You are on page 1of 9

bs_bs_banner

doi:10.1111/cga.12072 Congenital Anomalies 2015; 55, 17–25 17

REVIEW ARTICLE

Signaling regulating inner ear development: Cell fate determination,


patterning, morphogenesis, and defects
Yuji Nakajima
Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan

ABSTRACT The membranous labyrinth of the inner ear (cranial nerve VIII). During organogenesis, reciprocal signaling
is a highly complex organ that detects sound and balance. between the embryonic tissues successively occurs to establish
Developmental defects in the inner ear cause congenital hearing highly organized functional tissues and organs in a spatiotemporal-
loss and balance disorders. The membranous labyrinth consists restricted manner. Developmental arrest/alterations as well as
of three semicircular ducts, the utricle, saccule, and acquired injuries in the inner ear cause functional disorders for
endolymphatic ducts, and the cochlear duct. These complex hearing and balance. The aim of this review is to discuss signaling
structures develop from the simple otic placode, which is estab- that regulates the key developmental events responsible for the
lished in the cranial ectoderm adjacent to the neural crest at establishment of the complicated membranous labyrinth. Several
the level of the hindbrain at the early neurula stage. During excellent reviews on inner ear development have been published
development, the otic placode invaginates to form the otic (Torres and Giraldez 1998; Abelló and Alsina 2007; Bok et al.
vesicle, which subsequently gives rise to neurons for the 2007a; Driver and Kelley 2009; Groves and Fekete 2012;
vestibulocochlear ganglion, the non-sensory and sensory epithe- Magarinos et al. 2012; Neves et al. 2013).
lia of the membranous labyrinth that includes three ampullary
crests, two maculae, and the organ of Corti. Combined
paracrine and autocrine signals including fibroblast growth EMBRYOGENESIS OF THE INNER EAR AND
factor, Wnt, retinoic acid, hedgehog, and bone morphogenetic ITS MALFORMATIONS
protein regulate fate determination, axis formation, and
The membranous labyrinth and vestibulocochlear neurons are
morphogenesis in the developing inner ear. Juxtacrine signals
derived from an ectodermal thickening named the otic placode
mediated by Notch pathways play a role in establishing the
(OP), which develops from the cranial ectoderm immediately
sensory epithelium, which consists of mechanosensory hair cells
lateral to the neural crest at the level of the hindbrain at the neurula
and supporting cells. The highly differentiated organ of Corti,
stage. The OP invaginates and is then being pinched off from the
which consists of uniformly oriented inner/outer hair cells and
ectoderm, which results in the formation of the otic vesicle
specific supporting cells, develops during fetal development.
(otocyst). The posterior rim and its anterior domain of the OP give
Developmental alterations/arrest causes congenital malforma-
rise to non-sensory semicircular and cochlear epithelia, respectively
tions in the inner ear in a spatiotemporal-restricted manner. A
(Abelló et al. 2007). Epithelial cells in the anteromedial domain of
clearer understanding of the mechanisms underlying inner ear
the OP/otic vesicle further differentiate to the proneurosensory epi-
development is important not only for the management of
thelium, from which neural progenitors delaminate and give rise to
patients with congenital inner ear malformations, but also for
neuroblasts of the vestibulocochlear ganglion (Satoh and Fekete
the development of regenerative therapy for impaired function.
2005). After neural cell migration, the resulting prosensory domain
Key Words: development, growth factors, inner ear, malforma- later develops into five vestibular sensory patches and the organ of
tions, Notch Corti (basilar papilla in birds). The dorsal epithelium of the otic
vesicle expands to form the vertical pouch; its opposing epithelia at
the future anterior and posterior semicircular ducts later fuse to be
INTRODUCTION absorbed resulting in the formation of two superior semicircular
ducts and the common crus (Fig. 1, Martin and Swanson 1993;
The inner ear exists in the petrous part of the temporal bone and Chang et al. 2004). The lateral semicircular duct develops from the
consists of a membranous labyrinth and bony labyrinth. The mem- lateral pouch and the endolymphatic duct from the dorsomedial
branous labyrinth consists of at least six mechanosensory epithelia, epithelium of the otic vesicle. The five sensory patches that develop
which include the organ of Corti in the cochlea for sound, three in the vestibular labyrinth are three cristae in the anterior, posterior,
ampullary crests in the base of three semicircular ducts for angular and lateral ampulla; and two maculae in the utricle and saccule.
acceleration, and two maculae, macula utriculi and sacculi, for These cristae detect head rotation while the maculae maintain static
gravity/linear acceleration. Electrical signals, which are translated equilibrium. The organ of Corti, which develops specifically in the
from the mechanical signals evoked in these sensory epithelia, are mammalian cochlea, is a highly differentiated sensory epithelium
transmitted to the brain stem via the vestibulocochlear nerve for sound. The sensory epithelium basically consists of sensory hair
cells and supporting cells, while highly differentiated sensory and
Correspondence: Yuji Nakajima, MD, PhD, Department of Anatomy and supporting cells develop in the organ of Corti, that is, three lateral
Cell Biology, Graduate School of Medicine, Osaka City University, 1-4-3 rows of outer hair cells and a medial row of inner hair cells, with
Asahimachi, Abenoku, Osaka, 545-8585 Japan. Email: yuji@med.osaka- inner phalangeal cells (supporting inner hair cells), outer Deiters’
cu.ac.jp cells (supporting outer hair cells), and pillar cells (demarcating the
Received April 5, 2014; revised and accepted June 7, 2014. inner and outer hair cells and making the tunnel of Corti).

