Sie sind auf Seite 1von 25

REGULATION OF MANDIBULAR GROWTrH

AND MORPHOGENESIS
Mina Mina
Department of Pediatric Dentistry, School of Dental Medicine, University of Connecticut Health (enter, Farmington, al 06030; Mina@nsoL.uchc.edu

ABSTRACT: The development of the vertebrate face is a dynamic process that starts with the formation of facial
processes/prominences. Facial processes are small buds made up of mesenchymal masses enclosed by an epithelial layer that
surround the primitive mouth. The 2 maxillary processes, the 2 lateral nasal processes, and the frontonasal processes form the
upper jaw. The lower jaw is formed by the 2 mandibular processes. Although the question of the embryonic origin of facial struc-
tures has received considerable attention, the mechanisms that control differential growth of the facial processes and patterning
of skeletal tissues within these structures have been difficult to study and still are not well-understood. This has been partially
due to the lack of readily identifiable morphologically discrete regions in the developing face that regulate patterning of the face.
Nonetheless, in recent years there has been significant progress in the understanding of the signaling network controlling the pat-
terning and development of the face (for review, see Richman et al., 1991; Francis-West et al., 1998). This review focuses on cur-
rent understanding of the processes and signaling molecules that are involved in the formation of the mandibular arch.
Key words. Mandibular processes, branchial arches, signaling molecules, epithelial-mesenchymal interactions, transcription
factors, secreted factors, chick, mouse, growth, patterning, morphogenesis.

Introduction fate-mapping (Lumsden et al., 1991; Serbedzija et al., 1992; Couly


During normal development, the lower jaw is formed from et al.,
1993, 1996, 1998; Kontges and Lumsden, 1996).
the mandibular component of the first branchial arch. The The results of these studies indicate that, in contrast to the
mandibular processes are bilaterally symmetrical structures NCC of the rest of the body, the hindbrain CNCC migrate in 3
located below the future mouth cavity and composed of mes- non-mixing separate streams from RI and R2, from R4 and from
enchymal tissue enclosed by an epithelial layer of ectodermal R6 (Lumsden et al., 1991; Osumi-Yamashita et al., 1997). The for-
and endodermal origin. After their initial formation, these mation of 3 separated streams of migrating cells is due to lack of
processes undergo considerable outgrowth along all 3 axes, emigration and migration of CNCC from R3 and R5 (Lumsden
merge in the midline region, and give rise to the elongated tri- et al., 1991; Couly et al., 1996). The lack of emigration of CNCC
angular-shaped lower jaw. In addition to merging in the midline from R3 and R5 is mainly due to apoptotic elimination of NCC
region, mandibular processes also fuse/merge at the lateral cor- and is regulated by molecules such as BMP-4 and Msx2 and by
ners with the maxillary process and at their lower borders with interactions with neighboring rhombomeres (Graham et al.,
the hyoid process. 1996; Maden et al., 1997; Takahashi et al., 1998). More recent
The mesenchyme of the mandibular processes, as in other studies indicate that crest depletion from R3 and R5 is not
facial processes, is derived from cranial neural crest cells (CNCC) absolute and that a small number of NCC from R3 and R5 sur-
and paraxial mesoderm. CNCC give rise to the majority of the vive the apoptotic elimination and join migratory cells in adja-
craniofacial skeleton (for reviews, see Le Douarin et al., 1994; cent rhombomeres (Sechrist et al., 1993; Birgbauer et al., 1995;
Osumi-Yamashita et al., 1997; Schilling, 1997). On the other hand, Kontges and Lumsden, 1996).
the paraxial mesoderm forms the craniofacial muscles along Short-survival fate-mapping of CNCC in chick embryos
with some skeletal elements in the skull and vascular tissues showed that CNCC originating from R4 and R6 populate the
(Noden, 1983b, 1988; Couly et al., 1992, 1993). The skeletogenic second and third branchial arches, respectively (Lumsden et al.,
potential of neural crest cells (NCC) is restricted to the cephalic 1991; Couly et al., 1996, 1998). A mixed population of CNCC
region down to the level of somite 5. In addition to giving rise to from the posterior mesencephalon, Rl, and R2 (Lumsden et al.,
skeletal structures, CNCC give rise to the peripheral nervous 1991) colonizes the first branchial arch (maxillary and mandibu-
system and connective tissues associated with the facial muscles lar processes). Long-survival fate-mapping with use of the quail
(Noden, 1986, 1988; Couly et al., 1992; Kontges and Lumsden, marker extended these observations and showed the com-
1996; Osumi-Yamashita et al., 1997). pound nature of the mandibular skeleton in which CNCC origi-
The fate and final derivatives of CNCC cells have been nating from multiple rhombomeres (Rl, R2, and R4) participate
extensively analyzed in avian and mammalian embryos (for in the formation of the mandibular arch skeleton (Couly et al.,
reviews, see Le Douarin et al., 1994,1997; Osumi-Yamashita et al., 1996; Kontges and Lumsden, 1996). Interestingly, the mandibu-
1997; Teillet et al., 1999). In addition, after the identification of lar arch skeleton displays an organization that precisely reflects
rhombomeres (R), a series of segmented structures in the hind- the rostrocaudal order of crest emigration. The proximal ele-
brain, an elegant scheme of the migratory routes and derivatives ments (the major part of Meckel's cartilage, and dentary and
of CNCC has been generated through short-term and long-term splenial bones) are derived from the midbrain crest. The caudal

276Grit
276 Rev Oral Blot Med
Crit Rev Oral Biol Med
12(4)276-300 (2001)

12(4):276-300 (2001),
skeletal structures of the mandibular arch (elements around the nial NCC has remained controversial for many years, because no
lower jaw joints and close to the middle ear structures, such as other subpopulation of pre-migratory NCC other than those giv-
the articular and its internal process, angular, suprangular, and ing rise to the mandibular arch showed such pre-specification in
quadrate) are formed mainly by NCC from Rl and R2. Kontges the patterns of their derivatives. In addition, it was shown that
and Lumsden (1996) showed that the most caudal element of transplantation of mesencephalic NCC (which normally give
the lower jaw in chick embryos, the retroarticular process of the rise to skeletal elements in the frontonasal mass and maxillary
Meckel's cartilage, is derived from CNCC originating from R4 and peri-ocular region) into the presumptive second- and third-
and a small number of NCC from R3 and R5 that survive apop- arch NCC also gave rise to a duplicated subset of first-arch-spe-
tosis. On the other hand, Couly et al. (1996) found no contribu- cific structures (Noden, 1983a). Furthermore, these observations
tion of R4 to the caudal skeletal elements. More recent labeling are in contrast to the outcome of transplantation of the pre-
studies in chick (Sohal et al., 1999) and mouse (Chai et al., 2000) sumptive first-arch NCC into the rostral or more caudal regions
embryos showed that, in addition to the CNCC, Meckel's carti- which gave rise to normal head skeletal structures and did not
lage is populated by non-CNCC. Studies in chick embryos form mandibular arch structures (Noden, 1978, 1986).
showed that cells emigrating from the ventral region of the The first family of genes suggested to be involved in pre-
neural tube (VENT) also make a significant contribution to specification of CNCC and branchial arch development was
Meckel's cartilage (Sohal et al., 1999). the Hox genes (for reviews, see Hunt et al., 1991a,b; Krumlauf,
In addition, Kontges and Lumsden (1996) showed that the 1994; Schilling, 1997; Morrison, 1998). Analysis of the patterns
same groups of CNCC also give rise to the connective tissue of of expression of Hox genes in mouse embryos showed that 3
individual mandibular muscles and their respective attach- members of Hox A-D clusters are expressed in the hindbrain
ment sites to the skeletal elements in the mandibular arch and region before the formation of rhombomeres, and that later
the cranial base. While the CNCC from the posterior mesen- their patterns of expression respected rhombomeric bound-
cephalon and RI and R2 mix together, there is no mixing aries (Krumlauf, 1994; Wilkinson, 1995). In general, with the
between this population and those originating from R4 that exception of paralogous group 1 and Hoxa2, the paralogous
give rise to the retroarticular process and connective tissue of groups have similar anterior boundaries within the hind-
the hyoid arch muscle (Kontges and Lumsden, 1996). brain. These overlapping patterns of Hox expression have
Cell transplantation and DiI labeling experiments also been called the Hox codes.
showed similarities between the axial level of origin for the In addition, it was shown that at least part of the overlap-
paraxial mesoderm and the CNCC populating each branchial ping patterns of Hox expression (Hox code) in each rhom-
arch (Noden, 1983b, 1986, 1988; Trainor et al., 1994; Trainor and bomere is maintained in migratory CNCC as they populate the
Tam, 1995; Hacker and Guthrie, 1998). These studies in mouse branchial arches, so that the branchial arches end up with a
and chick embryos showed that cranial paraxial mesoderm distinct pattern of Hox expression that closely resembles the
migrates in overlapping streams into the nearest branchial Hox code of the rhombomeres from which they originated
arch. Mesoderm originating from somitomeres III and IV enters (Hunt et al., 1991a,b; Krumlauf, 1994; Wilkinson, 1995). Thus,
the developing mandibular and hyoid arch and colonizes their CNCC-derived mesenchyme of the third branchial arch (simi-
core, a region known to correspond to the muscle mass where lar to R6) expresses the second and third paralogous groups,
they express myogenic markers such as Myf5, MyoD, and myo- and the second branchial arch mesenchyme that is colonized
genin (Trainor and Tam, 1995; Hacker and Guthrie, 1998). by CNCC from R4 expresses only the second paralogous
In contrast to the regions occupied by paraxial mes- group. These observations provide strong support for the
enchyme, the migrating CNCC colonize the periphery of each involvement of a combinatorial Hox code in the pre-specifica-
branchial arch (Lumsden et al., 1991; Trainor and Tam, 1995; tion of CNCC giving rise to the skeletal elements of the more
Hacker and Guthrie, 1998). In the branchial arches, unlike caudal branchial arches (2nd and 3rd arches). Direct evidence
other regions in the face, there is a distinct segregation of these for roles of Hox genes in regulating the patterning of the
2 cell populations (Trainor and Tam, 1995). CNCC-derived skeleton of the caudal branchial arches comes
For many years, it has been thought that patterning of the from skeletal abnormalities in the third and fourth branchial
skeletal elements in the mandibular arch is the result of intrin- arches in knock-out mice lacking Hoxa3 (Chisaka and
sic morphogenetic specification in CNCC. This possibility was Capecchi, 1991; Manley and Capecchi, 1995, 1998). of the Hox
suggested by the results of a series of hetero- and isotopic graft- However, with the exception of Hoxa2, none
ing experiments in amphibian and avian embryos (Horstadius genes is expressed rostral to R3 (Wilkinson et al., 1989; Hunt et
and Sellman, 1946; Wagner, 1949; Noden, 1983a, 1988). The al., 1991b; Krumlauf, 1994; Prince and Lumsden, 1994; Richman,
quail-chick transplantation experiments demonstrated that 1995). Although Hoxa2 is expressed up to the R1/R2 boundary,
when presumptive first-arch CNCC (anterior region of the the most anterior Hoxa2-expressing CNCC populate the hyoid
hindbrain Rl and R2 and mesencephalic CNCC) are trans- arch but not the first arch. Therefore, the first branchial arch that
planted into the presumptive second-arch region (R4/R6), the was shown previously to be morphogenetically specified
grafted CNCC migrate ventrally to the second arch but subse- (Noden, 1983a) is devoid of any Hox gene expression.
quently give rise to a subset of first-arch-specific structures and The lack of Hox expression in the most rostral regions of
form a partially duplicated mandibular arch skeleton (Noden, the developing neural tube and in the CNCC of the first
1983a; Couly et al., 1998). One of the main conclusions drawn branchial arch suggested that the primary characteristic dis-
from these experiments is that, in contrast to trunk NCC, tinguishing the CNCC populating the first branchial arch from
CNCC are morphogenetically pre-specified, and the patterns of more caudal CNCC may be the absence of Hox gene products
the CNCC-derived skeleton are imprinted in the pre-migratory (Gendron-Maguire al., et 1993; Rijli al., 1993, 1998).
et
CNCC while they are still within the neural tube. A predication of this model is that removal of Hoxa2 func-
However, the concept of morphogenetic specification of cra- tion from the CNCC populating the second branchial arch

2001)
12(4) 276-300 (2001)
12(4):276-300
BioI Med

Crit Rev Oral Biol Med 277


would lead to re-specification of second-arch CNCC identity and gives rise to almost normal CNCC-derived skeletal com-
into that of the first arch. In fact, in Hoxa2 mutants, morphogen- ponents. For example, Hunt et al. (1998) showed that when the
esis of the first and second branchial arches is altered (Rijli et al., Hox-expressing R7 is placed in the Rl region, CNCC from the
1993; Gendron-Maguire et al., 1993). Hoxa2 mutants lack all of grafted Hox-expressing R7 together with the host mesen-
the skeletal elements of the second branchial arch and contain a cephalic crest cells migrate into the first branchial arch, become
mirror-image duplicated set of caudal mandibular skeletal ele- Hox-negative (match their new position in the first branchial
ments, including a truncated Meckel's cartilage, incus, malleus, arch), and give rise to normal first-arch skeletal components
tympanic and squamosal bones, and duplicated external audi- with some additional ectopic first-arch structures. The results
tory meatus (Gendron-Maguire et al., 1993; Rijli et al., 1993). of these studies suggest that the Hox code in the branchial arch-
However, the duplicated structures in the Hoxa2 mutants did es can be established independently of the hindbrain after
not include more proximal structures, such as the proximal por- CNCC migration into the branchial arches.
tion of Meckel's cartilage, dentary, maxilla, palatine, jugal The modification of Hox expression in the branchial arches
bones, and teeth. The phenotype of the Hoxa2 mutant is similar colonized with mixed populations of CNCC is thought to be
if not identical to the phenotype of the chick embryo after trans- mediated by "community effects", where the Hox code
plantation of the presumptive first-arch CNCC into the second- expressed by the majority of cells eventually predominates and
arch region (Noden, 1983a). results in either gain or loss of Hox expression (Saldivar et al.,
Based on the long-term fate-mapping studies in chick 1997; Hunt et al., 1998). Alternatively, it has been postulated that
embryos and established homologies between mammals and alteration in Hox expression may be mediated by environmen-
birds (Kontges and Lumsden, 1996), it was concluded that the tal influences emanating from adjacent tissues (i.e., neighboring
absence of Hoxa2 results in specific homeotic transformation in neural tube, mesoderm, and branchial arches). More recent
which the R4-derived mesenchymal cells in the hyoid arch results suggest that a Hox code, rather than being involved in
acquired the identity of the caudal region of mandibular mes- pre-specification of CNCC, may be involved in the maintenance
enchyme derived from Rl and R2, but not the identity of CNCC of appropriate registration of different structures within each
mesenchyme derived mainly from the midbrain (Gendron- branchial arch (Kontges and Lumsden, 1996; Hunt et al., 1998).
Maguire et al., 1993; Rijli et al., 1993, 1998; Kontges and Lumsden, Despite the lack of expression of 3' members of the antenna-
1996). The phenotypic abnormalities in Hoxa2 mutants suggest- pedia class of Hox genes, several other regulatory genes have
ed essential roles for Hoxa2 in the morphogenetic identity of the been shown to be expressed in presumptive CNCC in more ros-
second branchial arch skeletal elements consistent with postulat- tral regions of the brain and in distinct regions of CNCC-derived
ed roles of Hox codes in patterning of the CNCC-skeletal deriva- mesenchyme of the first branchial arch, suggestive of their
tives of the more caudal branchial arch. involvement in the initial formation and morphogenesis of the
One of the predications of the CNCC pre-specification first branchial arch. These genes include members of Msx, Dlx,
model, in which patterning information is imprinted by the Otx, Barx, Gsc, Pax, Hand, Pitx, and Prx genes. As transcription
presence absence of Hox expression in pre-migratory CNCC
or factors, they achieve this function by regulating the expression of
while still in the neural tube, is that experimental manipula- other regulatory genes.
tions of rhombomeres should result in colonization of the
branchial arch with CNCC expressing inappropriate Hox codes Outgrowth of the Mandibular Processes
and, thus, should give rise to inappropriate skeletal elements. Tissue recombination studies indicate that outgrowth of the
Although the original grafting experiments of Noden (1983a) mandibular processes is dependent on epithelial-mesenchy-
support this model, findings of more recent transplantation mal interactions (Wedden, 1987; Richman and Tickle, 1989;
studies in chick embryos have provided conflicting results. The Hall and Coffin-Collins, 1990; Mina et al., 1994). Following
differences in the results appear to be related to the axial level removal of the overlying epithelium, reduced outgrowth is
of origin of crest cell, the type of experimental manipulation, seen in the mandibular mesenchyme, suggesting that signals
and the time intervals in which Hox expression in the branchial derived from mandibular epithelium regulate growth of the
arches has been examined. underlying mesenchyme. In addition, tissue recombination
Studies in which the neural tube and pre-migratory studies in which epithelia and mesenchyme from different
CNCC are re-located within the hindbrain and patterns of Hox facial processes were exchanged suggest similarities in the sig-
expression in the branchial arches are examined early (one day naling cascades regulating outgrowth of all the facial process-
post-operatively) indicate that the grafted NCC populated the es (Richman and Tickle, 1989). Outgrowth of the mandibular
branchial arches according to their new position but main- mesenchyme is supported by epithelia covering other facial
tained the patterns of Hox gene expression appropriate for processes, and mandibular epithelium supports the outgrowth
their position of origin (Prince and Lumsden, 1994; Saldivar et of the mesenchyme from other facial processes. Although
al., 1996, 1997; Couly et al., 1998). The stability of the Hox code these recombination studies clearly indicated the mitogenic
in crest cells in ectopic locations in some, but not all, cases was roles of mandibular epithelium on the underlying mesen-
correlated with skeletal abnormalities. chyme, they did not identify any specific regions in the epithe-
On the other hand, analysis of embryos in which crest cells lium or mesenchyme that may be involved in regulating the
are re-routed by late deletion of adjacent crest cells and experi-
ments involving rotation of the entire hindbrain and its associ-
outgrowth of the developing mandible. Furthermore, the
mandibular arch, similar to other facial processes, lacks clear-
ated crest (R1-R7) indicates that the initial stable Hox code is ly identifiable morphological or functional regions.
modified within the branchial arches 2 or 3 days after trans- Studies in the developing chick mandible suggest that a
plantation (Hunt et al., 1995, 1998; Saldivar et al., 1997). In these population of cells in the medial region (see Fig. 1) plays a sig-
situations, the Hox code in CNCC in ectopic locations is modi- nificant role in the growth of the developing mandible. Dil
fied, matches their new location within the branchial arches, labeling studies indicated that cells in the medial region make
278
278 rlBoBiol Mede
e Oral
CiCrit Rev 12(4):276-300 (2001)
a greater contribution to the overall expansion of the develop-
ing mandible as compared with cells in the lateral region (A) Midline
(McGonnell et al., 1998). Cell proliferation studies showed a Medial (Proximal) 4 * Lateral (Caudal)
high percentage of S-phase-labeled cells in the medial region
(Mina et al., 1995; Barlow and Francis-West, 1997; McGonnell et Oral

al., 1998). Furthermore, when only the medial region of stage 24


mandibular arches is grown for 1 wk as a graft in the dorsal
surface of a chick wing, cartilage rods of substantial length
(60% of the length of the cartilage rods formed in the intact HyonmandiMar
cueft
mandible at stage 37) are formed (Richman and Tickle, 1989;
Mina et al., submitted manuscript).
Together, these observations suggest that the medial
region is a putative functional region involved in regulating
directional outgrowth of the developing mandible.
Interestingly, high levels of expression of many of the evolu- (B()
tionary conserved signaling factors and transcription factors
are detected in the epithelium and mesenchyme of the medi-
al region (see below). Direct evidence for the involvement of
some of these genes in the outgrowth of mandibular process-
es comes from abnormalities in the developing mandibles in CDP
homozygous null mutant mice lacking these genes (knock-
Ibone
out mice). Furthermore, some of these genes have been
shown to be candidate genes causing human craniofacial dis-
orders affecting mandibular growth and morphogenesis.
Transcription Factors
in Mandibular Morphogenesis
Msx GENES
The vertebrate Msx gene family was originally isolated by
homology to the Drosophila Msh gene (muscle segment Figure 1. (A) Line drawings illustrating a frontal view of the develop-
homeobox gene) (Hill et al., 1989; Robert et al., 1989; Davidson,
ing mandibular arch at early stages of development, indicating thle
anatomical landmarks and terminologies used in this paper. The
1995) and consists of 3 members that are not positioned in a medial (proximal region) is indicated by a hatched pattern. The lat-
single gene cluster. Studies in vertebrate embryos showed eral region is indicated by stippling, and an arrow indicates the
that Msxl and Msx2 are expressed throughout the embryo, region of the hyomandibular clef. (B) Line drawing illustratinq a lat-
including the rostral region of the developing head, suggest- eral view of the developing mandibular arch at late stages o devel-
ing their involvement in craniofacial development (Davidson, opment, indicating the anatomical structures referred to in this study.
1995; Maas and Bei, 1997, and references therein). The most The mandibular arch structures derived from the posterior midbrain,
interesting aspect of the vertebrate Msx family is their com- RI and R2, are indicated by a dotted pattern, and the skeletal ele-
plementary or overlapping patterns of expression in different ments of the hyaid arch (second branchial arch) that are derived
regions of the embryo where morphogenesis has been shown from R4 are idicated by hatched pattern. AP, articular process; CDP,
to be dependent on inductive epithelial-mesenchymal inter-
condylar process; CP, coronoid process; 1, incus; M, malleus; S,
stapes; Sq, squamosal. The letters in parentheses indicate avian
actions (Davidson, 1995; Maas and Bei, 1997). names for structures homologous in mammals in the middle ear
At early stages of development, the expression of Msxl region and have been adapted from Kontges and Lumsden (1996).
and Msx2 in the mandibular arch is limited to the mes- (A) articular; (Pq) pterygoquadrate; (Q) quadrate.
enchyme in the medial region and is excluded from the mes-
enchyme in the lateral region (MacKenzie et al., 1991, 1992;
Nishikawa et al., 1994; Mina et al., 1995; Barlow and Francis- mandible is truncated (Satokata and Maas, 1994).
West, 1997). In addition to its expression in the medial region, On the other hand, the domain of Msx2 expression in
Msxl is also expressed in the mesenchyme surrounding the the lower medial region of the mandibular mesenchyme
hyomandibular cleft. Tissue recombination and bead implan- (Wedden, 1991; Mina et al., 1995; Shen et al., 1997), similar
tation studies indicate that the expression of Msx genes in the to its expression in many organs, is correlated with areas of
developing mandible and other facial processes is dependent apoptosis (Graham et al., 1996; Macias et al., 1996; Maden et
on signals derived from the overlying epithelium (Takahashi al., 1997; Marazzi et al., 1997; Ferrari et al., 1998; Takahashi
et al., 1991; Brown et al., 1993, 1997; Jowett et al., 1993; Mina et et al., 1998). Studies in the developing chick mandible also
al., 1995; Chen et al., 1996; Bei and Maas, 1998; Wang et al., suggest that Msx genes may be involved in delineating the
1998b). In the developing mandible at early stages, the non-chondrogenic region at the midline region (symph-
expression of Msxl in the medial region is correlated with ysis). This possibility is supported by observations that, in
areas undergoing expansion which contain highly prolifera- contrast to control explants, explants from chick mandibu-
tive and undifferentiated mesenchyme cells (Mina et al., lar arches treated with Msx2 antisense oligonucleotides
1995). In Msxl-deficient mice, the medial part of the formed cartilage in the medial region, resulting in the

