Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

J CRITICAL

Dent Res 87(12) 2008


REVIEWS IN ORALDento-Craniofacial
BIOLOGY &Phenotypes in HED
MEDICINE 1089

Dento-Craniofacial Phenotypes and Underlying Molecular


Mechanisms in Hypohidrotic Ectodermal Dysplasia (HED):
a Review
F. Clauss1,4*, M.-C. Manière1, F. Obry1, Background, purpose, and perspectives
E. Waltmann1, S. Hadj-Rabia2, C. Bodemer2,
Y. Alembik3, H. Lesot4, and M. Schmittbuhl1,4 T he hypohidrotic ectodermal dysplasias (HED) form a large and
complex nosological group of congenital disorders, involving
all the Mendelian modes of inheritance, and are characterized by
1Department of Pediatric Dentistry, Dental Faculty, Louis Pasteur

University, National French Reference Center for Dental Manifestations the pathological development and morphogenesis of ectodermal
of Rare Diseases, University Hospital, 1, place de l’Hôpital, F-67000 structures (Pinheiro and Freire-Maia, 1994; Priolo and Lagana,
Strasbourg, France; 2Department of Dermatology, National French 2001; Lamartine, 2003; Itin and Fistarol, 2004). The main
Reference Center for Genodermatosis, Necker-Enfants Malades Hospital, syndromes of HED and their associated clinical signs and genetic
AP-HP, University Descartes-Paris V, France; 3Department of Medical
Genetics, University Hospital, Strasbourg, France; and 4INSERM UMR defects are summarized in Table 1. The most prevalent form of
595, Dental Faculty, Louis Pasteur University, Strasbourg, France; HED is X-linked hypohidrotic ectodermal dysplasia (XLHED),
*corresponding author, francois.clauss@chru-strasbourg.fr and autosomal forms of the disorder are also found with a lower
frequency (Vincent et al., 2001; Lind et al., 2006; Van der Hout et
J Dent Res 87(12):1089-1099, 2008 al., 2008).
Molecular etiologies of HED consist of mutations of the genes
ABSTRACT
involved in the Ectodysplasin (EDA)-NF-kB pathway. XLHED
The hypohidrotic ectodermal dysplasias (HED) belong to a corresponds to mutations of the EDA gene (Kere et al., 1996;
large and heterogenous nosological group of polymalfomative
Schneider et al., 2001; Vincent et al., 2001; RamaDevi et al., 2008).
syndromes characterized by dystrophy or agenesis of
Autosomal-dominant and -recessive forms of HED involve both
ectodermal derivatives. Molecular etiologies of HED consist
of mutations of the genes involved in the Ectodysplasin EDA Receptor and EDA Receptor-associated Death-Domain genes
(EDA)-NF-kB pathway. Besides the classic ectodermal (EDAR and EDARADD) (Chassaing et al., 2006; Bal et al., 2007;
signs, craniofacial and bone manifestations are associated Van der Hout et al., 2008). Syndromic HED with immunodeficiency
with the phenotypic spectrum of HED. The dental phenotype and osteopetrosis is associated with mutations in NF-kB Essential
of HED consists of various degrees of oligodontia with Modulator gene (NEMO) encoding Inhibitor Kappa Kinase g
other dental abnormalities, and these are important in the sub-unit (IKKg) (Dupuis-Girod et al., 2002; Vinolo et al., 2006)
early diagnosis and identification of persons with HED. (Table 1). EDA is a member of the Tumor Necrosis Factor (TNF)
Phenotypic dental markers of heterozygous females for EDA family, activating the NF-kB and Jun N-terminal Kinase (JNK)/
gene mutation—moderate oligodontia, conical incisors, c-jun/c-fos pathways (Kere et al., 1996; Mikkola et al., 1999;
and delayed dental eruption—are important for individuals Cui et al., 2002) (Fig. 1). This collagenous protein is implicated
giving reliable genetic counseling. Some dental ageneses in ectodermal structure and morphogenesis (Kere et al., 1996;
observed in HED are also encountered in non-syndromic Montonen et al., 1998), but is also implicated in osteogenesis
oligodontia. These clinical similarities may reflect possible (Montonen et al., 1998). Functional interactions between EDA and
interactions between homeobox genes implicated in early factors involved in migratory neural crest cell differentiation as Wnt
steps of odontogenesis and the Ectodysplasin (EDA)-NF- or BMP-Msx have been described and underline the role of the EDA
kB pathway. Craniofacial dysmorphologies and bone signaling pathway in craniofacial patterning and growth (Laurikkala
structural anomalies are also associated with the phenotypic et al., 2001; Pummila et al., 2007). The NF-kB transcriptional
spectrum of persons with HED patients. The corresponding factor is essential in a wide range of cellular processes, especially
molecular mechanisms involve altered interactions between osteoclastic differentiation, through Receptor Activator NF-kB
the EDA-NF-kB pathway and signaling molecules essential (RANK) signaling (Franzoso et al., 1997; Galibert et al., 1998;
in skeletogenic neural crest cell differentiation, migration, Asagiri and Takayanagi, 2007), and thus alterations of this complex
and osteoclastic differentiation. Regarding oral treatment lead to bone phenotypic modifications (Fig. 1).
of persons with HED, implant-supported prostheses are
The purposes of this review are to state precisely the dento-
used with a relatively high implant survival rate. Recently,
craniofacial and bone phenotypes associated with HED, from both
groundbreaking experimental approaches with recombinant
EDA or transgenesis of EDA-A1 were developed from the an analysis of the literature and from persons with HED, and also
perspective of systemic treatment and appear very promising. to point out the molecular mechanisms underlying these anomalies
All these clinical observations and molecular data allow for from recent progress in the understanding of the EDA-NF-kB
the specification of the craniofacial phenotypic spectrum in pathway.
HED and provide a better understanding of the mechanisms
involved in the pathogenesis of this syndrome. PERSONs & methods
Key words: Hypohidrotic ectodermal dysplasia, cranio­ Information for this review was gathered from the presentation of
facial dysmorphologies, bone density, NF-kB pathway. different individuals examined in the National French Reference
Center for Dental Manifestations of Rare Diseases at the Strasbourg
Received February 8, 2008; Last revision May 9, 2008; Accepted University Hospital (France). Of the 11 persons presenting,
July 8, 2008 all but the heterozygous female exhibited the classic features

Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.
1089
International and American Associations for Dental Research
1090 Clauss et al. J Dent Res 87(12) 2008

Table 1. Summary of the Main HED Syndromes

Disease Clinical Manifestations Genetic Etiologies

X-linked HED (XLHED) Alopecia, reduced number and dysfunction of sweat glands and Ectodysplasin (EDA) gene
mucous glands, oligodontia, dystrophic nails, hypoplastic skin,
craniofacial dysmorphic features.
Autosomal dominant and Same clinical signs as XLHED EDA Receptor (EDAR) and EDA Receptor
recessive forms of HED Less severe signs in autosomal-dominant forms of HED. Associated Death-Domain (EDARADD) genes
XLHED with immunodeficiency Ectodermal signs associated with immunodeficiency. NF-kB Essential Modulator (NEMO) gene
Osteopetrosis-lymphoedema-HED Ectodermal signs associated with osteopetrosis, lymphedema NEMO gene
and immunodeficiency.
Incontinentia pigmenti Ectodermal signs with skin lesions, neurological and retinal defects NEMO gene

Data derived from Pinheiro and Freire-Maia (1994), Priolo and Lagana (2001), Lamartine (2003), Itin and Fistarol (2004), Jonhson et al. (2002), Bal
et al. (2007), Dupuis-Girod et al. (2002).

of the XLHED phenotype. A rigorous clinical examination Epidemiological data and dental
was performed for each person, completed by photographs, phenotype analyses in PERSONS WITH HED
radiographs (panoramic, lateral projection, and, when possible, The differential dental phenotypic expressions of the genetic
CT examination), plaster casts, and molecular diagnosis (EDA defects associated with HED will be presented below.
gene sequencing). An overview of the main characteristics of
the persons with XLHED (sex, age, and genotype) is given in Tooth Number Anomalies
Table 2. Anomalies in Males Affected with X-linked HED
Primary Dentition
An average of 3.50 missing
teeth in the maxilla and 5.33
mandibular missing teeth has
been described (Barberia et al.,
2006) (Fig. 2a). This maxillary
and mandibular hypodontia in the
primary dentition of persons with
HED differs from non-syndromic
hypodontia, which exhibits an
opposite maxillary-mandibular
distribution of dental agenesis,
i.e., an average of 1.62 ageneses of
maxillary teeth vs. 0.25 mandibular
missing teeth (Barberia et al.,
2006). This maxillary-mandibular
discrepancy between HED and
non-syndromic hypodontia reveals
a more important disorder of the
molecular mechanisms responsible
for mandibular odontogenesis in
HED. Homeobox genes involved
in the patterning of the mandibular
odontogenic ectomesenchyme,
such as Msx-1, Msx-2, or Distal-
less Homeobox-2 (Dlx-2) and
Figure 1. Original overview of the main molecular interactions between the EDA pathway and the BMP-
belonging to the odontogenic
Msx, RANK-TRAF6-NF-kB, FGF, Wnt, and Activin-b pathways implicated in dental, craniofacial, and homeobox gene code (Jowett et al.,
bone phenotypes associated with HED. EDA activates NF-kB factor via its Receptor EDAR interacting 1993; Thomas and Sharpe, 1998),
with TRAF6, TAK1 Binding protein 2 (TAB2), TGF-b Activating Kinase 1 (TAK1) adapter molecules, and seem to be more influenced by
NEMO-IKKa/IKKb. Phosphorylation of the inhibitor factor IkB by IKKa/IKKb leads to its proteasomic
degradation and to the nuclear translocation of the dimeric NF-kB factor. NF-kB factor is involved in
EDA mutation. The most frequent
regulation of osteoclastic differentiation and ectodermal morphogenesis via Sonic Hedgehog (Shh) factor, tooth ageneses, with decreasing
Cyclin-D1, and Nuclear Factor of Activated T-cells (NFAT-1/2). Interactions between EDA-NF-kB and prevalence, are the mandibular
BMP-Msx are mediated by Ccn2-Connective Tissue Growth Factor (CTGF)/Follistatin BMP inhibitors. FGF incisors, lateral maxillary
activates Msx1-Pax9 expression via NF-kB factor. Functional interactions between EDA and RANKL-
RANK pathways are established through TRAF6 and NF-kB factors. EDA and EDAR transcriptions are
incisors, central maxillary
up-regulated by Wnt-b-catenin-Lef1 and Activin-b-smad pathways, respectively. incisors, mandibular molars, and
Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


