1–3) of ante- In zebrafish studies, fibroblast growth Concerted Action of Growth
rior hindbrain migrate into the first factor (FGF) signaling has been shown
and Transcription Factors in
branchial arch and, thereafter, reside to be critical for the development of
within the maxillary and mandibular pharyngeal endoderm itself and for Determining the Fate,
prominences (Osumi-Yamashita et the mediation of the endoderm to reg- Expansion, and Survival of
al., 1990; Serbedzija et al., 1992; Bron- ulate facial skeletal morphogenesis CNC Cells
ner-Fraser, 1993; Selleck et al., 1993; (Ruhin et al., 2003; Crump et al.,
Lumsden and Krumlauf, 1996). The 2004; Helms et al., 2005). On the other Recent studies have addressed the in-
migration of these rhombencephalic hand, a recent study has shown that fluence of growth factors on the fate of
crest cells is regulated by growth fac- pharyngeal endoderm is not critical multipotent progenitor cells, such as
tor signaling pathways and their for the normal development of the up- the neural crest, during embryogene-
downstream transcription factors be- per and lower face. Instead, it is the sis. It turns out that signaling centers,
fore the CNC cells become committed ectoderm that is critical for providing such as the isthmic organizer, rely on
to several different tissue types such the instructive information for facial growth factor signaling in regulating
as bone, cartilage, tooth, and cranial morphogenesis (Aoki et al., 2002). the fate of CNC cells. Specifically,
nerve ganglia (Noden, 1983, 1991; Furthermore, CNC cells also contain FGF8 signaling from the isthmic orga-
Lumsden, 1988; Graham and Lums- intrinsic information that can affect nizer can alter Hoxa2 expression and
den, 1993; Le Douarin et al., 1993; facial development, although this con- consequently control branchial arch
Echelard et al., 1994; Imai et al., 1996; tribution may depend upon the collec- patterning, demonstrating that neu-
Trainor and Krumlauf, 2000). Recent tive number of neural crest cells ral crest cells are patterned by envi-
studies provide strong supportive evi- present at a given time and position ronmental signals (Trainor et al.,
dence that the CNC cells are develop- (Schneider and Helms, 2003; Tucker 2002). Several growth factor signaling
mentally “plastic,” i.e., their fate is not and Lumsden, 2004). Overall, there is pathways have been shown to be crit-
predetermined before they reach their overwhelming evidence to support the ical for CNC fate determination, of
final destination; rather, these pro- notion that the pharyngeal endoderm which transforming growth factor-be-
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2359

ta/BMP (TGF-␤/BMP) signaling is a form dental and palatal mesenchyme, gratory neural crest cells. Noden’s ex-
classic example. Members of the respectively, suggesting an epistatic re- periment suggested that chick CNC
TGF-␤ superfamily of growth factors lationship between these two genes cells are predetermined according to
are expressed at sites where neural (Ferguson, 1994; Ito et al., 2003). In the their rostrocaudal origin in the neural
crest cells commit to form particular palatal mesenchyme, overexpression of tube (Noden, 1983). However, recent
cell types. TGF-␤ superfamily mem- TGF-␤ suppresses transcriptional activ- work has demonstrated that adjacent
bers promote alternative fates for ity of the Msx1 gene (Nugent and tissues, such as the isthmus, provide
trunk neural crest cells; BMP signal- Greene, 1998). We have shown recently instructive signals to regulate down-
ing promotes neurogenesis by induc- that compromised TGF-␤ signaling af- stream target genes to determine the
ing MASH1 expression, whereas fects the expression of Msx1, which in fate of CNC cells (Trainor et al., 2002).
TGF-␤ signaling favors smooth mus- turn controls the progression of the Furthermore, by providing critical
cle differentiation (Shah et al., 1996). CNC cell cycle by regulating cyclin D1 feedback to the surface ectoderm,
In contrast, CNC cells react to TGF-␤ expression (Ito et al., 2003). In parallel, CNC can provide species-specific pat-
signaling differently. TGF-␤ controls in vitro studies also suggest that Msx1 terning information during craniofa-
the differentiation of CNC cells to glial gene expression maintains cyclin D1 ex- cial development, highlighting the im-
cells, and TGF-␤, BMP, and Wnt to- pression and holds cells in an undiffer- portance of tissue–tissue interaction
gether control chondrocyte differenti- entiated state by promoting prolifera- in regulating organogenesis (Schnei-
ation. Apparently, one of the major tion (Hu et al., 2001). In general, TGF-␤ der and Helms, 2003; Helms et al.,
differences between CNC and TNC is signaling specificity in regulating down- 2005). In the mouse model, it has been
the expression of Hox genes, which may stream target gene expression is deter- demonstrated elegantly that CNC
account for the differential responsive- mined by the interaction with other fac- cells retain a remarkable degree of
ness between CNC and TNC to the tors (such as BMP, Wnt, FGF, and so plasticity, even after their migration
same environmental cues (Abzhanov et on) in a temporal- and spatial-specific into the branchial arch (Zhao et al.,
al., 2003). Furthermore, the function of manner (Massague, 2000). The close in- 2006). The second arch CNC cells
TGF-␤ in regulating neural crest cell tertwining of TGF-␤ signaling with have a cell-autonomous requirement
differentiation is sensitive to the TGF-␤ other pathways appears to be a critical for the Hoxa2 gene for their intrinsic
expression level, such that TGF-␤ may component of specific cell fate determi- patterning program (Santagati et al.,
promote alternative cell fates or induce nation mediated by TGF-␤ family mem- 2005). In vitro studies have also begun
apoptosis (Hagedorn et al., 2000). Dur- bers. to address the plasticity of postmigra-
ing early mouse craniofacial develop- In addition to regulating the fate of tory CNC cells (Zhao et al., 2006).
ment, TGF-␤ subtypes are present in CNC cells, the concerted action of Taken together, it is clear that the
the CNC-derived mesenchyme during growth and transcription factors also fate of mouse postmigratory CNC cells
critical epithelial–mesenchymal inter- controls cell proliferation and death. is determined through the reciprocal
actions related to the formation of var- For example, BMP signaling controls signaling between neural crest mesen-
ious organs (Lumsden, 1984; Hall et al., Msx gene expression by directly regu- chyme and the surrounding environ-
1992; Chai et al., 1994; Lumsden and lating Msx2 promoter activity (Brug- ment, where timing is an essential
Krumlauf, 1996; Chai et al., 2003). Tar- ger et al., 2004). There is also an ap- component.
geted null mutation of Tgfb2 or haplo- parent feedback for BMP signaling by
insufficiency of Smad2 results in a wide the Msx genes, because loss of Msx1 CRANIOFACIAL
range of developmental defects, includ- and Msx2 results in altered Bmp4 ex- PATTERNING AND TISSUE–
ing craniofacial malformations such as pression in the CNC cells (Ishii et al.,
small mandible, dysmorphic calvaria, 2005). Msx1 and Msx2 function redun-
TISSUE INTERACTION
and cleft palate (Sanford et al., 1997; dantly in regulating the survival and The principal goal of developmental bi-
Nomura and Li, 1998). Significantly, proliferation of CNC cells during ology is to understand how tissues are
many affected tissues have neural craniofacial development. This regu- induced and patterned to generate dif-
crest-derived components and simulate lation is most likely carried out ferent organs at the correct location and
neural crest deficiencies; thus, TGF-␤ through the control of cell cycle pro- time. Evidence suggests that signals
signaling may provide significant in- gression (Hu et al., 2001; Han et al., from the anterior visceral endoderm,
structive information to specify the fate 2003, Ishii et al., 2005). Recently, anterior neural ridge, and the meso-
of CNC during early craniofacial devel- studies in Xenopus have shown that a derm are required for the development
opment. Myc-mediated Id3 expression level is of head structures (Spemann, 1938;
Members of the TGF-␤ superfamily critical for determining whether the Shimamura and Rubenstein, 1997;
regulate the expression of transcription neural crest cells will proliferate or Beddington and Robertson, 1999; De-
factors to influence cell fate decisions die, thus determining the size of the pew et al., 2002a). Furthermore, the for-
instructively during embryogenesis neural crest population (Kee and mation of vertebrate “New Head” de-
(Shah et al., 1996; Dorsky et al., 2000). Bronner-Fraser, 2005; Light et al., pends upon the presence of CNC cells
For example, the expression patterns of 2005). It is currently unknown and additional sensory placodes. Multi-
Tgfb2 and transcription factor Msx1 whether Id3 plays a similar role dur- ple molecules have been identified as
have significant overlaps during early ing mouse development. critical regulators at these signaling
tooth and palate development when Finally, it is important to address centers. However, the challenge re-
CNC-derived cells become specified to the issue of prepatterning of premi- mains to determine how various signal-
2360 CHAI AND MAXSON

ing centers coordinate with each other ectoderm, FGF8 and BMP4 are criti- craniofacial structures. At this point,
and build complicated structures that cal for setting up the proximal– distal one cannot help but ask the question of
make the head. axis during development. In the upper what is responsible for setting up the
In the facial region, each branchial and middle face, where structures de- signaling domain in the ectoderm. It
arch contains a central blood vessel, rive from the frontonasal process, turns out that pharyngeal endoderm is
the aortic arch, which is surrounded Sonic hedgehog (Shh) and FGF signal- critical for this process.
by cells of paraxial mesoderm. The ing appear to be critical for setting up
mesoderm core is enveloped by the a boundary in the neural and surface
more peripherally located CNC cells. ectoderm (Helms et al., 2005). In Pharyngeal Endoderm
The inner surface of the branchial craniofacial patterning, definitive evi- The pharyngeal endoderm makes a
arch contains cells derived from the dence only supports the critical func- limited contribution to craniofacial
pharyngeal endoderm, while the outer tion of ectodermal FGF signaling, as development, but it serves as an in-
surface is covered by the ectoderm conditional inactivation of the Fgf8 dispensable inducer during tissue–
(Fig. 3). The spatial relationship of the signaling resulted in the disappear- tissue interactions, controlling
boundary of cells with different em- ance of the proximal portion of the craniofacial development (Fig. 3).
bryonic origin is crucial for normal or- mandible (Trumpp et al., 1999). More The traditional view that the neural
ganogenesis and may reflect the regu- recently, studies have shown that sig- crest plays the key role in patterning
latory interactions that control the naling through FGFR1 is critical for the branchial arches must be recon-
development of complex structures in the neural crest independent pattern- sidered, because studies have found
the craniofacial region. ing of the pharyngeal ectoderm. Ecto- that the early ablation of CNC cells
derm FGF signaling patterns the pha- did not affect the development and
ryngeal region to create a permissive function of endoderm cells. These
Ectoderm environment for the entry of CNC crestless branchial arches were pat-
Craniofacial ectoderm plays a critical cells (Trokovic et al., 2003, 2005). terned normally and had a sense of
role in regulating the fate of CNC cells Craniofacial ectoderm regulates the individual identity, strongly sug-
during craniofacial morphogenesis, expression of transcription factors to gesting that the patterning of
whereas the establishment of ecto- specify the fate of CNC cells. For ex- endoderm is not dependent on CNC
derm identity is independent of CNC ample, FGF8 signaling controls the cells (Veitch et al., 1999). Hox genes
cells. Later, the continuous and recip- expression of two Lim-homeobox do- are known to be critical for the iden-
rocal interaction between the ecto- main genes, Lhx6 and Lhx7, in the tity of neural crest cells. Hoxa1 and
derm and the CNC-derived ectomes- CNC-derived ectomesenchyme. Lhx6 Hoxb1 double mutant mice had pat-
enchyme controls the position, size, and Lhx7 are mainly expressed in the terning defects in the hindbrain;
and shape of craniofacial organs dur- oral side of ectomesenchyme, while specifically, rhombomere 4 lost its
ing embryogenesis. It is imperative to Gsc is expressed in the aboral region ability to generate neural crest cells.
appreciate the inductive capability of (Tucker et al., 1999). Endothelin-1 in Consequently, the second branchial
ectoderm and CNC cells in the context the mandibular epithelium controls the arch crest population was not gener-
of time. This concept is best illus- expression of Gsc. Mutations in either ated. Despite the neural crest defect,
trated by the seemingly contradictory Endothelin-1 or Gsc result in mandibu- the formation of the second branchial
conclusions regarding the ability of lar development defects, demonstrating arch endoderm and epithelial region-
the oral ectoderm or the CNC-derived the endothelin-mediated Gsc expres- alization was normal (Gavalas et al.,
ectomesenchyme to induce tooth mor- sion is critical for controlling the pat- 2001).
phogenesis. terning of mandible (Clouthier et al., Limited information is available re-
Tissue recombination experiments 1998). In addition, BMP signaling has garding the patterning of pharyngeal
show that the tooth inductive signal been shown to be a critical regulator for endoderm. Retinoic acid signaling
first resides in the oral ectoderm and Msx1 function, which is exclusively ex- clearly plays a crucial role in regulating
then shifts into the underlying CNC- pressed in the CNC-derived ectomesen- pharyngeal endoderm development as
derived ectomesenchyme at a later de- chyme (Chen et al., 1996; Tucker et al., inactivation of retinaldehyde-specific
velopmental stage. In mouse, dental 1998a). Conditional inactivation of dehydrogenase type 2 (Raldh2), the reti-
epithelium before embryonic day (E) Bmpr1a in the oral ectoderm results in noic acid synthetic enzyme, resulted in
12 is capable of inducing tooth forma- tooth agenesis (Andl et al., 2004). Inhi- the absence of all branchial arches cau-
tion when combined with nondental bition of BMP signaling by noggin dal to the first (Niederreither et al.,
mesenchyme, whereas dental mesen- causes ectopic Barx-1 expression in the 1999). Inhibition of retinoid action by
chyme after E12 can induce tooth for- distal, presumptive incisor ectomesen- pan-retinoic acid receptor (pan-RAR)
mation when combined with nonden- chyme and a transformation of tooth specifically perturbed the development
tal epithelium (such as the second identity from incisor to molar, thus of the third and fourth branchial
branchial arch epithelium; Mina and demonstrating the significance of BMP arches, although the first and second
Kollar, 1987). signaling in patterning of craniofacial branchial arches formed normally
Growth and transcription factors structures (Tucker et al., 1998b). Taken (Wendling et al., 2000). Recent studies
are responsible for establishing the together, it is clear that ectoderm- have shown that Raldh2 is expressed in
patterning of craniofacial develop- mediated signaling plays an important the lateral mesoderm flanking the
ment. In the first branchial arch oral role in determining the patterning of endoderm during early embryonic de-
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2361