© 2014 Japanese Teratology Society


18 Y. Nakajima

Fig. 1 Inner ear development and corresponding malformations. Modified from Jackler et al. (1987) and Yasuda et al. (2007). an, anterior semicircular duct;
c, cochlear duct; CS, Carnegie stage; SCC, semicircular canal (duct); e, endolymphatic duct; l, lateral SCC; p, posterior SCC; pl, primordium of the
lateral semicircular duct; v, vestibular pouch; W, age in weeks. Note that the superior SCC includes the anterior SCC and the posterior SCC.

Table 1 Classification of inner ear malformations based on PREPLACODE REGION


embryogenesis
The sensory nervous system in the head is derived from both the
Complete labyrinth aplasia (Michel) neural crest and placodes, while that in the trunk is derived from
the neural crest alone. At the late gastrula to early neurula stage, the
Cochlear malformations
developing ectoderm is subdivided into at least four distinct
Aplasia regions: the neural plate, neural crest, and epidermal ectoderm from
Common cavity the head to the trunk; and preplacode region (PPR) in only the head
Hypoplasia ectoderm. Therefore, the establishment of PPR may be precisely
regulated by surrounding tissues in a spatiotemporal-restricted
Incomplete partition (Mondini)
manner. Signals derived from the head and lateral mesoderm
Semicircular canal malformations (including the heart-forming mesoderm) are important for defining
Aplasia and establishing the PPR adjacent to the neural crest at the cranial
Small buds region (Fig. 2, Litsiou et al. 2005). Excess bone morphogenetic
Superior and lateral SCC dysplasia protein (BMP) and Wnt signals, which are secreted from the ecto-
derm and neural plate, respectively, act to suppress the expression
Lateral SCC aplasia/dysplasia of PPR marker genes, such as Six1, Six4, and Eya2, in the PPR-
Enlarged vestibular aqueduct forming ectoderm. Together with the BMP/Wnt signals, Cerberus
(BMP/Wnt antagonist) and fibroblast growth factor (FGF), which
Note that the superior semicircular canal (SCC) includes the are secreted from the mesoderm subjacent to the prospective PPR,
anterior SCC and the posterior SCC. The most common cochlear upregulate the expression of PPR genes (Fig. 2, Litsiou et al. 2005).
malformation is incomplete partition, and the most frequently diag- The anterior-posterior patterning of the PPR is regulated by mutual
nosed SCC malformation is lateral SCC dysplasia (Jackler et al. repression between Gbx2 and Otx2, that is, posteriorly localized
1987; Brenski and Arjmand 2003). Gbx2 is required for the formation of the OP, while anterior Otx2 is
required for that of the olfactory, lens, and trigeminal placode in the
anterior PPR (Steventon et al. 2012).
Developmental arrest in the membranous labyrinth leads to Six, Dach, and Eya are expressed in developing organs including
various congenital inner ear defects in temporally and spatially ear and eye (Li et al. 2003; Zou et al. 2004). The Six1 protein acts
restricted manners. These inner ear malformations include cochlear, as both a repressor and activator in a context-dependent manner.
semicircular canal, aqueduct, and combined malformations. Based The Six-Dach complex without Eya acts to repress target genes,
on embryogenesis and radiological findings, cochlear malforma- while the Six-Dach-Eya complex, in which Eya exhibits phos-
tions have been classified as aplasia, common cavity, hypoplasia, phatase activity to suppress the co-repressor complex, recruits
and incomplete partition, while semicircular canal malformations the co-activator to activate target genes for the promotion of
have been classified as aplasia, small buds, dysplasia of the three organogenesis (Li et al. 2003). Development of the inner ear, nose,
semicircular canals, and dysplasia of the lateral semicircular canal skeletal muscle, thymus, and kidney was shown to be severely
(Fig. 1, Table 1, Jackler et al. 1987; Sennaroglu and Saatci 2002; affected in Six1-null mice, (Ozaki et al. 2003). In this mutant, the
Brenski and Arjmand 2003). Developmental defects/arrest in the vestibular labyrinth is a single space with an expanded
inner ear and its surrounding structures are often diagnosed in endolymphatic duct, and ventral structures (cochlea and
children with sensorineural hearing loss; therefore, an accurate vestibulocochlear ganglion) are absent (Ozaki et al. 2003). A pre-
morphological assessment of the inner ear by high-resolution CT vious study reported that the initial cell fate determination and
(computed tomography) is important (Masuda et al. 2013). migration of vestibulocochlear neurons were unaffected in Eya- and