279
12(4) 27o 300 (2001
12(4)-.276-300 2001)1) Crit Rev Oral
Crit Rev Oral Biol
Biol
Med
Med 279
fusion of the 2 bilateral rods of cartilage at the midline mandibles of Dlx5 mutant mice are shortened, lack the coronoid
(Mina et al., 1996). More recently, it was shown that over- process, and contain misshapen condylar and angular processes.
expression of Msx2 also inhibits chondrogenesis in organ- The phenotypic abnormalities in the mandibular arches of Dlx5
cultured mouse mandibles (Semba et al., 2000). mutants suggest essential (non-redundant) roles of Dlx5 for
Although lack of Msxl does not appear to disturb the proper development of mandibular processes and the skeleton
mandibular symphysis, Msxl/Msx2 knock-outs display severe of the caudal region of the mandibular arch. Further analyses of
abnormalities in the developing mandible (Satokata et al., 2000). the branchial arches in Dlx5 mutants indicate that the absence of
In humans, mutation of one copy of MSX1 results in sin- Dlx5 did not affect cell proliferation or apoptosis but expanded
gle-tooth agenesis, and specific point mutations in the homeo- the territory of the proliferating cells within the first branchial
domain in one copy of MSX2 result in Boston-type craniosyn- arch (Acampora et al., 1999). In situ hybridization analysis indi-
ostosis (reviewed by Cohen, 2000). cated decreases of Goosecoid (Gsc) expression in the mesenchyme
of the frontonasal processes and in the mandibular and hyoid
DLX GENES mesenchyme of the Dlx5 mutants (Depew et al., 1999).
The Dlx genes constitute a highly conserved family of homeo- Unlike other members of the Dlx family, Dlx5 and Dlx6 are
box-containing genes, homologous to the Drosophila Distal-less also expressed in developing bones, cartilage, and teeth, sug-
(DlI) gene that is required for many processes, including the for- gesting that the Dlx5 and Dlx6 genes may play roles in the
mation of the mouth (Cohen et al., 1989). In the mouse, there are multi-step process of skeletal differentiation and/or morpho-
at least 6 Dlx genes arranged as pairs which are convergently genesis (Zhao et al., 1994a; Ferrari et al., 1995; Newberry et al.,
transcribed (Dlxl and Dlx2, Dlx3 and DIX7, Dlx5 and Dlx6) (see 1998; Acampora et al., 1999; Davideau et al., 1999; Depew et al.,
review by Kraus and Lufkin, 1999, and references therein). 1999). Dlx5 is also expressed at specific stages of osteoblast dif-
Members of the Dlx family are expressed in the craniofacial ferentiation in vitro and could repress osteocalcin gene expres-
region in both ectoderm and mesenchyme (Davideau et al., 1999; sion (Ryoo et al., 1997). Interestingly, lack of Dlx5 results in
Kraus and Lufkin, 1999, and other references therein). Among hypo-mineralization of calvaria (Acampora et al., 1999;
these, DIX2, DIX3, and Dlx5 are expressed at the junction of the Depew et al., 1999) and expression of osteocalcin in the perios-
neural plate and surface ectoderm, suggesting that they may be teum at birth (Acampora et al., 1999).
expressed by pre-migratory and migratory CNCC cells In humans, there is evidence implicating DLX5 and DLX6
(Robinson and Mahon, 1994; Qiu et al., 1997; Yang et al., 1998; genes as candidate genes for ectrodactyly (split hand/foot mal-
Acampora et al., 1999; Depew et al., 1999). formations, SHFM1) (Scherer et al., 1994; Crackower et al., 1996).
In situ hybridization studies on E9.5-E1O mouse embryos Often, these patients also have cleft palate and deafness
showed that Dlxl and Dlx2 are expressed in the mesenchyme of (Ignatius et al., 1996). A frame-shift mutation in DLX3 is also
both proximal and distal regions of all branchial arches, while associated with taurodontism and enamel hypoplasia in
DIX3, DIX5, and DIx6 are expressed predominantly in mes- humans with Tricho-dento-osseous syndrome (Price et al., 1998).
enchyme of the distal regions (reviewed by Kraus and Lufkin,
1999). For example, in the first branchial arch, Dlxl and Dlx2 are OTX- GENES
expressed in the maxillary processes (the proximally located Another family of evolutionary conserved homeodomain fac-
component) as well as in the mandibular processes (the distally tors with critical regulatory roles in the determination of head
located component) and Dlx3, Dlx5, and Dlx6 are expressed structures during development is the Otx genes, vertebrate
only in the mandibular processes. In the developing mandible, homologs to Orthodenticle in Drosophila (for review, see Bally-
Dlx genes are expressed in a lateral-to-medial gradient. Cuif and Boncinelli, 1997; Simeone, 1998; Acampora et al., 2000).
Similarly, in the second (hyoid) arch, Dlxl and DIx2 are The overlapping patterns of expression of Otx and Emx genes in
expressed throughout the proximo-distal axis, while DX3, Dlx5, the rostral region of the developing brain, together with func-
and Dlx6 are expressed only in the more distal (close to the mid- tional studies, have suggested that these genes, analogous to the
line) region of the second arch. These patterns of expression Hox code for hindbrain development, provide a combinatorial
suggested that Dlx genes play essential roles in proximo-distal code for rostral brain development (for reviews, see Bally-Cuif
patterning of the branchial arches (Qiu et al., 1997). and Boncinelli, 1997; Simeone, 1998; Acampora et al., 2000). Otx2
This possibility is supported by phenotypic abnormalities in homozygous null mutants die early in embryogenesis and fail
mice lacking Dlxl, Dlx2, Dlxl/Dlx2, and DIx5. Mice lacking Dlxl, to develop structures anterior to R3 (Acampora et al., 1995;
Dlx2, and Dlxl/Dlx2 exhibited abnormalities similar, but not Matsuo et al., 1995; Ang et al., 1996). The phenotypic abnormali-
identical, to those of maxillary and proximal (caudal) hyoid arch- ties in Otxl homozygous mutants indicated its essential role in
derived structures (Qiu et al., 1995, 1997; Thomas et al., 1997). the formation of cortex in the adult brain (Suda et al., 1996;
However, in these mutants, the skeletal components of the Acampora et al., 1996, 1998).
mandibular arch and distal hyoid arch appeared to be normal In addition to its expression in the rostral region of the
(Qiu et al., 1995, 1997; Thomas et al., 1997). On the other hand, developing brain, Otx2 is also expressed in neuroectoderm
one of the most noticeable abnormalities in the mice homozy- from the forebrain up to the mid-hind-brain isthmus (Matsuo
gous for a targeted deletion of Dlx5 is in the developing et al., 1995; Ang et al., 1996). Interestingly, Otx2 heterozygote
mandible (Acampora et al., 1995; Depew et al., 1999). At early animals exhibit otocephaly and abnormalities in midbrain
stages (E13-E14), the mandibular arch and the Meckel's cartilage crest derivatives of the first arch, suggesting a role for Otx2 in
of Dlx5 mutants are shorter than those in wild-type embryos pre-migratory CNCC originating from the midbrain region
and exhibit abnormalities in the caudal region of Meckel's carti- (Matsuo et al., 1995). In Otx2+/- mice, the elements that were
lage. At its caudal end, Meckel's cartilage in DIx5 mutants is shown to be derived solely from midbrain crest (distal
bifurcated and gives rise to an ectopic novel cartilage, which Meckel's cartilage, dentary, maxilla, and palatine) (Kontges
becomes surrounded by bone later in development. At birth, the and Lumsden, 1996) are lacking or severely reduced, suggest-
280 Crit
Crit
Rev Oral Biol
Rev Oral Biol Med
Med

12(4):276-300 (2001)
ing that defects in these animals may be due to deficiencies in branchial arch-including Msxl, Msx2, Gsc, Shh, Bmp2/4, Wnt5a,
CNNC derived from the midbrain region. However, in Otx2+/- and Pitx2-is unaltered in Pitxl null mutant mice (Szeto et al.,
mice, the hindbrain-derived, first-arch elements (malleus, 1999), suggesting that defects in the craniofacial structures simi-
incus, and pterygoid) are almost unaffected. Analysis of the lar to those observed in the hindlimb may be due to defects in
knock-in mice in which Otx2 was replaced with Otxl showed proliferation and/or abnormal chondrogenesis of mesenchymal
that most Otx2 functions, including the Otx2 haploinsufficien- cells (Lanctot et al., 1999; Szeto et al., 1999). Interestingly, it has
cy causing the otocephalic phenotype, are replaceable with been suggested that, in humans, mutant PITX1 alleles might be
those of Otxl (Suda et al., 1999). responsible for a subset of patients with Treacher-Collins syn-
drome (Crawford et al., 1997).
GOOSECOID Pitx2/RIEG was initially identified by positional cloning of the
Goosecoid (Gsc) encodes another evolutionary conserved paired- gene responsible for Rieger Syndrome in humans (Semina et al.,
like homeoprotein that acts as a transcription factor with essen- 1996; Gage and Camper, 1999; Meijlink et al., 1999). This autosomal-
tial roles in head development (for review, see Bally-Cuif and dominant haploinsufficiency syndrome is characterized by anteri-
Boncinelli, 1997). In vertebrates, Gsc is expressed transiently at or chamber ocular abnormalities and other features of various
the rostral end of the developing brain and then re-appears at degrees of severity, including dental hypodontia, umbilical abnor-
E9.5-E10.5 in many sites, including the mesenchyme of the malities, cardiac defects, and mild craniofacial dysmorphism.
branchial arches (Gaunt et al., 1993; Rivera-Perez et al., 1995, Pitx2 is expressed in many tissues during development,
1999; Yamada et al., 1995, 1997). In the mandibular arch, Gsc is including the craniofacial mesenchyme and epithelium of the
strongly expressed in the mesenchyme in the region of the first and second branchial arches (see review by Meijlink et al.,
hyomandibular cleft. 1999, and references therein). Experimental evidence indicated a
Gsc null mutants die soon after birth, with rib cage malfor- role for Pitx2 downstream of sonic hedgehog and nodal in a genetic
mations and multiple craniofacial defects, including abnormali- pathway regulating laterality of heart, gut, and other asymmetric
ties in the mandibular arch and middle ear structures (Rivera- organs (see review by Meijlink et al., 1999, and references therein).
Perez et al., 1995, 1999; Yamada et al., 1995,1997). In the mandibles The direct role of Pitx2 in left-right determination and devel-
of these mutants, although the condylar process appears normal, opment of other structures, including the development of the
the coronoid and angular processes are severely reduced in size, mandibular and maxillary processes, is provided by abnormalities
and the mandible is shortened in length. Furthermore, Meckel's in mice deficient for Pitx2 (Gage et al., 1999; Lin et al., 1999; Lu et al.,
cartilage is not enclosed by the mandibular bones but is embed- 1999b). Pitx2 mutant mice die at around E14-15 and exhibit lung
ded in a novel groove that extends along the entire length of the isomerization and defects in cardiac positioning and pituitary
mandible. Recent studies showed that abnormalities in the development (Lin et al., 1999; Lu et al., 1999b). In addition, Pitx2 null
mandible of Gsc null mutants are due to the absence of cells des- mutants exhibited cleft palate and abnormalities in the maxillary
tined to express Gsc in these mutants, suggesting the essential and mandibular arches, including severely hypoplastic mandible
roles of Gsc in the initial proliferation and/or survival of Gsc- and arrested tooth buds (Lin et al., 1999; Lu et al., 1999b). In situ
expressing cells (Rivera-Perez et al., 1999). hybridization analysis indicated the absence of Fgf8 and altered
domains of expression of Bmp4, Msxl, and Msx2 in the developing
PITX GENES facial processes in Pitx2 null mutants, suggesting that the facial
Pitxl (Ptxl /POTX) and Pitx2 (RIEG, Otx2, Otlx2, Brxl, Arpl) are abnormalities may be mediated by changes in the patterns of
2 members of a vertebrate multigene family with overlapping expression of these genes (Lin et al., 1999; Lu et al., 1999b).
and distinctive patterns of expression during embryogenesis (for
reviews, see Drouin et al., 1998; Gage et al., 1999; Meijlink et al., PAX GENES
1999, and references therein). Pitxl was originally identified as a Pax genes encode a family of transcription factors characterized
factor interacting with the pituitary-specific transcription factor by an evolutionary conserved paired box domain, a 384-bp DNA-
Pit-i and POMC promoter (see reviews). Pitxl is expressed in the binding motif that regulates embryonic development by the con-
pituitary gland throughout its development, in the lateral plate trol of target genes (for reviews, see Balling et al., 1996; Dahl et al.,
mesoderm of the caudal half of the embryo which leads to its 1997; Peters et al., 1998a; Mansouri et al., 1999, and references
expression exclusively in the hindlimb and not the forelimb, in therein). In mammals, the Pax gene family consists of 9 members
the first branchial arch and its derivatives, and in oral ectoderm that, based on the presence or absence of structural motifs, are
(Szeto et al., 1996; Lanctot et al., 1997; Meijlink et al., 1999). The pat- divided into 4 subgroups (see above reviews). Genes within an
terns of expression suggest that Pitxl is a critical transcription fac- individual group show a very high degree of sequence similarity
tor involved in specification of the hindlimb and development of within the paired domain and exhibit similar patterns of expres-
the pituitary gland and structures derived from the first branchial sion during embryogenesis (Dahl et al., 1997). The roles and func-
arch. This possibility was supported by phenotypic abnormalities tions of Pax genes in embryonic development have been eluci-
in null mutants for Pitxl and mis-expression of Pitxl in the chick dated by analysis of naturally occurring mouse mutants, targeted
wing bud (Lanctot et al., 1999; Logan and Tabin, 1999; Szeto et al., inactivation of several Pax genes in mice, and human syndromes
1999). Pitxl null mutants die immediately or shortly after birth (see above reviews). A common feature of all Pax mutants is
and are readily recognizable by their shortened mandibular arch. reduction in size and malformation or loss of specific organs.
These mutants exhibit abnormalities in the limb and in the deriv- Among all Pax genes, members of group I (Paxl, Pax 9),
atives of the first branchial arch, including cleft palate, signifi- group III (Pax3, Pax 7), and group IV (Pax 6) are expressed in the
cantly shortened tongue and mandible, a novel bone surround- developing facial processes (Peters et al., 1998a; Mansouri et al.,
ing Meckel's cartilage, and lack of gonial bones (Lanctot et al., 1999). Analyses of various mutants have indicated essential roles
1999; Szeto et al., 1999). In situ hybridization analysis indicated for Pax3, Pax6, and Pax7 in the development of CNCC-derived
that the expression of several markers expressed early in the first structures in the upper face (for reviews, see Peters et al., 1998a;

281
12(4) 276 300 (2001)
12(4)1:276-300 Crit
Crlf Rev Oral Biol Med
Rev Oral BiolMed
281
Mansouri et al., 1999; Francis-West et al., 1998). However, in these sation of Meckel's cartilage, suggesting that, in the absence of
mutants, no abnormalities were observed in CNCC-derived Prxl and Prx-2, the pre-chondrogenic condensation of Meckel's
structures of the lower face. On the other hand, phenotypic cartilage is initiated but is not maintained (Lu et al., 1999a). The
abnormalities in the Pax9 null mutant (Peters et al., 1998a,b) indi- phenotypic abnormalities in Prxl and Prxl/Prx2 mutants indi-
cate essential roles for Pax9 for the lower face. Pax9-deficient cate redundant but essential roles for Prxl and Prx2 in the sig-
mice lacked structures derived from pharyngeal pouches, such naling network regulating epithelial-mesenchymal interactions
as thymus and parathyroid glands. In the developing mandible, that promote outgrowth and skeletogenesis in the lower jaw.
in addition to abnormalities in the developing teeth, the alveolar Other members of the paired-related family of homeobox
ridge and the coronoid process are absent in Pax9 null mutants. genes in vertebrates include Alx3, Alx4, and Cartl (Meijlink et
al., 1999). These genes are also expressed in the distal part of
PRX GENES the mandibular arch (Zhao et al., 1994b; Qu et al., 1997a,b; ten
Prxl (previously called Mhox) and Prx2 (previously called S8) Berge et al., 1998). However, no phenotypic abnormalities in
are closely related members of the paired-related family of the developing mandible have been reported in mice lacking
homeobox genes that are co-expressed in a variety of sites, these genes (Zhao et al., 1994b; Qu et al., 1997a). It is possible
including the craniofacial mesenchyme (see review by that, similar to Prxl and Prx2, the absence of abnormalities in
Meijlink et al., 1999, and references therein). the mandibular arch in these knock-outs may be due to func-
Studies in developing mice and chickens indicate that Prxl tional redundancies.
and Prx2 are co-expressed in the CNCC-derived mesenchyme
of the frontonasal process, in the mesenchyme of the first and BARX GENES
second branchial arches, and in the pre-osteogenic areas. In the Barxl and Barx2 are 2 members of the vertebrate Bar class of
developing mandible, at early stages of development, high lev- homeobox-containing genes homologous to Drosophila BarHl
els of expression of both genes are detected in the mesenchyme and BarH2 (Higashijima et al., 1992a,b; Kojima et al., 1991). Three
of the medial region. In addition to the medial region, Prxl is mouse and 2 chick homologues of the Barx genes have been iso-
also expressed in the cells around the first branchial groove. As lated (Tissier-Seta et al., 1995; Jones et al., 1997; Saito et al., 1998;
development proceeds, the expression of both genes is down- Barlow et al., 1999; Smith and Tabin, 1999). Tissue distribution
regulated in the mandibular process but maintained in the studies showed that these genes are expressed in many sites,
maxillary and nasal processes (Lu et al., 1999a). including the facial processes (Tucker et al., 1989b; Tissier-Seta et
Although Prx2 null mutant mice show no obvious cranio- al., 1995; Jones et al., 1997; Mitsiadis et al., 1998b; Barlow et al.,
facial and skeletal abnormalities (ten Berge et al., 1998), Prxl null 1999; Smith and Tabin, 1999). Studies in developing mice
mutant mice show defects in skeletal elements derived from the showed that, in the developing maxilla and mandible, Barxl is
maxillary processes and the caudal part of the mandibular expressed in the mesenchyme (Tissier-Seta et al., 1995; Jones et al.,
processes, including hypoplastic coronoid, condylar, and angu- 1997; Mitsiadis et al., 1998b; Tucker et al., 1998b), and Barx2 is
lar processes and malformed malleus (Martin et al., 1995). In expressed in the overlying epithelium (Jones et al., 1997).
addition, Meckel's cartilage in Prxl mutants displayed abnor- However, studies in chick embryos indicate that, unlike in the
mal sigmoidal morphology (Martin et al., 1995). More recent mouse, Barxl is expressed in both epithelium and mesenchyme
studies (Lu et al., 1999a) indicate that, in the Prxl null mutants, of the maxillary and mandibular processes (Barlow et al., 1999).
cells fated to express Prxl are initially present in the regions that Furthermore, in chick embryos, Barx2b-which is 80% and 61%
give rise to the defective structures, but disappear later, sug- identical to mouse Barx2 and Barxl, respectively-is expressed
gesting that Prxl product may be required for the maintenance prominently in myogenic populations in the craniofacial region,
(proliferation and/or survival) of specific subpopulations of in the mesoderm of the branchial arches, and in areas of the
CNCC-derived mesenchymal cells in the branchial arches. forming bones (Smith and Tabin, 1999). At stage 25, Barx2b is
In contrast to single mutants, Prxl/Prx2 double-knock-out expressed at the tips of the outgrowing maxilla and mandible
mice exhibited severe craniofacial abnormalities, including but disappears by stage 30 (Smith and Tabin, 1999).
pointed snout, the absence of external ears, and severely short- In both the chick and mouse, the mesenchymal domain of
ened lower jaws (ten Berge et al., 1998; Lu et al., 1999a; Meijlink et Barxl expression is restricted to the lateral region, where Fg,f8
al., 1999). Double-mutant mice also had novel phenotypes, inclu- is expressed by the overlying epithelium (Tucker et al., 1998b;
ding abnormalities in the medial region of the developing Barlow et al., 1999). The expression of Barxl is excluded from
mandibles, lower incisors, and Meckel's cartilage (ten Berge et the mesenchyme in the medial region in which Bmp4 is
al., 1998; Lu et al., 1999a; Meijlink et al., 1999). Approximately 8% expressed in the overlying epithelium (Tucker et al., 1998b;
of the newborn double-mutants generated by ten Berge et al. Barlow et al., 1999). The expression of Barxl is also excluded
(1998) exhibited clefts in the mandible and tongue, whereas the from the central core corresponding to the regions forming
mandibular processes of the double-mutant mice generated by Meckel's cartilage and the muscle of the mandibular process
Lu et al. (1999a) lacked the midline symphysis and were fused. In (Tissier-Seta et al., 1995; Barlow et al., 1999).
these double-mutants, either a single incisor arrested in the bud These patterns of expression suggest the involvement of
stage or no incisors were present. The arrested incisor tooth buds signals derived from the overlying epithelium (Fibroblast
showed decreases in the expression of Pax9 and patched (Lu et al., growth factor-8 and Bone Morphogenetic proteins) in regulat-
1999a; Meijlink et al., 1999). Furthermore, in these double- ing the spatial patterns of Barxl expression in the mandibular
mutants, most of Meckel's cartilage was absent (ten Berge et al., mesenchyme. In fact, studies in mouse mandibles indicate that
1998; Lu et al., 1999a; Meijlink et al., 1999). In situ hybridization beads soaked in FGF8 can induce/maintain expression of Barxl
analysis showed that, in double-mutants, Sox9 (a marker of pre- in the lateral mandibular mesenchyme (Tucker et al., 1998b). On
chondrogenic mesenchyme of the branchial arches) was the other hand, beads soaked in BMP-4 inhibited expression of
expressed normally in the area of the pre-chondrogenic conden- Barxl in the lateral mandibular mesenchyme expression