velopment and may pattern the pha- become arrested evolutionarily as and the CNC is clearly established
ryngeal endoderm development somitomeres (Jacobson, 1963; Pack- from the moment that CNC cells enter
(Niederreither et al., 2003; Graham ard and Meier, 1983). Despite several the branchial arch and is maintained
et al., 2005). Tbx-1 is another impor- gene expression analyses suggesting throughout craniofacial development.
tant gene for pharyngeal endoderm some degree of regionalization in the In parallel, studies have also demon-
development. Tbx-1 mutant mice cranial paraxial mesoderm (such as strated that cranial mesoderm is ca-
show failure to form caudal pouches, Paraxis, Tbx1, and Hoxb-1), there is pable of providing crucial signals for
whereas the first pharyngeal pouch no evidence supporting the existence endoderm development.
develops normally (Jerome and Pa- of metameric patterns. The impor- To date, there is not a clear demon-
paioannou, 2001; Lindsay et al., tance of somitomeric head mesoderm stration of the patterning potential of
2001). Of interest, Tbx-1 is also ex- is yet to be determined (Noden and cranial paraxial mesoderm to regulate
pressed in the lateral mesoderm Trainor, 2005). At the molecular level, craniofacial development in mam-
flanking the endoderm before the genes that drive mesoderm segmenta- mals. This finding is largely because
patterning of the pharyngeal tion in the trunk are missing from the of (1) lack of information on molecular
endoderm (Brown et al., 2004). pre-otic mesoderm in the head. When signals involved in the interaction be-
The pharyngeal endoderm exerts its cranial paraxial mesoderm was tween cranial paraxial mesoderm and
regulatory function through tissue–tis- grafted to the trunk region, activation adjacent tissue and (2) our inability to
sue interactions. For example, the pha- of the myogenic program in this ec- generate tissue-specific gene inactiva-
ryngeal pouches generate several spe- topic position was inhibited, suggest- tions. Recently, using the Cre/loxp re-
cialized epithelial structures, such as ing that there are distinct regulatory combination approach, we have learned
the thyroid, parathyroid, and thymus cascades acting in the development of that Myf5-Cre- or Mesp1-Cre-mediated
(Graham et al., 2005). The development trunk and head muscles, possibly re- recombination can be used to generate a
of these organs involves the interaction flecting their distinct function and targeted gene inactivation in the cra-
between pharyngeal endoderm and its evolution (Mootoosamy and Dietrich, nial paraxial mesoderm-derived first
flanking cranial neural crest cells. Com- 2002). Of interest, however, hetero- branchial arch myogenic core (our un-
promised retinoic acid signaling from topic grafting of paraxial mesoderm published data). In Myf5-Cre;R26R
the pharyngeal mesoderm affects the cells to different regions of the cranial mouse embryos at E9.5, the first
development of pharyngeal endoderm, mesoderm in the mouse showed no re- branchial arch myoblasts are lacZ-
which in turn causes defects in CNC striction in cell potency in the cranio- positive, accurately reflecting their ori-
migration and development of pharyn- caudal axis, revealing considerable gin from the paraxial mesoderm (Fig.
geal pouch-derived organs, such as the plasticity in the fate of the cranial me- 4). Targeted gene inactivation in the
thymus and parathyroid glands (Nied- soderm (Trainor et al., 1994). Signals cranial paraxial mesoderm-derived
erreither et al., 2003). Clearly, the pha- from surface ectoderm and endoderm mesenchyme will reveal important in-
ryngeal endoderm plays an important as well as CNC may influence the fate formation on its patterning potential in
role in regulating the morphogenesis of of mesoderm cells (Trokovic et al., regulating craniofacial development.
pharyngeal arch derivatives. 2003). Collectively, it is clear that cranio-
In terms of patterning capability, facial development requires contin-
cranial paraxial mesoderm provides a ued interaction and contribution by
Mesoderm permissive substratum for the migrat- cell populations derived from the ec-
The cranial paraxial mesoderm has an ing CNC cells to populate the toderm, the neural crest, the parax-
organization different than the trunk branchial arch. Studies using chick ial mesoderm, and the endoderm.
paraxial mesoderm. The cranial parax- embryos suggest that cranial paraxial These interactions occur en route
ial mesoderm can be roughly divided mesoderm is able to direct CNC cell and at the terminal site of tissue and
into (1) preotic head mesoderm, which movement independently of their epi- organ genesis. There is no example
lacks any overt sign of segmentation thelial or mesenchymal organization of any craniofacial developmental
and never forms somites; and (2) occip- (Noden, 1986; Ferguson and Graham, event that can occur in a totally cell
ital somites, which are caudal to the otic 2004). Of interest, recent cell fate autonomous or completely depen-
vesicle and give rise to epaxial and hy- mapping analysis has suggested that dent manner. Such a dichotomous
paxial muscles of the neck, the pharyn- myoblast precursors contain posi- view can only impede our progress
geal and laryngeal muscles that develop tional identity inherited from their so- toward a better understanding of the
in the caudal branchial arches, and the matic mesenchymal stem cell precur- regulatory mechanism of craniofa-
tongue muscle (Fig. 3B; Noden, 1983; sors and can help to determine cial development. Instead, we need
Couly et al., 1992; Huang et al., 1999; skeletal homologies that are based on to focus our efforts to unveil the mo-
Mootoosamy and Dietrich, 2002). muscle attachments (Matsuoka et al., lecular signals involved in tissue–
There are profound morphological 2005). Conversely, CNC cells, which tissue interactions at each critical
and molecular differences between the provide most of the connective tissues time point and to gain a better under-
cranial and trunk paraxial mesoderm. and tendons in the head, may pattern standing of tissue boundary establish-
In comparison to the clearly seg- and shape the individual cranial mus- ment, because a disturbed tissue
mented somite development within cle (Noden, 1986; Kontges and Lums- boundary during early embryonic de-
the trunk mesoderm, studies have den, 1996). The intimate relationship velopment can lead to craniofacial mal-
suggested that head mesoderm has between cranial paraxial mesoderm formations.
2362 CHAI AND MAXSON

SIGNALING INTERACTIONS cells are the major driving forces for CNC cells have an intimate associa-
IN THE PATTERNING AND branchial arch development. As the tion with the oral ectoderm (Chai et
first branchial arch extends ventral– al., 2000). This CNC distribution pat-
MORPHOGENESIS OF
medially, it gives rise to both man- tern significantly overlaps with the
CRANIOFACIAL ORGANS dibular and maxillary prominences expression of two specific Lim-ho-
Recent studies have uncovered spe- (although recent study suggests that meobox domain genes, Lhx6 and
cific signaling cascades that play cru- the maxillary prominence has a sep- Lhx7, within the CNC-derived ecto-
cial roles in regulating the patterning arate origin from the mandibular mesenchyme, suggesting that they
and morphogenesis of craniofacial or- prominence in chicken; Lee et al., may have critical functions in direct-
gans. One of the best-studied models 2004). The primitive oral cavity ing CNC cells to reach their destina-
of craniofacial organogenesis is tooth forms as a consequence of the fusion tion (Grigoriou et al., 1998). In the
development, which involves contin- among intermaxillary segments of aboral region, the homeobox gene
uous tissue–tissue interactions be- the frontonasal prominence and the Goosecoid (Gsc) is expressed where
tween the ectoderm-derived enamel paired maxillary and mandibular Lhx6 and Lhx7 are excluded (Tucker
organ epithelium and the cranial prominences. The oral epithelium, in et al., 1999). Apparently, FGF8 is re-
neural crest-derived ectomesen- contrast to the aboral (suboral) epi- sponsible for regulating the expres-
chyme (Slavkin et al., 1968; Kollar, thelium, becomes thickened to form sion of both Lhx and Gsc genes. The
1972; Lumsden, 1988; Jernvall and the dental lamina, marking the time mandibular arch mesenchyme can be
Thesleff, 2000; Tucker and Sharpe, and location for tooth development roughly divided into the Lhx-positive
2004, and references therein). Multi- to begin. rostral (oral) domain and the Gsc-pos-
ple growth and transcription factors Before the initiation of tooth devel- itive caudal (aboral) domain (Fig. 5).
belonging to several signaling fami- opment, the mandibular ectoderm can Clearly, patterning of the mamma-
lies have been identified as critical be roughly divided into the proximal lian dentition is a three-dimensional
regulators at the initiation and domain, which expresses FGF8 and process. One aspect that is beginning
throughout all stages of tooth devel- gives rise to molars, and the distal to be explored is the determination of
opment (http://bite-it.helsinki.fi). domain, which expresses BMP4 and lingual– buccal (medial–lateral) den-
Furthermore, most of the signaling gives rise to incisors (Fig. 5). Signifi- tal cusp patterning (Fig. 5). Lingual
networks are used reiteratively cantly, FGF and BMP act antagonis- and buccal cusps of the molar are dif-
throughout tooth development and tically to restrict Barx1 and Dlx2 ex- ferent (in number and shape), and left
are common to the regulatory sys- pression to the proximal domain of the and right first molars, for example,
tems critical for governing the devel- first arch ectomesenchyme and Msx1 have a mirror image in patterning. It
opment of other organs (such as the and Msx2 expression to the distal do- will be fascinating to learn how sig-
development of feather, hair, mam- main, respectively. The biological sig- nals are set up to achieve this har-
mary gland, salivary gland, and pan- nificance of such a regional molecular mony during tooth development.
creas; see reviews by Nuckolls et al., specification has been demonstrated The development of each dental
1999; Shum et al., 2000). The grow- elegantly with inhibition of BMP sig- cusp is under the control of a transient
ing scientific evidence suggests a naling resulting in ectopic Barx-1 ex- signaling center known as the enamel
highly conserved biological mecha- pression in the distal, presumptive in- knot, which is a dense population of
nism to regulate the patterning and cisor mesenchyme and producing epithelial cells without any prolifera-
morphogenesis of craniofacial or- transformation of tooth form from in- tive activity (Fig. 6). The enamel knot
gans, but studies are now beginning cisor-form to molar-form (Tucker et contains multiple signaling molecules
to reveal the unique features of the al., 1998b). Recently, study has shown (such as Shh, BMP, and FGF) and
signaling network in regulating that Barx1 played a key role in the controls the size and shape of cusp
tooth morphogenesis. This topic will development of the dentition and di- formation. Thereafter, it is removed
be the focus of our discussion here. gestive system, which was vital for the by programmed cell death (Jernvall
Patterning of the evolution of mammals (Miletich et al., and Thesleff, 2000; Tucker and
2005). Overall, FGF and BMP growth Sharpe, 2004). Unlike the incisor
Mammalian Dentition factor gradients are critical determi- tooth organ, molar development relies
The patterning of dentition depends nants for specifying the initiation on additional enamel knots (second-
on the proper development of the oral sites of tooth formations as well as for ary and tertiary) to form multicusp
cavity, where maxillary and mandib- specifying the CNC-derived dental patterning.
ular teeth are housed and involves the mesenchyme to become incisor-form BMP signaling is involved in regu-
determination of location, shape, num- vs. molar-form tooth organs. lating the distance between adjacent
ber, and size of tooth development. Mul- Besides the proximal and distal do- secondary enamel knots to control the
tiple developmental decisions are mains within the oral ectoderm, the positioning of cusps. Two recently
made in the patterning and develop- mandibular arch mesenchyme can published studies have provided im-
ment of mammalian dentition. The also be divided into an oral and aboral portant information regarding the
development of the oral cavity be- (rostral– caudal) axis (Fig. 5). Signals contribution of BMP signaling inhibi-
gins with the establishment of the from the oral ectoderm appear to be tion to the regulation of cusp pattern-
frontonasal prominence and the first responsible for coordinating this divi- ing. Ectodin is a secreted BMP inhib-
branchial arch. The migrating CNC sion. Of interest, the postmigratory itor with an inverse expression
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2363

pattern to p21, a hallmark cell cycle in enlargement of the enamel knot, patterning occurs along the buccal
regulator expressed in the enamel highly altered cusp patterns, fusion of side of the cusps, suggesting that ec-
knot during tooth development (Kas- molars, and extra teeth. Interestingly, todin functions in the evolution of lat-
sai et al., 2005). Loss of ectodin results the most dramatic changes in cusp eral bias in teeth. Noggin is another
widely distributed BMP inhibitor (Chen
et al., 2004). Overexpression of Noggin
in the dental epithelium blocked the de-
velopment of all mandibular and max-
illary third molars at the bud stage and
severely altered the patterning of max-
illary molars (Plikus et al., 2005). The
differential defects between mandibu-
lar and maxillary molars suggest a re-
quirement for varying thresholds of
BMP signaling.