J Dent Res 87(12) 2008 Dento-Craniofacial Phenotypes in HED 1091

maxillary first molars (Vierucci et Table 2. Characteristics of Persons with XLHED Treated in the National French Reference Center for
al., 1994; Ruhin et al., 2001; Tarjan Dental Manifestations of Rare Diseases at the Strasbourg University Hospital (France)
et al., 2005). Anodontia—congenital
absence of primary and permanent Sex Age Genotype Figure
teeth—was also encountered in HED
(Beliakov et al., 1998; Acikgoz et al., Patient 1 male 4 yrs old c.756T>G exon 7 mutation of EDA gene Fig. 2a
2007), and corresponds to the most (alteration of EDA TNF homology sub-domain)
severe phenotype. Dental hypodontia Patient 2 male 35 yrs old Exon 3 deletion of EDA gene (alteration of Figs. 2b, 2c
or anodontia in the primary dentition EDA proteolytic cleavage site)
of persons with HED is a clinical Patient 3 female 41 yrs old Heterozygous for a 9-bp exon 8 deletion of Fig. 2d
predictor that should lead to further EDA gene (alteration of EDA TNF sub-domain) Fig. 3b
investigation (radiographic examination Patient 4 male 4 yrs old EDA gene exon 8 mutation Fig. 2e
and molecular analyses). Patient 5 male 7 yrs old EDA gene exon 3 mutation Fig. 2f
Patient 6 male 7 yrs old EDA gene exon 1 mutation (alteration of Fig. 2g
Permanent Dentition
EDA intracellular-transmembrane domain) Fig. 3d
Dental ageneses range between 11.2 Patient 7 male 6 yrs old R156C EDA gene exon 3 mutation Fig. 3a
and 16.4 absent teeth (Barberia et al., Patient 8 male 51 yrs old c.754C>A exon 7 mutation of EDA gene Fig. 3c
2006; Prager et al., 2006) (Figs. 2b, Fig. 3e
2c). In a sample of 30 persons with Patient 9 male 27 yrs old Undetermined genotype Fig. 3g
HED, oligodontia (absence of at least
6 teeth) and hypodontia occurred,
respectively, in 83% and 17% of
individuals (Prager et al., 2006). A
more severe dental phenotype was anodontia, present in 12% of
persons with HED, and oligodontia in all such persons has also
been reported (Yavuz et al., 2006). Frequency of tooth agenesis
seems to be higher for mandibular teeth than for maxillary
teeth (respectively, 7.9 and 6.8 missing teeth) (Prager et al.,
2006) (Fig. 2b). This more severe phenotype in the mandible
could be linked to specific effects of the EDA mutation that
may affect mandibular tooth organogenesis preferentially. The
most frequent missing teeth, in order of frequency, are: (in the
maxilla) lateral incisors (86%), second premolars (66%), and
second molars (53%); and (in the mandible) central incisors
(83%), lateral incisors (73%), second premolars (70%), and
second molars (53%) (Prager et al., 2006) (Fig. 2b). From these
clinical observations, the most posterior teeth of the incisor,
premolar, and molar series seem to be preferentially affected
by the EDA mutation, except for the mandibular central incisor
(Prager et al., 2006). The permanent teeth most likely to be
present are the maxillary central incisors (42%), maxillary first
molars (41%), mandibular first molars (39%), and maxillary
canines (22%) (Guckes et al., 1998). Some dental ageneses
observed in HED are also encountered in non-syndromic
oligodontia related to mutations of homeobox genes such as
Muscle Segment Homeobox-1 (Msx-1) and Paired Homeobox-9
genes (Pax-9) (Vastardis et al., 1996; Arte et al., 2001). The
similarities observed in dental agenesis may thus underline the
Figure 2. Dental phenotypes associated with XLHED (patients treated in
possible interactions between these genes and the Ectodysplasin the National French Reference Center for Rare Diseases, Strasbourg,
(EDA)-NF-kB pathway (Tucker et al., 1998; Ogawa et al., France). (a-c) Oligodontia in primary and permanent dentitions.
2006; Pummila et al., 2007). (a) The phenotype in the primary dentition consists of agenesis of
mandibular incisors and first primary molars, with absence of maxillary
Anomalies in Female Carriers of X-linked HED lateral primary and permanent incisors and the first left primary
Dental phenotype variability has been described in HED molar. (b-c) Permanent dentition lacking maxillary lateral incisors,
premolars, and molars (except for the right first molar), associated
heterozygous females, with moderate oligodontia (Fig. 2d)
with severe mandibular oligodontia (with only canines present). (d)
and delayed dental eruption observed in our department and Heterozygous female exhibiting lateral incisor agenesis, and slightly
reported in the literature (Cambiaghi et al., 2000; Nishibu et conical mandibular canines. (e-g) Different dental phenotypes
al., 2003; Lexner et al., 2007a). A higher frequency of dental observed in various mutations of the EDA gene. (e) Severe phenotype
agenesis was found in heterozygous HED females, particularly with mandibular anodontia and maxillary oligodontia, dysmorphic
developing permanent tooth germs of central maxillary incisors, and
involving the maxillary lateral incisors, mandibular incisors, dysmorphic primary canines (EDA gene exon 8 mutation). (f-g) Mild
and first molars; for these patients, the frequency of agenesis phenotypes with different degrees of oligodontia and taurodontism
ranged between 1 and 7 missing teeth (Cambiaghi et al., 2000; (respectively, EDA gene exon 3 and exon 1 mutations).

Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


1092 Clauss et al. J Dent Res 87(12) 2008

Nishibu et al., 2003; Lin et al., 2004; Lexner et al., 2007a). HED cases; external root resorption is present in 15% of children
heterozygous females thus exhibited moderate dental agenesis with HED (Yavuz et al., 2006). Persistence of primary teeth,
(Fig. 2d) compared with that observed in homozygous females, especially second primary molars and canines, corresponds to
because of the X-chromosome inactivation—a frequency of 11.8 another abnormality (Yavuz et al., 2006). Enamel dysplasias
tooth ageneses being observed in the most severe phenotypes linked to alterations in ameloblast cytodifferentiation (Ruhin
associated with homozygous female patients (Visinoni et al., et al., 2001) are also observed in EDA mutation. These clinical
2003; Prager et al., 2006). However, no correlations were manifestations are also encountered in the Tabby mouse model
established between the pattern of X-chromosome inactivation (XLHED experimental model) (Risnes et al., 2005).
and the frequency of agenesis (Vincent et al., 2001). Oligodontia
Anomalies in Female Carriers of X-linked HED
in the primary dentition encountered in females might predict an
EDA gene mutation at a probability of 1/50. In the permanent Cone-shaped incisors and microdontia are associated with EDA
dentition, this clinical feature could suggest an EDA mutation in mutation heterozygous females (Lexner et al., 2007a) (Fig. 2d).
1/500 cases (Spfaer, 1981). Oligodontia in the primary dentition A reduction in molar mesio-distal dimensions is observed in
appears to be an important clinical predictor sign of the EDA EDA mutation heterozygous females, which might be another
mutation in females (Na et al., 2004). These dental phenotypic interesting dental phenotypic marker of an EDA mutation gene
manifestations thus play a crucial role in the detection and carrier (Cambiaghi et al., 2000; Nishibu et al., 2003; Lexner
diagnosis of EDA mutation female carriers and should lead to et al., 2007a). These tooth morphological anomalies provide
further clinical, radiographic, and molecular investigation. clinical criteria for the detection of female carriers of the EDA
mutation (Glavina et al., 2001) and should be considered in
Anomalies in Autosomal Forms of HED genetic counseling.
Dental phenotypic manifestations in the primary and permanent
dentitions of HED autosomal-dominant and -recessive forms The genotypic heterogeneity in XLHED:
are associated with the EDAR-EDA Receptor Associated Death- a pathway toward dental phenotype
Domain (EDARADD) gene mutations (Aswegan et al., 1997; variablity
Kobielak et al., 2001a; Chassaing et al., 2006; Bal et al., 2007, A variablity affecting various exons of the EDA locus has been
RamaDevi et al., 2008; Van der Hout et al., 2008) and are also described in XLHED (Kobielak et al., 2001b; Schneider et al.,
characterized by multiple missing teeth (Aswegan et al., 1997). 2001; Hashiguchi et al., 2003; Lin et al., 2004; Huang et al.,
In the permanent dentition, ageneses of mandibular incisors seem 2006; RamaDevi et al., 2008). The most prevalent mutations
to be encountered with a high prevalence. Agenesis of maxillary were located between exons 3 and 9 of the EDA locus (94.4%)
incisors and agenesis of canines and premolars also occur with (Monreal et al., 1998); they clustered preferentially in three
decreasing prevalence (Aswegan et al., 1997). However, the functionally important molecular domains of EDA-A1 isoform:
oligodontia is more severe in the autosomal-recessive than in (1) the extracellular TNF homology sub-domain necessary for
-dominant forms of HED (Chassaing et al., 2006; Van der Hout receptor binding, which corresponds to exons 7-9 (Tariq et al.,
et al., 2008). If both maxillary incisor and canine ageneses in the 2007a; RamaDevi et al., 2008); (2) the collagen sub-domain
HED autosomal forms are confirmed by epidemiologic survey, (exons 5-6); and (3) the proteolytic cleavage site for a furin
this clinical sign would provide an interesting phenotypic marker protease (exon 3) (Pääkkönen et al., 2001; Schneider et al.,
of the EDAR mutation. 2001). Rare mutations involve exon 1 and affect intracellular,
transmembrane, and extracellular domains of EDA-A1 (Kere
Dental Dysmorphic Features Associated with HED
et al., 1996; Ferguson et al., 1998; Hashiguchi et al., 2003).
Anomalies in Males Affected with X-linked HED Different phenotypic expressions seem to be found in persons
Tooth dysmorphologies are associated with HED, and are affected by these various mutations (Figs. 2e, 2f, 2g). A milder
induced by alterations of the EDA-EDAR-NF-kB pathway phenotype, including moderate oligodontia and absence of both
morphogenetic functions during odontogenesis (Pispa et dermatological manifestations and infectious susceptibility,
al., 2004; Tucker et al., 2004; Tariq et al., 2007a,b) (Figs. was observed in a mutation of exon 9 (Hashiguchi et al., 2003).
2c-2e). Microdontia and cone-shaped teeth were frequent A similar expression characterized by moderate oligodontia,
morphological features observed in affected individuals (Fig. hypotrichosis, and hypohydrosis was found in EDA gene exon
2c) (Ruhin et al., 2001). Crown form abnormalities have been 1 mutation. However, hypodontia can be the only phenotypic
found in both primary and permanent dentitions (Prager et al., manifestation associated with a substitution mutation of
2006), and most frequently involve the anterior maxillary teeth exon 1 (Schneider et al., 2001). The more classic phenotype
(Yavuz et al., 2006). The conical crown results from the largest of XLHED, with dental abnormalities, hypohydrosis, and
mesio-distal diameter being displaced apically (Crawford craniofacial dysmorphologies, was also linked to mutated
et al., 1991). Moderate to severe taurodontism is known to exons 5 and 6 (Vincent et al., 2001). As indicated by these
affect preferentially the second primary mandibular molars in observations, the genetic variability seems to lead to various
some individuals with HED (Jorgenson, 1982; Crawford et dental phenotypes in XLHED (Figs. 2e, 2f, 2g). More precise
al., 1991; Glavina et al., 2001) (Figs. 2f, 2g). Nevertheless, dental phenotypic analyses could, therefore, allow for a better
this feature is also found in permanent molars and provides understanding of the genetic influence on the etiopathogenesis
a dental marker of developmental abnormality (Figs. 2f, of tooth anomalies in XLHED.
2g). Taurodontism may result from abnormal mesodermal-
ectodermal interactions linked to EDA mutations (Ruhin et Molecular mechanisms underlying dental
al., 2001). Other dental dysmorphologies are also observed in abnormalities in HED
HED. Root shape abnormalities are depicted in 30% of HED Specific interactions between EDA and BMP signaling

Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


J Dent Res 87(12) 2008 Dento-Craniofacial Phenotypes in HED 1093

have been demonstrated


(Pummila et al., 2007). EDA
induces the expression of two
BMP inhibitors, Ccn2/Ctgf
(Cyr61 CTGF Nov family
protein 2/connective tissue
growth factor) and follistatin
(Pummila et al., 2007) (Fig.
1). Nevertheless, an important
decrease in BMP expression is
required for placodal ectoderm
formation and neural crest
cell differentiation; these two
cellular groups are involved in
the initial steps of odontogenesis
(Tucker et al., 1998; Zhang
et al., 2003). The homeobox
genes Msx-1 and Msx-2 are
downstream components of
the BMP signaling pathway,
and are, respectively, involved
in the early differentiation of
maxillary incisor and molar
mesenchyme from neural-crest
cells (Bei and Maas, 1998;
Bei et al., 2000; Lidral and
Reising, 2002; Han et al., 2003;
Ramos and Robert, 2005). EDA
mutations are therefore likely to
alter EDA-BMP-Msx molecular
interactions, and could be
responsible for the disturbance
of odontogenesis.
The possibility for
convergence between EDA and
Fibroblast Growth Factor (FGF)
signaling pathways may be
evoked in normal odontogenesis
(Pispa et al., 1999) (Fig. 1). The
NF-kB heterotrimeric factor was
recently observed to mediate
FGF signaling and, particularly,
the FGF regulation of Msx-1
mesenchymal expression (Bei
and Maas, 1998; Bushdid et
al., 2001). In the Tabby mouse,
the FGF (Fgf-3 and Fgf-10)
level of expression appeared
to be decreased in the dental
mesenchyme, and Fgf-4 was
reduced in the enamel knot
of the molar cap (Pispa et al.,
1999). Fgf-10 has been shown to
stimulate cell division in dental
epithelium, and Fgf-4 stimulates Figure 3. Craniofacial and bone phenotypes associated with XLHED (persons treated in the National French
Reference Center for Rare Diseases, Strasbourg, France). (a-c) Craniofacial differences between persons with
cell proliferation in both dental XLHED and healthy individuals, highlighted by cephalometric radiographic analyses: (a) XLHED child, (b)
epithelial and mesenchymal heterozygous female, (c) XLHED adult. For each measurement, values are given for both persons with HED and
tissues (Jernvall et al., 1994; control individuals. Data from the control sample are derived from Johnson et al. (2002). Linear measurements
Kettunen and Thesleff, 1998). are expressed in mm and angles in degrees. (d-g) Bone structural changes in XLHED. Orthoradial CT slices of
the mandible showing (d) enlargement of the bone thickness and (e) hypermineralization of the medullary bone.
Alterations of FGF signaling (f-g) Bone densitometric profile. (f) Control individual: low density (D3-D4) in medullary bone (Lekholm, 1986)
induce defective development and higher density for mandibular cortex (D2-D1); (g) person with XLHED: characteristic hypermineralization of
of Tabby tooth germs, leading medullary bone (D2-D1) in the mandibular symphyseal area.
Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