282CrtRvOaBilMd1()7630(0)
282 Crit Rev Oral Biol Med 12(4):276-300 (2001)
(Tucker et al., 1998b). Inhibition of BMP4 signaling by applica- Secreted Factors in Mandibular Morphogenesis
tion of Noggin protein during the early stages of mandibular Secreted factors involved in mandibular outgrowth and mor-
development resulted in ectopic expression of Barxl in the mes- phogenesis include members of the fibroblast growth factor
enchyme in the medial region (Tucker et al., 1998b). Similar (FGFs), transforming growth factor beta (TGF3), and hedge-
antagonistic interactions between BMP and FGF signaling also hog families, Wnt genes, endothelin pathway, notch pathway,
restrict expression of Barxl to the maxillary mesenchyme in the and epidermal growth factor.
posterior region (Barlow et al., 1999), suggestive of the involve-
ment of Barxl in patterning of the facial processes. BONE MORPHOGENETIC PROTEINS (BMPS)
HAND GENES The transforming growth factor-beta (TGFI) superfamily is
composed of more than 30 members and has been divided into
dHAND and eHAND (Cserjesi et al., 1995; Srivastava et al., several subgroups based on the extent of homology in the
1997; Thomas et al., 1998), 2 members of the bHLH (basic helix- mature ligand (for review, see Hogan, 1996; Wozney, 1998). The
loop-helix) family of transcription factors, are co-expressed in largest subgroup constitutes the bone morphogenetic proteins
many regions, including the medial region of the developing (BMPs). In mammals, there are at least 12 BMPs that have been
mandible. Deletion of the dHAND gene in mice resulted in implicated in the development of almost all vertebrate organs
embryonic death at Ell secondary to cardiac failure and many and are thought to be important mediators of inductive tissue
abnormalities, including severely hypoplastic first and second interactions during embryogenesis (Hogan, 1996; Wozney,
branchial arches (Srivastava et al., 1997; Thomas et al., 1998). 1998). The actions of TGF3s are mediated by the TGF3 recep-
Molecular analyses indicated that although Prxl, Dlx2, eHand, tors. These serine/threonine receptor kinases fall into 2 classes,
and Msx2 were expressed at normal levels, Msxl expression type I and type II receptors (for review, see Kawabata et al.,
was not detectable in the medial region of the developing 1998; Massague, 1998; Raftery and Sutherland, 1999).
mandible of E9.5 dHAND null mutants (Thomas et al., 1998). Ligand binding leads to the formation of heterodimers of
These results also indicate that the hypoplastic mandible in the these 2 receptor types, phosphorylation of the type I receptor by
dHand-/- mutant is not due to defects in the migration of neur- the type II receptor, and subsequent propagation of the intracel-
al crest cells into the branchial arch and occurs secondary to lular signal. Seven type I or activin-like kinases (ALKs) and 5
programmed cell death. The unchanged patterns of expression type II receptors have been identified in vertebrates (Massague,
of eHAND in the dHAND mutant suggest that eHAND is 1998). Among these, BMPR1A (ALK-3), BMPR1B (ALK-6), and
unable to compensate fully for dHAND in the branchial arch ActR 1 (ALK-2) appear to be essential for BMP signals (Hogan,
and that these genes may have some unique roles in the devel- 1996; Massague, 1998). Different BMP ligands are capable of
opment of the developing mandible (Thomas et al., 1998). activating common signaling pathways (for review, see
Massagu6, 1998). Ligands in the TGFI superfamily are active as
OTHER TRANSCRIPTION FACTORS dimers, and the subunit composition of these molecules can
Other transcription factors that have been shown to have a role dramatically affect signaling activity. Although BMP het-
in facial development are twist, ski, and AP-2, Tbx2, Tbx3, and erodimers have yet to be identified in vivo, BMP heterodimers
TCOF1/Freacle. Twist is expressed at high levels in the mes- made in vitro (BMP-4/BMP-7 and BMP-2/BMP-7) show more
enchyme of the first branchial arch, and twist null mutants potent biological activity than BMP homodimers (Aono et al.,
exhibit abnormalities in branchial arch formation (Chen and 1995; Israel et al., 1996; Suzuki et al., 1997; Tsuji et al., 1998).
Behringer, 1995; Gitelman, 1997). AP-2 is expressed in the cra- In addition, Smads are essential for TGFJ-related signal
nial neural crest, and mice lacking Ap-2 had severe craniofacial transduction. Smad-1, Smad-5, and Smad-8 proteins (reviewed
abnormalities (for review, see Morriss-Kay, 1996; also see Shen by Kretzschmar and Massague, 1998; Miyazono, 1999) are
et al., 1997; Zhang et al., 1996). However, in Ap-2 knock-out, BMP receptor substrates, and Smad 2 and Smad 3 are sub-
similar to ski knock-out, the mandibular arch skeleton is less strates for the related TGF3 and activin receptors in vertebrate
affected than the skeleton of the upper face (reviewed by (Kretzschmar and Massague, 1998; Miyazono, 1999).
Morriss-Kay, 1996; Berk et al., 1997). Tbx2 and Tbx3 are 2 mem- Tissue distribution studies showed that multiple Bmps
bers of the T-box family that are expressed in the mesenchyme (Bmp-2, -4, -5, and -7) are expressed in the epithelium of the
of the maxillary and mandibular processes (Chapman et al., mandibular arch (Francis-West et al., 1994; Wall and Hogan,
1996; Gibson-Brown et al., 1998a,b; Isaac et al., 1998). 1995; Aberg et al., 1997; Barlow and Francis-West, 1997; Tucker
TCOF1/treacle was initially identified as the gene responsi- et al., 1998a; Wang et al., 1998b, 1999; Solloway and Robertson,
ble for Treacher-Collins syndrome, the most common autoso- 1999). At early stages, expression of Bmp-4 and Bmp-5 is
mal-dominant inherited human mandibulofacial disorder restricted to the epithelium of the medial region overlying the
characterized by cleft palate and abnormalities in the ear and Msxl and Msx2 expressing mesenchyme. In contrast to the
of some skeletal structures derived from the first arch restricted patterns of expression of these Bmps, Bmp-2 and
(Gladwin et al., 1996; Dixon et al., 1997a,b; Wise et al., 1997; Bmp-7 are expressed in a broader domain throughout the
Winokur and Shiang, 1998). The murine homologue of the epithelium of the branchial arches. Furthermore, similar to the
TCOF1 gene has been isolated (Dixon et al., 1997b; Paznekas et effects of mandibular epithelium in tissue recombination
al., 1997) and shown to be expressed in a wide variety of experiments, agarose beads soaked in BMP-2, -4, or -7 are able
embryonic and adult tissues of the mouse, including the to maintain cell proliferation and Msx expression in the medi-
developing branchial arches at the time of critical morpho- al mandibular mesenchyme (Barlow and Francis-West, 1997;
genetic events. The phenotypic abnormalities resulting from Wang et al., 1998b, 1999). When placed laterally, BMPs induced
the targeted inactivation of TCOF1 should reveal its role in cell proliferation and ectopic expression of Msx expression in
Treacher-Collins syndrome. the lateral mandibular mesenchyme, where Msx genes are not