Fig. 4. Two component genetic system for in-


delibly marking the fate of cranial neural crest-
(CNC), or mesoderm-, or ectoderm-derived
cells. A: Contribution of CNC cells during early
craniofacial development. CNC-derived cells
(blue) populate the frontonasal process (FN)
and the first branchial arch (arrow). B: Cross-
section of an embryonic day (E) 9.5 mouse em-
bryo (Wnt1-Cre/R26R) shows CNC-derived
cells (blue) in the first branchial arch. The arrow
indicates the paraxial mesoderm-derived myo-
Fig. 4. genic core in the first arch. The arrowhead in-
dicates ectoderm. C: Contribution of paraxial
mesoderm-derived cells during early craniofa-
cial development. The arrow indicates the
paraxial mesoderm-derived myogenic core
(blue) in the first branchial arch of the Myf5-Cre/
R26R sample. D: Cross-section of the first and
second branchial arch contains paraxial meso-
derm-derived myogenic cells (blue; arrow). No-
tice these cells are pink in B. E: Contribution of
ectodermal cells during craniofacial develop-
ment. Ectoderm-derived cells (blue) cover the
surface of the K14-Cre/R26R embryo. The ar-
row indicates the first branchial arch surface
ectoderm. F: Cross-section of the first branchial
arch shows ectoderm-derived cells (arrow) on
the surface.

Fig. 5. Patterning of the first branchial arch.


A: The frontal view of a scanning electronic
microscopic image of an embryonic day (E) 9.5
mouse embryo (25–29 somites) shows the fron-
tonasal process (FN), the maxillary (max), and
the mandibular (mand) process. B: Schematic
drawing of A. C: The first branchial arch can be
divided into proximal/distal, oral/aboral, and
buccal/lingual (see D) domains. In the distal
domain, bone morphogenetic protein (BMP)
signaling in the ectoderm regulates the expres-
sion of Msx1 and Msx2. It also provides inhibi-
tion for Barx1 expression in the cranial neural
crest (CNC) -derived mesenchyme in the prox-
imal domain. In the proximal domain, fibroblast
growth factor (FGF) signaling regulates the ex-
pression of multiple transcription factors (mod-
ified from Tucker and Sharpe, 2004). D: Molar
teeth (m1, m2, m3) show different cusp patterns
Fig. 5. along buccal and lingual aspects of the jaw.
2364 CHAI AND MAXSON

Control of Signaling Level lars, with “/” marking the front mid- Continued
and Tooth Development line]. Some rodents, such as mice, Epithelial–Mesenchymal
have a dental formula of 3.0.0.1/ Interactions and the Success
Recent studies show that local feed- 1.0.0.3. The initiation of each tooth
back provides a tightly controlled FGF of Tooth Morphogenesis
germ is marked by the formation of
and BMP gradient to ensure proper After the initiation of tooth develop-
dental lamina. Ectodysplasin (EDA)
tooth development. For example, Is- ment, there is continued interaction
signaling cascade molecules, which
let1 positively regulates the expres- between the dental epithelium and
belong to the tumor necrosis factor
sion of Bmp4, whereas a range of the CNC-derived ectomesenchyme
family of ligands, are critical regula-
Pitx2 levels can differentially regulate throughout tooth morphogenesis. The
the expression of Fgf8 and Bmp4 dur- tors for the determination of tooth
number during embryonic develop- initial tooth generating potential re-
ing initial tooth development (Lu et sides within the epithelium and is ca-
al., 1999; Mitsiadis et al., 2003; Liu et ment (Fig. 6). Mice with a compro-
mised EDA signaling cascade, such pable of inducing nontooth forming ec-
al., 2003). Pitx2 is restricted to the tomesenchyme to develop teeth (Mina
dental epithelium throughout tooth as Tabby (EDA), Downless (EDAR,
and Kollar, 1987; Jernvall and Thesleff,
morphogenesis (Fig. 6). Mutation of EDA receptor), and Crinkled (EDAR-
2000). Soon after development, this
the human PITX2 gene results in ADD, EDA intracellular adaptor
tooth-forming potential shifts to the
Rieger’s syndrome, an autosomal protein) mutations, show abnormal
dental mesenchyme. This shift of
dominant disorder, that leads to the tooth numbers and defects in the de-
tooth-forming potential coincides
absence of certain teeth and defects of velopment of other ectodermal or-
with a shift in BMP signaling from
the eye (Rieger, 1935; Semina et al., gans (hair follicles and exocrine the epithelium to the mesenchyme.
1996). Loss of Pitx2 results in re- glands; Sofaer, 1969, 1977; Headon Morphologically, at this time, tooth
tarded tooth development at the initi- et al., 2001; Pispa and Thesleff, development advances into the bud
ation/early bud stage, clearly demon- 2003; Tucker et al., 2004). On the stage (Fig. 6). Multiple signaling
strating the important function of other hand, overexpression of EDA molecules within the enamel organ
Pitx2 gene in regulating tooth devel- signaling molecules results in an ex- epithelium have been implicated in
opment (Lu et al., 1999). Significantly, pansion of the molar tooth field and the regulation of transcription fac-
loss of one copy of the Pitx2 gene can the formation of a supernumerary tors in the surrounding ectomesen-
also affect tooth development in mice tooth distal to the first molar (Mus- chyme during the bud to the cap
(Gage et al., 1999). Therefore, Pitx2 tonen et al., 2003; Tucker et al., stage transition (Fig. 6; Jernvall and
acts in a dosage-specific manner in hu- 2004). Of interest, the supernumer- Thesleff, 2000; Tucker and Sharpe,
mans and mice. The expression of ary tooth has the cusp patterning of 2004). Interestingly, individual null
Pitx2 gene is sensitive to the level of a premolar, suggesting that dental mutations of Msx1, Lef1, or Pax9 all
BMP4 or FGF8 signaling, indicating a result in retarded tooth development
cusp patterning is highly dependent
feedback loop among these signaling at the bud stage, indicating the crit-
on the exact position in the oral cav-
molecules (St. Amand et al., 2000). ical roles of these transcription fac-
ity where tooth development occurs.
Shh, another important epithelial tors during the transition from the
BMP signaling is another morpho-
signaling molecule, controls the prolif- bud to cap stage tooth development
regulator for the number, size, and
eration of enamel organ epithelial (Satokata and Maas, 1994; Kratoch-
cells during initiation of tooth devel- shape of tooth development. Loss of
BMP receptor 1a (Bmpr1a) caused wil et al., 1996; Peters et al., 1998;
opment (Fig. 6; Bitgood and McMa- Sasaki et al., 2005). Ectopic overex-
hon, 1995; Dassule and McMahon, retarded tooth development (Andl et
al., 2004). Overexpression of BMP pression and tissue recombination
1998; Hardcastle et al., 1998; Co- experiments have shown that these
bourne et al., 2001). The Wnt signal- inhibitor Noggin in the oral ecto-
transcription factors are regulated
ing pathway interacts with Shh signal- derm resulted in miniaturization of
by epithelial signals, can control the
ing to establish boundaries during tooth maxillary first and second molars,
expression of growth factors and
development. Specifically, Wnt-7b acts and retarded maxillary third and
other transcription factors in the ec-
to repress the expression of Shh in non- mandibular molar development at
tomesenchyme and provide instruc-
dental oral ectoderm, whereas Shh ex- the lamina stage (Plikus et al.,
tive feedback to the enamel organ
pression is restricted to the dental ecto- 2005). The selective loss of molars in epithelium. Thus, they are essential
derm and can instruct, permit, or Noggin-overexpressing mice, along determining factors during the cap
induce tooth bud formations (Sarkar et with the abnormal buccal cusp pat- stage tooth development.
al., 2000). terning in ectodin null mutant mice, To date, no null mutation experi-
strongly suggest that various tooth ments have resulted in failure of ini-
germs and dental cusps have differ- tiation of tooth development (thicken-
Determination of Tooth
ential requirements for the level of ing of the dental lamina). Some of the
Number BMP signaling. Modulation of BMP compound null mutations, however,
Humans have a dental formula of signaling may account for one of the have resulted in retardation of tooth
3.2.1.2/2.1.2.3 in the permanent den- mechanisms responsible for changes development at an early bud stage,
tition [two incisors, one canine, two in tooth number, size, and shape such as Msx1⫺/⫺/Msx2⫺/⫺, Dlx1⫺/⫺/
premolars (bicuspids) and three mo- through evolution. Dlx2⫺/⫺, and Gli2⫺/⫺/Gli3⫺/⫺.
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2365

Clearly, there is functional redun- tion for the future design and of the skull vault is of mesoderm ori-
dancy among different members of the fabrication of tooth regeneration. gin. In contrast, Couly and coworkers
same transcription factor family in (1993) concluded that the skull vault
regulating the advancement of tooth is entirely neural crest derived. In the
SKULL DEVELOPMENT
morphogenesis. Of interest, some of mouse model, it has been demon-
the null mutations reveal regional AND TISSUE BOUNDARY strated elegantly that frontal bones
specificity of certain signaling mole- The vertebral skull represents an ex- are neural crest-derived and parietal
cules in regulating tooth morphogene- cellent model for the investigation of bones are of mesoderm origin (Jiang et
sis. For example, only maxillary molar development and evolution. In mam- al., 2002). The posterior part of the
tooth organs were affected in Dlx1⫺/ mals, both CNC- and mesoderm-de- skull vault (supraoccipital and exoc-
⫺/⫺
⫺/Dlx2 double mutants. Further- rived cells contribute to the develop- cipital bones) derives from the occipi-
more, loss of Dlx1 and Dlx2 genes re- ment of the skull (Fig. 3D). Different tal somites (Fig. 3D). The dura mater
sulted in a change of cell fate in the elements of the skull are established that underlies the frontal and parietal
dental mesenchyme region from odon- by the formation of tissue boundaries, bones is also neural crest derived
togenic to chondrogenic (Thomas et which reflect an evolutionary contri- (Jiang et al., 2002; Ito et al., 2003;
al., 1997; Tucker and Sharpe, 1999). bution and are critical for the pre- and Sasaki et al., 2006). The coronal and
This finding also raises the possibility postnatal dynamic development of the sagittal sutures are of mesoderm ori-
that a subpopulation of CNC cells car- head and face. Recent studies are now gin (Morriss-Kay and Wilkie, 2005).
rying Dlx1/Dlx2 genes migrates into beginning to address the molecular Clearly, the skull vault elements are
the maxillary molar region and, upon regulation of patterning and size de- developed at the boundary between
receiving the proper instruction from termination of different elements of CNC- and mesoderm-derived tissue.
the dental epithelium, can contribute the skull. Multiple excellent reviews This boundary is of paramount impor-
to the formation of tooth organ. By have addressed the regulation of skull tance in mediating tissue–tissue in-
using the two-component genetic sys- vault development (Wilkie and Mor- teraction that control skull vault de-
tem (Wnt1-Cre/R26R) for indelibly riss-Kay, 2001; Santagati and Rijli, velopment. When there is mixing of
marking the progenies of CNC cells 2003; Morriss-Kay and Wilkie, 2005), the two cell populations as the result
(Fig. 4), it may be feasible to test the but there are very few reviews that of a gene mutation (such as in the
hypothesis that a subpopulation of address the molecular regulatory ephrin-B1 or Twist mutant), the
CNC cells designated for the maxil- mechanism of facial bone develop- boundary between CNC and the me-
lary molar region is affected in Dlx1/ ment. Here, using mandible and pal- soderm is lost, resulting in the prema-
Dlx2 double-mutant mice (Chai et al., ate development as examples, we ture fusion of cranial sutures known
2000; Han et al., 2003). The outcome summarize recent advancements to- as craniosynostosis (Twigg et al.,
of such experiments may address the ward the understanding of the regula- 2004; Merrill et al., 2006).
speculation that a predetermined sub- tory mechanism of craniofacial bone
population of CNC cells possesses in- development. MANDIBULAR
structive functions in the induction of The skull consists of the neurocra-
tooth development. nium and viscerocranium. The neuro-
MORPHOGENESIS
The activin ␤A null mutation re- cranium (skull vault and base) sur- Patterning of the branchial arches re-
veals another example of region-spe- rounds and protects the brain. In quires the establishment of both inter-
cific signaling in regulating tooth de- humans, eight bones compose the neu- branchial arch and intrabranchial
velopment. All teeth, except the rocranium: the paired temporal and arch identities (Depew et al., 2002a).
maxillary molars, are arrested at the parietal bones and the singular fron- It is well established that Hox, Pbx,
bud stage in activin ␤A⫺/⫺ mice, a tal, sphenoid, ethmoid, and occipital and Otx homeobox genes are critical
reverse phenotype compared with the bones. Fourteen bones compose the for the normal patterning of inter-
Dlx1/Dlx2 double mutant (Ferguson viscerocranium (the jaws and other branchial arch identities (Gendron-
et al., 1998). Because activin ␤A is pharyngeal arch derivatives): the Maguire et al., 1993; Rijli et al., 1993;
expressed in the CNC-derived ecto- paired nasal bones, maxillae, palatine Matsuo et al., 1995; Selleri et al.,
mesenchyme, it may help to specify bones, lacrimal bones, zygoma and in- 2001). For example, there clearly is a
the fate of CNC cells during tooth de- ferior nasal conchae, along with the Hox gene code that controls branchial
velopment. singular vomer and mandible. The arch development. To give rise to the
Overall, continuous and reciprocal viscerocranium derives from the neu- derivatives of the first branchial arch,
epithelial–mesenchymal interaction is ral crest, forms the face, and supports the structure needs to be Hox gene
the key for successful tooth organ mor- the functions of feeding and breath- negative. Until recently, however, less
phogenesis. From initiation to odonto- ing. information has been available about
blast/ameloblast differentiation to ma- Until recently, the tissue origin of the genetic control of the establish-
trix formation, this interaction provides the skull vault has been controversial ment of the intrabranchial arch iden-
precise communication between two ad- because of conflicting reports using tities. This is an important area in
jacent tissue types and governs the quail– chick grafting. Noden (1978, craniofacial development, because hu-
number, size, and shape of tooth forma- 1988) reported that the CNC only con- man mandibular dysmorphogenesis
tion. Our understanding of these biolog- tributes to the rostral portion of the appears to be a common malformation
ical processes may serve as a founda- frontal bones and that the remainder and appears in multiple congenital
2366 CHAI AND MAXSON