1094 Clauss et al. J Dent Res 87(12) 2008

to an absence of anteroconid and hypoconulid cusps as well as is also severely affected in HED, as indicated by the abnormal
third molars (Pispa et al., 1999). Interestingly, a partial recovery anterior and posterior cranial base lengths (respectively, S-Na
of morphogenesis associated with a stimulation of additional and Basion-S: Ba-S). While the anterior cranial base exhibits
cusp development was obtained in cultured Tabby molars by the a tendency to hypoplasia, the posterior cranial base undergoes
addition of exogenous Fgf-10 (Pispa et al., 1999). an anteroposterior hyperplasia (Figs. 3a-3c), an increased
The Ectodysplasin/NF-kB signaling pathway thus appears deviation from the norm for the corresponding length being
to be involved in tooth growth and morphogenesis at different registered (51.9 mm vs. 43.8 mm at ages 13-17 yrs) (Johnson
stages during development (Laurikkala et al., 2001; Courtney et al., 2002). Nevertheless, the total cranial base length seems
et al., 2005). Furthermore, the disturbance of this pathway to be within normal limits in some adults with HED, revealing
could lead to the dental phenotypic abnormalities associated a form of reciprocal compensation between the two areas of the
with HED. cranial base (Johnson et al., 2002). The flexure of the cranial
base is abnormally emphasized in HED, as demonstrated by the
Craniofacial-specific traits in PERSONS progressive reduction of the angle Ba-S-Na (123.5° vs. 128.1°,
WITH XLHED respectively, in adults with HED and healthy individuals)
Craniofacial dysmorphologies associated with HED have been (Johnson et al., 2002) (Fig. 3c). Moreover, a narrow cranial
documented by several studies, but their precise descriptions base seems to be another craniofacial change observed in
remain rare, since these studies were limited to case reports and HED (Skrinjarić et al., 2003). Paranasal sinuses sometimes
a few cephalometric studies (Johnson et al., 2002; Bondarets exhibit abnormal size variations in HED. Cases of reduced
et al., 2002; Ferrario et al., 2004; Yavuz et al., 2006; Arslan frontal sinuses as well as cases of hypertrophic maxillary
et al., 2007, Lexner et al., 2007b). The classic craniofacial sinuses have been reported (Ruhin et al., 2001). Nevertheless,
dysmorphic features described in HED include maxillary all the previous craniofacial abnormalities were probably
hypoplasia, mandibular prognathism, facial concavity, a underestimated when the cephalometric measurements were
depressed nasal bridge, and growth alteration of the cranial referred to Riolo’s cephalometric standards (1974) instead of
base (Figs. 3a-3c) (Ruhin et al., 2001; Bondarets et al., 2002; Johnson’s. For example, the mean length of the maxilla (ANS-
Johnson et al., 2002, Arslan et al., 2007; Lexner et al., 2007b). PNS) in children with HED children (5-12 yrs) was 42.1 mm,
Frontal prominence, reduced facial height (Figs. 3a-3c), cranial compared with 44.8 mm by Johnson’s standards and 53.4 mm
vertex hypoplasia, retroclined nasal bone, and antimongoloid by Riolo’s.
slant of the eyes are other reported craniofacial phenotypic Heterozygous females, carriers of XLHED, also exhibit
manifestations (Ruhin et al., 2001; Alcan et al., 2006; Lexner craniofacial phenotypic features that can be severe, despite a
et al., 2007b). Maxillary hypoplasia consists of reduced length moderate dental phenotype (Saksena and Bixler, 1990) (Fig.
and height of this bone. Maxillary hypodevelopment seems to 3b). Maxillary hypoplasia, mandibular hypoplasia, midface
be accentuated during growth, as indicated by the increasing hypoplasia, everted lips, saddle nose, and abnormal anterior
age-dependent differences in the maxillary dimensions for and total cranial base lengths can also be found (Cambiaghi
persons with HED compared with healthy individuals (Johnson et al., 2000) (Fig. 3b). A prognathic mandible can also be
et al., 2002). Angular measurements describing the sagittal associated with the phenotype of heterozygous females for
position of the maxilla (Sella turcica-Nasion-A point: SNA, EDA mutation, characterized by an increase in the Pog-Na-S
Anterior Nasal Spine-Nasion-Sella turcica: ANS-Na-S) angular measurement (Fig. 3b). These craniofacial traits could
confirm this morphological tendency (Bondarets et al., 2002) be considered as important clinical predictor signs contributing
(Figs. 3a-3c). The retrognathism of the maxilla appears to be to reliable genetic counseling leading to the diagnosis and
the most striking craniofacial feature, whatever the degree identification of heterozygous female carriers of the EDA gene
of hypodontia in XLHED males or heterozygous females mutation.
(Figs. 3a-3c). The transverse dimension of the maxilla is also The improvement in masticatory function through early
reduced (Saksena and Bixler, 1990). Interestingly, the palatal implant treatment and prosthesis allows for a partial recovery
width is not affected (Dellavia et al., 2006). In some persons, of facial growth and provides a limited rescue of the associated
the mandible exhibited a decreased length of the body (Fig. dysmorphology, as demonstrated by the slightly more
3a); despite this, it presents an increased relative prognathic normative cephalometric features in treated persons (Boj et al.,
position compared with the midface, as indicated by the angle 1993; Franchi et al., 1998; Johnson et al., 2002; Alcan et al.,
Pogonion-Nasion-Sella turcica (Pog-Na-S) (Fig. 3c) and the 2006).
distance S-Pog (Bondarets et al., 2002; Johnson et al., 2002). These observations emphasize the major role of genetic
On the whole, these cephalometric analyses of persons with factors in craniofacial development associated with XLHED.
HED indicated a class III tendency (Lexner et al., 2007b)
(Figs. 3a-3c). In a transverse plane, the lower facial width Craniofacial features in PERSONS
(bigonial diameter Gonion-Gonion: Go-Go) demonstrated WITH autosomal HED
a reduced value compared with that in control individuals In autosomal forms of HED, a craniofacial phenotypic
(Saksena and Bixler, 1990). Facial vertical dimensions were variability seems to be linked to different mutation sites
affected in HED, and the total anterior face height (Nasion- (Chassaing et al., 2006; Bal et al., 2007). Indeed, genetic
Menton: Na-Me), lower anterior face height (ANS-Me), and defects in autosomal-dominant and -recessive HED can
upper anterior face height (Na-ANS) were reduced in children involve both EDAR and EDARADD genes (Bal et al., 2007;
and adolescent groups (Johnson et al., 2002) (Fig. 3a). These RamaDevi et al., 2008; Van der Hout et al., 2008). Severe
differences seem to decrease in adults, since they were all craniofacial abnormalities with prominent forehead, depressed
within normal limits (Johnson et al., 2002). The cranial base nasal bridge, and small nose have been reported in autosomal-
Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


J Dent Res 87(12) 2008 Dento-Craniofacial Phenotypes in HED 1095

recessive EDAR mutations (Chassaing et al., 2006). A moderate et al., 2001a). The craniofacial dysmorphologies and alterations
craniofacial phenotype, including mild frontal bossing and of bone growth observed in HED (Motil et al., 2005) may be
zygomatic flattening, has generally been associated with emphasized by adenohypophysis dysfunction and reduced GH
autosomal-dominant forms (Aswegan et al., 1997; Chassaing secretion (Isgaard et al., 1986; Van Erum et al., 1997; Ramirez-
et al., 2006). More precise craniofacial phenotypic analyses Yanez et al., 2005).
are necessary to improve genotype-phenotype correlations in The NF-kB factor inactivation in developing ectoderm
autosomal forms of HED. In this context, experimental mouse leads to craniofacial phenotypic manifestations with alteration
models of HED, i.e., Tabby (XLHED experimental model), of palatal development, as observed in the keratin 5 (K5)-GR
Downless, or Crinkled (autosomal HED mouse models), transgenic mouse, which exhibits a phenotype of ectodermal
would be helpful for a better understanding of the X-linked dysplasia induced by ectoderm-targeted overexpression of the
and autosomal HED phenotypic spectrum and craniofacial glucocorticoid receptor (GR) (Cascallana et al., 2005).
variability.
All the present results allow for a better detection of Mandibular and maxillary bone
persons with HED, and underscore the use of anthropometric phenotypic modifications
approaches for screening HED carriers and selecting those associated with HED
individuals for molecular genetic testing. Bone structural modifications seem to be associated with
XLHED, as suggested by our craniofacial CT-scan
Molecular mechanisms suggested in THE examinations (Figs. 3d-3g). Areas of medullary bone
HED craniofacial phenotype hyperdensity–D1-D2 densities (Lekholm, 1986)—were
The EDA gene mutations and alterations in the developmental observed, particularly in the mandibular symphysis (Figs. 3d,
molecular function of EDA could be responsible for 3e). The high densitomeric profiles found with CT-examination
disturbances in craniofacial derivatives through alterations seem to confirm the medullary bone hypermineralization
of neural crest cell differentiation and migration. A (Fig. 3g). Variability in medullary bone hypermineralization
strong expression of EDA transcripts has been localized appears to exist among persons with XLHED, as demonstrated
in neuroectodermal cells involved in craniofacial skeletal by the different degrees of bone densities observed in the
patterning (Montonen et al., 1998; Kanzler et al., 2000). persons examined (Figs. 3d, 3e). A topographic variation of
Moreover, molecular dysfunction in interactions between EDA bone hypermineralization was also found in the mandible and
and Wnt-b-catenin- Lymphocyte Enhancer Factor-1 (Lef1) corresponded to high bone density in the inter-foraminal area
pathways (Fig. 1) is likely to contribute to other abnormalities and to subnormal bone densitometric profiles in posterior
of neuroectodermal derivatives (Laurikkala et al., 2001), since areas. The variations of bone densities may be linked to
Wnt factor is known to be a neural-crest inducer (Basch and genotypic heterogeneity in XLHED, as suggested by some
Bronner-Fraser, 2006). genotype-bone phenotype correlations. For example, a D1
EDA also influences BMP signaling, which is essential bone density was associated with EDA exon 7 mutation,
for the development of skeletogenic cranial neural crest cells whereas a D2 density was observed in an EDA exon 1 mutation
(Kanzler et al., 2000; Ashique et al., 2002; Haworth et al., (Figs. 3d, 3e). Other bone structural abnormalities consist of
2004; Knight and Schilling, 2006; Raible, 2006). Perturbation hypermineralization of mandibular cortical bone associated
of EDA alters BMP regulation via Ccn2/Ctgf and follistatin with an increased cortical thickness, as demonstrated by CT
(Pummila et al., 2007). A gradient of BMP activity specifies examinations and cortex measurements (Figs. 3d-3g).
the expression of Msx genes encoding homeobox proteins, Similar bone structural modifications have also been
essential for early cranial neural crest development (Ishimura et observed in the Tabby mouse experimental model of XLHED
al., 2000; Tribulo et al., 2003; Levi et al., 2006). (Hill et al., 2002). Micro-computed tomography analyses
Interaction between EDA and FGF has been demonstrated of the Tabby tibial trabecular structure showed a significant
(Pispa et al., 1999), and mutations of the EDA gene may increase in trabecular bone volume, trabecular connections,
influence the role of FGF signaling in later neural crest cell and trabecular numbers associated with a decrease in inter-
proliferation and craniofacial skeletal patterning. FGF activity trabecular spaces (Hill et al., 2002). This osteopetrotic
in the ectoderm and mesoderm plays a crucial role during phenotype could be linked to disturbance of the NF-kB pathway
craniofacial skeletogenesis, and FGF-dependent regulation of and osteoclastic differentiation (Montonen et al., 1998; Lomaga
Msx1 expression is regulated by the NF-kB factor, suggesting et al., 1999).
a molecular confluence between EDA and FGF signaling
pathways (Bushdid et al., 2001; Nie et al., 2006). FGF, acting Molecular mechanisms suggested in
downstream of Tumor Growth Factor-b (TGF-b) signaling, HED bone structural and metabolic
is involved in the expression of the developmental potential modifications
of neural crest cells and their osteogenic or chondrogenic EDA has been expressed not only in ectodermal structures
differentiation through induction of mesenchymal homeobox (Kere et al., 1996; Mikkola et al., 1999; Laurikkala et al., 2001;
gene expression as Bar subclass Homolog-1 (Barx-1), Dlx-1, Schmidt-Ullrich et al., 2006), but also in osteoblasts (Montonen
and Msx-1 in the craniofacial primordia (Bei and Maas, 1998; et al., 1998). EDA transcripts were intensively expressed during
Meyers et al., 1998; Barlow et al., 1999; Shigetani et al., 2000; human embryonic development from 12 wks of gestation in
Crump et al., 2004; Sasaki et al., 2006). rib osteoblasts; intense expression of EDAmRNA was also
Regulation of growth hormone (GH) secretion appears observed in calvarial bone osteoblasts at 16 wks of in utero
to be altered in HED. Indeed, EDA has been detected in the development (Montonen et al., 1998). These observations
developing adenohypophysis (Mikkola et al., 1999; Nordgarden highlight the role of EDA in osteogenesis and emphasize the
Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