283
12(4) 276 300 (2001
12(4):276-300 2001)) Oral Biol Med
Crit Rev C'ral
Grit Rev Biol Med
283
normally expressed (Barlow and Francis-West, 1997; Wang et Robertson, 1999). The first branchial arches in double-mutants
al., 1998b, 1999). However, unlike the mandibular epithelium, were smaller than those in the wild-type, and displayed
laterally or medially placed BMPs did not support the elonga- increased apoptosis in the surface ectoderm at the base of the
tion of stage 23 chick mandibular mesenchyme and Meckel's arch. Analysis of the patterns of expression of twist and AP2
cartilage (Wang et al., 1999). showed defects in the mesodermally derived tissues, suggest-
Further insight into the roles of BMPs in outgrowth and ing that Bmp5 and Bmp7 are necessary for the maintenance/
morphogenesis of the developing mandible comes from stud- survival of mesodermally derived cells in the first branchial
ies in which beads soaked in BMPs were implanted in vivo at arch (Solloway and Robertson, 1999).
different stages into different regions of the developing chick However, the negative effects of BMPs on the outgrowth
mandible (Barlow and Francis-West, 1997; Ekanayake and Hall, of the stage 20/22 chick mandibular arch, and their inability to
1997; Wang et al., 1999; Mina et al., submitted manuscript). support outgrowth of stage 23 chick mandibular mesenchyme
Although application of BMP-2, BMP-4, and BMP-7 for 7 to 10 (Barlow and Francis-West, 1997; Ekanayake and Hall, 1997;
days to the lateral region of a stage 23 chick mandible did not Wang et al., 1999), suggest that BMPs, similar to their roles in
affect the growth of the mandibular mesenchyme, it did induce outgrowth of the developing limb (Pizette and Niswander,
changes in the morphology and patterns of the skeletal ele- 1999), may in fact be involved in limiting the outgrowth of the
ments in the caudal region (jaw joint) of mandibles on the mandibular process, and that other growth factors may act
implanted side. In contrast to their effects at stage 23, in vivo cooperatively with BMPs to promote the directed outgrowth
implantation of beads soaked in similar concentrations of of the mandibular mesenchyme.
BMPs into the lateral region of stage 20/22 mandibles exerted Furthermore, since all BMPs also induced cell death in the
negative effects, resulting in reduced growth of the mandibular lateral mandibular mesenchyme (Ekanayake and Hall, 1997;
process and complete loss of Meckel's cartilage and its articu- Barlow and Francis-West, 1998; Wang et al., 1999), and BMP-2
lating elements on the implanted side (Barlow and Francis- inhibited chondrogenesis of the mandibular mesenchyme in
West, 1997; Ekanayake and Hall, 1997; Mina et al., submitted vitro (Ekanayake and Hall, 1997), it is possible that deficiencies
manuscript). On the other hand, implantation of beads soaked in the outgrowth of the mandibular arch may be either due to
in BMPs into a more medial position (closer to the Msx- cell death and/or secondary to the inhibitory effects of BMPs
expressing mesenchyme in the medial region) of stage 20/22 on chondrogenesis and elongation of Meckel's cartilage that
mandibles induced the formation of an additional outgrowth are thought to regulate the growth of the mandible
from the body of Meckel's cartilage (Barlow and Francis-West, (Ekanayake and Hall, 1997). Essential roles for members of the
1997, Mina et al., submitted manuscript). TGF3 family in mandibular morphogenesis are also support-
Analysis of these data indicates that chick mandibular ed by phenotypic abnormalities in Activin /3A and ActRcII
mesenchyme from different regions (lateral vs. medial) at dif- knock-outs (Matzuk et al., 1995a,b,c).
ferent stages (20 vs. 23) responds differently to BMPs. In con-
trast to the observations in the chick mandible, a recent study FAMILY OF FIBROBLAST GROWTH FACTORS (FGFs)
showed that application of beads soaked in 100 ng/pL for 6 The FGFs consist of a family of 18 low-molecular-weight
days to the lateral region of organ-cultured mouse mandibles polypeptide growth factors (for reviews, see Naski and Omitz,
(E10) induced ectopic cartilage (Semba et al., 2000). However, 1998; Szebenyi and Fallon, 1999). The actions of FGFs are medi-
BMP beads applied to the medial region did not induce ectopic ated by FGFRs, a family of 4 transmembrane receptors with lig-
cartilage (Semba et al., 2000). and-induced tyrosine kinase activity (for reviews, see Szebenyi
The overlapping patterns of expression of Bmps in man- and Fallon, 1999; Klint and Claesson-Welsh, 1999). The FGFRs
dibular epithelium and the similarities between the effects of are similar in their overall structural organization and consist of
BMPs on the mandibular mesenchyme suggest cooperative or an extracellular ligand-binding domain, a single transmem-
redundant roles for BMPs in signaling cascades mediating epi- brane domain, and an intracellular tyrosine kinase domain. The
thelial-mesenchymal interactions regulating outgrowth and extracellular ligand-binding domain contains 3 immuno-
morphogenesis of the developing mandible. However, the globulin-like (Ig-like) domains, a heparin-binding domain, and
roles of BMPs in the outgrowth of the mandibular arch remain a stretch of 7 conserved acidic amino acids (acidic box).
unclear. Alternative splicing of the FGF receptor mRNAs generates sev-
On the one hand, the formation of an ectopic outgrowth eral different variants of the receptors that are either soluble or
from the body of Meckel's cartilage after implantation of BMP- membrane-anchored and have different ligand-binding proper-
beads to the medial region at stage 20/22 suggests that BMPs, ties. The functionally critical splicing event results in the differ-
through their interactions with target genes such as Msxl and ential exon usage in the region coding the carboxyl-terminal
Msx2, may be involved in the outgrowth and morphogenesis half of the third Ig-like domain. The third Ig-like domain in all
of the tip of the developing mandible. The phenotypic abnor- but FGFR-4 is encoded by 3 exons designated as A-C that are
malities in the Bmp-5/Bmp-7 double-knock-out (Solloway and alternatively spliced. Receptors containing the region encoded
Robertson, 1999) provide direct evidence for the involvement by exon A are secreted and may interfere with FGF signal trans-
of BMPs in outgrowth of the mandibular arch. Previous stud- duction. Receptors containing the region encoded by exon B or
ies demonstrated that loss of either Bmp5 or Bmp7 has no C differ in their ligand-binding properties and tissue distribu-
apparent effect on the development of the mandibular arch tion. In general, the B form appears to be expressed in epithelial
(Kingsley et al., 1992; King et al., 1994; Dudley et al., 1995; Luo cells and the C form in mesenchymal cells.
et al., 1995; Dudley and Robertson, 1997). However, unlike the There is compelling experimental evidence that interac-
single null mutants, Bmp-5/Bmp-7 compound mutants display tions between the members of the FGF and FGFR families
abnormalities in many organs in which Bmp5 and Bmp7 are co- have important roles in mediating epithelial-mesenchymal
expressed, including the first branchial arch (Solloway and interactions during embryogenesis (for reviews, see Burke et
(2001)
284 (nt Rev C)ral
Crit
Biol Med
Rev Oral Biol Med 12(4):276-300 (2001)
al., 1998; Martin, 1998; Hogan, 1999). that Fgfr2c may not have an essential role in the initial forma-
Similar to the Bmps, several Fgfs, including Fgf-2, Fgf-4, Fgf- tion of mandibular processes (Arman et al., 1998; Xu et al.,
8, and Fgf-9, are expressed in facial processes and the mandibu- 1998). Despite the lack of detailed descriptions of the craniofa-
lar arch (Niswander and Martin, 1992; Heikinheimo et al., 1994; cial structure, apparently a normal mandibular process forms
Crossley and Martin, 1995; Mahmood et al., 1995; Wall and in E10 mice that express a hypomorphic allele of Fgfr2 (Xu et al.,
Hogan, 1995; Barlow and Francis-West, 1997; Richman et al., 1998). On the other hand, transgenic expression of the extracel-
1997; McGonnell et al., 1998; Kettunen and Thesleff, 1998; lular ligand-binding domain of Fgfr2b (KGER) (the isoform that
Kettunen et al., 1998; Colvin et al., 1999). Whereas Fg,f-2 and Fgf- is expressed in the epithelium of facial processes), which acts as
9 are expressed throughout the mandibular epithelium, Fgf-8 a dominant-negative receptor of FGFR2b, resulted in the devel-
and Fgf-4 are expressed in restricted regions. Although Fgf-1, opment of smaller faces, suggestive of the involvement of
Fgf-5, Fgf-12, and Fgf-10 are expressed in mouse craniofacial FGFR2b in craniofacial development (Celli et al., 1998).
mesenchyme, detailed expression patterns for these genes have Until recently, the roles of FGF-8 in mandibular morphogen-
not yet been reported (Hebert et al., 1990; Haub and Goldfarb, esis were not fully understood. Previous studies in mouse
1991; Hartung et al., 1997). Fgf-13, Fgf-17, and Fgf-18 are not embryos indicate that Fgf8 is expressed in regions of the embryos
expressed in the facial processes of mouse embryos (Hartung et where epithelium is known to direct outgrowth (Crossley and
al., 1997; Maruoka et al., 1998). Martin, 1995). Studies in the chick face also showed correlations
Fgr genes are also expressed in the developing mandible of the patterns of expression of Fgf-8 in the epithelium of devel-
(Orr-Urtreger et al., 1991, 1993; Peters et al., 1992, 1993; Wilke et oping facial processes with areas where mesenchyme undergoes
al., 1997; Kettunen et al., 1998). Fgfr2b (KGFR) is expressed in the the greatest expansion and which express high levels of Msxl
mandibular epithelium, and Fgfrlc, Fg2c, both isoforms of (McGonnell et al., 1998). However, unlike its patterns of expres-
Fgfr3, and Fgfr4 in the mandibular mesenchyme. At early stages, sion in other facial processes, Fgf8 is expressed in the epithelium
both isoforms of Fffrl (flg, cek-1), Fgfr3 (cek-2), and Fgr2c (bek, in the lateral but not the medial region of the developing
cek-3) are expressed uniformly throughout the mandibular mes- mandible. Studies in mice suggest that FGF-8 may be involved in
enchyme. However, at later stages of development, Fgfr-2c and the establishment of the oral-aboral polarity of the developing
Fgfr3 are no longer expressed uniformly and are expressed in mandible (Tucker et al., 1999). A recent study in which Fgf8 was
restricted regions (Orr-Urtreger et al., 1991; Wilke et al., 1997; inactivated in the epithelium of the first branchial arch of trans-
Mina et al., submitted manuscript). High levels of expression of genic mice by means of Cre/lox technology has provided direct
both genes are seen in the mesenchyme in the lower border of evidence for roles of FGF8 in the development and morphogene-
the mandible that surrounds the hyomandibular cleft. sis of the lateral (but not medial) regions of the developing
Expression of Fgfr2c but not Fgfr3 also extends to the mes- mandible (Trumpp et al., 1999). This study provided direct evi-
enchyme in the medial region. At the time of initiation of chon- dence that the mandibular process is specified by at least 2 dis-
drogenesis, Fgfr2c and Ffr-3 are expressed in the condensing tinct functional regions: 2 large lateral (proximal) regions, where
mesenchyme of Meckel's cartilage, and Fe4 is expressed in the morphogenesis is dependent on and controlled by FGF-8, and a
developing muscles (Kettunen et al., 1998). small medial region, in which morphogenesis is independent of
Similar to BMPs, FGFs can also mimic many effects of FGF-8 and is controlled by other signaling molecules. In these
mandibular epithelium on the mandibular mesenchyme in mutants, the symphysial portion of Meckel's cartilage and the
vitro. In the absence of mandibular epithelium, FGF-2 and mandibular incisors and their associated bones were invariably
FGF-4 can maintain expression of Msxi and Msx2 in the mes- present. On the other hand, with the exception of the presence of
enchyme of the medial region (Mina et al., 1999, submitted a vestigial malleus, the skeletal elements in the lateral regions of
manuscript). In vitro studies indicate that, in contrast to BMPs, the developing mandible were absent (Trumpp et al., 1999).
in the absence of mandibular epithelium, locally applied FGFs Molecular analyses indicated that Fgf8;Nes-cre mutants fail to
can partially or completely support outgrowth of the express Lhx6 and, for the most part, Barxl. In addition, although
mandibular mesenchyme and Meckel's cartilage (Richman et ET-1 expression was not detected in most of the epithelium of the
al., 1997; Mina et al., 1999, submitted manuscript). Application developing mandible, it was detected in the epithelium of the
of beads soaked in recombinant FGF-2 and FGF-4 to the medi- hyomandibular cleft. In these mutants, expression of Gsc was
al (but not lateral) region of isolated stage 24 mandibular mes- seen only in a restricted region in which cells expressing Barxl
enchyme stimulated increases in the length of Meckel's carti- and ET-1 were found. These observations indicate that expression
lage. The cartilage rods formed in FGF-treated grafts were 60% of Lhx6, Barxl, Etl, and Gsc in the mandibular arch are depen-
or almost the same length as those formed in homotypic dent, directly or indirectly, on FGF8 signaling. However, expres-
recombinations. Taken together, these observations suggest sion of Dlxl, Dlx2, Dlx5, and Lhx7 in the mesenchyme of the lat-
essential roles for FGFs/FGFR2c (the only receptor with high eral regions and Pitxl in the mandibular epithelium was
levels of expression in the mesenchyme in this region) in the unchanged. Although Pax9 was not detected in the molar region
mediation of epithelial-mesenchymal interactions in the medi- of the mutant embryos, it was detected in the regions of pre-
al region of the developing mandible. sumptive mandibular incisors. The absence of abnormalities in
Based on their patterns of expression in vivo, FGF-2, FGF-4, the small medial region and the unaltered patterns of expression
and FGF-9 (but not FGF-8) are likely endogenous candidates for of Bmp-4, dHand, eHand, Msxl, and Msx2 (genes that are normal-
mediating the growth-promoting effects of mandibular epithe- ly expressed in the medial region) indicate that morphogenesis of
lium from the medial region. Furthermore, it has been shown the small medial region depends on signals other than FGF8
that Fgfr2c binds with high affinity and transmits mitogenic sig- (Trumpp et al., 1999). The similarities between the phenotypic
nals from FGF-1, -2, -4, and -9 (for review, see Szebenyi and abnormalities in the Fgf8;Nes-cre mutants and abnormalities in
Fallon, 1999). the human first-arch syndromes characterized by agnathia/
However, the phenotypes of Fgfr2 null mutants suggest micrognathia raise the possibility that mutations in Fgf8 or in
285
2001)
12)4) 276-300 (2001)
12(4):276-300 Crit
Crit Rev Oral Biol
Rev Oral Blot
Med
Med 285
genes directly upstream or downstream of it might cause some of expressed in the developing mandible in similar domains in
these human syndromes (Trumpp et al., 1999). regions near the cells expressing Shh (Platt et al., 1997).
Roles for FGFs and FGFRs in craniofacial development are The phenotypic abnormalities in Gli2, Gli-3, and
also supported by abnormalities in human syndromes associ- Gli2/Gli3 double-mutants indicated that Gli2 and Gli3 have
ated with mutations in FGFRs (for reviews, see Muenke and specific as well as redundant functions in the development
Schell, 1995; Yamaguchi and Rossant, 1995; Webster and of mandibular arch. Both Gli2 and Gli3 mutant mice die at
Donoghue, 1997; Wilkie, 1997; Naski and Ornitz, 1998). In gen- birth and have severe skeletal and facial abnormalities
eral, the disorders caused by mutations in FGFRs can be clas- (Vortkamp et al., 1991, 1992; Hui and Joyner, 1993; Mo et al.,
sified into 2 groups: (1) dwarfing conditions and (2) conditions 1997). Loss of Gli2 is associated with truncation of the prox-
with craniosynostosis. While dwarfing conditions have been imal regions of the maxilla and mandible, resulting in the
shown to be associated with mutations in FGFR3, the second absence of maxillary and mandibular incisors (Mo et al.,
group has been associated with mutations in FGFR2. 1997). On the other hand, Gli3 mutants displayed abnormal-
ities in the maxillary region but not in the mandible (Hui et
HEDGEHOG FAMILY al., 1994; Mo et al., 1997). In contrast to the single mutant,
The Hedgehog (Hh) family of secreted proteins is involved in mandibles of Gli2-/Gli2;Gli3 -/+ mutants were severely short-
the regulation of a wide range of developmental processes ened and had hypoplastic coronoid, condylar, angular, and
during vertebrate development (for reviews, see Ingham, alveolar bones.
1995; Hammerschmidt et al., 1997). The roles of Shh in craniofacial development have also
There are 3 known mammalian homologs of Hh: sonic hedge- been elucidated in more recent studies in chick embryos
hog (Shh), Indian hedgehog (11h), and desert hedgehog (Dhh). (Helms et al., 1997; Ahlgren and Bronner-Fraser, 1999; Hu and
Among these, Shh has been shown to be expressed in numerous Helms, 1999). Ahlgren and Bronner-Fraser (1999) showed that
sites of epithelial-mesenchymal interactions (Bitgood and inhibition of Shh signaling after neural tube closure resulted in
McMahon, 1995) and is thought to play an important role in many a transient decrease in neural tube cell proliferation and an
developmental processes, including dorso-ventral patterning of extensive increase in cell death in the neural tube and neural
the neural tube and somites, anterio-posterior patterning of the crest. These changes, in turn, resulted in decreased head size
limb buds, morphogenesis of the hindgut, and craniofacial devel- and loss of branchial arch structures. The phenotypes
opment (reviewed by Bumcrot and McMahon, 1995; Bronner- observed after reduction of Shh in chick embryos are similar to
Fraser and Fraser, 1997; Weed et al., 1997; Schneider et al., 1999). those observed after cranial neural crest ablation.
Most of the details of components of the Hh signaling Mice homozygous for a disruption of Shh die at birth with
pathway have been elucidated from studies in Drosophila. A defects in all tissues, including holoprosencephaly, which is
similar signaling cascade is also present in vertebrates (for characterized by abnormal forebrain and facial development.
reviews, see Alcedo and Noll, 1997; Ingham, 1998; Johnson Although the branchial arches appear to be normal at E9.5, cra-
and Scott, 1998; Murone et al., 1999). These studies indicate niofacial bones are severely affected in Shh null mutants and
that Hh signaling is transduced by a heterodimeric receptor are almost entirely absent (Chiang et al., 1996). Mice homozy-
complex that includes 2 transmembrane proteins: Smoothened gous for the Ptc deletion also die during embryogenesis and
(Smo), a member of the Frizzled (Fz) family of receptors, and have open and overgrown neural tubes (Goodrich et al., 1997).
Patched (Ptc). Hedgehog proteins bind specifically with high Facial abnormalities in human syndromes associated with
affinity to Ptc without any direct involvement of Smo. mutations in the components of the Hh signaling pathway also
However, Smo, which does not directly interact with Shh, is support the involvement of the Hh signaling pathway in
the signaling subunit of the receptor complex. mandibular and craniofacial morphogenesis (for reviews, see
The current model for the Hh signaling pathway sug- Roessler et al., 1996; Ming and Muenke, 1998; Ming et al., 1998).
gests that, in the absence of ligand, the Hh receptor complex In humans, haplo-insufficiency of Ptc results in Gorlin syn-
is inactive, because Ptc interacts with Smo, resulting in drome. Partial loss of Gli3 function results in Greig cephalopoly-
repression of Smo signal-transducing activity. Binding of Hh syndactyly syndrome (GCPS), and a point mutation in Shh
to Ptc causes a conformation change that alleviates this results in autosomal-dominant holoprosencephaly, one of the
repressive interaction and results in the release and activa- most common developmental defects of the forebrain and face.
tion of Smo from the complex (Murone et al., 1999). After its
activation, Smo activates the transcription of downstream WNT GENES
target genes via cubitus interruptus (Ci), a zinc-finger-con- The Wnt gene family also constitutes another group of signal-
taining protein, which appears to act as the final component ing factors with important roles in many developmental
in the Hh signaling pathway (Murone et al., 1999). processes, including specifications of the dorso-ventral axis of
Homologous to Ci, in vertebrates there are 3 Gli genes: Gli the developing limb (for reviews, see Johnson and Tabin, 1997;
(Glil), Gli2, and Gli3 (for review, see Ingham, 1998). Sokol, 1999) and modulation of chondrocyte differentiation in
The possible involvement of this pathway in mandibular the limb and face (Rudnicki and Brown, 1997; Kawakami et al.,
morphogenesis is suggested by the patterns of expression of 1999). This family consists of at least 15 developmentally reg-
genes in the Hh signaling pathway in the developing mouse ulated genes that encode cysteine-rich glycoproteins (Cadigan
mandible. Shh is expressed at low levels in the endoderm of and Nusse, 1997; Wodarz and Nusse, 1988).
the hyomandibular cleft (Platt et al., 1997; Hardcastle et al., Studies in mice indicate that Wnt5a, -1Qa, -1Ob, and -11 (but
1998). At early stages, Gli2, Gli3, and Smo are expressed not Wnt-1, and Wnt -2) are expressed in the facial mesenchyme
throughout the mandibular mesenchyme (Walterhouse et al., (Gavin et al., 1990; Dealy et al., 1993; Parr and McMahon, 1995;
1993; Hui et al., 1994; Hardcastle et al., 1998). Similar to their Takada et al.., 1994; Tanda et al., 1995; Christiansen et al., 1995;
patterns of expression in other tissues, Glil and Ptc are Wang and Shackleford, 1996; Yamaguchi et al., 1999).

286 Crit Rev Oral Biol


Crit Rev Oral Biol Med
Med

12(4):276-300 (2001)
Expression of Wnt5a in the facial processes is best-document- of Meckel's cartilage. The mandibular bone was hypoplastic
ed. Wnt5a expression is first detected at the time of formation and highly disorganized, with abnormal articulation to the
of the facial processes (Takada et al., 1994; Yamaguchi et al., maxillary arch and the base of the skull. Furthermore, the
1999), and later in a graded fashion (being highest at the tips tongue was severely hypoplastic. Teeth were intact but embed-
of the processes) in the frontonasal mass, and in maxillary and ded in loose mesenchyme instead of mandibular bone.
mandibular processes (Yamaguchi et al., 1999). The graded The defects observed in ETA-deficient embryos suggest that
pattern of expression of Wnt-5a in the facial processes is simi- the mutation disrupts the development of CNCC arising from
lar to its pattern of expression in the developing limb, in which the posterior midbrain as well as Ri, R2, R4, and R6. This sug-
high levels of Wnt5a are expressed in the Apical Ectodermal gests that ET-1 /ETA signaling may be required for cranial NCC
Ridge (AER) and mesenchymal cells underneath the AER migration and/or proliferation (Clouthier et al., 1998;
(Dealy et al., 1993; Kawakami et al., 1999; Yamaguchi et al., Yanagisawa et al., 1998). While migration of neural crest cells into
1999). These patterns of expression suggest a conserved role the arches of ETA-mutant embryos appears unaffected, the
for Wnt5a in the morphogenesis of many structures whose absence of ETA signaling appears to affect the proliferation/dif-
development requires extension from the primary body axis ferentiation of post-migratory crest cells and some apoptosis in
(Yamaguchi et al., 1999). This possibility is supported by phe- the branchial arch mesenchyme. In situ hybridization analysis of
notypes of the homozygous Wnt5a null mutants which have E9.5 and E10.5 ETA-/- null mutants indicated either the absence
morphological defects in outgrowing tissues, including limb, or a significant reduction in the expression of several transcrip-
maxilla, and mandible (Yamaguchi et al., 1999). tion factors, including Gsc, Dlx2, Dlx3, dHAND, and eHAND in
the post-migratory ectomesenchymal cells of the first and second
ENDOTHELIN PATHWAY branchial arches. Interestingly, while Barxl expression was
Ligands of the endothelin pathway consist of 3 structurally absent in the second branchial arch, its expression was unaffect-
related small peptide ligands, called ET-1, ET-2, and ET-3, that ed in the first branchial arch of E10.5 ETA-/- embryos (Clouthier
bind to 2 of the G-protein-coupled endothelin receptors, the et al., 1998, 2000). On the other hand, in these mutants, the
endothelin-A receptor (ETA) and the endothelin-B receptor expression of other transcription factors-including Prxl, Hoxa2,
(ETB) (for a recent review, see Kurihara et al., 1999, and refer- CRABP1, and Ufdl-was unaffected, suggesting that these fac-
ences therein). The endothelin pathway also includes 2 tors either act upstream of ETA signaling or belong to different
isozymes of endothelin-converting enzyme (ECE), ECE-1 and or parallel genetic pathways involved in mandibular develop-
ECE-2 (Kurihara et al., 1999, and references therein). ment (Clouthier et al., 2000). Analyses of ET1-/- null mutants
In situ hybridization analysis indicates that the ETA recep- showed that dHand and eHand are down-regulated in the
tor is expressed by migratory CNNC originating from the hind- branchial arches of these mutants (Thomas et al., 1998).
brain and ECE-1 in the head mesenchyme (reviewed by The phenotype of ETA null mutant mice resembles the
Kurihara et al., 1999). In the branchial arches, the spatial pat- human conditions collectively termed CATCH 22 or velocar-
terns of ET-1 and ETA expression are complementary, in that diofacial syndrome, and includes severe craniofacial deformi-
ETA is expressed by the CNCC-derived mesenchymal cells of ties and defects in the cardiovascular outflow tract (Wilson et
branchial arches and the ET-1 ligand is expressed in the surface al., 1993; Clouthier et al., 1998). However, the patterns of expres-
epithelium of arches 1, 2, and 3 and in the paraxial mesodermal sion of Ufdl, a gene shown to be deleted in CATCH 22 patients,
core of the first branchial arch (Barni et al., 1998; Clouthier et al., was unchanged in ETA-/- null mutants (Clouthier et al., 2000).
1998, 2000; Kurihara et al., 1999). ECE-1 is expressed in both the
mesenchyme and surface epithelium of branchial arches NOTCH SIGNALING PATHWAY
(Yanagisawa et al., 1998). The Notch signaling pathway is an evolutionary conserved
Several components of the endothelin pathway have been pathway with essential roles in mammalian embryonic devel-
inactivated in mice by homologous recombination (Baynash et opment (for reviews, see Gridley, 1997; Greenwald, 1998;
al., 1994; Hosoda et al., 1994; Kurihara et al., 1994; Clouthier et al., Weinmaster, 1998; Artavanis-Tsakonas et al., 1999). This path-
1998; Yanagisawa et al., 1998). These studies indicate that 2 way is characterized by interactions of the receptor Notch with
endothelin-mediated cell-cell signaling pathways (ET-1/ETA the Delta and Serrate/Jagged families of ligands. Studies in
and ET-3/ETB) are independently involved in the development Drosophila showed that the Notch gene encodes large trans-
of 4 distinct groups of NCC-derived tissues (for review, see membrane receptors that interact with membrane-bound lig-
Kurihara et al., 1999). Mice deficient in ET-3 and ETB exhibited ands encoded by Delta and Serrate genes (Greenwald, 1998;
similar abnormalities and revealed essential roles for ET-3/ETB- Artavanis-Tsakonas et al., 1999; Gray et al., 1999). Genes
mediated signaling in the development of epidermal and homologous to members of the Notch signaling pathway have
choroidal melanocytes and enteric neurons (Baynash et al., 1994; been cloned in mammals and include the 4 genes encoding the
Hosoda et al., 1994). On the other hand, mice deficient in ETA ligands: Jagged-1 (Serrate-1), Jagged 2 (Serrate 2), Delta-like 1
and ET-1 showed striking craniofacial and cardiovascular (Dlll), and Delta-like 3 (D113) (Bettenhausen et al., 1995;
abnormalities that were virtually identical to each other and to Lindsell et al., 1995; Shawber et al., 1996; Dunwoodie et al.,
those observed in mice deficient for ECE-1 (Kurihara et al., 1994, 1997; Gray et al., 1999).
1999; Clouthier et al., 1998, 2000; Yanagisawa et al., 1998). The importance of the Notch signaling pathway in develop-
In ET-1, ETA, and ECE-1 null mutants, numerous struc- ment is also implicated by the findings that 3 human diseases-
tures derived from branchial arches 1, 2, and 3 were affected T-cell acute lymphoblastic leukemia/lymphoma, the Alagille
(Kurihara et al., 1994, 1999; Clouthier et al., 1998; Yanagisawa et syndrome, and CADASIL (Cerebral Autosomal Dominant
al., 1998). The most striking phenotype in all 3 mutants was the Arteriopathy with subcortical Infarcts and Leukoencepha-
poorly formed mandible that sometimes lacked the midline lopathy)-are associated with mutations in genes encoding
fusion. The mutant mandibles also exhibited complete absence Notch-1, Jagged-1, and Notch-3, respectively (for reviews, see
287
112(4) 276-300 ((200h
2(4):276-300 2001 ) Crit Oral Biol Med
Rev Oral Biol
Grit Rev Med
287
Gridley, 1997; Joutel and Tournier-Lasserve, 1998). ed by observations showing that mitogenic growth factors that
Among these, Alagille syndrome exhibits autosomal-domi- signal through the Erk-MAP kinases pathway, such as EGF, can
nant inheritance and is one of the major forms of chronic liver antagonize the BMP-induced responses by phosphorylation of
disease in childhood (Krantz et al., 1997). Accompanying features Smad-1 at multiple serines in the linker region (Kretzschmar et
of this syndrome include a characteristic facial appearance, al., 1997). Although Erk-mediated phosphorylation of Smad-1
including a pointed mandible (Krantz et al., 1997). In the majori- does not directly interfere with the association of Smad-1 with
ty of patients, mutations in JAG1 result in the formation of trun- Smad-4, it prevents the translocation of Smad-I to the nucleus,
cated protein, suggesting that Alagille syndrome is due to the thereby inhibiting BMP-4 signal transduction (Kretzschmar et
haploinsufficiency of JAG1 (Li et al., 1997; Oda et al., 1997; Krantz al., 1997; Attisano and Wrana, 1998; Kretzschmar and
et al., 1998; Yuan et al., 1998). However, studies in Drosophila sup- Massague, 1998). This possibility is supported by the recent
port alternative dominant-negative roles for truncated forms of observations in micromass cultures (Nonaka et al., 1999) that
Serrate proteins (Hukriede et al., 1997; Sun and Artavanis- showed that the addition of EGF inhibited BMP-induced
Tsakonas, 1997). Smadl nuclear translocation and accumulation.
Studies in developing mice indicate that, whereas Jaggedl The roles of EGF/EGFrs in craniofacial morphogenesis
(Serratel) is expressed in the maxillary and mandibular mes- were confirmed by craniofacial abnormalities in Egfr null
enchyme, Jagged2 (similar to Notchl) is expressed in the epithe- mutants (Miettinen et al., 1999). The abnormalities in Egfr null
lium of the branchial arches (Mitsiadis et al., 1995, 1997, 1998a; mutants included a pointed snout, high incidence of cleft
Shawber et al., 1996; Sidow et al., 1997; Valsecchi et al., 1997; Jiang palate, short mandibles, and poorly developed Meckel's carti-
et al., 1998). Mice homozygous for Jaggedl null mutations die lage (Miettinen et al., 1999). Furthermore, in Egfr null mutants,
early in embryogenesis, and mice heterozygous for the Jaggedl there were decreases in the amount of matrix metallopro-
null allele exhibit abnormalities in the eye but not in the cranio- teinases (MMPs). Inactivation of MMPs in explanted wild-
facial region (Xue et al., 1999). Null mutants of Jagged2 (Jiang et type mandibles resulted in defects similar to those in Egfr null
al., 1998) and expression of a hypomorphic allele of Jagged2 mutants, suggesting that MMPs may act downstream of the
(Sidow et al., 1997) exhibit abnormalities in many Jagged2 EGF/EGFR signaling pathway (Miettinen et al., 1999).
(Serrate2)-expressing tissues, including defects in the craniofa-
cial region. However, abnormalities in the developing mandible PLATELET-DERIVED GROWTH FACTORS (PDGFs)
have not been reported. Mice homozygous for a targeted deletion of the PDGFot recep-
tor that is expressed by CNNC-derived mesenchyme of the
EPIDERMAL GROWTH FACTOR (EGF) branchial arches (Orr-Urterger and Lonai, 1992; Schatteman et
The EGF-superfamily has been implicated in inductive interac- al., 1992; Chai et al., 1998) and can bind to all PDGF (for
tions in the first branchial arch, including those involved in review, see Westermark et al., 1989) exhibit significant defects
odontogenesis, bone and cartilage formation, and outgrowth of in the skull, most noticeable in the nasal bone (Soriano, 1997).
the maxillary and mandibular arches. EGF and EGFR are However, PDGFRot mutants have relatively normal maxillary
expressed in the developing mandible (Partanen et al., 1985; and mandibular arches (Soriano, 1997). Increased apoptosis
Partanen and Thesleff, 1987; Snead et al., 1989; Kronmiller et al., observed along the pathway of CNNC migration and mes-
1991; Shum et al., 1993). Treatment of explants of mouse enchyme of the branchial arches in these mutants suggested
mandibular arches with EGF antisense oligonucleotides results roles for PDGFs and PDGFRo- in the survival of non-neuronal
in the formation of bilaterally symmetrical Fussilli-formed dys- derivatives of CNNC (Soriano, 1997).
morphic Meckel's cartilage, and inhibition of EGFR kinase
activity caused overall retardation in mandibular growth and MERGING OF THE TWO MANDIBULAR PROCESSES
the complete absence of Meckel's cartilage (Shum et al., 1993). In Another essential feature of facial morphogenesis is the union
addition, it has been shown that EGF can substitute for the mito- of the facial prominences/processes that occurs either by
genic effects of mandibular epithelium on the underlying mes- fusion or by merging of the adjacent processes (for review, see
enchyme, suggesting a role for EGF and EGF-like protein in Ten Cate, 1998). In general, the process of fusion seen in the
epithelial-mesenchymal interaction involved in the outgrowth palatal processes involves initial contact and subsequent
of the developing mandible (Coffin-Collins and Hall, 1989; Hall fusion of the surface epithelia of the neighboring processes,
and Coffin-Collins, 1990). A recent study by Nonaka et al. (1999) removal of the epithelial cells from the fusion region, and
showed that exogenous EGF inhibited the BMP-4-induced merging of the mesenchymal cells of the neighboring process-
ectopic cartilage in organ-cultured mouse and chick mandibles. es (reviewed by Shuler, 1995, and references therein). On the
It has also been shown that application of EGF-releasing other hand, union of other facial processes, such as the
beads to mouse and chick mandibular mesenchyme induces a mandibular processes, appears to involve the merger of 2
translucent zone and extensive cell proliferation, and prevents incompletely separated processes. During this merger, the fur-
apoptosis (Vaahtokari et al., 1996; Wang et a!., 1998a, 1999). row between these processes is eliminated (Ten Cate, 1998).
Although EGF beads are not able to induce expression of Msxl, The mechanisms for removal of the epithelial cells that results
Msx2, and Bmp4 in these mesenchymes, they were able to mod- in mesenchymal continuity in facial processes are thought to
ulate the effects of BMPs on the expression of Msxl and Msx2 be mediated by either programmed cell death (apoptosis),
(Wang et al., 1998a, 1999). These observations suggest the pos- epithelial-mesenchymal transformation, or migration to adja-
sibility that EGF, by modulating the effects of BMPs on the cent epithelia (Shuler, 1995). Although in many regions of the
expression of Msxl and Msx2 in mandibular mesenchyme, may developing face, cell death occurs in the epithelium at sites of
be involved in mediating inductive interactions during the ini- fusion, analysis of available datas suggest that union of the 2
tiation phase of odontogenesis and during outgrowth of the mandibular processes does not involve epithelial cell death or
mandibular arch (Wang et al., 1999). This possibility is support- epithelial-mesenchymal transformation. Rather, it involves