birth defect syndromes, ranging from ing (such as BMP) from the ectoderm Similar to the proximal– distal do-
agnathia (agenesis of the jaw) to mi- and become committed to give rise to mains within the ectoderm of the first
crognathia to patterning malforma- structures associated with the distal branchial arch, there are discrete pop-
tions. portion of the first arch. As embryonic ulations within the underlying CNC-
Based on the mouse model, we know development progresses, there is a derived mesenchyme. Dlx genes are
now that the first branchial arch shift of the instructive capability for known to play important roles in reg-
(mandibular arch) becomes apparent patterning the mandible from the ec- ulating the patterning of the develop-
at E8.0 –E8.5 (6 – 8 somites) as small toderm to the CNC-derived mesen- ing jaw. Six Dlx genes (Dlx1, Dlx2,
swellings on the side of the developing chyme. The patterning of the proxi- Dlx3, Dlx5, Dlx6, and Dlx7) have been
head. As CNC cells migrate into and mal– distal domain of the first described in mice (Dolle et al., 1992;
proliferate within the mandibular branchial arch is achieved through Robinson and Mahon, 1994; Simeone
arch, it develops rapidly toward the the antagonistic interaction between et al., 1994; Qiu et al., 1995, 1997;
ventral midline. The CNC-derived BMP and FGF, which controls the Stock et al., 1996). During the devel-
cells are localized immediately subja- mesenchymal expression of signaling opment of the branchial arches, Dlx
cent to the covering epithelium (Chai molecules, such as Msx1 and Barx1, genes show nested expression patterns
et al., 2000). Mandibular development respectively (Tucker et al., 1998b; and play important roles in establishing
depends upon the interaction between Tucker and Sharpe, 2004). BMP sig- the identity along the proximodistal
the oral ectoderm and the CNC-de- naling clearly has an important role in axis within each branchial arch (Depew
rived mesenchyme within the first regulating mandibular morphogene- et al., 2002a, 2005). Specifically, Dlx1/2
branchial arch. Within the oral ecto- sis. Specifically, loss of BMP signaling are expressed in the CNC-derived ecto-
derm, signaling molecules, such as in the oral ectoderm and the pharyn- mesenchyme both in the proximal and
BMP, TGF-␤, and FGF, are expressed geal endoderm results in extreme phe- distal region of the branchial arch,
in a region-specific manner (Chai et notypes, ranging from an almost com- whereas Dlx5/6 and Dlx3/7 show pro-
al., 1994, 1997; Trumpp et al., 1999; pletely missing mandible to severe gressively restricted expression do-
Ito et al., 2002; Liu et al., 2005). They defects in the distal region (Liu et al., mains toward the distal region of the
may regulate homeobox-containing 2005). BMP target genes Msx1 and branchial arch.
genes (such as Dlx, Lhx, and Gsc) Msx2 have differential dose require- Recent studies have shown that a
within the CNC-derived mesenchyme ments for BMP4 signaling, and they combinatorial Dlx code regulates the
to generate early polarity and are re- act in a functionally redundant man- establishment of the distinct skeletal
sponsible for patterning of the first ner. Both Msx1 and Msx2 are ex- elements within a given branchial
branchial arch. pressed in the midline region, and loss arch unit (Qiu et al., 1995, 1997; De-
Ectodermal FGF signaling is criti- of BMP4 signaling results in compro- pew et al., 2002a,b; Cobourne and
cal for CNC cell survival in the man- mised Msx gene expression. Mutation Sharpe, 2003). Loss of Dlx5 and Dlx6
dibular arch as conditional inactiva- of Msx1 and Msx2 genes results in results in a homeotic transformation
tion of Fgf8 in the ectoderm caused midline cleft of the first arch and se- of the lower jaw into an upper jaw,
increased apoptotic activity and a dra- vere defects in mandibular morpho- supporting a model of patterning
matic loss of all first branchial arch genesis (Satokata et al., 2000; Ishii et within the branchial arch that relies
skeletal structures (except the most dis- al., 2005; our unpublished data). on a nested pattern of Dlx gene ex-
tal portion of the mandible; Trumpp et Mandibular development requires pression (Depew et al., 2002b). In ad-
al., 1999). During early embryonic de- proper modulation of BMP and FGF dition, jaw development is sensitive to
velopment, however, FGF signaling is signaling in the ectoderm. This modu- the dosage of Dlx genes, as haploinsuf-
not required for cell proliferation and lation is achieved through the antag- ficiency of single or multiple Dlx genes
survival, but instead is required for cell onistic interaction between BMP and has a gradient effect on mandibular
migration. Therefore, FGF signaling FGF signaling, the inhibition of BMP development (Depew et al., 2005).
has differential functional specificity in signaling by Noggin or Chordin and by This observation clearly suggests that
different developmental contexts (Sun the feedback from the underlying CNC- the expression of Dlx genes must be
et al., 1999). derived mesenchyme (Stottmann et al., tightly regulated. To date, little infor-
In the proximal domain of the first 2001; Wilson and Tucker, 2004; Liu et mation is available about the direct
branchial arch, FGF signaling directly al., 2005). For example, BMP signaling regulation on Dlx gene expression. Al-
or indirectly regulates the expression is required for maintaining FGF signal- though FGF8-soaked beads placed in
of homeobox genes in the mesenchyme ing in the proximal domain of the man- the first arch epithelium are able to
to control the development of mandi- dibular arch, but it represses FGF sig- induce Dlx2 and Dlx5 expression in
ble (see previous discussion). In the naling in the distal domain (Liu et al., the mandibular mesenchyme, loss of
distal domain of the first branchial 2005). Furthermore, BMP signaling is Fgf8 in the ectoderm shows unaltered
arch, BMP signaling appears to be a subject to multiple points of regulation, Dlx2 and Dlx5 expression in the first
crucial regulator for mandibular mor- at the ligand, receptor, Smad, and tran- branchial arch (Trumpp et al., 1999).
phogenesis. BMP4 is expressed within scription complex level (Massague et Similarly, BMP-soaked beads are able
the distal ectoderm that covers the al., 2005). Clearly, tightly controlled to induce Dlx gene expression, but the
first branchial arch mesenchyme at BMP and FGF signaling is of para- endogenous BMP and Dlx expression
E9.5. The postmigratory CNC cells mount importance for mandibular mor- patterns do not suggest a direct regu-
are exposed to the instructive signal- phogenesis. latory relationship. Overall, Dlx genes
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2367

are clearly important for intrabranchial


arch patterning, and further studies in
this area will advance our understand-
ing of their role and the regulatory
mechanism in this process.

PALATOGENESIS AND THE


MOLECULAR MECHANISM
OF CLEFT PALATE
The mammalian palate develops from
two primordia: the primary palate and
the secondary palate. The primary
palate represents only a small part of
the adult hard palate. The secondary
palate is the primordium of the hard
and soft parts of the palate. Palate
development is a multistep process that

Fig. 6.

Fig. 6. Schematic drawing of reciprocal and


reiterative signaling in regulating epithelial–
mesenchymal interactions throughout sequen-
tial stages of tooth morphogenesis. A precise
spatial and temporal orchestration of multiple
growth and transcription factors (as listed
within each box) is critical for the initiation as
well as patterning of tooth germ development.
These signaling molecules tightly regulate both
the enamel organ epithelia and the cranial neu-
ral crest-derived dental mesenchyme. Specifi-
cally, when tooth development is initiated with
formation of dental lamina, its underlying mes-
enchyme is almost entirely populated with cra-
nial neural crest (CNC) -derived cells (dark
blue). As tooth germ develops from the bud to
the cap stage, CNC-derived cells are concen-
trated (dark blue) at the interface with the
enamel organ epithelium, whereas the periph-
eral portion of the dental sac is populated with
both CNC- and non–CNC-derived cells (light
blue). Molecular signaling residing within the
enamel organ epithelium and the CNC-derived
dental mesenchyme are engaged in a constant
and reciprocal dialogue to mediate tooth mor-
phogenesis. The dark red dot represents
enamel knot-a signaling center within the
enamel organ epithelia, whereas light pink rep-
resents enamel organ epithelium (modified from
Jernvall and Thesleff, 2000; Chai et al., 2000).

Fig. 7. Anatomy of palatogenesis and five cat-


egories of palatal shelf defects that result in
cleft palate. A: Mouse palatogenesis starts at
embryonic day (E) 11.5. By E13.5, palatal
shelves (P) are on both sides of the tongue (T).
Each palatal shelf can be divided into the me-
dial (m) and lateral (l) aspects. t, tooth germ.
B,C: Between E13.5 and E14.0, palatal shelves
turn horizontally above the tongue and face
each other along the midline. D: At E14.5, pal-
atal fusion begins to take place. The arrow in-
dicates the midline epithelial seam. n, nasal
epithelium; o, oral epithelium. E,F: From E15.5
to E16.5, palatal fusion is completed through-
out the entire palate. Five different categories of
palatal shelf defects that result in cleft palate
Fig. 7. (also see Zhang et al., 2002). P, palatal shelf.
2368 CHAI AND MAXSON

involves palatal shelf growth, elevation, ternative fate for the MEE is migra- butions to our understanding of the
midline fusion of palatal shelves, and tion along the midline toward the na- gene pathways involved in palate de-
the disappearance of midline epithelial sal and oral epithelia, resulting in the velopment and the nature of signaling
seam (Fig. 7). The palatal structures loss of MEE (Carette and Ferguson, molecules that act in a tissue-specific
are composed of the CNC-derived ecto- 1992, Hilliard et al., 2005). Studies manner at critical stages of embryonic
mesenchyme and pharyngeal ectoderm from multiple laboratories have sug- development. Interestingly, however,
(Ferguson, 1988; Shuler, 1995). The ep- gested that epithelial–mesenchymal most of genetic mutations cause mul-
ithelia that cover the palatal shelves transformation (EMT) is an important tiple structural and functional defects
are regionally divided into oral, nasal, cellular mechanism for the disappear- in the developing embryo. Conse-
and medial edge epithelium (Fig. 7D). ance of MEE (Fitchett and Hay, 1989; quently, assessment of the role of a
The nasal and oral epithelia differenti- Griffith and Hay, 1992; Shuler et al., particular gene in regulating palato-
ate into pseudostratified and squamous 1992). These studies were largely genesis has been a challenge. To fur-
epithelia, respectively, whereas the me- based on cell lineage tracing using ther complicate the issue, cleft lip
dial edge epithelium (MEE) is removed membrane-intercalating dye and epi- and/or palate is a complex trait caused
from the fusion line by means of pro- thelial and mesenchymal cellular by multiple genetic and environmen-
grammed cell death and cell migration markers. Recently, however, studies tal factors (Murray, 2002). Neverthe-
(Martinez-Alvarez et al., 2000; Vaziri using genetic cellular markers have less, these animal models have ad-
Sani et al., 2005). challenged the theory of EMT during vanced our understanding of some
Mouse palatogenesis initiates at palatal fusion. Both in vivo and in important gene functions in human
E11.5 as marked by the formation of vitro studies show that EMT does not palate development.
palatal shelves extending from the in- occur during palatal fusion (Cuervo et One of the important discoveries
ternal aspects of maxillae. After initi- al., 2002; Vaziri Sani et al., 2005; Xu has been the existence of genetic het-
ation, the palatal shelves project et al., 2006). Apoptosis and cell migra- erogeneity along the anterior–poste-
downward on each side of the tongue tion may be sufficient to account for rior and medial–lateral axes of the de-
between E12.5 and E13.5 (Fig. 7A,B). the cellular mechanism for the disap- veloping palate (for review, see
As the jaws develop, the tongue de- pearance of MEE cells. Hilliard et al., 2005). This heterogene-
scends, thus providing space to accom- CNC cells are critical for palatogen- ity may provide differential regula-
modate the horizontal apposition of esis. Until recently, however, little tory mechanisms for the fusion of the
palatal shelves above the tongue (Fig. has been known about the fate of the anterior vs. posterior region of the pal-
7C). The fusion of palatal shelves oc- CNC-derived palatal mesenchyme or ate. For example, MEE cells begin to
curs at E14.5, resulting in the forma- the molecular mechanisms that regu- undergo apoptosis at different times
tion of continuous palate (Fig. 7D). late the epithelial–mesenchymal in- during palatal fusion, depending on
Both the elevation and fusion of the teractions during palate development. their location. It has been shown that
palatal shelves occur in an anterior to The lack of information is largely apoptosis of MEE cells is triggered by
posterior sequence. The complete fu- due to the difficulty in CNC cell la- palatal shelf contact in the anterior
sion of palatal shelves results in the beling and fate analysis in the palate. region, whereas it is initiated before
separation of the oral cavity from the Significantly, using the Wnt1-Cre/ any contact between the opposing
nasal cavity (Fig. 7E,F). R26R model, we have systematically shelves in the posterior region (Cu-
followed the migration, proliferation, ervo et al., 2002). This may be the
and differentiation of CNC cells result of differential molecular signals
Fate of Medial Edge throughout embryogenesis. We show in the palatal mesenchyme along the
Epithelial Cells and the that the CNC-derived ectomesen- anteroposterior (A-P) axis that in-
Contribution of Cranial chyme contribute significantly to the struct different fates to the palatal ep-
palatal mesenchyme and that there is ithelium (Ferguson and Honig, 1984).
Neural Crest Cells During
a dynamic distribution of these CNC More recent studies have demon-
Palatogenesis cells during palatogenesis (Chai et al., strated that constant and reciprocal
There has been a tremendous interest 2000; Ito et al., 2003). This two- interactions between the palatal epi-
in the fate of medial edge epithelial component genetic system provides thelium and the CNC-derived mesen-
cells during palatal fusion. Apoptosis the opportunity to integrate analysis chyme are responsible for setting up
is clearly one of the important mecha- of the fate and function of the mam- this genetic heterogeneity along the
nisms for eliminating MEE cells dur- malian neural crest with mouse mo- A-P axis and are crucial for normal
ing palatal fusion (Farbman, 1968; lecular genetics in both normal and palatal development and fusion
Saunders, 1966; Dudas and Kaarti- abnormal embryonic development. (Zhang et al., 2002; Murray and
nen, 2005). At the molecular level, Schutte, 2004; Rice et al., 2004).
TGF-␤ and RA are critical inducers for Mouse Models for Multiple genes have been found to
apoptosis of the MEE. The dosage of Investigating the Molecular be critical for the development of the
these signaling molecules may differ- Regulatory Mechanism of anterior region of the palate. For ex-
entially control the progression of the ample, Msx1, Bmp4, Bmp2, Fgf10,
cell cycle in the MEE (Cuervo et al.,
Palatogenesis and Shox2 show restricted expression
2002; Martinez-Alvarez et al., 2000; Multiple genetically mutated mouse patterns in the anterior region of the
Cuervo and Covarrubias, 2004). An al- models have made significant contri- palate (Rice et al., 2004; Hilliard et al.,
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2369