1096 Clauss et al. J Dent Res 87(12) 2008

potential bone alterations linked to EDA mutations. Recombinant proteins containing the receptor-binding domain
Endocrine disturbance, expressed as a reduction in of EDA fused to the C terminus of an IgG1 Fc domain
parathyroid hormone (PTH), is another feature of EDA mutation (Fc:EDA1) have been engineered to cross the placental barrier,
(Söderholm and Kaitila, 1985). Since PTH is a major regulator and were administered to pregnant Tabby mice. This treatment
of calcium homeostasis and osteoclastic differentiation, permanently rescued the Tabby phenotype in the offspring. The
especially via Receptor Activator NF-kB Ligand (RANKL) jaw and molars regained both their normal sizes and the classic
activation (Fu et al., 2002), the EDA mutation could be wild-type pattern of sharp cusps (Gaide and Schneider, 2003).
responsible for disorders of bone remodeling, characteristic of However, Fc:EDA1-treated mice did not correct hypodontia in
osteopetrosis. offspring (Gaide and Schneider, 2003).
The NF-kB transcriptional factor is a major regulator of Transgenic Tabby mice bearing the mouse EDA-A1
skeletal development and osteoclastic differentiation, requiring isoform show almost complete restoration of the wild-type
activation of the RANKL-RANK-TNF Receptor Associated phenotype. The wild-type number of 3 molars was observed
Factor-6 (TRAF6) complex (Franzoso et al., 1997; Aradhya in transgenic mice, but these teeth were still generally smaller
and Nelson, 2001; Courtois et al., 2001; Smahi et al., 2002; and exhibited fewer and flattened cusps. Incisors regained their
Chaisson et al., 2004; Asagiri and Takayanagi, 2007; Kurihara wild-type size (Srivastava et al., 2001).
et al., 2007). EDA activates the NF-kB transduction pathway Both reversion of oligodontia and dental dysmorphologies
via the EDA receptor (EDAR), TRAF, and NF-kB Essential were obtained by post-natal single or multiple EDA-A1
Modulator-Inhibitor Kappa Kinase a-Inhibitor Kappa Kinase intravenous administrations in XLHED dogs (Casal et al.,
b (NEMO-IKK-a-IKK-b) complex (Courtney et al., 2005; 2007). All the teeth showed normal development, confirmed
Morlon et al., 2005) (Fig. 1). An alteration of the NF-kB by clinical and radiographic examinations. The present model,
transduction pathway linked to the EDA mutation might developed to approach the conditions of future therapeutic use
interfere with EDA-TRAF6-RANK molecular interaction and of recombinant EDA-A1 in humans, could show the ability of
thus with osteoclastic differentiation, leading to osteopetrosis recombinant EDA-A1 to correct the pathological features of
(Galibert et al., 1998; Dupuis-Girod et al., 2002). Such XLHED (Casal et al., 2007).
molecular interactions are emphasized by the TRAF6-deficient
mice exhibiting osteopetrosis, with an increase in long bone CONCLUSION
and vertebral body radiopacities (Lomaga et al., 1999). A In conclusion, this review, supplemented by our phenotypic
defect in osteoclast function, rather than differentiation, seems analyses of persons with HED, adds to our understanding of the
to be responsible for osteopetrosis. The NF-kB factor also plays dental and craniofacial manifestations of this syndrome and its
a critical role in the maturation of osteoclasts. Abnormalities phenotypic variability. Craniofacial bone hypermineralizations
of the present complex, as in NF-kB p50 and p52 subunit are an integral part of the clinical picture of HED, and need
inactivation, lead to a lack of mature osteoclasts in the mouse to be considered in all persons with HED, but especially
model, thus exhibiting osteopetrosis (Franzoso et al., 1997) when dental implant therapy is planned. Changes in molecular
interaction between EDA-NF-kB and BMP-Msx, as well as
Therapeutic aspects and perspectives in HED FGF pathways linked to an absence, reduced level, or non-
Endosteal Dental Implant Therapy functional extracellular EDA (Fig. 1), seem to be involved in
the odontogenic abnormalities observed in HED. The EDA-
Oral treatment of persons with HED exhibiting severe
NF-kB pathway is also implicated in skeletogenic neural crest
phenotypes of oligodontia or anodontia benefits increasingly
cell differentiation, through BMP-Msx gradient regulation (Fig.
from implant-supported prostheses (Guckes et al., 2002;
1), and its molecular dysfunction seems to lead to craniofacial
Umberto et al., 2007). However, clinical difficulties
dysmorphologies and bone remodeling associated with HED.
encountered in these therapies are partly linked to alveolar
Oral treatment of persons with HED benefits increasingly from
bone hypotrophy (Kramer et al., 2007). Early implant therapy
implant-supported prostheses, with a relatively high implant
in children has been described as not affecting the growth
survival rate. However, the lack of development of the alveolar
and development of the craniofacial complex (Kearns et al.,
ridge in the maxilla and the bone structural hypermineralization
1999; Becktor et al., 2001; Alcan et al., 2006), and a relatively
seem to lead to some implant failure. Recently, groundbreaking
high implant survival rate (85%) was observed (Guckes et al.,
experimental approaches with recombinant EDA or transgenesis
2002). In adults, the 24-month implant survival rate was 95%,
of EDA-A1 have been developed from the perspective of a
with consistently higher survival for implants placed in the
systemic treatment in HED and seem to provide promising
mandible than for those placed in the maxilla. Nevertheless,
results.
27% of the recipients had a failed implant whatever their age
or the anatomical location of the implant (Guckes et al., 2002). ACKNOWLEDGMENTS
These negative clinical outcomes may be linked to the lack of
We thank Bruno Grollemund, DDS (orthodontist, Department
development of the alveolar ridge in the maxilla and to bone
of Orthodontics, Dental Faculty, Strasbourg, France) for his
structural hypermineralization, as suggested by our observations
assistance in cephalometric analysis. We are grateful to Olivia
(Figs. 3d-3g). Indeed, the thickened mandibular cortex appears
Niclas (President of the French Association of Ectodermal
to be difficult to drill, leading to potential surgical thermic
Dysplasia). We thank Professor Roger K. Hall (OAM, MDSc,
trauma with alterations in implant osteointegration.
FRACDS, FICD, FADI, Department of Pharmacology,
Treatment Perspectives University of Melbourne, Royal Children’s Hospital,
Recombinant EDA-A1 administration provides the basis for Melbourne, Australia) for his kind and helpful review of the
possible treatment in XLHED (Gaide and Schneider, 2003). manuscript.
Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