288288CrtRvOaBilMd1(:7600(01 Crit Rev Oral Biol Med 1 2(4):276-300 (2001 )


epithelial cell migration (Shuler et al., 1992; Shuler, 1995; Chai thelial-mesenchymal interactions. Based on patterns of gene
et al., 1997; McGonnell et al., 1998). expression and on functional studies, the mandibular arch can be
Studies in the mouse embryo indicate that merger of the 2 divided into 3 overlapping functional regions (Fig. 1A): a small
mandibular processes starts at E9.5 and is completed within 24 medial region, 2 large lateral regions, and the regions of the hyo-
to 36 hrs at Ell (Chai et al., 1997). During this process, the mandibular clefts located in the lower border of the lateral
epithelium covering the tips of the mandibular processes con- regions. Available evidence suggests that each of these regions
sists of 2 cell layers that adhere to each other and subsequent- contains signaling centers. Signaling centers are defined as
ly fuse to form an epithelial seam about 3 or 4 cell layers thick. groups of cells that regulate the behavior of the surrounding cells
Disappearance of the epithelial seam in the midline is first by producing positive and negative intercellular signaling mole-
seen at E10.5 and is completed at Ell, at which time the mes- cules, including FGFs, BMPs, Hedgehogs, Wnts, and EGFs
enchymal cells of 2 mandibular processes mix together and (Hogan, 1996).
become continuous. In contrast to the region of merging in the The 3 functional regions can be characterized by restricted and
midline, fusion/merging of the mandibular process to the region-specific expression of many of the evolutionary conserved
maxillary process and hyoid process involves epithelial apop- signaling factors and transcription factors. During recent years,
tosis (McGonnell et al., 1998). Interestingly, mesenchymal cells it has also become clear that the restricted patterns of expression
around these sites undergo expansion and bi-directional cell of many regulatory molecules in the mesenchyme of these
movements that result in a mixing of the mesenchymal cells regions (e.g., Pax9, Barxl, Msxl, Msx2, Gsc) are established by
between the 2 mandibular process-
es and mandibular processes and
neighboring processes (McGonnell
et al., 1998).
Conclusions
This review attempts to provide an
overview of the current under-
standing of the putative regions
and signaling cascades involved in
mandibular outgrowth and mor-
phogenesis. Classic tissue recom-
bination studies have demonstrat-
ed that the epithelium is required
for outgrowth and correct skeleto-
genesis in the mandibular arch.
However, these studies did not
identify specific regions in the
epithelium or mesenchyme that
regulate the outgrowth of the
developing mandible.
As was discussed in this re-
view, insight into putative regions
and signaling molecules involved in
mandibular outgrowth and mor-
phogenesis has come through stud-
ies examining: patterns of expres-
sion of signaling molecules, func- Figure 2. Summary of multiple genetic pathways that are involved in mandibular morphogenesis.
tional studies examining the simi- The differences in t e signaling cascades in the medial vs. lateral regions of the developing mandible
larities and differences between the were shown by Trumpp et al. (1 999). Morphogenesis of the large lateral region is dependent on and
effects of signaling molecules and controlled by FGF8. In the lateral region, Fgf8 expression requires Pitx2 [indicated by (1),thesee Lin et
mandibular epithelium, abnormali-
al., 1999; Lu et al., 1 999b], and the expression of Pax9 and Lhx6 in the oral side and expres-
sion of Barxl in both the oral and aboral regions of the hyomandibular cleft is dependent on FGF8
ties in the developing mandible signaling, which also regulates the expression of ET-1 in the mandibular epithelium [indicated by (2),
caused by targeted inactivation of see Trumpp et al., 1999].
candidate molecules, and identifica- In the region of the hyomandibular cleft, the expression of Gsc and Dlx3 is dependent on ET-
tion of mutated genes associated 1 /ETA signaling [indicatedby (3) and (A), see Clouthier et al., 1998, 2000]. Gsc expression in this
with craniofacial abnormalities the signaling
region is also dependent on DIx5 [indicated by (5), see Depew et al., 1999]. However,factors
pathway regulating the expression of other signaling molecules and transcription in this
affecting the mandible in humans.
Collectively, these studies show that region has not been fully identified (indicated by .).
signaling factors involved in the Unlike the lateral region, morphogenesis of the small medial region depends on signals other
outgrowth and morphogenesis of than FGF8. In this region, the expression of dHand and Msxl is dependent on ET-1 /ETA signalingis
[indicated by (6), Thomas et al., 1998]. The expression of Msxl and Msx2 in the medial region
the mandibular arch are similar to also de encent on BMP and FGF signaling secreted by the epithelium of medial region [indicated by
those involved in morphogenesis of (7) and (8), see references in the text]. However, the hierarchy of the signaling molecules in this
many other organs in which mor- region and their roles in regulating other genes expressed in the medial region are still not fully
phogenesis is dependent on epi- understood (indicated by ?).

2001)
12(4) 276-300 (2001) Crit Rev Oral Biol Med
Oral Biol Med
289
12(4):276-300
stage-specific antagonistic interactions of BMP-4 and FGF-8 sig- downstream target genes including ET-1, Lhx6, and Barxl, is
naling in the lateral and medial regions (Neubuser et al., 1997; involved in mediating the interactions that regulate survival,
Thomas et al., 1998; Tucker et al., 1998b). outgrowth, and morphogenesis of this region (Fig. 2).
The small medial region (Fig. 1A), where the 2 mandibular The hyomandibular cleft region separating the mandibular
processes merge, is characterized by the restricted patterns of and hyoid arches (Fig. 1A) gives rise to middle ear structures (for
expression of Bmp4 in the epithelium and Msxl, Msx2, HAND, review, see Mallo, 1998). In these regions, high levels of expres-
Bmp2, and Fgfr2 in the underlying mesenchyme. In addition, Dlx, sion of many regulatory genes-including Shh, Fgfs, Ffr2c, Ff3,
Prxl, Prx2, and Wnt5a genes are expressed at high levels in the Msxl, Gsc, Prxl, Et-1, Glil, and ptc-are detected in the epitheli-
mesenchyme of this region. This region gives rise to the medial um and mandibular mesenchyme surrounding these regions
region of the mandibular arch containing incisor teeth, the most (Mallo et al., 1998), suggestive of the intriguing possibility that
medially (proximally) located skeletal elements including the these additional signaling centers are involved in mandibular as
symphysis that separates the 2 rods of Meckel's cartilage. Ex- well as middle ear morphogenesis. This possibility is further sup-
perimental evidence indicates that the medial region is derived ported by abnormalities in the developing mandible and middle
exclusively from CNCC originating from the midbrain (Fig. 1B), ear structures of homozygous null mice lacking these genes
contains highly proliferative mesenchymal cells that, for the most (Mallo et al., 1998). Furthermore, the mirror-image duplication of
part, lack in vitro chondrogenic potential, and makes a significant caudal mandibular arch skeletal elements in the Hoxa-2 null
contribution to the overall growth of the developing mandible. mutants suggests that inductive signals from the region of the
Their proliferative activity and the expression of several genes in hypomandibular cleft may be involved in the establishment of
the mesenchyme of the medial region are dependent on signals the polarity of the skeletal elements derived from the caudal
derived from the overlying epithelium. The mesenchymal cells in region of the mandibular arch, including the middle ear struc-
the lower medial region also contain apoptotic cells. Analysis of tures (Rijli et al., 1993). Furthermore, branchial arches with normal
the available data suggests that epithelial-mesenchymal interac- skeletal polarity develop in embryos in which short segments of
tions in the medial region may be involved in directional out- the cranial neural tube were rotated along the rostrocaudal axis or
growth of the developing mandible, and that ET-1/ETA, dorsoventral axis (Noden, 1978, 1983a,b; Hall, 1982). This sug-
FGFs/FGFR2C, BMPs/BMPRs, EGF/EGFR, ET-11ETA, and Wnt5a, gests the existence of putative signaling centers (polarizing sig-
through their interactions with downstream target genes (inclu- nals) in the local environment into which CNCC migrate and
ding Msx, Prx, and dHand) are a component of the signaling path- which regulate the polarities of the skeletal tissues within the
way mediating the interactions in this signaling center (Fig. 2). mandible arch. Although these observations suggest that sig-
The possible involvement of the medial region in regulating nal(s) from the hyomandibular cleft may be involved in pattern-
the directional outgrowth of the developing mandible is support- ing the skeletal elements within the mandible, the roles of the
ed by the expression of mitogens in this region and by abnormal- hyomandibular cleft region in the development of the mandibu-
ities in mandibular outgrowth in mice lacking regulatory genes lar arch are poorly understood.
that are either specifically expressed in this region (e.g., dHand-/- Experimental evidence suggests that expression of Gsc in
and Msx-1-/-; for more details, see text) or their expression the ectomesenchyme of the hyomandibular cleft region is
extends into the medial region (e.g., Prxl/Prx2 compound dependent on FGF8 and ET-1/ETA signaling from the overlying
mutants, ET-1-/-, ETA-/-, ETC-1-/-; for more details, see text). epithelium (Clouthier et al., 1998, 2000; Trumpp et al., 1999) (Fig.
The medial region may also be involved in inhibiting chon- 2). These studies also indicate that FGF-8 acts upstream of ET-1
drogenesis at the midline (symphysis), thus preventing the 2 in this pathway (Trumpp et al., 1999). The abnormalities in the
rods of Meckel's cartilage from fusing and allowing for contin- mandibular arch in Gsc null mutants are similar to, although
ued growth and morphogenesis of the mandible and Meckel's less severe than, abnormalities in Dlx5 null mutants, suggesting
cartilage. The abnormalities in Prxl/Prx2 double-knock-outs, that these gene products may directly or indirectly interact and
and in mice lacking 3 members of the endothelin pathway function in the same pathway. In situ hybridization studies
(ETA, ET-1, ECE-1), suggest essential roles for these genes in the showed decreased Gsc expression at E10.5 in Dlx5 - null
formation of midline structures in the developing mandible. mutants (Depew et al., 1999).
The 2 larger lateral regions (Fig. 1B), where chondrogen- The absence of abnormalities in the medial region in FGF8
esis and osteogenesis are initiated in vivo, are derived from mutants also provides clear evidence that, unlike the lateral
CNNC originating from the midbrain and from R2-4 (Fig. region, the small medial region depends on signals other than
2A), and give rise to the regions containing the molar teeth, FGF8 (Trumpp et al., 1999) (Fig. 2).
most of the skeletal components of the mandibular arch The similarities in the mandibular phenotypes in dHAND-
(including the joint elements), and the middle ear structures. I-, ETA-I-, ETi-l-, Prxl/Prx2 double-knock-outs, and Msxl-/-
The lateral region is characterized by restricted expression of mutants also suggest that direct or indirect interactions
Fgf8 in the epithelium and high levels of expression of a vari- between products of these genes are involved in morphogene-
ety of genes, including Dlx genes, Barxl, Lhx-6/7, Pitxl, Pax sis of the medial region. Molecular analyses of dHAND-/-,
genes, and Wnt5a. Knock-outs of many of the genes expressed ETA-!-, ET1-/- mutants indicate that the "ET-1-dHAND-Msxl
in the lateral region also result in mandibular hypoplasia of pathway" constitutes one of the genetic pathways regulating
various degrees and/or skeletal abnormalities in the articu- mandibular outgrowth. This pathway suggests that ET-1/ETA
lating ends of the mandibular arch (coronoid, condylar, and signaling is required for high levels of expression of dHAND in
angular processes), indicating that the lateral regions also the underlying mesenchyme, which, in turn, regulates expres-
contain signaling centers regulating mandibular morphogen- sion of Msxl (but not Msx2) (Fig. 2). However, the unchanged
esis (Fig. 2). The abnormalities in the lateral but not the medi- patterns of expression of Prxl in the mandibles of dHAND-/-
al regions of the mandibles of the Fgf8 mutants provide direct and ETI-1- mutants (Clouthier et al., 1998, 2000; Thomas et al.,
evidence that FGF-8 signaling, through interactions with 1998) indicate that Prxl is not regulated by ET-1 and either acts
290
290 Crit
Crit Rev Oral Biol
Rev Oral Biol Med
Med
12(4):276-300

12(4):276-300 (2001)
upstream of dHAND or belongs to different or parallel genet- anterior neuroectoderm specification during gastrulation.
ic pathways. Based on the patterns of expression of mRNAs, Development 121:3279-3290.
BMP-4, FGF2, FGF4, and FGF9 are also likely candidates Acampora D, Mazan S, Avantaggiato V, Barone P, Tuorto F,
involved in regulating morphogenesis of the medial region. Lallemand Y, et al. (1996). Epilepsy and brain abnormali-
In addition to Bmps, ET-1, and Fgfs, mandibular epithelium ties in mice lacking the Otxl gene. Nat Genet 14:218-222.
also expresses other regulatory genes, including Jagged2, Pitx2, Acampora D, Mazan S, Tuorto F, Avantaggiato V, Tremblay JJ,
PDGFA, and Egf and Egfr. In situ hybridization analysis shows a Lazzaro D, et al. (1998). Transient dwarfism and hypogo-
lack of Fgf8 in the mandibular epithelium of Pitx2 knock-outs, nadism in mice lacking Otxl reveal prepubescent stage-spe-
placing Pitx2 upstream of FGF-8 (Fig. 2). Although the roles of cific control of pituitary levels of GH, FSH and LH.
some of these gene products in the outgrowth and morphogene- Development 125:1229-1239.
sis of the mandibular arch have been elucidated by phenotypic Acampora D, Merlo GR, Paleari L, Zerega B, Postiglione MP,
abnormalities in knock-outs, with few exceptions, the effects of Mantero S, et al. (1999). Craniofacial, vestibular and bone
many of these gene products on the expression of the target genes defects in mice lacking the distal-less-related gene dlx5.
in the mandibular mesenchyme have not been fully examined. Development 126:3795-3809.
In summary, abnormalities in the developing mandibles of Acampora D, Gulisano M, Simeone A (2000). Genetic and mol-
various mouse mutants caused by targeted inactivation of indi- ecular roles of Otx homeodomain proteins in head devel-
vidual candidate molecules indicate that multiple factors form a opment. Gene 246:23-35.
combinatorial code which regulates outgrowth and morphogen- Ahlgren SC, Bronner-Fraser M (1999). Inhibition of Sonic
esis of the mandibular arch and its skeletal elements. In addition, hedgehog signaling in vivo results in craniofacial neural
the similarities between the mandibular phenotypes of various crest cell death. Curr Biol 9:1304-1314.
mouse mutants suggest that these gene products regulate Alcedo J, Noll M (1997). Hedgehog and its patched-
mandibular outgrowth (and morphogenesis) through a common smoothened receptor complex: a novel signalling mecha-
pathway by acting on similar populations of cells. These mutants nism at the cell surface. Biol Chem 378:583-590.
provide powerful tools for examining the hierarchy of signaling Ang SL, Jin 0, Rhinn M, Daigle N, Stevenson L, Rossant J
molecules involved in mandibular morphogenesis. Furthermore, (1996). A targeted mouse Otx2 mutation leads to severe
the generations of compound mutants (mice bearing mutations defects in gastrulation and formation of axial mesoderm
in different pairs of related genes) provide strong evidence for and to deletion of rostral brain. Development 122:243-252.
shared and/or unique functions for these genes and are valuable Aono A, Hazama M, Notoya K, Taketomi S, Yamasaki H,
for placing them within the genetic pathways. Further insight Tsukuda R, et al. (1995). Potent ectopic bone-inducing
into the roles of these gene products will also come from pheno- activity of bone morphogenetic protein-4/7 heterodimer.
typic analysis of animals in which candidate genes are over- Biochem Biophys Res Commun 210:670-677.
expressed in the developing mandible. An essential requirement Arman E, Haffner-Krausz R, Chen Y, Heath JK, Lonai P (1998).
for the performance of these studies in transgenic mice will be the Targeted disruption of fibroblast growth factor (FGF)
identification of promoter elements, which specifically direct receptor 2 suggests a role for FGF signaling in pregastrula-
gene expression to the mandibular arch. Furthermore, accessibil- tion mammalian development. Proc Natl Acad Sci USA
ity of the developing face in the chick embryo for experimental 95:5082-5087.
manipulations and infection by means of retrovirus vectors, Artavanis-Tsakonas S, Rand MD, Lake RJ (1999). Notch sig-
together with detailed analysis of chick mutants with abnormali- naling: cell fate control and signal integration in develop-
ties in the lower jaw, will continue to provide invaluable infor- ment. Science 284:770-776.
mation in the area of craniofacial biology. Elucidating the molec- Attisano L, Wrana JL (1998). Mads and Smads in TGF beta sig-
ular pathways and mechanisms regulating development and nalling. Curr Opin Cell Biol 10:188-194.
morphogenesis of the mandibular arch is fundamental to an Balling R, Helwig U, Nadeau J, Neubuser A, Schmahl W, Imai K
understanding of the pathogenesis of the numerous human con- (1996). Pax genes and skeletal development. Ann NY Acad Sci
genital syndromes which manifest severe abnormalities of first 785:27-33.
branchial arch derivatives, including first-arch syndromes, Bally-Cuif L, Boncinelli E (1997). Transcription factors and
Treacher-Collins syndrome, and Pierre Robin sequence. head formation in vertebrates. Bioessays 19:127-135.
Barlow AJ, Francis-West PH (1997). Ectopic application of
Acknowledgments recombinant BMP-2 and BMP-4 can change patterning of
The author thanks Drs. Edward Kollar, William Upholt, Dimitrios Velonis, developing chick facial primordia. Development 124:391-398.
and Y.-H. Wang, for their helpful discussions and critical comments on the Barlow AJ, Bogardi JP, Ladher R, Francis-West PH (1999).
manuscript. The work in the author's laboratory was supported by NIH Expression of chick Barx-1 and its differential regulation
Grant DE08682 by FGF-8 and BMP signaling in the maxillary primordia.
Dev Dyn 214:291-302.
Barni T, Fantoni G, Gloria L, Maggi M, Peri A, Balsi E, et al.
REFERENCES (1998). Role of endothelin in the human craniofacial mor-
Aberg T, Wozney J, Thesleff I (1997). Expression patterns of phogenesis. J Craniofac Genet Dev Biol 18:183-194.
bone morphogenetic proteins (Bmps) in the developing Baynash AG, Hosoda K, Giaid A, Richardson JA, Emoto N,
mouse tooth suggest roles in morphogenesis and cell dif- Hammer RE, et al. (1994). Interaction of endothelin-3 with
ferentiation. Dev Dyn 210:383-396. endothelin-B receptor is essential for development of epi-
Acampora D, Mazan S, Lallemand Y, Avantaggiato V, Maury dermal melanocytes and enteric neurons. Cell 79:1277-1285.
M, Simeone A, et al. (1995). Forebrain and midbrain Bei M, Maas R (1998). FGFs and BMP4 induce both Msxl-inde-
regions are deleted in Otx2-/- mutants due to a defective pendent and Msxl-dependent signaling pathways in