2005). Loss of Msx1 results in a cell dient in the palatal shelf. Signifi- For example, mutations in Msx1 and
proliferation defect in the CNC-de- cantly, mutation of the Osr2 gene re- Lhx8 and conditional inactivation of
rived palatal mesenchyme in the an- sults in the compromised development Tgfbr2 in CNC cells or Shh in the ep-
terior region of secondary palate. of the medial aspect of palatal shelf ithelium all result in retarded palatal
BMP4 functions downstream of Msx1 development and retards palatal shelf shelf development (Satokata and
and controls the expression of Shh in elevation (Lan et al., 2004). The ex- Maas, 1994; Zhao et al., 1999; Ito et
the palatal epithelium. Shh in turn pression of Fgfr2 is focused on the me- al., 2003; Rice et al., 2004). (5) Persis-
regulates the expression of Bmp2 in dial aspect of the developing palatal tence of medial edge epithelium (Fig.
the mesenchyme to promote cell prolif- shelf, suggesting a possible functional 7). In Tgfb3 or Egfr mutant mice,
eration (Zhang et al., 2002). Meanwhile, significance in regulating the develop- there is an alteration of the fate of
FGF10 is expressed in the anterior pal- ment and elevation of palatal shelf. MEE cells (Kaartinen et al., 1995;
atal mesenchyme and functions in a In analyzing different mutant ani- Proetzel et al., 1995; Miettinen et al.,
paracrine manner through its receptor mal models with cleft palate, we pro- 1999). In Tgfb3 null mutant mice,
FGFR2 in the palatal epithelium to me- pose to divide the palatal shelf devel- MEE cells fail to undergo apoptosis
diate Shh expression, which in turn reg- opment defects into the following five and persist along the midline to pre-
ulates Bmp2 expression in the mesen- categories. (1) Failure of palatal shelf vent normal fusion. In addition to its
chyme to promote cell proliferation. So formation (Fig. 7). Although this is a function in regulating the fate of
the BMP and FGF signaling pathways severe type of palatal shelf develop- MEE, TGF-␤3 is also critical for
converge on Shh signaling in the epithe- ment defect, it has a rare occurrence. proper proliferation of the CNC-de-
lium to control the growth of the ante- Mutation of activin-␤A causes a se- rived palatal mesenchyme (our un-
rior region of the palatal shelf. The vere facial primordia development de- published data). Furthermore, muta-
Shox2 gene is exclusively expressed in fect, which is likely responsible for the tions in TGF-␤3 have been associated
the anterior palatal mesenchyme. Loss retardation of palatal shelf develop- with cleft palate in humans, under-
of Shox2 results in an incomplete cleft ment and complete cleft palate (Mat- scoring the crucial function of TGF-␤
of the anterior hard palate, whereas fu- zuk et al., 1995). The Fgfr2 mutation signaling in regulating palatogenesis
sion of the posterior palate is normal also affects the initial development of (Lidral et al., 1998).
(Yu et al., 2005). Significantly, this the palatal shelf and results in com-
study clearly demonstrates that there plete cleft palate (Rice et al., 2004). (2)
are different regulatory mechanisms Fusion of the palatal shelf with the
PROSPECTUS
that control the development and fusion tongue or mandible (Fig. 7). For exam- Each year, approximately 250,000 in-
of the anterior and posterior parts of the ple, loss of function mutation of Fgf10 fants born in the United States have
palate and that a successful fusion of results in anterior palatal shelf fusion some mental or physical defects.
the posterior part of the palate can oc- with the tongue, whereas the middle Three fourths of all malformations
cur, despite that there is a failure of and posterior part of the palatal shelf seen at birth involve craniofacial dys-
anterior palate fusion. adheres to the mandible, thus pre- morphogenesis, affecting the develop-
We know less about the specific venting the elevation of the palatal ment of head, face, or neck. These
gene expression patterns in the poste- shelf (Alappat et al., 2005). In hu- malformations are particularly devas-
rior region of the palate. Fgfr2 is ex- mans, mutations in TBX22 have been tating, as our faces are our identity—
pressed in the epithelium and the reported in families with X-linked they are how we see ourselves and
CNC-derived mesenchyme in the mid- cleft palate and ankyloglossia (Bray- how others see us. In recent years,
dle and posterior palate. FGF8 signal- brook et al., 2001). Similarly, Tbx22 is research has progressed so that we
ing selectively induces the expression expressed in the developing palate know the precise genetic error that
of Pax9 in the posterior region of pal- and tongue in mice, suggesting an im- leads to many craniofacial birth de-
atal mesenchyme. Loss of Pax9 results portant role of Tbx22 in regulating fects.
in a palatal shelf development defect palate and tongue development (Bush At the conclusion of a recent Gordon
and cleft palate (Peters et al., 1998; et al., 2002). (3) Failure of palatal Research Conference on Craniofacial
Hilliard et al., 2005). To date, there is shelf elevation (Fig. 7). Studies have Morphogenesis and Tissue Regenera-
little known about the regulation of shown that mutations of Pax9, Pitx1, tion (Ventura, CA), it was clear that
Pax9 expression in the developing pal- or Osr2 can lead to failed palatal shelf there has been tremendous interest
ate. Therefore, the biological signifi- elevation and cleft palate defect (Pe- and development in recent years to-
cance of FGF8-mediated Pax9 expres- ters et al., 1998; Szeto et al., 1999; Lan ward a better understanding of the
sion during palatogenesis remains to et al., 2004). The cellular defect is molecular regulatory mechanism of
be determined. In addition to the dif- mainly associated with the CNC-de- craniofacial development. Develop-
ferential gene expression patterns rived palatal mesenchyme, suggesting mental and evolutionary biologists as
along the A-P axis of the developing important functions of these tran- well as tissue engineers are working
palate, there is also mesenchymal het- scription factors in regulating the fate together to investigate and compare
erogeneity between the medial and of CNC cells during palatogenesis. (4) the tissue origin, patterning, and
lateral regions of the palatal shelf Failure of palatal shelves to meet af- growth of various craniofacial organs
(Fig. 7A). For example, the odd ter elevation (Fig. 7). By far, this is the in an effort to reproduce and/or repair
skipped-related genes Osr1 and Osr2 most common type of cleft palate de- defective tissue in the craniofacial re-
are expressed in a medial–lateral gra- fect documented in animal studies. gion.
2370 CHAI AND MAXSON

To date, only approximately one developmental and evolutionary biol- publications were not cited due to
third of mouse mutations associated ogy (Chai and Slavkin, 2003). The re- space limitations. Both Dr. Chai’s lab
with craniofacial malformations have cent convergence of the human ge- and Dr. Maxson’s lab were supported
been linked to mutations of the or- nome project and scientific advances by the NIH.
thologous human genes with similar toward understanding the molecular
dysmorphogenesis (Wilkie and Mor- regulation of craniofacial morphogen-
riss-Kay, 2001). It is not known what esis, stem cell biology, and biotechnol- REFERENCES
defects might arise from mutations of ogy offer unprecedented opportunities Abzhanov A, Tzahor E, Lassar AB, Tabin
the other two thirds as some of these to realize craniofacial tissue regener- CJ. 2003. Dissimilar regulation of cell
mutations in mice result in early em- ation. One of the next critical steps differentiation in mesencephalic (cra-
bryonic lethality. This lethality dem- will be to apply our knowledge of mo- nial) and sacral (trunk) neural crest cells
onstrates the important function of lecular regulation of tooth morpho- in vitro. Development 130:4567–4579.
Abzhanov A, Protas M, Grant BR, Grant
these genes in early embryonic devel- genesis, for example, to manipulate PR, Tabin CJ. 2004. Bmp4 and morpho-
opment but makes it impossible to in- adult stem cells toward an odonto- logical variation of beaks in Darwin’s
vestigate the functional significance of genic phenotype. finches. Science 305:1462–1465.
that particular gene in regulating Significant progress has been made Alappat SR, Zhang Z, Suzuki K, Zhang X,
Liu H, Jiang R, Yamada G, Chen Y.
craniofacial development. It is con- in stem cell biological research, which
2005. The cellular and molecular etiol-
ceivable that the equivalent muta- has advanced our understanding in ogy of the cleft secondary palate in Fgf10
tions in human will also result in a the area of hematopoiesis, tissue engi- mutant mice. Dev Biol 277:102–113.
lethal phenotype. Nevertheless, an neering, and biomaterials (Lovell- Andl T, Ahn K, Kairo A, Chu EY, Wine-Lee
important conclusion from studies us- Badge, 2001). Recently, studies have L, Reddy ST, Croft NJ, Cebra-Thomas
JA, Metzger D, Chambon P, Lyons KM,
ing various animal models suggests shown that adult stem cells have a Mishina Y, Seykora JT, Crenshaw EB
that there are no “craniofacial genes.” much higher degree of developmental III, Millar SE. 2004. Epithelial Bmpr1a
The same morphogen (BMP, TGF-␤, potential than previously thought, regulates differentiation and prolifera-
FGF, Shh, and so on) that controls and this has prompted considerations tion in postnatal hair follicles and is es-
sential for tooth development. Develop-
limb development, for example, also to explore the potential of stem cell
ment 131:2257–2268.
plays a crucial role in regulating mediated muscle, bone, cartilage, and Aoki TO, David NB, Minchiotti G, Saint-
craniofacial morphogenesis. Clearly, dentin regeneration (Gronthos et al., Etienne L, Dickmeis T, Persico GM,
the functional specificity and the de- 2000; Bianco et al., 2001; Bianco and Strahle U, Mourrain P, Rosa FM. 2002.
velopmental outcome of a signaling Robey, 2001). Stem cells are truly re- Molecular integration of casanova in the
Nodal signalling pathway controlling
molecule is determined by the cell markable. They have the potential to endoderm formation. Development 129:
upon which the morphogen acts, as grow into an array of specialized cells 275–286.
different cell types are exposed to a and hold great promise for treating Baker CV, Bronner-Fraser M. 2001. Verte-
different combination of growth and medical and dental conditions. Sys- brate cranial placodes I. Embryonic in-
transcription factors and may respond temically injected mouse bone marrow duction. Dev Biol 232:1–61.
Basch ML, Garcia-Castro MI, Bronner-
differently to the same growth factor derived cells have given rise to mus- Fraser M. 2004. Molecular mechanisms
signaling (“Ask not what TGF-␤ can cle, cartilage, bone, liver, heart, brain, of neural crest induction. Birth Defects
do for the cell, ask what the cell can do lung, alveolar epithelium, intestine, Res C Embryo Today 72:109 –123.
with TGF-␤,” Joan Massague, per- and, of course, hematocytes (Ferrari et Beddington RS, Robertson EJ. 1999. Axis
development and early asymmetry in
sonal communication). Another im- al., 1998; Lagasse et al., 2000; Wood-
mammals. Cell 96:195–209.
portant lesson from animal studies is bury et al., 2000; Kotton et al., 2001). Bianco P, Robey PG. 2001. Stem cells in
the available dosage of an important Although most of these animal studies tissue engineering. Nature 414:118 –121.
regulatory gene, as haploinsufficiency serve now as precursors for future hu- Bianco P, Riminucci M, Gronthos S, Robey
or gain of function of a critical gene is man clinical trials in treating certain PG. 2001. Bone marrow stromal stem
cells: nature, biology, and potential ap-
often associated with congenital mal- medical and dental conditions, the ba- plications. Stem Cells 19:180 –192.
formations in humans. Overall, stud- sic scientific principles learned from Bitgood MJ, McMahon AP. 1995. Hedge-
ies using mutant mice have signifi- these current analyses certainly have hog and Bmp genes are coexpressed at
cantly improved our understanding of advanced our understanding of the bi- many diverse sites of cell– cell interac-
human embryogenesis in general and ological regulation of tissue engineer- tion in the mouse embryo. Dev Biol 172:
126 –138.
craniofacial development in particu- ing. The 21st century provides us with Braybrook C, Doudney K, Marcano AC, Ar-
lar. It is of paramount importance tremendous opportunities to enable nason A, Bjornsson A, Patton MA, Good-
that there are constant interactions biological solutions to biological prob- fellow PJ, Moore GE, Stanier P. 2001.
among researchers involved in both lems. The T-box transcription factor gene
TBX22 is mutated in X-linked cleft pal-
human and animal studies as this will
ate and ankyloglossia. Nat Genet 29:179 –
help to ensure the rapid progress to- 183.
ward a better understanding, treat- ACKNOWLEDGMENTS Bronner-Fraser M. 1993. Neural crest cell
ment, and prevention of human con- We thank Pablo Bringas, Jr., Jun migration in the developing embryo.
genital malformations. Han, Ryoichi Hosokawa, Julie Mayo, Trends Cell Biol 3:392–397.
Brown CB, Wenning JM, Lu MM, Epstein
Tissue regeneration is another area Kyoko Oka, and Xun Xu for their as- DJ, Meyers EN, Epstein JA. 2004. Cre-
with many exciting new developments sistance with this manuscript. We mediated excision of Fgf8 in the Tbx1
based on research advancements in apologize to those colleagues whose expression domain reveals a critical role
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2371