J Dent Res 87(12) 2008 Dento-Craniofacial Phenotypes in HED 1097

REFERENCES absence of inhibitor of kappa B kinase alpha. J Biol Chem 279:54841-


54848.
Acikgoz A, Kademoglu O, Elekdag-Türk S, Karagoz F (2007). Hypohidrotic Chassaing N, Bourthoumieu S, Cossee M, Calvas P, Vincent MC (2006).
ectodermal dysplasia with true anodontia of the primary dentition. Mutations in EDAR account for one-quarter of non-ED1-related
Quintessence Int 38:853-858. hypohidrotic ectodermal dysplasia. Hum Mutat 27:255-259.
Alcan T, Basa S, Kargul B (2006). Growth analysis of a patient with Courtney JM, Blackburn J, Sharpe PT (2005). The Ectodysplasin and
ectodermal dysplasia treated with endosseous implants: 6-year follow- NFkappaB signalling pathways in odontogenesis. Arch Oral Biol
up. J Oral Rehabil 33:175-182. 50:159-163.
Aradhya S, Nelson D (2001). NF-kappaB signaling and human disease. Curr Courtois G, Smahi A, Israel A (2001). NEMO/IKK gamma linking
Opin Genet Dev 11:300-306. NF-kappa B to human disease. Trends Mol Med 7:427-430.
Arslan S, Kama J, Ozer T, Yavuz I (2007). Craniofacial and upper airway Crawford PJ, Aldred MJ, Clarke A (1991). Clinical and radiographic dental
cephalometric in hypohidrotic ectodermal dysplasia. Dentomaxillofac findings in X linked hypohidrotic ectodermal dysplasia. J Med Genet
Radiol 36:478-483. 28:181-185.
Arte S, Nieminen P, Apajalahti S, Haavikko K, Thesleff I, Pirinen S (2001). Crump JG, Maves L, Lawson ND, Weinstein BM, Kimmel CB (2004). An
Characteristics of incisor-premolar hypodontia in families. J Dent Res essential role for Fgf in endodermal pouch formation influences later
80:1445-1450. craniofacial skeletal patterning. Development 131:5703-5716.
Asagiri M, Takayanagi H (2007). The molecular understanding of osteoclast Cui CY, Durmowicz M, Tanaka TS, Hartung AJ, Tezuka T, Hashimoto K,
differentiation. Bone 40:251-264. et al. (2002). EDA targets revealed by skin gene expression profiles of
Ashique AM, Fu K, Richman J (2002). Signalling via type IA and wild-type, Tabby and Tabby EDA-A1 transgenic mice. Hum Mol Genet
type IB bone morphogenetic protein receptors (BMPR) regulates 11:1763-1773.
intramembranous bone formation, chondrogenesis and feather formation Dellavia C, Sforza C, Malerba A, Strohmenger L, Ferrario VF (2006).
in the chicken embryo. Int J Dev Biol 46:243-253. Palatal size and shape in 6-year old affected by hypohidrotic ectodermal
Aswegan AL, Josephson KD, Mowbray R, Pauli RM, Spritz RA, Williams dysplasia. Angle Orthod 76:978-983.
MS (1997). Autosomal dominant hypohidrotic ectodermal dysplasia in Dupuis-Girod S, Corradini N, Hadj-Rabia S, Fournet JC, Faivre L, Le Deist
a large family. Am J Med Genet 72:462-467. F, et al. (2002). Osteopetrosis, lymphedema, anhidrotic ectodermal
Bal E, Baala L, Cluzeau C, El Kerch F, Ouldim K, Hadj-Rabia S, et al. dysplasia, and immune-deficiency in a boy and incontinentia pigmenti
(2007). Autosomal dominant anhidrotic ectodermal dysplasias at the in his mother. Pediatrics 109:97-103.
EDARADD locus. Hum Mutat 28:703-709. Ferguson BM, Thomas NS, Munoz F, Morgan D, Clarke A, Zonana J
Barberia E, Saavedra D, Arenas M, Maroto M (2006). Multiple agenesis (1998). Scarcity of mutations detected in families with X linked
and anhidrotic ectodermal dysplasia: a comparative longitudinal study hypohidrotic ectodermal dysplasia: diagnostic implications. J Med
of dental similarities and genetic differences in two groups of children. Genet 35:112-115.
Eur J Paediatr Dent 7:113-121. Ferrario V, Dellavia C, Serrao G, Sforza C (2004). Soft-tissue facial areas
Barlow AJ, Bogardi JP, Ladher R, Francis-West PH (1999). Expression and volumes in individuals with ectodermal dysplasia. Am J Med Genet
of chick Barx-1 and its differential regulation by FGF-8 and BMP A 126:253-260.
signaling in the maxillary primordia. Dev Dyn 214:291-302. Franchi L, Branchi R, Tollaro I (1998). Craniofacial changes following early
Basch ML, Bronner-Fraser M (2006). Neural crest inducing signals. Adv prosthetic treatment in a case of hypohidrotic ectodermal dysplasia with
Exp Med Biol 589:24-31. complete anodontia. ASDC J Dent Child 65:116-121.
Becktor KB, Becktor JP, Keller EE (2001). Growth analysis of a patient Franzoso G, Carlson L, Xing L, Poljac L, Shores E, Brown K, et al. (1997).
with ectodermal dysplasia treated with endosseous implants: a case Requirement for NF-kB in osteoclast and B-cell development. Genes
report. Int J Oral Maxillofac Implants 16: 864-874. Dev 11:3482-3496.
Bei M, Maas R (1998). FGFs and BMP4 induce both Msx-1-independent Fu Q, Jilka RL, Manolagas SC, O’Brien CA (2002). Parathyroid hormone
and Msx-1-dependent signalling pathways in early tooth development. stimulates receptor activator of NFkappa B ligand and inhibits
Development 125: 4325-4333. osteoprotegerin expression via protein kinase A activation of cAMP-
Bei M, Kratochwil K, Maas RL (2000). BMP4 rescues a non-cell- response element-binding protein. J Biol Chem 277:48868-48875.
autonomous function of Msx1 in tooth development. Development Gaide O, Schneider P (2003). Permanent correction of an inherited
127:4711-4718. ectodermal dysplasia with recombinant EDA. Nat Med 9:614-618.
Beliakov A, Elizarova VM, Smirnova TA, Krotov V (1998). Primary Galibert L, Tometsko ME, Anderson DM, Cosman D, Dougall WC (1998).
anodontia in X-linked hypohidrotic ectodermal dysplasia. Stomatologia The involvement of multiple Tumor Necrosis Factor (TNFR)-associated
77:29-31. factors in the signaling mechanisms of Receptor Activator of NF-kB, a
Boj JR, Duran J, Cortada M, Jimenez A, Golobart J (1993). Cephalometric member of the TNFR superfamily. J Biol Chem 273:34120-34127.
changes in a patient with ectodermal dysplasia after placement of Glavina D, Majstorović M, Lulić-Dukić O, Jurić H (2001). Hypohidrotic
dentures. J Clin Pediatr Dent 17:217-220. ectodermal dysplasia: dental features and carriers detection. Coll
Bondarets N, Jones RM, McDonald F (2002). Analysis of facial growth in Antropol 25:303-310.
subjects with syndromic ectodermal dysplasia: a longitudinal analysis. Guckes A, Roberts M, McCarthy G (1998). Pattern of permanent teeth
Orthod Craniofac Res 5:71-84. present in individual with ectodermal dysplasia and severe hypodontia
Bushdid PB, Chen CL, Brantley DM, Yull F, Raghow R, Kerr LD, et suggests treatment with dental implants. Pediatr Dent 20:278-280.
al. (2001). NF-kappaB mediates FGF signal regulation of msx-1 Guckes A, Scurria M, King T, McCarthy G, Brahim J (2002). Prospective
expression. Dev Biol 237:107-115. clinical trial of dental implants in persons with ectodermal dysplasia. J
Cambiaghi S, Restano L, Paakkonen K, Caputo R, Kere J (2000). Clinical Prosthet Dent 88:21-25.
findings in mosaic carriers of hypohidrotic ectodermal dysplasia. Arch Han J, Ito Y, Yeo JY, Sucov HM, Maas R, Chai Y (2003). Cranial neural
Dermatol 136:217-224. crest-derived mesenchymal proliferation is regulated by Msx-1
Casal ML, Lewis JR, Mauldin EA, Tardivel A, Ingold K, Favre M, et al. mediated p19 (INK4d) expression during odontogenesis. Dev Biol
(2007). Significant correction of disease after postnatal administration 261:183-196.
of recombinant ectodysplasin A in a canine X-linked ectodermal Hashiguchi T, Yotsumoto S, Kanzaki T (2003). Mutations in the ED1 gene
dysplasia. Am J Hum Genetic 81:1050-1056. in Japanese families with X-linked hypohidrotic ectodermal dysplasia.
Cascallana JL, Bravo A, Donet E, Leis H, Lara MF, Paramio JM, et Exp Dermatol 12:518-522.
al. (2005). Ectoderm-targeted overexpression of the glucocorticoid Haworth KE, Healy C, Morgan P, Sharpe PT (2004). Regionalisation of
receptor induces hypohidrotic ectodermal dysplasia. Endocrinology early head ectoderm is regulated by endoderm and prepatterns the
146:2629-2638. orofacial epithelium. Development 131:4797-4806.
Chaisson ML, Branstetter DG, Derry JM, Armstrong AP, Tometsko ME, Hill NL, Laib A, Duncan MK (2002). Mutation of the ectodysplasin-A gene
Takeda K, et al. (2004). Osteoclast differentiation is impaired in the results in bone defects in mice. J Comp Pathol 126:220-225.

Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


1098 Clauss et al. J Dent Res 87(12) 2008

Huang C, Yang A, Ke T, Wang H, Wang X, Shen J, et al. (2006). A novel Lexner MO, Bardow A, Bjorn-Joergensen J, Hertz JM, Almer L, Kreiborg S
de-novo frame-shift mutation of the EDA gene in a Chinese Han family (2007b). Anthropometric and cephalometric measurements in X-linked
with hypohidrotic ectodermal dysplasia. J Hum Genet 51:1133-1137. hypohidrotic ectodermal dysplasia. Orthod Craniofac Res 10:203-215.
Isgaard J, Nilsson A, Lindahl A, Jansson JO, Isaksson OG (1986). Effects of Lidral AC, Reising BC (2002). The role of MSX1 in human tooth agenesis. J
local administration of GH and IGF-1 on longitudinal bone growth in Dent Res 81:274-278.
rats. Am J Physiol 250(4 Pt 1):367-372. Lin TK, Huang CY, Lin MH, Chao SC (2004). A novel 7-bp deletion
Ishimura A, Maeda R, Takeda M, Kikkawa M, Daar I, Maeno M (2000). mutation in a Taiwanese family with X-linked hypohidrotic ectodermal
Involvement of BMP-4/Msx-1 and FGF pathways in neural induction dysplasia. Clin Exp Dermatol 29:536-538.
in the xenopus embryo. Dev Growth Differ 42:307-316. Lind LK, Stecksén-Blicks C, Lejon K, Schmitt-Egenolf M (2006). EDAR
Itin PH, Fistarol SK (2004). Ectodermal dysplasias. Am J Med Genet C mutation in autosomal dominant hypohidrotic ectodermal dysplasia in
Semin Med Genet 131:45-51. two Swedish families. BMC Med Genet 7:80.
Jernvall J, Kettunen P, Karavanova I, Martin LB, Thesleff I (1994). Evidence Lomaga MA, Yeh WC, Sarosi I, Duncan GS, Furlonger C, Ho A, et
for the role of the enamel knot as a control center in mammalian tooth al. (1999). TRAF6 deficiency results in osteopetrosis and defective
cusp formation: non-dividing cells express growth stimulating Fgf-4 interleukin-1, CD40, and LPS signaling. Genes Dev 13:1015-1024.
gene. Int J Dev Biol 38:463-469. Meyers EN, Lewandoski M, Martin GR (1998). An Fgf8 mutant allelic
Johnson EL, Roberts MW, Guckes AD, Bailey LJ, Phillips CL, Wright series generated by Cre- and Flp- mediated recombination. Nat Genet
JT (2002). Analysis of craniofacial development in children with 18:136-141.
hypohidrotic ectodermal dysplasia. Am J Med Genet 112:327-334. Mikkola ML, Pispa J, Pekkanen M, Paulin L, Nieminen P, Kere J, et al.
Jorgenson RJ (1982). The conditions manifesting taurodontism. Am J Med (1999). Ectodysplasin, a protein required for epithelial morphogenesis,
Genet 11:435-442. is a novel TNF homologue and promotes cell-matrix adhesion. Mech
Jowett AK, Vainio S, Ferguson MW, Sharpe PT, Thesleff I (1993). Dev 88:133-146.
Epithelial-mesenchymal interactions are required for msx 1 and msx Monreal AW, Zonana J, Ferguson B (1998). Identification of a new splice
2 gene expression in the developing murine molar tooth. Development form of the EDA1 gene permits detection of nearly all X-linked
117:461-470. hypohidrotic ectodermal dysplasia mutations. Am J Hum Genet 63:380-
Kanzler B, Foreman R, Labosky P, Mallo M (2000). BMP signaling is 389; erratum in Am J Hum Genet 63:1253-1255, 1998.
essential for development of skeletogenic and neurogenic cranial neural Montonen O, Ezer S, Saarialho-Kere UK, Herva R, Karjalainen-Lindsberg
crest. Development 127:1096-1104. ML, Kaitila I, et al. (1998). The gene defective in anhidrotic ectodermal
Kearns G, Sharma A, Perrott D, Schmidt B, Kaban L, Vargervik K (1999). dysplasia is expressed in the developing epithelium, neuroectoderm,
Placement of endosseous implants in children and adolescents with thymus, and bone. J Histochem Cytochem 46:281-289.
hereditary ectodermal dysplasia. Oral Surg Oral Med Oral Pathol Oral Morlon A, Munnich A, Smahi A (2005). TAB2, TRAF6 and TAK1 are
Radiol Endod 88:5-10. involved in NF-kappaB activation induced by the TNF-receptor, Edar
Kere J, Srivastava AK, Montonen O, Zonana J, Thomas N, Ferguson B, et and its adaptator Edaradd. Hum Mol Genet 14:3751-3757.
al. (1996). X-linked anhidrotic (hypohidrotic) ectodermal dysplasia Motil K, Fete T, Fraley J, Schultz R, Foy T, Ochs U, et al. (2005). Growth
is caused by mutation in a novel transmembrane protein. Nat Genet characteristics of children with ectodermal dysplasia syndromes.
13:409-416. Pediatrics 116:229-234.
Kettunen P, Thesleff I (1998). Expression and function of FGFs-4, -8, and Na GY, Kim DW, Lee SJ, Chung SL, Park DJ, Kim JC, et al. (2004).
-9 suggest functional redundancy and repetitive use as epithelial signals Mutation in the ED1 gene, Ala349Thr, in a Korean patient with
during tooth morphogenesis. Dev Dyn 211:256-268. X-linked hypohidrotic ectodermal dysplasia developing de novo.
Knight RD, Schilling TF (2006). Cranial neural crest and development of Pediatr Dermatol 21:568-572.
the head skeleton. Adv Exp Med Biol 589:120-133. Nie X, Luukko K, Kettunen P (2006). FGF signalling in craniofacial
Kobielak A, Kobielak K, Wisniewski SA, Midro AT, Trzeciak WH (2001a). development and developmental disorders. Oral Dis 12:102-111.
Sequence polymorphisms of the EDA and the DL genes in the patients Nishibu A, Hashiguchi T, Yotsumoto S, Takahashi M, Nakamura K,
with an X-linked and an autosomal forms of anhidrotic ectodermal Kanzaki T, et al. (2003). Frameshift mutation of the ED1 gene in
dysplasia. Folia Histochem Cytobiol 39:113-114. sibling cases with X-linked hypohidrotic ectodermal dysplasia.
Kobielak K, Kobielak A, Roszkiewicz J, Wierzba J, Limon J, Trzeciak WH Dermatology 207:178-181.
(2001b). Mutations in the EDA gene in three unrelated families reveal Nordgarden H, Reintoft I, Nolting D, Fischer-Hansen, Kjaer I (2001a).
no apparent correlation between phenotype and genotype in the patients Craniofacial tissues including tooth buds in fetal hypohidrotic
with an X-linked anhidrotic ectodermal dysplasia. Am J Med Genet ectodermal dysplasia. Oral Dis 7:163-170.
100:191-197. Nordgarden H, Jensen JL, Storhaug K (2001b). Oligodontia is associated
Kramer F, Baethge C, Tschernitschek H (2007). Implants in children with with extra-oral ectodermal symptoms and low whole salivary flow
ectodermal dysplasia: a case report and literature review. Clin Oral rates. Oral Dis 7:226-232.
Impl Res 18:140-146. Ogawa T, Kapadia H, Feng JQ, Raghow R, Peters H, D’Souza RN (2006).
Kurihara N, Hiruma Y, Zhou H, Subler MA, Dempster DW, Singer FR, Functional consequences of interactions between Pax9 and Msx1
et al. (2007). Mutation of the sequestosome 1 (p62) gene increases genes in normal and abnormal tooth development. J Biol Chem
osteoclastogenesis but does not induce Paget disease. J Clin Invest 281:18363-18369.
117:133-142. Pääkkönen K, Cambiaghi S, Novelli G, Ouzts LV, Penttinen M, Kere J,
Lamartine J (2003). Towards a new classification of ectodermal dysplasias. et al. (2001). The mutation spectrum of the EDA gene in X-linked
Clin Exp Dermatol 28:351-355. anhidrotic ectodermal dysplasia. Hum Mutat 17:349.
Laurikkala J, Mikkola M, Mustonen T, Aberg T, Koppinen P, Pispa J, et al. Pinheiro M, Freire-Maia N (1994). Ectodermal dysplasias: a clinical
(2001). TNF signaling via the ligand-receptor pair ectodysplasin and classification and a causal review. Am J Med Genet 53:153-162.
edar controls the function of epithelial signaling centers and is regulated Pispa J, Jung HS, Jernvall J, Kettunen P, Mustonen T, Tabata MJ, et al.
by Wnt and activin during tooth organogenesis. Dev Biol 229:443-455. (1999). Cusp patterning defect in Tabby mouse teeth and its partial
Lekholm U (1986). Osseointegrated implants in clinical practice. J Oral rescue by FGF. Dev Biol 216:521-534.
Implantol 12:357-364. Pispa J, Mustonen T, Mikkola ML, Kangas AT, Koppinen P, Lukinmaa
Levi G, Mantero S, Barbieri O, Cantatore D, Paleari L, Beverdam A, PL, et al. (2004). Tooth patterning and enamel formation can be
et al. (2006). Msx1 and Dlx5 act independently in development of manipulated by misexpression of TNF receptor Edar. Dev Dyn
craniofacial skeleton, but converge on the regulation of Bmp signaling 231:432-440.
in palate formation. Mech Dev 123:3-16. Prager TM, Finke C, Miethke RR (2006). Dental findings in patients with
Lexner MO, Bardow A, Hertz JM, Nielsen LA, Kreiborg S (2007a). ectodermal dysplasia. J Orofac Orthop 67:347-355.
Anomalies of tooth formation in hypohidrotic ectodermal dysplasia. Int Priolo M, Lagana C (2001). Ectodermal dysplasias: a new clinical-genetic
J Paediatr Dent 17:10-18. classification. J Med Genet 38:579-585.

Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research


J Dent Res 87(12) 2008 Dento-Craniofacial Phenotypes in HED 1099

Pummila M, Fliniaux I, Jaatinen R, James MJ, Laurikkala J, Schneider P, et and sweat glands in Tabby mice. Hum Mol Genet 10:2973-2981.
al. (2007). Ectodysplasin has a dual role in ectodermal organogenesis: Tariq M, Wasif N, Ayub M, Ahmad W (2007a). A novel 4-bp insertion
inhibition of Bmp activity and induction of Shh expression. in EDA1 gene in a Pakistani family with X-linked hypohidrotic
Development 134:117-125. ectodermal dysplasia. Eur J Dermatol 17:209-212.
Raible DW (2006). Development of the neural crest: achieving specificity Tariq M, Wasif N, Ahmad W (2007b). A novel deletion mutation in
in regulatory pathways. Curr Opin Cell Biol 18:698-703. the EDAR gene in a Pakistani family with autosomal recessive
RamaDevi AR, Reddy EC, Ranjan S, Bashyam MD (2008). Molecular hypohidrotic ectodermal dysplasia. Br J Dermatol 157:207-209.
genetic analysis of patients from India with hypohidrotic ectodermal Tarjan I, Gabris K, Rozsa N (2005). Early prosthetic treatment of patients
dysplasia reveals novel mutations in the EDA and EDAR genes. Br J with ectodermal dysplasia: a clinical report. J Prosthet Dent 93:419-
Dermatol 158:163-167. 424.
Ramirez-Yanez GO, Smid JR, Young WG, Waters MJ (2005). Influence of Thomas BL, Sharpe PT (1998). Patterning of the murine dentition by
growth hormone on the craniofacial complex of transgenic mice. Eur J homeobox genes. Eur J Oral Sci 106(Suppl 1):48-54.
Orthod 27:494-500. Tribulo C, Aybar M, Nguyen V, Mullins M, Mayor R (2003). Regulation of
Ramos C, Robert B (2005). msh/Msx gene family in neural development. Msx genes by a BMP gradient is essential for neural crest specification.
Trends Genet 21:624-632. Development 130:6441-6452.
Riolo ML (1974). An atlas of craniofacial growth. Michigan: Center for Tucker AS, Al Khamis A, Sharpe PT (1998). Interactions between Bmp-4
Human Growth and Development, 379 pp. and Msx-1 act to restrict gene expression to odontogenic mesenchyme.
Risnes S, Peterkova R, Lesot H (2005). Distribution and structure of dental Dev Dyn 212:533-539.
enamel in incisors of Tabby mice. Arch Oral Biol 50:181-184. Tucker AS, Headon DJ, Courtney JM, Overbeek P, Sharpe PT (2004.)
Ruhin B, Martinot V, Lafforgue P, Catteau B, Manouvrier-Hanu S, Ferri J The activation level of the TNF family receptor, Edar, determines
(2001). Pure ectodermal dysplasia: retrospective study of 16 cases and cusp number and tooth number during tooth development. Dev Biol
literature review. Cleft Palate Craniofac J 38:504-518. 268:185-194.
Saksena SS, Bixler D (1990). Facial morphometrics in the identification Umberto G, Maiorana C, Ghiglione V, Marzo G, Santoro F, Szabo G
of gene carriers of X-linked hypohidrotic ectodermal dysplasia. Am J (2007). Osseointegration and guided bone regeneration in ectodermal
Med Genet 35:105-114 dysplasia patients. J Craniofac Surg 18:1296-1304.
Sasaki T, Ito Y, Bringas P Jr, Chou S, Urata MM, Slavkin H, et al. (2006). Van der Hout AH, Oudesluijs GG, Venema A, Verheij JB, Mol BG, Rump
TGFb-mediated FGF signaling is crucial for regulating cranial neural B, et al. (2008). Mutation screening of the Ectodysplasin-A receptor
crest cell proliferation during frontal bone development. Development gene EDAR in hypohidrotic ectodermal dysplasia. Eur J Hum Genet
133:371-381. 16:673-679.
Schmidt-Ullrich R, Tobin DJ, Lenhard D, Schneider P, Paus R, Scheidereit Van Erum R, Mulier M, Carels C, Verbeke G, de Zegher F (1997).
C (2006). NF-kappaB transmits Eda A1/EdaR signalling to activate Craniofacial growth in short children born small for gestational age:
Shh and cyclin D1 expression, and controls post-initiation hair placode effect of growth hormone treatment. J Dent Res 76:1579-1586.
down growth. Development 133:1045-1057. Vastardis H, Karimbux N, Guthua SW, Seidman JG, Seidman CE (1996).
Schneider P, Street SL, Gaide O, Hertig S, Tardivel A, Tschopp J, et A human MSX1 homeodomain missense mutation causes selective
al. (2001). Mutations leading to X-linked hypohidrotic ectodermal tooth agenesis. Nat Genet 13:417-421.
dysplasia affect three major functional domains in the tumor necrosis Vierucci S, Baccetti T, Tollaro I (1994). Dental and craniofacial findings in
factor family member ectodysplasin-A. J Biol Chem 276:18819-18827. hypohidrotic ectodermal dysplasia during the primary dentition phase.
Shigetani Y, Nobusada Y, Kuratani S (2000). Ectodermally derived FGF8 J Clin Pediatr Dent 18:291-297.
defines the maxillomandibular region in the early chick embryo: Vincent MC, Biancalana V, Ginisty D, Mandel JL, Calvas P (2001).
epithelial-mesenchymal interactions in the specification of the Mutational spectrum of the ED1 gene in X-linked hypohidrotic
craniofacial ectomesenchyme. Dev Biol 228:73-85. ectodermal dysplasia. Eur J Hum Genet 9:355-363.
Skrinjarić I, Skrinjarić K, Vranić DN, Majstorović M, Glavina D (2003). Vinolo E, Sebban H, Chaffotte A, Israël A, Courtois G, Véron M, et
Craniofacial anthropometric pattern profile in hypohidrotic ectodermal al. (2006). A point mutation in NEMO associated with anhidrotic
dysplasia-application in detection of gene carriers. Coll Antropol ecto­dermal dysplasia with immunodeficiency pathology results in
27:753-759. destabilization of the oligomer and reduces lipopolysaccharide- and
Smahi A, Courtois G, Rabia SH, Doffinger R, Bodemer C, Munnich A, Tumor Necrosis Factor-mediated NF-kappaB activation. J Biol Chem
et al. (2002). The NF-kappaB signalling pathway in human diseases: 281:6334-6348.
from incontinentia pigmenti to ectodermal dysplasias and immune- Visinoni AF, De Souza RL, Freire-Maia N, Gollop TR, Chautard-Freire-
deficiency syndromes. Hum Mol Genet 11:2371-2375. Maia EA (2003). X-linked hypohidrotic ectodermal dysplasia
Söderholm AL, Kaitila I (1985). Expression of X-linked hypohidrotic mutations in Brazilian families. Am J Med Genet A 122:51-55.
ectodermal dysplasia in six males and in their mothers. Clin Genet Yavuz I, Baskan Z, Ulku R, Dulgergil TC, Dari O, Ece A, et al. (2006).
28:136-144. Ectodermal dysplasia: retrospective study of fifteen cases. Arch Med
Spfaer JA (1981). A dental approach to carrier screening in X linked Res 37:403-409.
hypohidrotic ectodermal dysplasia. J Med Genet 18:459-460. Zhang Y, Wang S, Song Y, Han J, Chai Y, Chen Y (2003). Timing of
Srivastava A, Durmowicz M, Hartung A, Hudson J, Ouzts L, Donovan D, odontogenic neural crest cell migration and tooth-forming capability in
et al. (2001). Ectodysplasin-A1 is sufficient to rescue both hair growth mice. Dev Dyn 226:713-718.

Downloaded from jdr.sagepub.com at BRIGHAM YOUNG UNIV on January 27, 2015 For personal use only. No other uses without permission.

International and American Associations for Dental Research

You might also like