© 2014 Japanese Teratology Society


Inner ear development 19

Fig. 3 Signals regulating otic placode (OP) formation. Head mesoderm-


secreted FGF, the expression of which is induced by the pharyngeal
endoderm (Pha) FGF8, defines the posterior preplacode region
(PPR, pink) and upregulates the expression of Wnt8a and FGF in
the hindbrain (HB). Anterior hindbrain-derived FGF and Wnt8a
Fig. 2 Signals regulating the preplacode region (PPR). In the late gastrula specify the PPR to OP (green). Wnt8a suppresses the epibranchial
to early neurula embryo, PPR (pink) is determined in the head placode (Epi, purple) gene, Foxi2, to demarcate the area of the OP.
ectoderm (Ect), which is immediately lateral to the neural crest
(NCr, green) and dorsal to the heart/head mesoderm. Excess bone
morphogenetic protein (BMP) and Wnt signals, which are derived
from the ectoderm (yellow) and neural plate (NP, blue), respec-
tively, suppress PPR genes. Fibroblast growth factor (FGF) and Fgf3 is expressed in the hindbrain adjacent to the prospective OP
Cerberus (Cer, a BMP/Wnt antagonist), which are secreted from the at the level of rhombomere 4–6, as well as in the OP itself, at the
heart/head mesoderm, can upregulate the expression of PPR genes. onset of OP formation in the mouse. Fgf10 was shown to be
expressed in the head mesenchyme underlying the prospective
OP-forming ectoderm (Wright and Mansour 2003; Alvarez et al.
Six1-null mutant mice; however, early neurogenesis was affected in 2003). Gain-of-function experiments demonstrated that the ectopic
the mutant inner ear (Zou et al. 2004). Genes expressed in the expression of Fgf10 in the hindbrain was capable of inducing an
ventral otic vesicle of the Six1-null developing inner ear, including OP-like structure, whereas Fgf3 only exhibited faint inducible
Otx1, Otx2, Lunatic fringe (Lfng), Fgf3, Fgf10, Bmp4, Gata3, and activity (Alvarez et al. 2003). The Fgf10- and Fgf3-null mutants
Nkx5.1, were found to be suppressed (Ozaki et al. 2003; Zou et al. displayed only mild defects during formation of the otic vesicle;
2004). In humans, an impaired EYA1-SIX1 regulatory network is however, double knockout mutant had a hypoplastic otic vesicle, in
one of the candidates involved in branchio-oto-renal syndrome, which the expression of some otic marker genes, such as Pax2,
which is associated with the incomplete partition of the cochlear Dlx5, and Sox9, was absent (Wright and Mansour 2003; Alvarez
duct as well as semicircular canal dysplasia (Propst et al. 2005; et al. 2003). These findings indicated the redundant role of FGFs
Kochhar et al. 2007; Senel et al. 2009). and also that head mesoderm-derived FGF10 acts as one of the
inductive signals for OP formation in the mouse. Human LAMM
(labyrinth aplasia, microtia, and microdontia) syndrome, which is
THE OTIC PLACODE caused by homoallelic mutations in FGF3, presents as a series of
inner ear malformations including complete labyrinth aplasia as
The otic placode (OP) is induced by combined signaling that is well as common cavity and incomplete partitions (Ramsebner et al.
mediated by the head mesoderm and hindbrain in vertebrates 2010).
(Fig. 3). During the inner ear development, mesodermal tissue adja-
cent to the hindbrain expresses FGF, which defines the posterior
PPR (precursor region of OP and epibranchial placode) and induces
ANTERIOR-POSTERIOR PATTERNING
the expression of Wnt8a and FGF in the hindbrain. Signaling medi-
ated by anterior hindbrain-derived Wnt8a and FGF was shown to At the early otic placode/vesicle stage, cells possessing both neu-
involve the differentiation of OP (Ladher et al. 2000, 2010; Urness rogenic and sensory lineages in the anteromedial domain express
et al. 2010; Groves and Fekete 2012). An explantation experiment Fgf10 and Sox3 (Alsina et al. 2004; Abelló et al. 2007; 2010). Later
revealed that endoderm-derived FGF8 acts as an upstream signal of these cells express Lim homeodomain transcription factor, Islet1
FGF in the chick mesoderm (Ladher et al. 2005). and homeobox transcription factor, Prox1 (Stone et al. 2003;
The expression of Fgf10 in the mesoderm subjacent to prospective Radde-Gallwitz et al. 2004). Neurogenin1 and NeuroD are later
OP ectoderm of Fgf3-null and hypomorphic Fgf8 mouse embryo was upregulated in neurogenic cells in this domain. The sensory marker
reduced, which resulted in the failed formation of the OP, suggesting gene Lfng was shown to be co-expressed in this neurogenic epithe-
that FGF8/3 acts as an upstream signal of Fgf10 in the head meso- lium (Cole et al. 2000). Lineage tracing experiments with fluores-
derm during the induction of the OP. Taken together with these cent dye in chick otic cup (16–18 somite stage) clarified that the
findings, pharyngeal endoderm-derived FGF8 plays an upstream anterior domain of the otic cup, posterior rim of the otic cup, and
role in the FGF signaling cascade to induce the OP in both the chick posteromedial ventral region of the otic cup gave rise to
and mouse (Ladher et al. 2005). In addition, hindbrain-derived Wnt neurosensory domain, non-sensory epithelium of the semicircular
signaling was reported to suppress the expression of the epibranchial ducts, and non-sensory epithelium of the cochlear duct, respectively
placode gene, Foxi2, which indicated that Wnt signaling also defined (Abelló et al. 2007). A more detailed lineage analysis revealed
the area of the otic region (Freter et al. 2008) (Fig. 3). that the prosensory region was located more peripheral to the

© 2014 Japanese Teratology Society


20 Y. Nakajima

(Frenz et al. 1996). The most prominent abnormality in this model


was the absence/hypoplasia of the vestibulocochlear ganglion, in
which neural cells are developed from the anterior domain of the
otic vesicle, which suggested that excess levels of RA may alter
anterior positional information to posterior positional information,
resulting in a reduction in neuronal differentiation.