for Fgf8 in cardiovascular development phalic mesoderm in quail-chick chime- crest and paraxial mesoderm cells. Dev
in the mouse. Dev Biol 267:190 –202. ras. Development 114:1–15. Dyn.
Brugger SM, Merrill AE, Torres-Vazquez Couly GF, Coltey PM, Le Douarin NM. Farbman AI. 1968. Electron microscope
J, Wu N, Ting MC, Cho JY, Dobias SL, Yi 1993. The triple origin of skull in higher study of palate fusion in mouse embryos.
SE, Lyons K, Bell JR, Arora K, Warrior vertebrates: a study in quail-chick chi- Dev Biol 18:93–116.
R, Maxson R. 2004. A phylogenetically meras. Development 117:409 –429. Ferguson MW. 1988. Palate development.
conserved cis-regulatory module in the Couly G, Creuzet S, Bennaceur S, Vincent Development 103(Suppl):41– 60.
Msx2 promoter is sufficient for BMP-de- C, Le Douarin NM. 2002. Interactions Ferguson MW. 1994. Craniofacial malfor-
pendent transcription in murine and between Hox-negative cephalic neural mations: towards a molecular under-
Drosophila embryos. Development 131: crest cells and the foregut endoderm in standing. Nat Genet 6:329 –330.
5153–5165. patterning the facial skeleton in the ver- Ferguson CA, Graham A. 2004. Redefining
Bush JO, Lan Y, Maltby KM, Jiang R. tebrate head. Development 129:1061– the head-trunk interface for the neural
2002. Isolation and developmental ex- 1073. crest. Dev Biol 269:70 –80.
pression analysis of Tbx22, the mouse Creuzet S, Couly G, Le Douarin NM. 2005. Ferguson MW, Honig LS. 1984. Epithelial-
homolog of the human X-linked cleft pal- Patterning the neural crest derivatives mesenchymal interactions during verte-
ate gene. Dev Dyn 225:322–326. during development of the vertebrate brate palatogenesis. Curr Top Dev Biol
Carette MJ, Ferguson MW. 1992. The fate head: insights from avian studies. J Anat 19:138 –164.
of medial edge epithelial cells during pal- 207:447–459. Ferguson CA, Tucker AS, Christensen L,
atal fusion in vitro: an analysis by DiI Crump JG, Maves L, Lawson ND, Wein- Lau AL, Matzuk MM, Sharpe PT. 1998.
labelling and confocal microscopy. Devel- stein BM, Kimmel CB. 2004. An essen- Activin is an essential early mesenchy-
opment 114:379 –388. tial role for Fgfs in endodermal pouch mal signal in tooth development that is
Chai Y, Slavkin HC. 2003. Prospects for formation influences later craniofacial required for patterning of the murine
tooth regeneration in the 21st century: a skeletal patterning. Development 131: dentition. Genes Dev 12:2636 –2649.
perspective. Microsc Res Tech 60:469 – 5703–5716. Ferrari G, Cusella-De Angelis G, Coletta
479. Cuervo R, Covarrubias L. 2004. Death is M, Paolucci E, Stornaiuolo A, Cossu G,
Chai Y, Mah A, Crohin C, Groff S, Bringas the major fate of medial edge epithelial Mavilio F. 1998. Muscle regeneration by
P Jr, Le T, Santos V, Slavkin HC. 1994. cells and the cause of basal lamina deg- bone marrow-derived myogenic progeni-
Specific transforming growth factor-beta radation during palatogenesis. Develop- tors. Science 279:1528 –1530.
subtypes regulate embryonic mouse ment 131:15–24. Fitchett JE, Hay ED. 1989. Medial edge
Meckel’s cartilage and tooth develop- Cuervo R, Valencia C, Chandraratna RAS, epithelium transforms to mesenchyme
ment. Dev Biol 162:85–103. Covarrubias L. 2002. Programmed cell after embryonic palatal shelves fuse.
death is required for palate shelf fusion Dev Biol 131:455–474.
Chai Y, Sasano Y, Bringas P Jr, Mayo M,
and is regulated by retinoic acid. Dev Gage PJ, Suh H, Camper SA. 1999. Dosage
Kaartinen V, Heisterkamp N, Groffen J,
Biol 245:145–156. requirement of Pitx2 for development of
Slavkin H, Shuler C. 1997. Characteriza-
tion of the fate of midline epithelial cells Dassule HR, McMahon AP. 1998. Analysis multiple organs. Development 126:4643–
during the fusion of mandibular promi- of epithelial-mesenchymal interactions 4651.
nences in vivo. Dev Dyn 208:526 –535. in the initial morphogenesis of the mam- Gavalas A, Trainor P, Ariza-McNaughton
malian tooth. Dev Biol 202:215–227. L, Krumlauf R. 2001. Synergy between
Chai Y, Jiang X, Ito Y, Bringas P Jr, Han J,
Dahmann C, Basler K. 1999. Compartment Hoxa1 and Hoxb1: the relationship be-
Rowitch DH, Soriano P, McMahon AP,
boundaries: at the edge of development. tween arch patterning and the genera-
Sucov HM. 2000. Fate of the mammalian
Trends Genet 15:320 –326. tion of cranial neural crest. Development
cranial neural crest during tooth and
Depew MJ, Tucker AS, Sharpe PT. 2002a. 128:3017–3027.
mandibular morphogenesis. Develop-
Craniofacial development. In: Mouse de- Gendron-Maguire M, Mallo M, Zhang M,
ment 127:1671–1679.
velopment. Orlando: Academic Press. p Gridley T. 1993. Hoxa-2 mutant mice ex-
Chai Y, Ito Y, Han J. 2003. TGF-beta sig- 421– 498. hibit homeotic transformation of skeletal
naling and its functional significance in
Depew MJ, Lufkin T, Rubenstein JL. elements derived from cranial neural
regulating the fate of cranial neural
2002b. Specification of jaw subdivisions crest. Cell 75:1317–1331.
crest cells. Crit Rev Oral Biol Med 14:
by Dlx genes. Science 298:381–385. Graham A, Lumsden A. 1993. The role of
78 –88.
Depew MJ, Simpson CA, Morasso M, segmentation in the development of the
Chen Y, Bei M, Woo I, Satokata I, Maas R. Rubenstein JL. 2005. Reassessing the branchial region of higher vertebrate
1996. Msx1 controls inductive signaling Dlx code: the genetic regulation of embryos. Birth Defects Orig Artic Ser
in mammalian tooth morphogenesis. De- branchial arch skeletal pattern and de- 29:103–112.
velopment 122:3035–3044. velopment. J Anat 207:501–561. Graham A, Okabe M, Quinlan R. 2005. The
Chen D, Zhao M, Mundy GR. 2004. Bone Dolle P, Price M, Duboule D. 1992. Expres- role of the endoderm in the development
morphogenetic proteins. Growth Factors sion of the murine Dlx-1 homeobox gene and evolution of the pharyngeal arches.
22:233–241. during facial, ocular and limb develop- J Anat 207:479 –487.
Clouthier DE, Hosoda K, Richardson JA, ment. Differentiation 49:93–99. Griffith CM, Hay ED. 1992. Epithelial-
Williams SC, Yanagisawa H, Kuwaki T, Dorsky RI, Moon RT, Raible DW. 2000. mesenchymal transformation during
Kumada M, Hammer RE, Yanagisawa Environmental signals and cell fate palatal fusion: carboxyfluorescein traces
M. 1998. Cranial and cardiac neural specification in premigratory neural cells at light and electron microscopic
crest defects in endothelin-A receptor- crest. Bioessays 22:708 –716. levels. Development 116:1087–1099.
deficient mice. Development 125:813– Dudas M, Kaartinen V. 2005. Tgf-beta su- Grigoriou M, Tucker AS, Sharpe PT, Pach-
824. perfamily and mouse craniofacial devel- nis V. 1998. Expression and regulation of
Cobourne MT, Sharpe PT. 2003. Tooth and opment: interplay of morphogenetic pro- Lhx6 and Lhx7, a novel subfamily of LIM
jaw: molecular mechanisms of pattern- teins and receptor signaling controls homeodomain encoding genes, suggests
ing in the first branchial arch. Arch Oral normal formation of the face. Curr Top a role in mammalian head development.
Biol 48:1–14. Dev Biol 66:65–133. Development 125:2063–2074.
Cobourne MT, Hardcastle Z, Sharpe PT. Echelard Y, Vassileva G, McMahon AP. Gronthos S, Mankani M, Brahim J, Robey
2001. Sonic hedgehog regulates epithe- 1994. Cis-acting regulatory sequences PG, Shi S. 2000. Postnatal human dental
lial proliferation and cell survival in the governing Wnt-1 expression in the devel- pulp stem cells (DPSCs) in vitro and in
developing tooth germ. J Dent Res 80: oping mouse CNS. Development 120:2213– vivo. Proc Natl Acad Sci U S A 97:13625–
1974 –1979. 2224. 13630.
Couly GF, Coltey PM, Le Douarin NM. Evans DJ, Noden DM. 2006. Spatial rela- Gross JB, Hanken J. 2005. Cranial neural
1992. The developmental fate of the ce- tions between avian craniofacial neural crest contributes to the bony skull vault
2372 CHAI AND MAXSON