12(4) 276 3.O (2001)


2001) Crit Rev Oral Biol Med
Oral Biol Med
291
12(4):276-300
early tooth development. Development 125:4325-4333. Chisaka 0, Capecchi MR (1991). Regionally restricted devel-
Berk M, Desai SY, Heyman HC, Colmenares C (1997). Mice opmental defects resulting from targeted disruption of the
lacking the ski proto-oncogene have defects in neurula- mouse homeobox gene hox-1.5. Nature 350:473-479.
tion, craniofacial, patterning, and skeletal muscle devel- Christiansen JH, Dennis CL, Wicking CA, Monkley SJ, Wilkinson
opment. Genes Dev 11:2029-2039. DG, Wainwright BJ (1995). Murine Wnt-11 and Wnt-12 have
Bettenhausen B, Hrabe de Angelis M, Simon D, Guenet JL, temporally and spatially restricted expression patterns dur-
Gossler A (1995). Transient and restricted expression dur- ing embryonic development. Mech Dev 51:341-350.
ing mouse embryogenesis of Dll1, a murine gene closely Clouthier DE, Hosoda K, Richardson JA, Williams SC,
related to Drosophila delta. Development 121:2407-2418. Yanagisawa H, Kuwaki T, et al. (1998). Cranial and cardiac
Birgbauer E, Sechrist J, Bronner-Fraser M, Fraser S (1995). neural crest defects in endothelin-A receptor-deficient
Rhombomeric origin and rostrocaudal reassortment of mice. Development 125:813-824.
neural crest cells revealed by intravital microscopy. Clouthier DE, Williams SC, Yanagisawa H, Wieduwilt M,
Development 121:935-945. Richardson JA, Yanagisawa M (2000). Signaling pathways
Bitgood MJ, McMahon AP (1995). Hedgehog and Bmp genes crucial for craniofacial development revealed by endothe-
are coexpressed at many diverse sites of cell-cell interac- lin-A receptor-deficient mice. Dev Biol 217:10-24.
tion in the mouse embryo. Dev Biol 172:126-138. Coffin-Collins PA, Hall BK (1989). Chondrogenesis of man-
Bronner-Fraser M, Fraser SE (1997). Differentiation of the ver- dibular mesenchyme from the embryonic chick is inhibit-
tebrate neural tube. Curr Opin Cell Biol 9:885-891. ed by mandibular epithelium and by epidermal growth
Brown JM, Wedden SE, Millburn GH, Robson LG, Hill RE, factor. Int J Dev Biol 33:297-311.
Davidson DR, et al. (1993). Experimental analysis of the Cohen MM Jr (2000). Craniofacial disorders caused by muta-
control of expression of the homeobox-gene Msx-I in the tions in homeobox genes MSX1 and MSX2. J Craniofac
developing limb and face. Development 119:41-48. Genet Dev Biol 20:19-25.
Brown JM, Robertson KE, Wedden SE, Tickle C (1997). Cohen SM, Bronner G, Kuttner F, Jurgens G, Jackle H (1989).
Alterations in Msx 1 and Msx 2 expression correlate with Distal-less encodes a homeodomain protein required for
inhibition of outgrowth of chick facial primordia induced limb development in Drosophila. Nature 338:432-434.
by retinoic acid. Anat Embryol 195:203-207. Colvin JS, Feldman B, Nadeau JH, Goldfarb M, Ornitz DM
Bumcrot DA, McMahon AP (1995). Somite differentiation. (1999). Genomic organization and embryonic expression
Sonic signals somites. Curr Biol 5:612-614. of the mouse fibroblast growth factor 9 gene. Dev Dyn
Burke D, Wilkes D, Blundell TL, Malcolm S (1998). Fibroblast 216:72-88.
growth factor receptors: lessons from the genes. Trends Couly GF, Coltey PM, Le Douarin NM (1992). The develop-
Biochem Sci 23:59-62. mental fate of the cephalic mesoderm in quail-chick
Cadigan KM, Nusse R (1997). Wnt signaling: a common theme chimeras. Development 114:1-15.
in animal development. Genes Dev 11:3286-3305. Couly GF, Coltey PM, Le Douarin NM (1993). The triple origin
Celli G, LaRochelle WJ, Mackem S, Sharp R, Merlino G (1998). of skull in higher vertebrates: a study in quail-chick
Soluble dominant-negative receptor uncovers essential chimeras. Development 117:409-429.
roles for fibroblast growth factors in multi-organ induc- Couly G, Grapin-Botton A, Coltey P, Le Douarin NM (1996).
tion and patterning. EMBO J 17:1642-1655. The regeneration of the cephalic neural crest, a problem
Chai Y, Sasano Y, Bringas P Jr, Mayo M, Kaartinen V, revisited: the regenerating cells originate from the con-
Heisterkamp N, et al. (1997). Characterization of the fate of tralateral or from the anterior and posterior neural fold.
midline epithelial cells during the fusion of mandibular Development 122:3393-3407.
prominences in vivo. Dev Dyn 208:526-535. Couly G, Grapin-Botton A, Coltey P, Ruhin B, Le Douarin NM
Chai Y, Bringas P Jr, Mogharei A, Shuler CF, Slavkin HC (1998). (1998). Determination of the identity of the derivatives of
PDGF-A and PDGFR-alpha regulate tooth formation via the cephalic neural crest: incompatibility between Hox
autocrine mechanism during mandibular morphogenesis gene expression and lower jaw development. Development
in vitro. Dev Dyn 213:500-511. 125:3445-3459.
Chai Y, Jiang X, Ito Y, Bringas P, Han J, Rowitch DH, et al. (2000). Crackower MA, Scherer SW, Rommens JM, Hui CC, Poorkaj P,
Fate of the mammalian cranial neural crest during tooth and Soder S, et al. (1996). Characterization of the split
mandibular morphogenesis. Development 127:1671-1679. hand/split foot malformation locus SHFM1 at 7q21.3-
Chapman DL, Garvey N, Hancock S, Alexiou M, Agulnik SI, q22.1 and analysis of a candidate gene for its expression
Gibson-Brown JJ, et al. (1996). Expression of the T-box family during limb development. Hum Mol Genet 5:571-579.
genes, Tbxl-Tbx5, during early mouse development. Dev Dyn Crawford MJ, Lanctot C, Tremblay JJ, Jenkins N, Gilbert D,
206:379-390. Copeland N, et al. (1997). Human and murine PTX1/Ptxl
Chen Y, Bei M, Woo I, Satokata I, Maas R (1996). Msxl controls gene maps to the region for Treacher Collins syndrome.
inductive signaling in mammalian tooth morphogenesis. Mamm Genome 8:841-845.
Development 122:3035-3044. Crossley PH, Martin GR (1995). The mouse Fgf8 gene encodes
Chen ZF, Behringer RR (1995). twist is required in head mes- a family of polypeptides and is expressed in regions that
enchyme for cranial neural tube morphogenesis. Genes direct outgrowth and patterning in the developing
Dev 9:686-699. embryo. Development 121:439-451.
Chiang C, Litingtung Y, Lee E, Young KE, Corden JL, Cserjesi P, Brown D, Lyons GE, Olson EN (1995). Expression of
Westphal H, et al. (1996). Cyclopia and defective axial the novel basic helix-loop-helix gene eHAND in neural
patterning in mice lacking Sonic hedgehog gene function. crest derivatives and extraembryonic membranes during
Nature 383:407-413. mouse development. Dev Biol 170:664-678.

292 Crit Rev


Crit Oral Biol Med
Revo Oral Biol Med
12(4)276-300 (2001)

12(4):276-300 (2001)
Dahl E, Koseki H, Balling R (1997). Pax genes and organogen- gene family in development. Mamm Genome 10:197-200.
esis. Bioessays 19:755-765. Gaunt SJ, Blum M, De Robertis EM (1993). Expression of the
Davideau JL, Demri P, Gu TT, Simmons D, Nessman C, Forest mouse goosecoid gene during mid-embryogenesis may
N, et al. (1999). Expression of DLX5 during human embry- mark mesenchymal cell lineages in the developing head,
onic craniofacial development. Mech Dev 81:183-186. limbs and body wall. Development 117:769-778.
Davidson D (1995). The function and evolution of Msx genes: Gavin BJ, McMahon JA, McMahon AP (1990). Expression of
pointers and paradoxes. Trends Genet 11:405-411. multiple novel Wnt-1/int-l-related genes during fetal and
Dealy CN, Roth A, Ferrari D, Brown AM, Kosher RA (1993). adult mouse development. Genes Dev 4:2319-2332.
Wnt-5a and Wnt-7a are expressed in the developing chick Gendron-Maguire M, Mallo M, Zhang M, Gridley T (1993).
limb bud in a manner suggesting roles in pattern forma- Hoxa-2 mutant mice exhibit homeotic transformation of
tion along the proximodistal and dorsoventral axes. Mech skeletal elements derived from cranial neural crest. Cell
Dev 43:175-186. 75:1317-1331.
Depew MJ, Liu JK, Long JE, Presley R, Meneses JJ, Pedersen Gibson-Brown JJ, Agulnik SI, Silver LM, Papaioannou VE
RA, et al. (1999). Dlx5 regulates regional development of (1998a). Expression of T-box genes Tbx2-Tbx5 during
the branchial arches and sensory capsules. Development chick organogenesis. Mech Dev 74:165-169.
126:3831-3846. Gibson-Brown JJ, Agulnik SI, Silver LM, Niswander L,
Dixon J, Hovanes K, Shiang R, Dixon MJ (1997a). Sequence Papaioannou VE (1998b). Involvement of T-box genes
analysis, identification of evolutionary conserved motifs Tbx2-Tbx5 in vertebrate limb specification and develop-
and expression analysis of murine tcofl provide further ment. Development 125:2499-2509.
evidence for a potential function for the gene and its Gitelman 1 (1997). Twist protein in mouse embryogenesis. Dev
human homologue, TCOF1. Hum Mol Genet 6:727-737. Biol 189:205-214.
Dixon J, Edwards SJ, Anderson I, Brass A, Scambler PJ, Dixon Gladwin AJ, Dixon J, Loftus SK, Edwards S, Wasmuth JJ,
MJ (1997b). Identification of the complete coding sequence Hennekam RC, et al. (1996). Treacher Collins syndrome
and genomic organization of the Treacher Collins syn- may result from insertions, deletions or splicing muta-
drome gene. Genome Res 7:223-234. tions, which introduce a termination codon into the gene.
Drouin J, Lamolet B, Lamonerie T, Lanctot C, Tremblay JJ (1998). Hum Mol Genet 5:1533-1538.
The PTX family of homeodomain transcription factors dur- Goodrich LV, Milenkovic L, Higgins KM, Scott MP (1997).
ing pituitary developments. Mol Cell Endocrinol 140:31-36. Altered neural cell fates and medulloblastoma in mouse
Dudley AT, Robertson EJ (1997). Overlapping expression patched mutants. Science 277:1109-1113.
domains of bone morphogenetic protein family members Graham A, Koentges G, Lumsden A (1996). Neural crest apop-
potentially account for limited tissue defects in BMP7 tosis and the establishment of craniofacial pattern: an hon-
deficient embryos. Dev Dyn 208:349-362. orable death. Mol Cell Neurosci 8:76-83.
Dudley AT, Lyons KM, Robertson EJ (1995). A requirement for Gray GE, Mann RS, Mitsiadis E, Henrique D, Carcangiu ML,
bone morphogenetic protein-7 during development of the Banks A, et al. (1999). Human ligands of the Notch recep-
mammalian kidney and eye. Genes Dev 9:2795-2807. tor. Am I Pathol 154:785-794.
Dunwoodie SL, Henrique D, Harrison SM, Beddington RS Greenwald I (1998). LIN-12/Notch signaling: lessons from
(1997). Mouse D113: a novel divergent delta gene which worms and flies. Genes Dev 12:1751-1762.
may complement the function of other delta homologues Gridley T (1997). Notch signaling in vertebrate development
during early pattern formation in the mouse embryo. and disease. Mol Cell Neurosci 9:103-108.
Development 124:3065-3076. Hacker A, Guthrie S (1998). A distinct developmental pro-
Ekanayake S, Hall BK (1997). The in vivo and in vitro effects of gramme for the cranial paraxial mesoderm in the chick
bone morphogenetic protein-2 on the development of the embryo. Development 125:3461-3472.
chick mandible. Int J Dev Biol 41:67-81. Hall BK (1982). Mandibular morphogenesis and craniofacial
Ferrari D, Sumoy L, Gannon J, Sun H, Brown AM, Upholt WB, malformations. J Craniofac Genet Dev Biol 2:309-322.
et al. (1995). The expression pattern of the Distal-less Hall BK, Coffin-Collins PA (1990). Reciprocal interactions
homeobox-containing gene Dlx-5 in the developing chick between epithelium, mesenchyme, and epidermal growth
limb bud suggests its involvement in apical ectodermal factor (EGF) in the regulation of mandibular mitotic activ-
ridge activity, pattern formation, and cartilage differentia- ity in the embryonic chick. J Craniofac Genet Dev Biol
tion. Mech Dev 52:257-264. 10:241-261.
Ferrari D, Lichtler AC, Pan ZZ, Dealy CN, Upholt WB, Kosher Hammerschmidt M, Brook A, McMahon AP (1997). The world
RA (1998). Ectopic expression of Msx-2 in posterior limb according to hedgehog. Trends Genet 13:14-21.
bud mesoderm impairs limb morphogenesis while induc- Hardcastle Z, Mo R, Hui CC, Sharpe PT (1998). The Shh sig-
ing BMP-4 expression, inhibiting cell proliferation, and nalling pathway in tooth development: defects in Gli2 and
promoting apoptosis. Dev Biol 197:12-24. Gli3 mutants. Development 125:2803-2811.
Francis-West PH, Tatla T, Brickell PM (1994). Expression pat- Hartung H, Feldman B, Lovec H, Coulier F, Birnbaum D,
terns of the bone morphogenetic protein genes Bmp-4 and Goldfarb M (1997). Murine FGF-12 and FGF-13: expres-
Bmp-2 in the developing chick face suggest a role in out- sion in embryonic nervous system, connective tissue and
growth of the primordia. Dev Dyn 201:168-178. heart. Mech Dev 64:31-39.
Francis-West P, Ladher R, Barlow A, Graveson A (1998). Haub 0, Goldfarb M (1991). Expression of the fibroblast
Signalling interactions during facial development. Mech growth factor-5 gene in the mouse embryo. Development
Dev 75:3-28. 112:397-406.
Gage PJ, Suh H, Camper SA (1999). The bicoid-related Pitx Hebert JM, Basilico C, Goldfarb M, Haub 0, Martin GR (1990).

12(4) 276 300 (2001)