DORSAL-VENTRAL PATTERNING
After development of the otic vesicle is completed, establishing the
dorsal-ventral (DV) axis is necessary to further generate the
complex inner ear morphology consisting of the dorsal vestibular
Fig. 4 Anterior-posterior (AP) axis formation in the otic placode. At the labyrinth and ventral cochlea. The early otic vesicle is located
early otic placode/cup stage, the retinoic acid (RA)-synthesizing closely to the hindbrain (rhombomere 4–6) as well as the midline
enzyme, Raldh2 (retinaldehyde dehydrogenese2), is expressed structure, the notochord. The ablation of the floor plate in the
in the mesoderm posterior to the otic placode (OP), while hindbrain and notochord, from which Shh (Sonic hedgehog) is
RA-catabolizing enzyme (Cyp26) is expressed in the ectoderm, secreted, caused a defective cochlea in chick embryos, and inhibit-
which is anterior to the OP. RA inhibits expression of the anterior
genes of the OP, which suggests that the posterior-to-anterior gra-
ing Shh activity led to the same result (Bok et al. 2005). The
dient of RA regulates the AP axis of the OP. FGF8, which is experimental inversion of the DV axis in the hindbrain with the
expressed in the ectoderm anterior to the placode, upregulates the notochord resulted in an inverted DV axis in the otic vesicle, in
expression of anterior otic genes. Note that a dorsal view of the which the expression of ventral marker genes including NeuroD,
cranial half of the neurula embryo is shown. Lfng, and Six1 is shifted dorsally (Bok et al. 2005). These results
indicated that Shh from the floor plate and notochord plays a critical
role in establishing the DV axis in the inner ear.
The ventral structures of the inner ear, such as the cochlea and
neurogenic region, but also that some neurons and sensory cells
vestibulocochlear ganglion, are absent in Shh-null mutant mice.
developed from common progenitors (Satoh and Fekete 2005).
Ventral markers including Otx1/2 and Pax2 were reported to be
At the early otic cup stage in the chick (11–15 somite stage),
downregulated in this mutant, while the dorsal marker gene Dlx5
experiments demonstrated that AP (anterior–posterior) inversion of
was upregulated, which suggested that the Shh signal may be
the otic cup (epithelium) alone did not affect the expression of
required to specify the ventral structure of the otic vesicle
anterior placode genes, while inversion with the associated preotic
(Riccomagno et al. 2002). The neurogenic markers, Neurogenin1
ectoderm altered the expression of anterior marker genes in the
and NeuroD, were also downregulated; therefore, the
posterior otic cup, which suggested that signaling from tissue sur-
vestibulocochlear ganglion was absent in the mutant inner ear.
rounding the otic cup defined the AP polarity of the otic cup (Bok
However, expression of the sensory markers, Bmp4, Lfng, and Fgf3
et al. 2011). Retinoic acid (RA) is known to regulate the AP axis
in the crests and maculae remained unaltered, which suggested that
in the embryonic body and organs. At the early OP/cup stage in
Shh is required for neurogenic specification, but not for sensory
the chick embryo, the RA-synthesizing enzyme, retinaldehyde
lineage during inner ear development (Riccomagno et al. 2002).
dehydrogenese2 (Raldh2) is expressed in the mesoderm just poste-
Gli3 is one of the transcriptional mediators for the Shh signal and
rior to the otic region, while the P450-associated RA-catabolizing
is thought to act not only as a transcriptional activator, but also as a
enzyme, Cyp26, is expressed in the ectoderm anterior to the OP,
transcriptional repressor that is dependent on Shh protein levels
suggesting an posterior-to-anterior gradient of RA in the developing
(Jacob and Briscoe 2003). Shh-dependent Gli3 functions are
placode. Bead implantation experiments in chick embryos revealed
required for pattern formations during development, such as the DV
that the posterior-to-anterior gradient of RA activity defined, at least
axis in the spinal cord and neocortex in the brain (Komada 2012).
partly, AP polarity in the inner ear (Fig. 4; Bok et al. 2011).
Using several combinations of mutant alleles for Shh, Gli2, and
Pax2 and Sox3 were found to be expressed in the otic and
Gli3, Bok et al. (2007b) reported that in addition to the Gli2/Gli3-
epibranchial ectoderm at the onset of otic development at the 5
dependent activator signal, Gli3-mediated repression was required
somite stage. The expression of Sox3 subsequently becomes
for the morphology of the semicircular ducts (Fig. 5). In addition,
restricted to the anteromedial (neurosensory) domain of the otic
formation of the distal-most cochlear duct at the ventral region
region at the 10 somite stage by FGF8, which is expressed in the
requires a strong Shh activator signal, while the proximal cochlear
ectoderm anterior to the otic ectoderm (Abelló et al. 2010). Sox3
and saccule requires a weaker Shh signal controlled by Gli2-
can induce anterior neurogenic markers, such as Sox2 and Delta1,
mediated activator and Gli3-mediated repressor signals (Bok et al.
and was shown to repress the non-neurogenic posterior gene, Lmx1
2007b).
(Abelló et al. 2010). Therefore, in addition to RA, FGF8 plays a
role in the establishment of AP polarity in the OP (Fig. 4). Once the
neurosensory region is determined in the anterior domain, several
ONSET OF NEURAL AND SENSORY CELL
Notch-related genes are expressed in a region-specific manner; that
DEVELOPMENT
is, Lfng (a Notch modulator), Delta1 (ligand), and Hes5 (repressor)
in the anterior region of the placode, and Serrate1 (ligand) and Neurosensory progenitors develop in the anterior domain of the otic
Hairy1 (Hey1, repressor) in the posterior domain. Notch1 (receptor) cup at the onset of neural development in the inner ear. They express
is expressed in the entire otic region. the HMG-box transcription factor Sox2, which is thought to main-
Various types of cochlear and vestibular malformations as well as tain the multipotency of tissue-specific stem cells including neurons
a defective cartilaginous otic capsule were induced in mouse (Takahashi and Yamanaka 2006). A previous study demonstrated
embryos exposed to maternally administered excess levels of RA that the expression of Sox2 is regulated by a signaling cascade