in adult Xenopus laevis: insights from Ito Y, Bringas P Jr, Mogharei A, Zhao J, the factors behind evolutionary novelty.
cell labeling studies. J Exp Zool B Mol Deng C, Chai Y. 2002. Receptor-regu- J Anat 205:335–347.
Dev Evol 304:169 –176. lated and inhibitory Smads are critical in Kuratani S. 2005. Developmental studies
Hagedorn L, Floris J, Suter U, Sommer L. regulating transforming growth factor of the lamprey and hierarchical evolu-
2000. Autonomic neurogenesis and apo- beta-mediated Meckel’s cartilage devel- tionary steps towards the acquisition of
ptosis are alternative fates of progenitor opment. Dev Dyn 224:69 –78. the jaw. J Anat 207:489 –499.
cell communities induced by TGFbeta. Ito Y, Yeo JY, Chytil A, Han J, Bringas P Kuratani S, Nobusada Y, Horigome N,
Dev Biol 228:57–72. Jr, Nakajima A, Shuler CF, Moses HL, Shigetani Y. 2001. Embryology of the
Hall BD, Cadle RG, Golabi M, Morris CA, Chai Y. 2003. Conditional inactivation of lamprey and evolution of the vertebrate
Cohen MM Jr. 1992. Beare-Stevenson Tgfbr2 in cranial neural crest causes jaw: insights from molecular and devel-
cutis gyrata syndrome. Am J Med Genet cleft palate and calvaria defects. Devel- opmental perspectives. Philos Trans R
44:82–89. opment 130:5269 –5280. Soc Lond B Biol Sci 356:1615–1632.
Han J, Ito Y, Yeo JY, Sucov HM, Maas R, Jacobson AG. 1963. The determination and LaBonne C, Bronner-Fraser M. 1999. Mo-
Chai Y. 2003. Cranial neural crest-de- positioning of the nose, lens and ear. I. lecular mechanisms of neural crest for-
rived mesenchymal proliferation is regu- Interactions within the ectoderm, and mation. Annu Rev Cell Dev Biol 15:81–
lated by Msx1-mediated p19(INK4d) ex- between the ectoderm and underlying 112.
pression during odontogenesis. Dev Biol tissues. J Exp Zool 154:273–283. Lagasse E, Connors H, Al-Dhalimy M, Re-
261:183–196. Jeffery WR, Strickler AG, Yamamoto Y. itsma M, Dohse M, Osborne L, Wang X,
Hardcastle Z, Mo R, Hui CC, Sharpe PT. 2004. Migratory neural crest-like cells Finegold M, Weissman IL, Grompe M.
1998. The Shh signalling pathway in form body pigmentation in a urochordate 2000. Purified hematopoietic stem cells
tooth development: defects in Gli2 and embryo. Nature 431:696 –699. can differentiate into hepatocytes in
Gli3 mutants. Development 125:2803– Jernvall J, Thesleff I. 2000. Reiterative sig- vivo. Nat Med 6:1229 –1234.
2811. naling and patterning during mamma- Lan Y, Ovitt CE, Cho ES, Maltby KM,
Haworth KE, Healy C, Morgan P, Sharpe lian tooth morphogenesis. Mech Dev 92: Wang Q, Jiang R. 2004. Odd-skipped re-
PT. 2004. Regionalisation of early head 19 –29. lated 2 (Osr2) encodes a key intrinsic
ectoderm is regulated by endoderm and Jerome LA, Papaioannou VE. 2001. Di- regulator of secondary palate growth and
prepatterns the orofacial epithelium. De- George syndrome phenotype in mice mu- morphogenesis. Development 131:3207–
velopment 131:4797–4806. tant for the T-box gene, Tbx1. Nat Genet 3216.
Headon DJ, Emmal SA, Ferguson BM, 27:286 –291. Le Douarin NM, Ziller C, Couly GF. 1993.
Tucker AS, Justice MJ, Sharpe PT, Zo- Jiang X, Iseki S, Maxson RE, Sucov HM, Patterning of neural crest derivatives in
nana J, Overbeek PA. 2001. Gene defect Morriss-Kay GM. 2002. Tissue origins the avian embryo: in vivo and in vitro
in ectodermal dysplasia implicates a and interactions in the mammalian skull studies. Dev Biol 159:24 –49.
death domain adapter in development. vault. Dev Biol 241:106 –116. Le Douarin NM, Creuzet S, Couly G, Dupin
Nature 414:913–916. Jollie M. 1981. Segment theory and the E. 2004. Neural crest cell plasticity and
Helms JA, Cordero D, Tapadia MD. 2005. homologizing of cranial bone. Am Nat its limits. Development 131:4637–4650.
New insights into craniofacial morpho- 118:785–802. Le Lievre CS. 1978. Participation of neural
genesis. Development 132:851–861. Kaartinen V, Voncken JW, Shuler C, War- crest-derived cells in the genesis of the
Hilliard SA, Yu L, Gu S, Zhang Z, Chen YP. burton D, Bu D, Heisterkamp N, Groffen skull in birds. J Embryol Exp Morphol
2005. Regional regulation of palatal J. 1995. Abnormal lung development and 47:17–37.
growth and patterning along the anteri- cleft palate in mice lacking TGF-beta 3 Lee SH, Bedard O, Buchtova M, Fu K,
or-posterior axis in mice. J Anat 207:655– indicates defects of epithelial-mesenchy- Richman JM. 2004. A new origin for the
667. mal interaction. Nat Genet 11:415–421. maxillary jaw. Dev Biol 276:207–224.
Holland LZ, Laudet V, Schubert M. 2004. Kassai Y, Munne P, Hotta Y, Penttila E, Lidral AC, Romitti PA, Basart AM,
The chordate amphioxus: an emerging Kavanagh K, Ohbayashi N, Takada S, Doetschman T, Leysens NJ, Daack-Hir-
model organism for developmental biol- Thesleff I, Jernvall J, Itoh N. 2005. Reg- sch S, Semina EV, Johnson LR, Machida
ogy. Cell Mol Life Sci 61:2290 –308. ulation of mammalian tooth cusp pat- J, Burds A, Parnell TJ, Rubenstein JL,
Hu G, Lee H, Price SM, Shen MM, Abate- terning by ectodin. Science 309:2067– Murray JC. 1998. Association of MSX1
Shen C. 2001. Msx homeobox genes in- 2070. and TGFB3 with nonsyndromic clefting
hibit differentiation through upregula- Kee Y, Bronner-Fraser M. 2005. To prolif- in humans. Am J Hum Genet 63:557–
tion of cyclin D1. Development 128:2373– erate or to die: role of Id3 in cell cycle 568.
2384. progression and survival of neural crest Light W, Vernon AE, Lasorella A, Iavarone
Huang R, Zhi Q, Izpisua-Belmonte JC, progenitors. Genes Dev 19:744 –755. A, LaBonne C. 2005. Xenopus Id3 is re-
Christ B, Patel K. 1999. Origin and de- Kollar EJ. 1972. Histogenetic aspects of quired downstream of Myc for the forma-
velopment of the avian tongue muscles. dermal-epidermal interactions. In: De- tion of multipotent neural crest progeni-
Anat Embryol (Berl) 200:137–152. velopmental aspects of oral biology. New tor cells. Development 132:1831–1841.
Imai H, Osumi-Yamashita N, Ninomiya Y, York: Academic Press. p 126 –149. Lindsay EA, Vitelli F, Su H, Morishima M,
Eto K. 1996. Contribution of early-emi- Kontges G, Lumsden A. 1996. Rhombence- Huynh T, Pramparo T, Jurecic V, Ogun-
grating midbrain crest cells to the dental phalic neural crest segmentation is pre- rinu G, Sutherland HF, Scambler PJ,
mesenchyme of mandibular molar teeth served throughout craniofacial ontogeny. Bradley A, Baldini A. 2001. Tbx1 haplo-
in rat embryos. Dev Biol 176:151–165. Development 122:3229 –3242. insufficieny in the DiGeorge syndrome
Ishii M, Merrill AE, Chan YS, Gitelman I, Kotton DN, Ma BY, Cardoso WV, Sander- region causes aortic arch defects in mice.
Rice DP, Sucov HM, Maxson RE Jr. son EA, Summer RS, Williams MC, Fine Nature 410:97–101.
2003. Msx2 and Twist cooperatively con- A. 2001. Bone marrow-derived cells as Liu W, Selever J, Lu MF, Martin JF. 2003.
trol the development of the neural crest- progenitors of lung alveolar epithelium. Genetic dissection of Pitx2 in craniofa-
derived skeletogenic mesenchyme of the Development 128:5181–5188. cial development uncovers new functions
murine skull vault. Development 130: Kratochwil K, Dull M, Farinas I, Galceran in branchial arch morphogenesis, late
6131–6142. J, Grosschedl R. 1996. Lef1 expression is aspects of tooth morphogenesis and cell
Ishii M, Han J, Yen HY, Sucov HM, Chai Y, activated by BMP-4 and regulates induc- migration. Development 130:6375–6385.
Maxson RE Jr. 2005. Combined deficien- tive tissue interactions in tooth and hair Liu W, Selever J, Murali D, Sun X, Brugger
cies of Msx1 and Msx2 cause impaired development. Genes Dev 10:1382–1394. SM, Ma L, Schwartz RJ, Maxson R, Fu-
patterning and survival of the cranial Kuratani S. 2004. Evolution of the verte- ruta Y, Martin JF. 2005. Threshold-spe-
neural crest. Development 132:4937– brate jaw: comparative embryology and cific requirements for Bmp4 in mandib-
4950. molecular developmental biology reveal ular development. Dev Biol 283:282–293.
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2373

Lovell-Badge R. 2001. The future for stem Mitsiadis TA, Angeli I, James C, Lendahl cial malformations. Cleft Palate Craniofac
cell research. Nature 414:88 –91. U, Sharpe PT. 2003. Role of Islet1 in the J 36:12–26.
Lu MF, Pressman C, Dyer R, Johnson RL, patterning of murine dentition. Develop- Nugent P, Greene RM. 1998. MSX-1 gene
Martin JF. 1999. Function of Rieger syn- ment 130:4451–4460. expression and regulation in embryonic
drome gene in left-right asymmetry and Mootoosamy RC, Dietrich S. 2002. Distinct palatal tissue. In Vitro Cell Dev Biol
craniofacial development. Nature 401: regulatory cascades for head and trunk Anim 34:831–835.
276 –278. myogenesis. Development 129:573–583. Osumi-Yamashita N, Noji S, Nohno T,
Lumsden A. 1984. Tooth morphogenesis: Morriss-Kay GM, Wilkie AO. 2005. Growth Koyama E, Doi H, Eto K, Taniguchi S.
contributions of the cranial neural crest of the normal skull vault and its alter- 1990. Expression of retinoic acid recep-
in mammals. In: Tooth morphogenesis ation in craniosynostosis: insights from tor genes in neural crest-derived cells
and differentiation. Paris: Colloq. IN- human genetics and experimental stud- during mouse facial development. FEBS
SERM 125. ies. J Anat 207:637–653. Lett 264:71–74.
Lumsden AG. 1988. Spatial organization of Murray JC. 2002. Gene/environment causes Packard DS Jr, Meier S. 1983. An experi-
the epithelium and the role of neural of cleft lip and/or palate. Clin Genet 61: mental study of the somitomeric organi-
crest cells in the initiation of the mam- 248 –256. zation of the avian segmental plate. Dev
malian tooth germ. Development Murray JC, Schutte BC. 2004. Cleft palate: Biol 97:191–202.
103(Suppl):155–169. players, pathways, and pursuits. J Clin Peters H, Neubuser A, Kratochwil K, Ball-
Lumsden A, Krumlauf R. 1996. Patterning Invest 113:1676 –1678. ing R. 1998. Pax9-deficient mice lack
the vertebrate neuraxis. Science 274: Mustonen T, Pispa J, Mikkola ML, Pum- pharyngeal pouch derivatives and teeth
1109 –1115. mila M, Kangas AT, Pakkasjarvi L, and exhibit craniofacial and limb abnor-
Martinez-Alvarez C, Tudela C, Perez- Jaatinen R, Thesleff I. 2003. Stimulation malities. Genes Dev 12:2735–2747.
Miguelsanz J, O’Kane S, Puerta J, Fer- of ectodermal organ development by Ec- Pispa J, Thesleff I. 2003. Mechanisms of
guson MW. 2000. Medial edge epithelial todysplasin-A1. Dev Biol 259:123–136. ectodermal organogenesis. Dev Biol 262:
cell fate during palatal fusion. Dev Biol Niederreither K, Subbarayan V, Dolle P, 195–205.
220:343–357. Chambon P. 1999. Embryonic retinoic Plikus MV, Zeichner-David M, Mayer JA,
Massague J. 2000. How cells read TGF- acid synthesis is essential for early Reyna J, Bringas P, Thewissen JG,
beta signals. Nat Rev Mol Cell Biol 1: mouse post-implantation development. Snead ML, Chai Y, Chuong CM. 2005.
169 –178. Nat Genet 21:444 –448. Morphoregulation of teeth: modulating
Massague J, Seoane J, Wotton D. 2005. Niederreither K, Vermot J, Le Roux I, the number, size, shape and differentia-
Smad transcription factors. Genes Dev Schuhbaur B, Chambon P, Dolle P. 2003. tion by tuning Bmp activity. Evol Dev
19:2783–2810. The regional pattern of retinoic acid syn- 7:440 –457.
Matsuo I, Kuratani S, Kimura C, Takeda thesis by RALDH2 is essential for the
N, Aizawa S. 1995. Mouse Otx2 func- Proetzel G, Pawlowski SA, Wiles MV, Yin
development of posterior pharyngeal M, Boivin GP, Howles PN, Ding J, Fer-
tions in the formation and patterning of arches and the enteric nervous system.
rostral head. Genes Dev 9:2646 –2658. guson MW, Doetschman T. 1995. Trans-
Development 130:2525–2534. forming growth factor-beta 3 is required
Matsuoka T, Ahlberg PE, Kessaris N, Noden DM. 1975. An analysis of migratory for secondary palate fusion. Nat Genet
Iannarelli P, Dennehy U, Richardson behavior of avian cephalic neural crest 11:409 –414.
WD, McMahon AP, Koentges G. 2005. cells. Dev Biol 42:106 –130.
Neural crest origins of the neck and Qiu M, Bulfone A, Martinez S, Meneses JJ,
Noden DM. 1978. The control of avian ce- Shimamura K, Pedersen RA, Rubenstein
shoulder. Nature 436:347–355.
phalic neural crest cytodifferentiation. I. JL. 1995. Null mutation of Dlx-2 results
Matzuk MM, Kumar TR, Vassalli A, Bick- Skeletal and connective tissues. Dev Biol
enbach JR, Roop DR, Jaenisch R, Brad- in abnormal morphogenesis of proximal
67:296 –312. first and second branchial arch deriva-
ley A. 1995. Functional analysis of ac-
Noden DM. 1983. The role of the neural tives and abnormal differentiation in the
tivins during mammalian development.
crest in patterning of avian cranial skel- forebrain. Genes Dev 9:2523–2538.
Nature 374:354 –356.
etal, connective, and muscle tissues. Dev Qiu M, Bulfone A, Ghattas I, Meneses JJ,
Merrill AE, Bochukova EG, Brugger SM,
Biol 96:144 –165. Christensen L, Sharpe PT, Presley R,
Ishii M, Pitz DT, Wall SA, Lyons KM,
Wilkie AM, Maxson RE. 2006. Cell mix- Noden DM. 1986. Origins and patterning Pedersen RA, Rubenstein JLR. 1997.
ing at a neural crest-mesoderm bound- of craniofacial mesenchymal tissues. J Role of the Dlx homeobox genes in proxi-
ary and deficient ephrin-Eph signaling Craniofac Genet Dev Biol Suppl 2:15–31. modistal patterning of the branchial
in the pathogenesis of craniosynostosis. Noden DM. 1988. Interactions and fates of arches: mutations of Dlx-1, Dlx-2, and
Hum Mol Genet 15:1319 –1328. avian craniofacial mesenchyme. Devel- Dlx-1 and -2 alter morphogenesis of
Meulemans D, Bronner-Fraser M. 2004. opment 103(Suppl):121–140. proximal skeletal and soft tissue struc-
Gene-regulatory interactions in neural Noden DM. 1991. Cell movements and con- tures derived from the first and second
crest evolution and development. Dev trol of patterned tissue assembly during arches. Dev Biol 185:165–184.
Cell 7:291–299. craniofacial development. J Craniofac Rice R, Spencer-Dene B, Connor EC, Gritli-
Miettinen PJ, Chin JR, Shum L, Slavkin Genet Dev Biol 11:192–213. Linde A, McMahon AP, Dickson C,
HC, Shuler CF, Derynck R, Werb Z. Noden DM, Trainor PA. 2005. Relations Thesleff I, Rice DPC. 2004. Disruption of
1999. Epidermal growth factor receptor and interactions between cranial meso- Fgf10/Fgfr2b-coordinated epithelial-mes-
function is necessary for normal cranio- derm and neural crest populations. J enchymal interactions causes cleft palate.
facial development and palate closure. Anat 207:575–601. J Clin Invest 113:1692–1700.
Nat Genet 22:69 –73. Nomura M, Li E. 1998. Smad2 role in me- Rieger HH. 1935. Beitrage zur kenntnis
Miletich I, Sharpe PT. 2004. Neural crest soderm formation, left-right patterning seltener Missbildungen der Iris. Graefes
contribution to mammalian tooth forma- andcraniofacialdevelopment.Nature393: Arch Clin Exp Ophthalmol 133:602–635.
tion. Birth Defects Res C Embryo Today 786 –790. Rijli FM, Mark M, Lakkaraju S, Dierich A,
72:200 –212. Northcutt RG. 2005. Taste bud develop- Dolle P, Chambon P. 1993. A homeotic
Miletich I, Buchner G, Sharpe PT. 2005. ment in the channel catfish. J Comp transformation is generated in the ros-
Barx1 and evolutionary changes in feed- Neurol 482:1–16. tral branchial region of the head by dis-
ing. J Anat 207:619 –622. Northcutt RG, Gans C. 1983. The genesis ruption of Hoxa-2, which acts as a selec-
Mina M, Kollar EJ. 1987. The induction of of neural crest and epidermal placodes: a tor gene. Cell 75:1333–1349.
odontogenesis in non-dental mesen- reinterpretation of vertebrate origins. Q Robinson GW, Mahon KA. 1994. Differen-
chyme combined with early murine man- Rev Biol 58:1–28. tial and overlapping expression domains
dibular arch epithelium. Arch Oral Biol Nuckolls GH, Shum L, Slavkin HC. 1999. of Dlx-2 and Dlx-3 suggest distinct roles
32:123–127. Progress toward understanding craniofa- for Distal-less homeobox genes in cranio-
2374 CHAI AND MAXSON