12(4):276-300
Oral Biol
Crit Rev Oral Biol Med
Med
293
Isolation of cDNAs encoding four mouse FGF family Hunt P, Clarke JD, Buxton P, Ferretti P, Thorogood P (1998).
members and characterization of their expression patterns Stability and plasticity of neural crest patterning and
during embryogenesis. Dev Biol 138:454-463. branchial arch Hox code after extensive cephalic crest
Heikinheimo M, Lawshe A, Shackleford GM, Wilson DB, rotation. Dev Biol 198:82-104.
MacArthur CA (1994). Fgf-8 expression in the post-gastru- Ignatius J, Knuutila S, Scherer SW, Trask B, Kere J (1996). Split
lation mouse suggests roles in the development of the face, hand/split foot malformation, deafness, and mental retar-
limbs and central nervous system. Mech Dev 48:129-138. dation with a complex cytogenetic rearrangement involv-
Helms JA, Kim CH, Hu D, Minkoff R, Thaller C, Eichele G ing 7q21.3. J Med Genet 33:507-510.
(1997). Sonic hedgehog participates in craniofacial mor- Ingham PW (1995). Signalling by hedgehog family proteins in
phogenesis and is down-regulated by teratogenic doses of Drosophila and vertebrate development. Curr Opin Genet
retinoic acid. Dev Biol 187:25-35. Dev 5:492-498.
Higashijima S, Kojima T, Michiue T, Ishimaru S, Emori Y, Saigo Ingham PW (1998). Transducing Hedgehog: the story so far.
K (1992a). Dual Bar homeo box genes of Drosophila EMBO J 17:3505-3511.
required in two photoreceptor cells, Rl and R6, and pri- Isaac A, Rodriguez-Esteban C, Ryan A, Altabef M, Tsukui T,
mary pigment cells for normal eye development. Genes Patel K, et al. (1998). Tbx genes and limb identity in chick
Dev 6:50-60. embryo development. Development 125:1867-1875.
Higashijima S, Michiue T, Emori Y, Saigo K (1992b). Subtype Israel DI, Nove J, Kerns KM, Kaufman RJ, Rosen V, Cox KA, et
determination of Drosophila embryonic external sensory al. (1996). Heterodimeric bone morphogenetic proteins
organs by redundant homeo box genes BarHl and BarH2. show enhanced activity in vitro and in vivo. Growth
Genes Dev 6:1005-1018. Factors 13:291-300.
Hill RE, Jones PF, Rees AR, Sime CM, Justice MJ, Copeland NG, Jiang R, Lan Y, Chapman HD, Shawber C, Norton CR,
et al. (1989). A new family of mouse homeo box-containing Serreze DV, et al. (1998). Defects in limb, craniofacial,
genes: molecular structure, chromosomal location, and and thymic development in Jagged2 mutant mice. Genes
developmental expression of Hox-7.1. Genes Dev 3:26-37. Dev 12:1046-1057.
Hogan BL (1996). Bone morphogenetic proteins in develop- Johnson RL, Scott MP (1998). New players and puzzles in the
ment. Curr Opin Genet Dev 6:432-438. Hedgehog signaling pathway. Curr Opin Genet Dev 8:450-
Hogan BL (1999). Morphogenesis. Cell 96:225-233. 456.
Horstadius S, Sellman S (1946). Experimentelle Untersuchun- Johnson RL, Tabin CJ (1997). Molecular models for vertebrate
gen uber die Determination des knorpeligen Kopfskelettes limb development. Cell 90:979-990.
bei Urodelen. Nov Act Reg Soc Scient Ups (Ser IV) 13:1-170. Jones FS, Kioussi C, Copertino DW, Kallunki P, Holst BD,
Hosoda K, Hammer RE, Richardson JA, Baynash AG, Cheung Edelman GM (1997). Barx2, a new homeobox gene of the
JC, Giaid A, et al. (1994). Targeted and natural (piebald- Bar class, is expressed in neural and craniofacial structures
lethal) mutations of endothelin-B receptor gene produce during development. Proc Natl Acad Sci USA 94:2632-2637.
megacolon associated with spotted coat color in mice. Cell Joutel A, Tournier-Lasserve E (1998). Notch signalling path-
79:1267-1276. way and human diseases. Semin Cell Dev Biol 9:619-625.
Hu D, Helms JA (1999). The role of sonic hedgehog in normal Jowett AK, Vainio S, Ferguson MW, Sharpe PT, Thesleff I
and abnormal craniofacial morphogenesis. Development (1993). Epithelial-mesenchymal interactions are required
126:4873-4884. for msx 1 and msx 2 gene expression in the developing
Hui CC, Joyner AL (1993). A mouse model of greig murine molar tooth. Development 117:461-470.
cephalopolysyndactyly syndrome: the extra-toesJ muta- Kawabata M, Imamura T, Miyazono K (1998). Signal trans-
tion contains an intragenic deletion of the Gli3 gene. Nat duction by bone morphogenetic proteins. Cytokine Growth
Genet 3:241-246. Factor Rev 9:49-61.
Hui CC, Slusarski D, Platt KA, Holmgren R, Joyner AL (1994). Kawakami Y, Wada N, Nishimatsu SI, Ishikawa T, Noji S, Nohno
Expression of three mouse homologs of the Drosophila T (1999). Involvement of Wnt-5a in chondrogenic pattern
segment polarity gene cubitus interruptus, Gli, Gli-2, and formation in the chick limb bud. Dev Growth Differ 41:29-40.
Gli-3, in ectoderm- and mesoderm-derived tissues sug- Kettunen P, Thesleff 1 (1998). Expression and function of FGFs-
gests multiple roles during postimplantation develop- 4, -8, and -9 suggest functional redundancy and repetitive
ment. Dev Biol 162:402-413. use as epithelial signals during tooth morphogenesis. Dev
Hukriede NA, Gu Y, Fleming RJ (1997). A dominant-negative Dyn 211:256-268.
form of Serrate acts as a general antagonist of Notch acti- Kettunen P, Karavanova I, Thesleff I (1998). Responsiveness of
vation. Development 124:3427-3437. developing dental tissues to fibroblast growth factors:
Hunt P, Gulisano M, Cook M, Sham MH, Faiella A, Wilkinson expression of splicing alternatives of FGFR1, -2, -3, and of
D, et al. (1991a). A distinct Hox code for the branchial FGFR4; and stimulation of cell proliferation by FGF-2, -4,
region of the vertebrate head. Nature 353:861-864. -8, and -9. Dev Genet 22:374-385.
Hunt P, Wilkinson D, Krumlauf R (1991b). Patterning the ver- King JA, Marker PC, Seung KJ, Kingsley DM (1994). BMP5 and
tebrate head: murine Hox 2 genes mark distinct subpopu- the molecular, skeletal, and soft-tissue alterations in short
lations of premigratory and migrating cranial neural crest. ear mice. Dev Biol 166:112-122.
Development 112:43-50. Kingsley DM, Bland AE, Grubber JM, Marker PC, Russell LB,
Hunt P, Ferretti P, Krumlauf R, Thorogood P (1995). Copeland NG, et al. (1992). The mouse short ear skeletal
Restoration of normal Hox code and branchial arch mor- morphogenesis locus is associated with defects in a bone
phogenesis after extensive deletion of hindbrain neural morphogenetic member of the TGF beta superfamily. Cell
crest. Dev Biol 168:584-597. 71:399-410.
294
1241:26-30 (001
294 Crit Rev Oral Biol Med 12(4):276-300 (2001)
Klint P, Claesson-Welsh L (1999). Signal transduction by fibro- et al. (1999a). prx-I functions cooperatively with another
blast growth factor receptors. Front Biosci 4:D165-177. paired-related homeobox gene, prx-2, to maintain cell
Kojima T, Ishimaru S, Higashijima S, Takayama E, Akimaru H, fates within the craniofacial mesenchyme. Development
Sone M, et al. (1991). Identification of a different-type 126:495-504.
homeobox gene, BarHl, possibly causing Bar (B) and Lu MF, Pressman C, Dyer R, Johnson RL, Martin JF (1999b).
Om(1D) mutations in Drosophila. Proc Natl Acad Sci USA Function of Rieger syndrome gene in left-right asymmetry
88:4343-4347. and craniofacial development. Nature 401:276-278.
Kontges G, Lumsden A (1996). Rhombencephalic neural crest Lumsden A, Sprawson N, Graham A (1991). Segmental origin
segmentation is preserved throughout craniofacial and migration of neural crest cells in the hindbrain region
ontogeny. Development 122:3229-3242. of the chick embryo. Development 113:1281-1291.
Krantz ID, Piccoli DA, Spinner NB (1997). Alagille syndrome. Luo G, Hofmann C, Bronckers AL, Sohocki M, Bradley A,
J Med Genet 34:152-157. Karsenty G (1995). BMP-7 is an inducer of nephrogenesis,
Krantz ID, Colliton RP, Genin A, Rand EB, Li L, Piccoli DA, et and is also required for eye development and skeletal pat-
al. (1998). Spectrum and frequency of jaggedl (JAG1) terning. Genes Dev 9:2808-2820.
mutations in Alagille syndrome patients and their fami- Maas R, Bei M (1997). The genetic control of early tooth devel-
lies. Am J Hum Genet 62:1361-1369. opment. Crit Rev Oral Biol Med 8:4-39.
Kraus P, Lufkin T (1999). Mammalian Dlx homeobox gene con- Macias D, Ganan Y, Ros MA, Hurle JM (1996). In vivo inhibi-
trol of craniofacial and inner ear morphogenesis. J Cell tion of programmed cell death by local administration of
Biochem Suppl 32/33:133-140. FGF-2 and FGF-4 in the interdigital areas of the embryon-
Kretzschmar M, Massague J (1998). SMADs: mediators and ic chick leg bud. Anat Embryol 193:533-541.
regulators of TGF-beta signaling. Curr Opin Genet Dev MacKenzie A, Leeming GL, Jowett AK, Ferguson MW, Sharpe
8:103-111. PT (1991). The homeobox gene Hox 7.1 has specific region-
Kretzschmar M, Doody J, Massague J (1997). Opposing BMP al and temporal expression patterns during early murine
and EGF signalling pathways converge on the TGF-beta craniofacial embryogenesis, especially tooth development
family mediator Smadl. Nature 389:618-622. in vivo and in vitro. Development 111:269-285.
Kronmiller JE, Upholt WB, Kollar EJ (1991). EGF antisense MacKenzie A, Ferguson MW, Sharpe PT (1992). Expression
oligodeoxynucleotides block murine odontogenesis in patterns of the homeobox gene, Hox-8, in the mouse
vitro. Dev Biol 147:485-488. embryo suggest a role in specifying tooth initiation and
Krumlauf R (1994). Hox genes in vertebrate development. Cell shape. Development 115:403-420.
78:191-201. Maden M, Graham A, Gale E, Rollinson C, Zile M (1997).
Kurihara Y, Kurihara H, Suzuki H, Kodama T, Maemura K, Positional apoptosis during vertebrate CNS development
Nagai R, et al. (1994). Elevated blood pressure and cranio- in the absence of endogenous retinoids. Development
facial abnormalities in mice deficient in endothelin-1. 124:2799-2805.
Nature 368:703-710. Mahmood R, Bresnick J, Hombruch A, Mahony C, Morton N,
Kurihara H, Kurihara Y, Nagai R, Yazaki Y (1999). Endothelin Colquhoun K, et al. (1995). A role for FGF-8 in the initia-
and neural crest development. Cell Mol Biol 45:639-651. tion and maintenance of vertebrate limb bud outgrowth.
Lanctot C, Lamolet B, Drouin J (1997). The bicoid-related Curr Biol 5:797-806.
homeoprotein Ptxl defines the most anterior domain of Mallo M (1998). Embryological and genetic aspects of middle
the embryo and differentiates posterior from anterior lat- ear development. Int J Dev Biol 42:11-22.
eral mesoderm. Development 124:2807-2817. Manley NR, Capecchi MR (1995). The role of Hoxa-3 in
Lanctot C, Moreau A, Chamberland M, Tremblay ML, Drouin mouse thymus and thyroid development. Development
J (1999). Hindlimb patterning and mandible development 121:1989-2003.
require the Ptxl gene. Development 126:1805-1810. Manley NR, Capecchi MR (1998). Hox group 3 paralogs regu-
Le Douarin NM, Dupin E, Ziller C (1994). Genetic and epige- late the development and migration of the thymus, thy-
netic control in neural crest development. Curr Opin Genet roid, and parathyroid glands. Dev Biol 195:1-15.
Dev 4:685-695. Mansouri A, Goudreau G, Gruss P (1999). Pax genes and their
Le Douarin NM, Catala M, Batini C (1997). Embryonic neural role in organogenesis. Cancer Res 59:1707s-1709s; discus-
chimeras in the study of vertebrate brain and head devel- sion 1709s-1710s.
opment. Int Rev Cytol 175:241-309. Marazzi G, Wang Y, Sassoon D (1997). Msx2 is a transcription-
Li L, Krantz ID, Deng Y, Genin A, Banta AB, Collins CC, et al. al regulator in the BMP4-mediated programmed cell
(1997). Alagille syndrome is caused by mutations in death pathway. Dev Biol 186:127-138.
human Jaggedl, which encodes a ligand for Notchl. Nat Martin GR (1998). The roles of FGFs in the early development
Genet 16:243-251. of vertebrate limbs. Genes Dev 12:1571-1586.
Lin CR, Kioussi C, O'Connell S, Briata P, Szeto D, Liu F, et al. Martin JF, Bradley A, Olson EN (1995). The paired-like homeo
(1999). Pitx2 regulates lung asymmetry, cardiac positioning box gene MHox is required for early events of skeletogen-
and pituitary and tooth morphogenesis. Nature 401:279-282. esis in multiple lineages. Genes Dev 9:1237-1249.
Lindsell CE, Shawber CJ, Boulter J, Weinmaster G (1995). Maruoka Y, Ohbayashi N, Hoshikawa M, Itoh N, Hogan BLM,
Jagged: a mammalian ligand that activates Notchl. Cell Furuta Y (1998). Comparison of the expression of three
80:909-917. highly related genes, Fgf8, Fgfl7 and Fgfl8, in the mouse
Logan M, Tabin CJ (1999). Role of Pitxl upstream of Thx4 in embryo. Mech Dev 74:175-177.
specification of hindlimb identity. Science 283:1736-1739. Massague J (1998). TGF-beta signal transduction. Annu Rev
Lu MF, Cheng HT, Kern MJ, Potter SS, Tran B, Diekwisch TG, Biochem 67:753-791.

(2001)
12(4) 276-300 (2001)
12(4),.276-300
Oral Biol
Crit Rev Oral Biol Med
Med
295
Matsuo I, Kuratani S, Kimura C, Takeda N, Aizawa S (1995). Morrison AD (1998). 1 + 1 = r4 and much much more. Bioessays
Mouse Otx2 functions in the formation and patterning of 20:794-797.
rostral head. Genes Dev 9:2646-2658. Morriss-Kay GM (1996). Craniofacial defects in AP-2 null
Matzuk MM, Kumar TR, Vassalli A, Bickenbach JR, Roop DR, mutant mice. Bioessays 18:785-788.
Jaenisch R, et al. (1995a). Functional analysis of activins Muenke M, Schell U (1995). Fibroblast-growth-factor recep-
during mammalian development. Nature 374:354-356. tor mutations in human skeletal disorders. Trends Genet
Matzuk MM, Kumar TR, Bradley A (1995b). Different pheno- 11:308-313.
types for mice deficient in either activins or activin recep- Murone M, Rosenthal A, de Sauvage FJ (1999). Sonic hedgehog
tor type II. Nature 374:356-360. signaling by the patched-smoothened receptor complex.
Matzuk MM, Lu N, Vogel H, Sellheyer K, Roop DR, Bradley A Curr Biol 9:76-84.
(1995c). Multiple defects and perinatal death in mice defi- Naski MC, Ornitz DM (1998). FGF signaling in skeletal devel-
cient in follistatin. Nature 374:360-363. opment. Front Biosci 3:D781-794.
McGonnell IM, Clarke JD, Tickle C (1998). Fate map of the Neubuser A, Peters H, Balling R, Martin GR (1997).
developing chick face: analysis of expansion of facial pri- Antagonistic interactions between FGF and BMP signal-
mordia and establishment of the primary palate. Dev Dyn ing pathways: a mechanism for positioning the sites of
212:102-118. tooth formation. Cell 90:247-255.
Meijlink F, Beverdam A, Brouwer A, Oosterveen TC, Berge DT Newberry EP, Latifi T, Towler DA (1998). Reciprocal regulation
(1999). Vertebrate aristaless-related genes. Int I Dev Biol of osteocalcin transcription by the homeodomain proteins
43:651-663. Msx2 and Dlx5. Biochemistry 37:16360-16368.
Miettinen PJ, Chin JR, Shum L, Slavkin HC, Shuler CF, Nishikawa K, Nakanishi T, Aoki C, Hattori T, Takahashi K,
Derynck R, et al. (1999). Epidermal growth factor receptor Taniguchi S (1994). Differential expression of homeobox-
function is necessary for normal craniofacial development containing genes Msx-1 and Msx-2 and homeoprotein
and palate closure. Nat Genet 22:69-73. Msx-2 expression during chick craniofacial development.
Mina M, Upholt WB, Kollar EJ (1994). Enhancement of avian Biochem Mol Biol Int 32:763-771.
mandibular chondrogenesis in vitro in the absence of Niswander L, Martin GR (1992). Fgf-4 expression during gas-
epithelium. Arch Oral Biol 39:551-562. trulation, myogenesis, limb and tooth development in the
Mina M, Gluhak J, Upholt WB, Kollar EJ, Rogers B (1995). mouse. Development 114:755-768.
Experimental analysis of Msx-I and Msx-2 gene expression Noden DM (1978). The control of avian cephalic neural crest
during chick mandibular morphogenesis. Dev Dyn cytodifferentiation. I. Skeletal and connective tissues. Dev
202:195-214. Biol 67:296-312.
Mina M, Gluhak J, Rodgers B (1996). Downregulation of Msx- Noden DM (1983a). The role of the neural crest in patterning
2 expression results in chondrogenesis in the medial of avian cranial skeletal, connective, and muscle tissues.
region of the avian mandible. Connect Tissue Res 35:79-84. Dev Biol 96:144-165.
Ming JE, Muenke M (1998). Holoprosencephaly: from Homer Noden DM (1983b). The embryonic origins of avian cephalic
to Hedgehog. Clin Genet 53:155-163. and cervical muscles and associated connective tissues.
Ming JE, Roessler E, Muenke M (1998). Human developmen- Am J Anat 168:257-276.
tal disorders and the Sonic hedgehog pathway. Mol Med Noden DM (1986). Patterning of avian craniofacial muscles.
Today 4:343-349. Dev Biol 116:347-356.
Mitsiadis TA, Lardelli M, Lendahl U, Thesleff I (1995). Noden DM (1988). Interactions and fates of avian craniofacial
Expression of Notch 1, 2 and 3 is regulated by epithelial- mesenchyme. Development 103(Suppl):121-140.
mesenchymal interactions and retinoic acid in the devel- Nonaka K, Shum L, Takahashi I, Takahashi K, Ikura T, Dashner
oping mouse tooth and associated with determination of R, et al. (1999). Convergence of the BMP and EGF signal-
ameloblast cell fate. J Cell Biol 130:407-418. ing pathways on Smadl in the regulation of chondrogen-
Mitsiadis TA, Henrique D, Thesleff I, Lendahl U (1997). Mouse esis. Int J Dev Biol 43:795-807.
Serrate-i (Jagged-i): expression in the developing tooth is Oda T, Elkahloun AG, Pike BL, Okajima K, Krantz ID, Genin A,
regulated by epithelial-mesenchymal interactions and et al. (1997). Mutations in the human Jaggedl gene are
fibroblast growth factor-4. Development 124:1473-1483. responsible for Alagille syndrome. Nat Genet 16:235-242.
Mitsiadis TA, Hirsinger E, Lendahl U, Goridis C (1998a). Orr-Urtreger A, Lonai P (1992). Platelet-derived growth factor-
Delta-notch signaling in odontogenesis: correlation with A and its receptor are expressed in separate, but adjacent
cytodifferentiation and evidence for feedback regulation. cell layers of the mouse embryo. Development 115:1045-1058.
Dev Biol 204:420-431. Orr-Urtreger A, Givol D, Yayon A, Yarden Y, Lonai P (1991).
Mitsiadis TA, Mucchielli ML, Raffo S, Proust JP, Koopman P, Developmental expression of two murine fibroblast
Goridis C (1998b). Expression of the transcription factors growth factor receptors, flg and bek. Development
Otlx2, Barxl and Sox9 during mouse odontogenesis. Eur i 113:1419-1434.
Oral Sci 106(Suppl 1):112-116. Orr-Urtreger A, Bedford MT, Burakova T, Arman E, Zimmer Y,
Miyazono K (1999). Signal transduction by bone morphogenetic Yayon A, et al. (1993). Developmental localization of the
protein receptors: functional roles of Smad proteins. Bone splicing alternatives of fibroblast growth factor receptor-2
25:91-93. (FGFR2). Dev Biol 158:475-486.
Mo R, Freer AM, Zinyk DL, Crackower MA, Michaud J, Heng Osumi-Yamashita N, Ninomiya Y, Eto K (1997). Mammalian
HH, et al. (1997). Specific and redundant functions of Gli2 craniofacial embryology in vitro. Int J Dev Biol 41:187-194.
and Gli3 zinc finger genes in skeletal patterning and devel- Parr BA, McMahon AP (1995). Dorsalizing signal Wnt-7a
opment. Development 124:113-123. required for normal polarity of D-V and A-P axes of
296 Crit Rev Oral Biol
Crit Rev Oral Biol Med
Med

12(4):276-300 (2001)
mouse limb. Nature 374:350-353. makes headway. Curr Biol 5:345-348.
Partanen AM, Thesleff I (1987). Localization and quantitation Richman JM, Tickle C (1989). Epithelia are interchangeable
of 1251-epidermal growth factor binding in mouse embry- between facial primordia of chick embryos and morpho-
onic tooth and other embryonic tissues at different devel- genesis is controlled by the mesenchyme. Dev Biol
opmental stages. Dev Biol 120:186-197. 136:201-210.
Partanen AM, Ekblom P, Thesleff I (1985). Epidermal growth Richman JM, Rowe A, Brickell PM (1991). Signals involved in
factor inhibits morphogenesis and cell differentiation in patterning and morphogenesis of the embryonic face. Prog
cultured mouse embryonic teeth. Dev Biol 111:84-94. Clin Biol Res 373:117-131.
Paznekas WA, Zhang N, Gridley T, Jabs EW (1997). Mouse Richman JM, Herbert M, Matovinovic E, Walin J (1997). Effect
TCOF1 is expressed widely, has motifs conserved in of fibroblast growth factors on outgrowth of facial mes-
nucleolar phosphoproteins, and maps to chromosome 18. enchyme. Dev Biol 189:135-147.
Biochem Biophys Res Commun 238:1-6. Rijli FM, Mark M, Lakkaraju S, Dierich A, Dolle P, Chambon P
Peters KG, Werner S, Chen G, Williams LT (1992). Two FGF (1993). A homeotic transformation is generated in the ros-
receptor genes are differentially expressed in epithelial tral branchial region of the head by disruption of Hoxa-2,
and mesenchymal tissues during limb formation and which acts as a selector gene. Cell 75:1333-1349.
organogenesis in the mouse. Development 114:233-243. Rijli FM, Gavalas A, Chambon P (1998). Segmentation and
Peters K, Ornitz D, Werner S, Williams L (1993). Unique specification in the branchial region of the head: the role of
expression pattern of the FGF receptor 3 gene during the Hox selector genes. Int J Dev Biol 42:393-401.
mouse organogenesis. Dev Biol 155:423-430. Rivera-Perez JA, Mallo M, Gendron-Maguire M, Gridley T,
Peters H, Neubuser A, Balling R (1998a). Pax genes and Behringer RR (1995). Goosecoid is not an essential compo-
organogenesis: Pax9 meets tooth development. Eur J Oral nent of the mouse gastrula organizer but is required for cra-
Sci 106(Suppl 1):38-43. niofacial and rib development. Development 121:3005-3012.
Peters H, Neubuser A, Kratochwil K, Balling R (1998b). Pax9- Rivera-Perez JA, Wakamiya M, Behringer RR (1999). Goosecoid
deficient mice lack pharyngeal pouch derivatives and acts cell autonomously in mesenchyme-derived tissues dur-
teeth and exhibit craniofacial and limb abnormalities. ing craniofacial development. Development 126:3811-3821.
Genes Dev 12:2735-2747. Robert B, Sassoon D, Jacq B, Gehring W, Buckingham M (1989).
Pizette S, Niswander L (1999). BMPs negatively regulate struc- Hox-7, a mouse homeobox gene with a novel pattem of
ture and function of the limb apical ectodermal ridge. expression during embryogenesis. EMBO J 8:91-100.
Development 126:883-894. Robinson GW, Mahon KA (1994). Differential and overlapping
Platt KA, Michaud J, Joyner AL (1997). Expression of the expression domains of Dlx-2 and Dlx-3 suggest distinct
mouse Gli and Ptc genes is adjacent to embryonic sources roles for Distal-less homeobox genes in craniofacial devel-
of hedgehog signals suggesting a conservation of path- opment. Mech Dev 48:199-215.
ways between flies and mice. Mech Dev 62:121-135. Roessler E, Belloni E, Gaudenz K, Jay P, Berta P, Scherer SW, et
Price JA, Wright JT, Kula K, Bowden DW, Hart TC (1998). A al. (1996). Mutations in the human Sonic Hedgehog gene
common DLX3 gene mutation is responsible for tricho- cause holoprosencephaly. Nat Genet 14:357-360.
dento-osseous syndrome in Virginia and North Carolina Rudnicki JA, Brown AM (1997). Inhibition of chondrogenesis
families. J Med Genet 35:825-828. by Wnt gene expression in vivo and in vitro. Dev Biol
Prince V, Lumsden A (1994). Hoxa-2 expression in normal and 185:104-118.
transposed rhombomeres: independent regulation in the Ryoo HM, Hoffmann HM, Beumer T, Frenkel B, Towler DA,
neural tube and neural crest. Development 120:911-923. Stein GS, et al. (1997). Stage-specific expression of Dlx-5
Qiu M, Bulfone A, Martinez S, Meneses JJ, Shimamura K, during osteoblast differentiation: involvement in regula-
Pedersen RA, et al. (1995). Null mutation of Dlx-2 results tion of osteocalcin gene expression. Mol Endocrinol
in abnormal morphogenesis of proximal first and second 11:1681-1694.
branchial arch derivatives and abnormal differentiation in Saito T, Sawamoto K, Okano H, Anderson DJ, Mikoshiba K
the forebrain. Genes Dev 9:2523-2538. (1998). Mammalian BarH homologue is a potential regula-
Qiu M, Bulfone A, Ghattas I, Meneses JJ, Christensen L, Sharpe tor of neural bHLH genes. Dev Biol 199:216-225.
PT, et al. (1997). Role of the Dlx homeobox genes in proxi- Saldivar JR, Krull CE, Krumlauf R, Ariza-McNaughton L,
modistal patterning of the branchial arches: mutations of Bronner-Fraser M (1996). Rhombomere of origin deter-
Dlx-1, Dlx-2, and Dlx-1 and -2 alter morphogenesis of mines autonomous versus environmentally regulated
proximal skeletal and soft tissue structures derived from expression of Hoxa-3 in the avian embryo. Development
the first and second arches. Dev Biol 185:165-184. 122:895-904.
Qu S, Li L, Wisdom R (1997a). Alx-4: cDNA cloning and char- Saldivar JR, Sechrist JW, Krull CE, Ruffins S, Bronner-Fraser M
acterization of a novel paired-type homeodomain protein. (1997). Dorsal hindbrain ablation results in rerouting of
Gene 203:217-223. neural crest migration and changes in gene expression, but
Qu S, Niswender KD, Ji Q, van der Meer R, Keeney D, normal hyoid development. Development 124:2729-2739.
Magnuson MA, et al. (199Tb). Polydactyly and ectopic Satokata I, Maas R (1994). Msxl deficient mice exhibit cleft
ZPA formation in Alx-4 mutant mice. Development palate and abnormalities of craniofacial and tooth devel-
124:3999-4008. opment. Nat Genet 6:348-356.
Raftery LA, Sutherland DJ (1999). TGF-beta family signal Satokata I, Ma L, Ohshima H, Bei M, Woo I, Nishizawa K, et al.
transduction in Drosophila development: from Mad to (2000). Msx2 deficiency in mice causes pleiotropic defects
Smads. Dev Biol 210:251-268. in bone growth and ectodermal organ formation. Nat
Richman JM (1995). Head development. Craniofacial genetics Genet 24:391-395.