© 2014 Japanese Teratology Society


Inner ear development 21

Fig. 5 Dorsal-ventral (DV) patterning of the inner ear. The otic vesicle,
which is located adjacent to the hindbrain (HB), gives rise to the
ventral cochlea duct (CD) and dorsal semicircular ducts. At this
time, the floor plate (FP) and notochord (NCh) secrete Shh (Sonic
hedgehog) to establish the DV axis of the inner ear and neural tube.
The Shh signal is converted to Gli2/3 activator and Gli3 repressor
activities in a Shh concentration-dependent manner. ES,
endolymphatic duct; LSCD, lateral semicircular duct; SSCD, supe-
rior semicircular duct; S, saccule; U, utricle
Fig. 6 Neurosensory development. In inner ear development, neural cells
and sensory cells develop from the proneurosensory domain, which
is located in the anteromedial region of the otic vesicle and
including FGF, BMP, Wnt, and Tbx6 (Takemoto 2013). At the onset expresses Sox2. At the onset of neural development, Sox2 initiates
of neural development in the OP, prospective neuroblasts are speci- the expression of Neurogenin1 (Ngn1), which then suppresses Sox2
fied in the neurosensory epithelium and then delaminate to generate and upregulates the expression of NeuroD. Proneural cells also
neurons for the vestibulocochlear ganglion, from which bipolar express the Notch ligand, Delta1 (Dl1), which activates Notch
signaling in the neighboring cells to inhibit a neural fate, thereby
neurons will connect hair cells to the brain via the vestibulocochlear maintaining Sox2-positive prosensory cells by lateral inhibition.
nerve. Delaminated neurons lose their expression of Sox2 as well as Once the prosensory cells are determined, the Notch ligand,
proliferative activity. At the onset of neural determination, prospec- Jagged1 (Jag1), acts to maintain these prosensory cells probably by
tive neuroblasts express Neurogenin1, the expression of which is lateral induction. In some prosensory cells, Sox2 induces Atoh1 for
positively regulated by Sox2. Neurogenin1 has been reported to hair cell differentiation. Prospective hair cells express Dl1, which
repress the expression of Sox2 by a negative feedback loop (Evsen binds to the Notch receptors of neighboring cells to maintain Sox2-
positive supporting cells by lateral inhibition.
et al. 2013). Therefore, negative feedback inhibition to Sox2
by Neurogenin1 (also by NeuroD) is required for progressive
neurogenesis (Fig. 6). In cultured otic vesicles, FGF10 was found to
reduce the proliferation of neurosensory progenitors, and, conse- fashion (Neves et al. 2012). Supporting cells still express Sox2 and
quently, the expansion of NeuroD-expressing neural cells. On the proliferating characteristics, which suggests that Sox2 may be
other hand, SU5402 (FGF receptor inhibitor) stimulates the prolif- involved in the maintenance of stem cell characteristics as well as
eration of neurosensory progenitors, thereby reducing the number regenerative capacity.
of mature neurons. Therefore, the FGF signal acts on the progenitor
cells, resulting in their exit from the cell cycle and commitment to
SEMICIRCULAR DUCTS
a neural fate (Alsina et al. 2004). Neurogenin1 and/or Sox3 also
upregulate the expression of Delta1 (Notch ligand) in neurogenic Three-dimensional morphogenesis has been examined in the inner
cells. This then activates Notch signaling in neighboring cells, ear by a paint-fill method (Bissonnette and Fekete 1996; Morsli
which inhibits Neurogenin1 by Hes-mediated lateral inhibition, et al. 1998; Mansour and Schoenwolf 2005). After completion of
which suppresses neurogenesis and the maintenance of Sox2 the pear-shaped otic vesicle (otocyst), two distinct bulges are
expression, resulting in the preservation of undifferentiated sensory formed, the pars superior (dorsal bulge) and pars inferior (ventral
progenitors (Jeon et al. 2011, Fig. 6). Once the prosensory domains bulge), from which the semicircular ducts/utricle and cochlear duct/
are established, Jagged1 (another Notch ligand) acts to maintain the saccule will later develop, respectively. The endolymphatic duct
expression of Sox2 probably by Hesr-mediated lateral induction simultaneously emerges from the dorsal-medial surface of the otic
(Hayashi et al. 2008a; Neves et al. 2011, Fig. 6). Prosensory pro- vesicle. Two distinct pouches, the dorsal pouch and lateral pouch,
genitors later differentiate to Sox2-negative hair cells and Sox2- subsequently develop from the pars superior. The dorsal pouch
positive supporting cells (Neves et al. 2007). Nascent hair cells gives rise to two superior (vertical) ducts, the anterior and posterior
express Delta1 during this process to accelerate hair cell fate in a semicircular ducts, while the lateral pouch gives rise to the lateral
cell-autonomous manner, while Delta1 activates Notch signaling in (horizontal) semicircular duct. At the onset of semicircular duct
the neighboring cells to suppress Atoh1 via Hes1/5 to maintain formation, the opposing epithelia in the center of each pouch fuse to
supporting cells in a non-cell-autonomous manner (Chrysostomou form the epithelial fusion plate, which is thereafter resorbed by
et al. 2012). Sox2 has been shown to exhibit both positive and epithelial-mesenchymal transition and apoptosis, resulting in the
negative regulatory functions for Atoh1 in a stage-dependent formation of tube-like semicircular ducts (Kobayashi et al. 2008).