facial development. Mech Dev 48:199 – Serbedzija GN, Bronner-Fraser M, Fraser man AS, Izpisua-Belmonte JC, Rosen-
215. SE. 1992. Vital dye analysis of cranial feld MG. 1999. Role of the Bicoid-related
Ruhin B, Creuzet S, Vincent C, Benouaiche neural crest cell migration in the mouse homeodomain factor Pitx1 in specifying
L, Le Douarin NM, Couly G. 2003. Pat- embryo. Development 116:297–307. hindlimb morphogenesis and pituitary
terning of the hyoid cartilage depends Shah NM, Groves AK, Anderson DJ. 1996. development. Genes Dev 13:484 –494.
upon signals arising from the ventral fo- Alternative neural crest cell fates are in- Thomas BL, Tucker AS, Qui M, Ferguson
regut endoderm. Dev Dyn 228:239 –246. structively promoted by TGFbeta super- CA, Hardcastle Z, Rubenstein JL,
Sanford L, Ormsby I, Gittenberger-de family members. Cell 85:331–343. Sharpe PT. 1997. Role of Dlx-1 and Dlx-2
Groot A, Sariola H, Friedman R, Boivin Shigetani Y, Sugahara F, Kawakami Y, genes in patterning of the murine denti-
G, Cardell E, Doetschman T. 1997. TG- Murakami Y, Hirano S, Kuratani S. tion. Development 124:4811–4818.
Fbeta2 knockout mice have multiple de- 2002. Heterotopic shift of epithelial-mes- Thyagarajan T, Totey S, Danton MJ,
velopmental defects that are non-over- enchymal interactions in vertebrate jaw Kulkarni AB. 2003. Genetically altered
lapping with other TGFbeta knockout evolution. Science 296:1316 –1319. mouse models: the good, the bad, and the
phenotypes. Development 124:2659 – Shimamura K, Rubenstein JL. 1997. In- ugly. Crit Rev Oral Biol Med 14:154 –
2670. ductive interactions direct early region- 174.
Santagati F, Rijli FM. 2003. Cranial neural alization of the mouse forebrain. Devel- Trainor PA, Tan SS, Tam PP. 1994. Cra-
crest and the building of the vertebrate opment 124:2709 –2718. nial paraxial mesoderm: regionalisation
head. Nat Rev Neurosci 4:806 –818. Shuler CF. 1995. Programmed cell death of cell fate and impact on craniofacial
Santagati F, Minoux M, Ren SY, Rijli FM. and cell transformation in craniofacial development in mouse embryos. Devel-
2005. Temporal requirement of Hoxa2 in development. Crit Rev Oral Biol Med 6: opment 120:2397–2408.
cranial neural crest skeletal morphogen- 202–217. Trainor PA, Krumlauf R. 2000. Patterning
esis. Development 132:4927–4936. Shuler CF, Halpern DE, Guo Y, Sank AC. the cranial neural crest: hindbrain seg-
Sarkar L, Cobourne M, Naylor S, Smalley 1992. Medial edge epithelium fate traced mentation and Hox gene plasticity. Nat
M, Dale T, Sharpe PT. 2000. Wnt/Shh by cell lineage analysis during epithelial- Rev Neurosci 1:116 –124.
interactions regulate ectodermal bound- mesenchymal transformation in vivo. Trainor PA, Ariza-McNaughton L, Krum-
ary formation during mammalian tooth Dev Biol 154:318 –330. lauf R. 2002. Role of the isthmus and
development. Proc Natl Acad Sci U S A Shum L, Takahashi K, Takahashi I, Na- FGFs in resolving the paradox of neural
97:4520 –4524. gata M, Tan DP, Semba I, Tanaka O, crest plasticity and prepatterning. Sci-
Sasaki T, Ito Y, Xu X, Han J, Bringas P Jr, Bringas P, Nuckolls G, Slavkin HC. ence 295:1288 –1291.
Maeda T, Slavkin HC, Grosschedl R, 2000. Embryogenesis and the classifica- Trokovic N, Trokovic R, Mai P, Partanen J.
Chai Y. 2005. LEF1 is a critical epithe- tion of craniofacial dysmorphogenesis. 2003. Fgfr1 regulates patterning of the
lial survival factor during tooth morpho- In: Oral and maxillofacial surgery. Phil- pharyngeal region. Genes Dev 17:141–
genesis. Dev Biol 278:130 –143. adelphia: WB Saunders Co. p 149 –194. 153.
Sasaki T, Ito Y, Bringas P Jr, Chou S, Simeone A, Acampora D, Pannese M, Trokovic N, Trokovic R, Partanen J. 2005.
Urata MM, Slavkin H, Chai Y. 2006. D’Esposito M, Stornaiuolo A, Gulisano Fibroblast growth factor signalling and
TGF{beta}-mediated FGF signaling is M, Mallamaci A, Kastury K, Druck T, regional specification of the pharyngeal
crucial for regulating cranial neural Huebner K. 1994. Cloning and charac- ectoderm. Int J Dev Biol 49:797–805.
crest cell proliferation during frontal terization of two members of the verte- Trumpp A, Depew MJ, Rubenstein JLR,
bonedevelopment.Development133:371– brate Dlx gene family. Proc Natl Acad Bishop JM, Martin GR. 1999. Cre-medi-
381. Sci U S A 91:2250 –2254. ated gene inactivation demonstrates
Satokata I, Maas R. 1994. Msx1 deficient Slavkin HC, Beierle J, Bavetta LA. 1968. that FGF8 is required for cell survival
mice exhibit cleft palate and abnormali- Odontogenesis: cell– cell interactions in and patterning of the first branchial
ties of craniofacial and tooth develop- vitro. Nature 217:269 –270. arch. Genes Dev 13:3136 –3148.
ment. Nat Genet 6:348 –356. Sofaer JA. 1969. The genetics and expres- Tucker AS, Lumsden A. 2004. Neural crest
Satokata I, Ma L, Ohshima H, Bei M, Woo sion of a dental morphological variant in cells provide species-specific patterning
I, Nishizawa K, Maeda T, Takano Y, the mouse. Arch Oral Biol 14:1213–1223. information in the developing branchial
Uchiyama M, Heaney S, Peters H, Tang Sofaer JA. 1977. The teeth of the “sleek” skeleton. Evol Dev 6:32–40.
Z, Maxson R, Maas R. 2000. Msx2 defi- mouse. Arch Oral Biol 22:299 –301. Tucker AS, Sharpe PT. 1999. Molecular
ciency in mice causes pleiotropic defects Spemann H. 1938. Embryonic develop- genetics of tooth morphogenesis and pat-
in bone growth and ectodermal organ ment and induction. New Haven: Yale terning: the right shape in the right
formation. Nat Genet 24:391–395. University Press. place. J Dent Res 78:826 –834.
Saunders JW Jr. 1966. Death in embryonic St. Amand TR, Zhang Y, Semina EV, Zhao Tucker AS, Sharpe P. 2004. The cutting-
systems. Science 154:604 –612. X, Hu Y, Nguyen L, Murray JC, Chen Y. edge of mammalian development; how
Schneider RA, Helms JA. 2003. The cellu- 2000. Antagonistic signals between the embryo makes teeth. Nat Rev Genet
lar and molecular origins of beak mor- BMP4 and FGF8 define the expression of 5:499 –508.
phology. Science 299:565–568. Pitx1 and Pitx2 in mouse tooth-forming Tucker AS, Al Khamis A, Sharpe PT.
Selleck MA, Scherson TY, Bronner-Fraser anlage. Dev Biol 217:323–332. 1998a. Interactions between Bmp-4 and
M. 1993. Origins of neural crest cell di- Stock DW, Ellies DL, Zhao Z, Ekker M, Msx-1 act to restrict gene expression to
versity. Dev Biol 159:1–11. Ruddle FH, Weiss KM. 1996. The evolu- odontogenic mesenchyme. Dev Dyn 212:
Selleri L, Depew MJ, Jacobs Y, Chanda SK, tion of the vertebrate Dlx gene family. 533–539.
Tsang KY, Cheah KS, Rubenstein JL, Proc Natl Acad Sci U S A 93:10858 – Tucker AS, Matthews KL, Sharpe PT.
O’Gorman S, Cleary ML. 2001. Require- 10863. 1998b. Transformation of tooth type in-
ment for Pbx1 in skeletal patterning and Stottmann RW, Anderson RM, Klingen- duced by inhibition of BMP signaling.
programming chondrocyte proliferation smith J. 2001. The BMP antagonists Science 282:1136 –1138.
and differentiation. Development 128:3543– Chordin and Noggin have essential but Tucker AS, Yamada G, Grigoriou M, Pach-
3557. redundant roles in mouse mandibular nis V, Sharpe PT. 1999. Fgf-8 determines
Semina EV, Reiter R, Leysens NJ, Alward outgrowth. Dev Biol 240:457–473. rostral-caudal polarity in the first
WL, Small KW, Datson NA, Siegel-Bar- Sun X, Meyers EN, Lewandoski M, Martin branchial arch. Development 126:51–61.
telt J, Bierke-Nelson D, Bitoun P, Zabel GR. 1999. Targeted disruption of Fgf8 Tucker AS, Headon DJ, Courtney JM,
BU, Carey JC, Murray JC. 1996. Cloning causes failure of cell migration in the Overbeek P, Sharpe PT. 2004. The acti-
and characterization of a novel bicoid- gastrulating mouse embryo. Genes Dev vation level of the TNF family receptor,
related homeobox transcription factor 13:1834 –1846. Edar, determines cusp number and tooth
gene, RIEG, involved in Rieger syn- Szeto DP, Rodriguez-Esteban C, Ryan AK, number during tooth development. Dev
drome. Nat Genet 14:392–399. O’Connell SM, Liu F, Kioussi C, Gleiber- Biol 268:185–194.
RECENT ADVANCES IN CRANIOFACIAL MORPHOGENESIS 2375

Twigg SR, Kan R, Babbs C, Bochukova EG, Wilson J, Tucker AS. 2004. Fgf and Bmp neural crest evolution. Dev Dyn 225:289 –
Robertson SP, Wall SA, Morriss-Kay signals repress the expression of Bapx1 297.
GM, Wilkie AO. 2004. Mutations of eph- in the mandibular mesenchyme and con- Yu L, Gu S, Alappat S, Song Y, Yan M,
rin-B1 (EFNB1), a marker of tissue trol the position of the developing jaw Zhang X, Zhang G, Jiang Y, Zhang Z,
boundary formation, cause craniofronto- joint. Dev Biol 266:138 –150. Zhang Y, Chen Y. 2005. Shox2-deficient
nasal syndrome. Proc Natl Acad Sci U S Woodbury D, Schwarz EJ, Prockop DJ, mice exhibit a rare type of incomplete
A 101:8652–8657. Black IB. 2000. Adult rat and human clefting of the secondary palate. Develop-
Vaziri Sani F, Hallberg K, Harfe BD, Mc- bone marrow stromal cells differentiate ment 132:4397–4406.
Mahon AP, Linde A, Gritli-Linde A. into neurons. J Neurosci Res 61:364 – Zhang Z, Song Y, Zhao X, Zhang X, Fermin
2005. Fate-mapping of the epithelial 370. C, Chen Y. 2002. Rescue of cleft palate in
seam during palatal fusion rules out ep- Wu P, Jiang TX, Suksaweang S, Widelitz Msx1-deficient mice by transgenic Bmp4
ithelial-mesenchymal transformation. RB, Chuong CM. 2004. Molecular shap- reveals a network of BMP and Shh sig-
Developmental Biology 285:490 –495. naling in the regulation of mammalian
ing of the beak. Science 305:1465–1466.
palatogenesis. Development 129:4135–
Veitch E, Begbie J, Schilling TF, Smith Xu X, Han J, Ito Y, Bringas P Jr, Chai Y. 4146.
MM, Graham A. 1999. Pharyngeal arch 2006. Cell autonomous requirement for Zhao Y, Guo YJ, Tomac AC, Taylor NR,
patterning in the absence of neural crest. Tgfbr2 in the disappearance of Medial Grinberg A, Lee EJ, Huang S, Westphal
Curr Biol 9:1481–1484. Edge Epithelium during palatal fusion. H. 1999. Isolated cleft palate in mice
Wendling O, Dennefeld C, Chambon P, Dev Biol. (in press). with a targeted mutation of the LIM ho-
Mark M. 2000. Retinoid signaling is es- Yelick PC, Schilling TF. 2002. Molecular meobox gene lhx8. Proc Natl Acad Sci U
sential for patterning the endoderm of dissection of craniofacial development S A 96:15002–15006.
the third and fourth pharyngeal arches. using zebrafish. Crit Rev Oral Biol Med Zhao H, Bringas P Jr, Chai Y. 2006. An in
Development 127:1553–1562. 13:308 –322. vitro model for characterizing the post-
Wilkie AO, Morriss-Kay GM. 2001. Genet- Yu JK, Holland ND, Holland LZ. 2002. An migratory cranial neural crest cells of
ics of craniofacial development and mal- amphioxus winged helix/forkhead gene, the first branchial arch. Dev Dyn 235:
formation. Nat Rev Genet 2:458 –468. AmphiFoxD: insights into vertebrate 1433–1440.

You might also like