297
I2(4.276-3OO
1 2(4):276-300 l(200L
2001l) Crit Rev Oral Biol Med
Crit Rev Oral Biol Med
297
Schatteman GC, Morrison-Graham K, van Koppen A, Weston Sokol SY (1999). Wnt signaling and dorso-ventral axis specifi-
JA, Bowen-Pope DF (1992). Regulation and role of PDGF cation in vertebrates. Curr Opin Genet Dev 9:405-410.
receptor alpha-subunit expression during embryogenesis. Solloway MJ, Robertson EJ (1999). Early embryonic lethality in
Development 115:123-131. Bmp5;Bmp7 double-mutant mice suggests functional redun-
Scherer SW, Poorkaj P, Massa H, Soder S, Allen T, Nunes M, et dancy within the 60A subgroup. Development 126:1753-1768.
al. (1994). Physical mapping of the split hand/split foot Soriano P (1997). The PDGF alpha receptor is required for
locus on chromosome 7 and implication in syndromic neural crest cell development and for normal patterning
ectrodactyly. Hum Mol Genet 3:1345-1354. of the somites. Development 124:2691-2700.
Schilling TF (1997). Genetic analysis of craniofacial develop- Srivastava D, Thomas T, Lin Q, Kirby ML, Brown D, Olson EN
ment in the vertebrate embryo. Bioessays 19:459-468. (1997). Regulation of cardiac mesodermal and neural crest
Schneider RA, Hu D, Helms JA (1999). From head to toe: con- development by the bHLH transcription factor, dHAND.
servation of molecular signals regulating limb and cranio- Nat Genet 16:154-160.
facial morphogenesis. Cell Tissue Res 296:103-109. Suda Y, Matsuo I, Kuratani S, Aizawa S (1996). Otxl function
Sechrist J, Serbedzija GN, Scherson T, Fraser SE, Bronner-Fraser overlaps with Otx2 in development of mouse forebrain
M (1993). Segmental migration of the hindbrain neural and midbrain. Genes to Cells 1:1031-1044.
crest does not arise from its segmental generation. Suda Y, Nakabayashi J, Matsuo I, Aizawa S (1999). Functional
Development 118:691-703. equivalency between Otx2 and Otxl in development of
Semba I, Nonaka K, Takahashi I, Takahashi K, Dashner R, Shum the rostral head. Development 126:743-757.
L, et al. (2000). Positionally-dependent chondrogenesis Sun X, Artavanis-Tsakonas S (1997). Secreted forms of DELTA
induced by BMP4 is co-regulated by sox9 and msx2. Dev Dyn and SERRATE define antagonists of Notch signaling in
217:401-414. Drosophila. Development 124:3439-3448.
Semina EV, Reiter R, Leysens NJ, Alward WL, Small KW, Suzuki A, Kaneko E, Maeda J, Ueno N (1997). Mesoderm
Datson NA, et al. (1996). Cloning and characterization of a induction by BMP-4 and -7 heterodimers. Biochem Biophys
novel bicoid-related homeobox transcription factor gene, Res Commun 232:153-156.
RIEG, involved in Rieger syndrome. Nat Genet 14:392-399. Szebenyi G, Fallon JF (1999). Fibroblast growth factors as mul-
Serbedzija GN, Bronner-Fraser M, Fraser SE (1992). Vital dye tifunctional signaling factors. Int Rev Cytol 185:45-106.
analysis of cranial neural crest cell migration in the mouse Szeto DP, Ryan AK, O'Connell SM, Rosenfeld MG (1996). P-
embryo. Development 116:297-307. OTX: a PIT-i-interacting homeodomain factor expressed
Shawber C, Boulter J, Lindsell CE, Weinmaster G (1996). during anterior pituitary gland development. Proc Natl
Jagged2: a serrate-like gene expressed during rat embryo- Acad Sci USA 93:7706-7710.
genesis. Dev Biol 180:370-376. Szeto DP, Rodriguez-Esteban C, Ryan AK, O'Connell SM, Liu
Shen H, Wilke T, Ashique AM, Narvey M, Zerucha T, Savino F, Kioussi C, et al. (1999). Role of the Bicoid-related homeo-
E, et al. (1997). Chicken transcription factor AP-2: cloning, domain factor Pitxl in specifying hindlimb morphogene-
expression and its role in outgrowth of facial prominences sis and pituitary development. Genes Dev 13:484-494.
and limb buds. Dev Biol 188:248-266. Takada S, Stark KL, Shea MJ, Vassileva G, McMahon JA,
Shuler CF (1995). Programmed cell death and cell transforma- McMahon AP (1994). Wnt-3a regulates somite and tailbud
tion in craniofacial development. Crit Rev Oral Biol Med formation in the mouse embryo. Genes Dev 8:174-189.
6:202-217. Takahashi K, Nuckolls GH, Tanaka 0, Semba I, Takahashi I,
Shuler CF, Halpem DE, Guo Y, Sank AC (1992). Medial edge Dashner R, et al. (1998). Adenovirus-mediated ectopic
epithelium fate traced by cell lineage analysis during expression of Msx2 in even-numbered rhombomeres
epithelial-mesenchymal transformation in vivo. Dev Biol induces apoptotic elimination of cranial neural crest cells
154:318-330. in ovo. Development 125:1627-1635.
Shum L, Sakakura Y, Bringas P Jr, Luo W, Snead ML, Mayo M, Takahashi Y, Bontoux M, Le Douarin NM (1991). Epithelio-
et al. (1993). EGF abrogation-induced fusilli-form dys- mesenchymal interactions are critical for Quox 7 expres-
morphogenesis of Meckel's cartilage during embryonic sion and membrane bone differentiation in the neural crest
mouse mandibular morphogenesis in vitro. Development derived mandibular mesenchyme. EMBO 1 10:2387-2393.
118:903-917. Tanda N, Ohuchi H, Yoshioka H, Noji S, Nohno T (1995). A
Sidow A, Bulotsky MS, Kerrebrock AW, Bronson RT, Daly MJ, chicken Wnt gene, Wnt-11, is involved in dermal develop-
Reeve MP, et al. (1997). Serrate2 is disrupted in the mouse ment. Biochem Biophys Res Commun 211:123-129.
limb-development mutant syndactylism. Nature 389:722-725. Teillet MA, Ziller C, Le Douarin NM (1999). Quail-chick
Simeone A (1998). Otxl and Otx2 in the development and evo- chimeras. Meth Mol Biol 97:305-318.
lution of the mammalian brain. EMBO J 17:6790-6798. ten Berge D, Brouwer A, Korving J, Martin JF, Meijlink F
Smith DM, Tabin CJ (1999). Chick Barx2b, a marker for myo- (1998). Prxl and Prx2 in skeletogenesis: roles in the cra-
genic cells also expressed in branchial arches and neural niofacial region, inner ear and limbs. Development
structures. Mech Dev 80:203-206. 125:3831-3842.
Snead ML, Luo W, Oliver P, Nakamura M, Don-Wheeler G, Ten Cate AR (1998). Oral histology, development, structure,
Bessem C, et al. (1989). Localization of epidermal growth and function. St. Louis, MO: C.V. Mosby, pp. 24-49.
factor precursor in tooth and lung during embryonic Thomas BL, Tucker AS, Qiu M, Ferguson CA, Hardcastle Z,
mouse development. Dev Biol 134:420-429. Rubenstein JL, et al. (1997). Role of Dlx-I and Dlx-2 genes
Sohal GS, Ali MM, Ali AA, Dai D (1999). Ventrally emigrating in patterning of the murine dentition. Development
neural tube cells contribute to the formation of Meckel's 124:4811-4818.
and quadrate cartilage. Dev Dyn 216:37-44. Thomas T, Kurihara H, Yamagishi H, Kurihara Y, Yazaki Y,

298 Grit
Crit Rev
Rev Oral
Oral Biol Med
Biol Med
12(4):276-300

12(4):276-300 (2001)
Olson EN, et al. (1998). A signaling cascade involving of embryos and in adults. Oncogene 13:1537-1544.
endothelin-1, dHAND and msxl regulates development of Wang YH, Kollar EJ, Upholt WB, Mina M (1998a). EGF does
neural-crest-derived branchial arch mesenchyme. not induce Msx-1 and Msx-2 in dental mesenchyme. Eur J
Development 125:3005-3014. Oral Sci 106(Suppl 1):100-103.
Tissier-Seta JP, Mucchielli ML, Mark M, Mattei MG, Goridis C, Wang YH, Upholt WB, Sharpe PT, Kollar EJ, Mina M (1998b).
Brunet JF (1995). Barxl, a new mouse homeodomain tran- Odontogenic epithelium induces similar molecular responses
scription factor expressed in cranio-facial ectomes- in chick and mouse mandibular mesenchyme. Dev Dyn
enchyme and the stomach. Mech Dev 51:3-15. 213:386-397.
Trainor PA, Tam PP (1995). Cranial paraxial mesoderm and Wang YH, Rutherford B, Upholt WB, Mina M (1999). Effects of
neural crest cells of the mouse embryo: co-distribution in BMP-7 on mouse tooth mesenchyme and chick mandibu-
the craniofacial mesenchyme but distinct segregation in lar mesenchyme. Dev Dyn 216:320-335.
branchial arches. Development 121:2569-2582. Webster MK, Donoghue DJ (1997). FGFR activation in skeletal
Trainor PA, Tan SS, Tam PP (1994). Cranial paraxial mesoderm: disorders: too much of a good thing. Trends Genet 13:178-182.
regionalisation of cell fate and impact on craniofacial devel- Wedden SE (1987). Epithelial-mesenchymal interactions in the
opment in mouse embryos. Development 120:2397-2408. development of chick facial primordia and the target of
Trumpp A, Depew MJ, Rubenstein JL, Bishop JM, Martin GR retinoid action. Development 99:341-351.
(1999). Cre-mediated gene inactivation demonstrates that Wedden SE (1991). Effects of retinoids on chick face develop-
FGF8 is required for cell survival and patterning of the ment. J Craniofac Genet Dev Biol 11:326-337.
first branchial arch. Genes Dev 13:3136-3148. Weed M, Mundlos S, Olsen BR (1997). The role of sonic hedge-
Tsuji K, Ito Y, Noda M (1998). Expression of the hog in vertebrate development. Matrix Biol 16:53-58.
PEBP2alphaA/AML3/CBFAl gene is regulated by BMP4/7 Weinmaster G (1998). Notch signaling: direct or what? Curr
heterodimer and its overexpression suppresses type I colla- Opin Genet Dev 8:436-442.
gen and osteocalcin gene expression in osteoblastic and Westermark B, Claesson-Welsh L, Heldin CH (1989). Structural
nonosteoblastic mesenchymal cells. Bone 22:87-92. and functional aspects of the receptors for platelet-derived
Tucker AS, Al Khamis A, Sharpe PT (1998a). Interactions growth factor. Prog Growth Factor Res 1:253-266.
between Bmp-4 and Msx-1 act to restrict gene expression Wilke TA, Gubbels S, Schwartz J, Richman JM (1997).
to odontogenic mesenchyme. Dev Dyn 212:533-539. Expression of fibroblast growth factor receptors (FGFR1,
Tucker AS, Matthews KL, Sharpe PT (1998b). Transformation FGFR2, FGFR3) in the developing head and face. Dev Dyn
of tooth type induced by inhibition of BMP signaling. 210:41-52.
Science 282:1136-1138. Wilkie AO (1997). Craniosynostosis: genes and mechanisms.
Tucker AS, Yamada G, Grigoriou M, Pachnis V, Sharpe PT Hum Mol Genet 6:1647-1656.
(1999). Fgf-8 determines rostral-caudal polarity in the first Wilkinson DG (1995). Genetic control of segmentation in the
branchial arch. Development 126:51-61. vertebrate hindbrain. Perspect Dev Neurobiol 3:29-38.
Vaahtokari A, Aberg T, Thesleff I (1996). Apoptosis in the Wilkinson DG, Bhatt S, Cook M, Boncinelli E, Krumlauf R (1989).
developing tooth: association with an embryonic signal- Segmental expression of Hox-2 homeobox- containing genes
ing center and suppression by EGF and FGF-4. in the developing mouse hindbrain. Nature 341:405-409.
Development 122:121-129. Wilson DI, Burn J, Scambler P, Goodship J (1993). DiGeorge
Valsecchi C, Ghezzi C, Ballabio A, Rugarli El (1997). syndrome: part of CATCH 22. J Med Genet 30:852-856.
JAGGED2: a putative Notch ligand expressed in the apical Winokur ST, Shiang R (1998). The Treacher Collins syndrome
ectodermal ridge and in sites of epithelial-mesenchymal (TCOF1) gene product, treacle, is targeted to the nucleolus
interactions. Mech Dev 69:203-207. by signals in its C-terminus. Hum Mol Genet 7:1947-1952.
Vortkamp A, Gessler M, Grzeschik KH (1991). GLI3 zinc-finger Wise CA, Chiang LC, Paznekas WA, Sharma M, Musy MM,
gene interrupted by translocations in Greig syndrome Ashley JA, et al. (1997). TCOF1 gene encodes a putative
families. Nature 352:539-540. nucleolar phosphoprotein that exhibits mutations in
Vortkamp A, Franz T, Gessler M, Grzeschik KH (1992). Treacher Collins Syndrome throughout its coding region.
Deletion of GLI3 supports the homology of the human Proc Natl Acad Sci USA 94:3110-3115.
Greig cephalopolysyndactyly syndrome (GCPS) and the Wodarz A, Nusse R (1998). Mechanisms of Wnt signaling in
mouse mutant extra toes (Xt). Mamm Genome 3:461-463. development. Annu Rev Cell Dev Biol 14:59-88.
Wagner G (1949). Die Bedeutung der Neuralleiste fur die Wozney JM (1998). The bone morphogenetic protein family:
kopfgestaltung der Amphibienlarven. Rev Suisse Zool multifunctional cellular regulators in the embryo and
56:519-620. adult. Eur J Oral Sci 106(Suppl 1):160-166.
Wall NA, Hogan BL (1995). Expression of bone morphogenet- Xu X, Weinstein M, Li C, Naski M, Cohen RI, Ornitz DM, et al.
ic protein-4 (BMP-4), bone morphogenetic protein-7 (1998). Fibroblast growth factor receptor 2 (FGFR2)-medi-
(BMP-7), fibroblast growth factor-8 (FGF-8) and sonic ated reciprocal regulation loop between FGF8 and FGF1O
hedgehog (SHH) during branchial arch development in is essential for limb induction. Development 125:753-765.
the chick. Mech Dev 53:383-392. Xue Y, Gao X, Lindsell CE, Norton CR, Chang B, Hicks C, et
Walterhouse D, Ahmed M, Slusarski D, Kalamaras J, Boucher al. (1999). Embryonic lethality and vascular defects in
D, Holmgren R, et al. (1993). gli, a zinc finger transcription mice lacking the Notch ligand Jaggedl. Hum Mol Genet
factor and oncogene, is expressed during normal mouse 8:723-730.
development. Dev Dyn 196:91-102.0 Yamada G, Mansouri A, Torres M, Stuart ET, Blum M, Schultz
Wang J, Shackleford GM (1996). Murine WntlOa and WntlOb: M, et al. (1995). Targeted mutation of the murine goosecoid
cloning and expression in developing limbs, face and skin gene results in craniofacial defects and neonatal death.

299
12(4) 276 300 (2001)
1 2(4):276-300 ( 2001 ) Crit Rev Oral Biol Med
Grit Rev Oral Biol Med

299
Development 121:2917-2922. defines the rostral boundary of the neural plate. J Neurosci
Yamada G, Ueno K, Nakamura S, Hanamure Y, Yasui K, 18:8322-8330.
Uemura M, et al. (1997). Nasal and pharyngeal abnormal- Yuan ZR, Kohsaka T, Ikegaya T, Suzuki T, Okano S, Abe J, et al.
ities caused by the mouse goosecoid gene mutation. (1998). Mutational analysis of the Jagged 1 gene in Alagille
Biochem Biophys Res Commun 233:161-165. syndrome families. Hum Mol Genet 7:1363-1369.
Yamaguchi TP, Rossant J (1995). Fibroblast growth factors in Zhang J, Hagopian-Donaldson S, Serbedzija G, Elsemore J,
mammalian development. Curr Opin Genet Dev 5:485-491. Plehn-Dujowich D, McMahon AP, et al. (1996). Neural
Yamaguchi TP, Bradley A, McMahon AP, Jones S (1999). A tube, skeletal and body wall defects in mice lacking tran-
Wnt5a pathway underlies outgrowth of multiple struc- scription factor AP-2. Nature 381:238-241.
tures in the vertebrate embryo. Development 126:1211-1223. Zhao GQ, Zhao S, Zhou X, Eberspaecher H, Solursh M, de
Yanagisawa H, Yanagisawa M, Kapur RP, Richardson JA, Crombrugghe B (1994a). rDlx, a novel distal-less-like
Williams SC, Clouthier DE, et al. (1998). Dual genetic path- homeoprotein is expressed in developing cartilages and
ways of endothelin-mediated intercellular signaling discrete neuronal tissues. Dev Biol 164:37-51.
revealed by targeted disruption of endothelin converting Zhao GQ, Eberspaecher H, Seldin MF, de Crombrugghe B
enzyme-i gene. Development 125:825-836. (1994b). The gene for the homeodomain-containing protein
Yang L, Zhang H, Hu G, Wang H, Abate-Shen C, Shen MM Cart-i is expressed in cells that have a chondrogenic poten-
(1998). An early phase of embryonic Dlx5 expression tial during embryonic development. Mech Dev 48:245-254.

300 Crit
Crit Rev
Rev Oral
Oral Biol
Biol Med
Med
12(4):276-300 (2001)

12(4):276-300 (2001)

Das könnte Ihnen auch gefallen