© 2014 Japanese Teratology Society


22 Y. Nakajima

Fig. 8 Epithelial differentiation of the organ of Corti. At early mammalian


cochlear development, the medial cochlea epithelium is subdivided
into three distinct regions from the medial (neural) to the lateral
(abneural) site, i.e., Kölliker’s organ (neural side of the cochlear
duct), the prosensory auditory domain, and the outer sulcus. Bmp4
is expressed in the outer epithelial region, and, thus, the establish-
ment of a BMP signaling gradient, which is required to form three
distinct epithelial regions. FGF20, the expression of which is
directly regulated by Notch signals, is required for differentiation of
the auditory sensory epithelium, which consists of hair cells and
supporting cells. Inner hair cell-secreted FGF8 locally controls the
Fig. 7 Semicircular duct development. Wnt/β-catenin signaling stimulates differentiation of pillar cells from FGFR3-positive supporting cells.
the expression of BMP4/FGF10 in the cristae (yellow domain), The Wnt-PCP pathway regulates not only the planar cell polarity of
which then induce the expression of Bmp2 in the prospective semi- the hair cells, but also extension of the cochlear duct.
circular duct (SCD) epithelium. BMP2 induces the expression of
Dlx5 in the perimeter rim of the pouch (future SCD) to inhibit
Netrin1 expression, thereby maintaining the epithelial structure of
the SCD. In the fusion plate of the pouch, Netrin1 is maintained to polarity (PCP) pathways including Rho-associated coiled-coil
expand the surrounding mesenchyme, which then pushes the facing kinase (ROCK) and non-muscle myosin II activity (Yamamoto
epithelia to fuse (fusion plate), resulting in the absorption of the et al. 2009; Fritzsch et al. 2011). The cochlear duct did not grow in
fusion plate. mutant mice in which Shh pathways related to fate determination/
cell proliferation were affected, which resulted in a common cavity
or hypoplastic cochlea with or without lateral semicircular duct
Each semicircular duct connects with the utricle, and the two supe- malformation (Riccomagno et al. 2002; Bok et al. 2007b). A short-
rior (anterior and posterior) ducts possess a common duct, the crus ened cochlea with a larger number of hair cell rows and other inner
membranous commune. The anterior and posterior ends of the ear malformations occur in the Foxg1 mutant, in which embryonic
vertical semicircular ducts (opposite sides of the common crus) and morphogenesis including cell fate determination and proliferation is
the anterior end of the lateral duct have their own ampulla connect- affected (Pauley et al. 2006). Mutant mice, in which the Wnt-PCP
ing to the utricle. A mutant mouse with normal semicircular ducts pathways are affected, were found to have a shortened cochlea with
and no sensory crista has not been yet generated, which suggests a larger number of disorganized hair cell rows (Yamamoto et al.
that the crista is involved in the formation of the semicircular ducts 2009). A shortened cochlea reflects the incomplete partition of
(Chang et al. 2004). In the developing crista (Fig. 7), Wnt/β-catenin cochlea (Mondini) in human inner ear malformations (Table 1).
signaling stimulates the expression of BMP4 and FGF10, which Mice in which the Wnt-PCP pathways were deleted also displayed
then induces the expression of Bmp2 in the prospective semicircular other congenital defects, such as conotruncal heart defects, skeletal
duct epithelium adjacent to the cristae (Chang et al. 1999, 2002, defects, and social abnormalities (Etheridge et al. 2008). Cochlear
2004; Gerlach et al. 2000). BMP2 has been shown to induce the elongation and hair cell development are coordinated in a context-
expression of Dlx5 in the perimeter rim of the canal pouch to inhibit dependent manner because hair cell development was shown to be
the expression of Netrin1, thereby maintaining the epithelial struc- affected in the Aho1 conditional mutant while almost normal
ture of the semicircular ducts (Chang et al. 2004, 2008; Rakowiecki growth was observed in the cochlear duct (Pan et al. 2011). A
and Epstein 2013). The expression of Netrin1 is maintained in the shortened cochlea with a smaller number of turns (Mondini) repre-
fusion plate of the canal pouch in order to expand the surrounding sents a common inner ear malformation diagnosed in children with
mesenchyme, which pushes the center of the epithelial pouch to congenital sensorineural hearing loss, and syndromes associated
generate the fusion plate. The fusion plate will then dissociate via with this type of defect include trisomy 21, DiGeorge, CHARGE,
apoptosis and epithelia-mesenchymal transition, leaving tube-like Pendred, congenital cytomegalovirus infection, and congenital
semicircular ducts (Salminen et al. 2000; Kobayashi et al. 2008). rubella (Brenski and Arjmand 2003).
During early mammalian cochlear development, the medial
thickened cochlea epithelium is subdivided into three distinct
COCHLEA
regions from the medial (neural) to lateral (abneural) site along the
At the onset of cochlear formation, cells in the medioanterior region short axis of the cochlear duct, that is, Kölliker’s organ (neural side
of the otic vesicle proliferate and the cochlear bud elongates to form of the cochlear duct), the prosensory auditory domain, and the outer
the cochlear duct, in which the specific sensory epithelium, the sulcus. At this time, Bmp4 is expressed in the outer sulcus region;
organ of Corti, develops. Once the fate of hair cells is determined, thus, a BMP signal gradient is established in the developing sensory
future hair cells exit the cell cycle; therefore, elongation/growth of domain and is high at the lateral site and low at the neural site
the cochlear duct is thought to occur via convergent extension (Fig. 8, Ohyama et al. 2010). The prosensory domain and outer
movements of cochlear cells, which is regulated by Wnt-planar cell sulcus region did not form in the Alk3-CKO;Alk6+/− mutant, in

© 2014 Japanese Teratology Society


Inner ear development 23

which BMP signals were conditionally deleted in the developing which are unique to the mammalian cochlea (Fig. 8, Colvin et al.
inner ear. In the cultured cochlea, a high dose of the BMP4 protein 1996; Puligilla et al. 2007). Low-frequency sensorineural hearing
was found to upregulate the expression of outer sulcus markers and loss has been reported in a mouse model of human Muenke syn-
downregulate the Kölliker’s organ’s genes, while an intermediate drome carrying Fgfr3P244R, and the organ of Corti in this mutant has
dose induced a large number of prosensory cells (Ohyama et al. not only extra pillar cells and fewer Deiters’ cells, but also extra hair
2010). These findings suggested that BMP signaling with a lateral- cells in the apical cochlear duct (Mansour et al. 2009). A genetic
to-medial gradient is required for the patterning of the cochlear reduction in the expression of FGF10, which normally activates
sensory epithelium along the short axis. FGFR2b or FGFR1b, can reverse the cochlear disorders associated
Previous studies reported defects in hair cells and supporting with this model (Mansour et al. 2013). In the chick developing
cells in the developing cochlear duct of mutant mice with the basilar papilla (homolog of the organ of Corti), FGF signaling
tissue-specific deletion of Fgfr1 or Jagged1-Notch signaling mediated by FGFR3 was shown to be required for maintaining
(Kiernan et al. 2001; Pirvola et al. 2002). The expression of Fgf20 supporting cells, which are known to act as a stem cell pool for the
and its receptor Fgfr1 precedes the differentiation of hair cells and regeneration of injured hair cells (Jacques et al. 2012). A previous
supporting cells in the developing cochlea duct. In a cultured study demonstrated that mammalian cochlear supporting cells were
murine developing inner ear, SU5402 (FGF receptor inhibitor) or capable of proliferating and differentiating into hair cells when
the anti-FGF20 neutralizing antibody were found to effectively cultured. This finding suggests that the regenerative mechanisms
inhibit the formation of the sensory epithelium, including both hair observed in adult avians may be conserved in the developing mam-
cells and supporting cells, when added to the medium before/during malian inner ear (White et al. 2006).
the formation of the sensory epithelium. These findings suggested After the specification of hair cell development in the organ of
that signaling mediated by FGF20/FGFR1 may play a role in gen- Corti, hair cells begin to be oriented as a set of three rows of outer
erating the sensory domain of the developing cochlear duct (Fig. 8, hair cells and a single row of inner hair cells. The hair cells then
Hayashi et al. 2008b). Recent experiments showed that the FGFR1- generate a V-shaped bundle of stereocilia on their apical surface, on
mediated MAP kinase activation through FGFR substrate (Frs) 2/3 which the vertex points to the lateral site. Cells specified to the
is necessary for the maintenance of Sox2-positive sensory progeni- organ of Corti no longer proliferate; thus, a defined number of
tors and their subsequent commitment to the sensory cell differen- postmitotic cells undergo convergent extension movement to elon-
tiation (Ono et al. 2014). The Notch ligand Jagged1 is expressed gate the cochlear duct in a manner basal to apical direction. The
earlier than that of Fgf20 in the developing inner ear. The expres- planar cell polarity of hair cells as well as polarized cochlear exten-
sion of Fgf20 was absent in Jagged1-conditional mutant mice, and sion are known to be regulated by the Wnt-PCP pathway (Fig. 8,
the Fgf20 promoter region possessed an Rbpj-binding domain, Curtin et al. 2003; Montcouquiol et al. 2003; Wang et al. 2005). In
which indicated that the expression of Fgf20 is directly regulated by the V-shaped hair cell bundle, which possesses uniform medio-
Notch signaling (Munnamalai et al. 2012). Not only the expression lateral polarity on the apical surface of hair cells, a polarity/mitotic
of Fgf20, but also the generation of hair cells and supporting cells spindle-associated protein complex consisting of mInsc (mamma-
was inhibited in a cultured cochlea duct treated with DAPT (Notch lian Inscuteable), LGN (vertebrate partner of Insc [Pins]), and Gαi
inhibitor) before the onset of hair cell specification, and this inhibi- (heteromeric G protein) is localized in a lateral microvilli-free
tory effect was reversed by adding the FGF20 protein to the region of the apical surface of each hair cell. Loss-of-function
medium (Munnamalai et al. 2012). These findings suggested that experiments on the mInsc/LGN/Gαi complex revealed irregular
FGF20 may be involved in the specification and/or maintenance of bundles of stereocilia, which suggested that the mInsc/LGN/Gαi
the prosensory domain under the control of Notch signaling path- complex plays a role in the establishment of uniform bundles of
ways The sensitivity of the prosensory epithelium to DAPT is stereocilia on the hair cell surface (Tarchini et al. 2013). Therefore,
known to be stage-dependent; treating the early cochlea (ED12.5 in at least two pathways involving planer cell polarity are required for
mouse embryo) with DAPT led to a reduction in the number of hair the establishment of a mature sensory epithelium in the organ of
cells and supporting cells because of the blockade of lateral induc- Corti.
tion, whereas, conversely, the treatment with DAPT at a later stage
(ED13.5) increased the number of hair cells because of the block-
ade of lateral inhibition. At the earlier stage, Notch signaling-
PERSPECTIVES
dependent Sox2 maintains the prosensory region; therefore,
Notch-induced FGF20 may amplify/maintain Sox2 to specify the One of the most intriguing issues related to inner ear developmental
prosensory domain, from which hair cells and supporting cells will biology is a clearer understanding of the mechanisms regulating the
develop (Dabdoub et al. 2008; Munnamalai et al. 2012). In the regeneration of hair cells. Adult mammalian hair cells do not regen-
organ of Corti, hair cell differentiation progresses in a medial-to- erate after injury; therefore, hair cell injury after birth causes irre-
lateral direction by an, as yet, unidentified mechanism. Fgf20-null versible deafness. The loss of regenerative activity in the adult
mutant mice lack not only outer hair cells, but also differentiated sensory epithelium has been attributed to an age-dependent reduc-
supporting cells, such as Deiters’ cells (Huh et al. 2012). Therefore, tion in the number of multi-potent progenitor/stem cells due to a
a fine-tuned balance between FGF and BMP signaling may be a loss of multipotency and proliferative activity. Therefore, elucidat-
prerequisite for establishing the proper number of hair cell rows. ing the genetic and epigenetic mechanisms underlying such
Phospho-Erk, a downstream signal of FGF, was shown to phospho- quiescence in the adult sensory epithelium may facilitate the devel-
rylate the linker region of Smad1 to disrupt BMP-mediated Smad1 opment of strategies for inner ear regeneration. At the onset of
signaling (Pera et al. 2003). After the formation of hair cells in the sensory epithelium development, the prosensory domain exits the
cochlea, inner hair cells expressed Fgf8 and outer hair cells and cell cycle to commit to a lineage for mechanosensory hair cells.
supporting cells expressed Fgfr3 (Jacques et al. 2007). Mutant mice However, the cochlear epithelium in the neonatal rodent possesses
without FGF8/FGFR3 signaling have defective pillar cells and potent regenerative activity, which gradually decreases in an age-
deafness, which indicated that inner hair cell-derived FGF8 locally dependent manner. In the adult chick inner ear, the sensory epithe-
controls the differentiation of supporting cells into pillar cells, lium maintains regenerative activity throughout life; therefore, the

© 2014 Japanese Teratology Society


24 Y. Nakajima

sensory epithelium in adult birds can proliferate and differentiate to Colvin JS, Bohne BA, Harding GW, McEwen DG, Ornitz DM. 1996.
hair cells after injury. Further investigations are necessary to under- Skeletal overgrowth and deafness in mice lacking fibroblast growth factor
stand the structural, cellular, and molecular mechanisms responsi- receptor 3. Nat Genet 12:390–397.
ble for the regenerative activity observed in the sensory epithelium Curtin JA, Quint E, Tsipouri V et al. 2003. Mutation of Celsr1 disrupts
planar polarity of inner ear hair cells and causes severe neural tube defects
in neonatal mammals and avians.
in the mouse. Curr Biol 13:1129–1133.
Dabdoub A, Puligilla C, Jones JM et al. 2008. Sox2 signaling in prosensory
ACKNOWLEDGMENTS domain specification and subsequent hair cell differentiation in the devel-
oping cochlea. Proc Natl Acad Sci U S A 105:18396–18401.
The author thanks S Uoya for technical assistance. This work was Driver EC, Kelley MW. 2009. Specification of cell fate in the mammalian
supported by a JSPS Grant-in-Aid for Scientific Research (C) cochlea. Birth Defects Res C Embryo Today 87:212–221.
25460273. Etheridge SL, Ray S, Li S et al. 2008. Murine dishevelled 3 functions in
redundant pathways with dishevelled 1 and 2 in normal cardiac outflow
tract, cochlea, and neural tube development. Plos Genet 4:e1000259.
DISCLOSURE Evsen L, Sugahara S, Uchikawa M, Kondoh H, Wu DK. 2013. Progression
of neurogenesis in the inner ear requires inhibition of Sox2 transcription
None. by neurogenin1 and neurod1. J Neurosci 33:3879–3890.
Frenz DA, Liu W, Galinovic-Schwartz V, Van De Water TR. 1996. Retinoic
REFERENCES acid-induced embryopathy of the mouse inner ear. Teratology 53:292–303.
Freter S, Muta Y, Mak SS, Rinkwitz S, Ladher RK. 2008. Progressive
Abelló G, Alsina B. 2007. Establishment of a proneural field in the inner ear. restriction of otic fate: the role of FGF and Wnt in resolving inner ear
Int J Dev Biol 51:483–493. potential. Development 135:3415–3424.
Abelló G, Khatri S, Giraldez F, Alsina B. 2007. Early regionalization of the Fritzsch B, Jahan I, Pan N, Kersigo J, Duncan J, Kopecky B. 2011. Dissect-
otic placode and its regulation by the Notch signaling pathway. Mech Dev ing the molecular basis of organ of Corti development: where are we
124:631–645. now? Hear Res 276:16–26.
Abelló G, Khatri S, Radosevic M, Scotting PJ, Giraldez F, Alsina B. 2010. Gerlach LM, Hutson MR, Germiller JA, Nguyen-Luu D, Victor JC, Barald
Independent regulation of Sox3 and Lmx1b by FGF and BMP signaling KF. 2000. Addition of the BMP4 antagonist, noggin, disrupts avian inner
influences the neurogenic and non-neurogenic domains in the chick otic ear development. Development 127:45–54.
placode. Dev Biol 339:166–178. Groves AK, Fekete DM. 2012. Shaping sound in space: the regulation of
Alsina B, Abelló G, Ulloa E, Henrique D, Pujades C, Giraldez F. 2004. FGF inner ear patterning. Development 139:245–257.
signaling is required for determination of otic neuroblasts in the chick Hayashi T, Kokubo H, Hartman BH, Ray CA, Reh TA,
embryo. Dev Biol 267:119–134. Bermingham-McDonogh O. 2008a. Hesr1 and Hesr2 may act as early
Alvarez Y, Alonso MT, Vendrell V et al. 2003. Requirements for FGF3 and effectors of Notch signaling in the developing cochlea. Dev Biol 316:87–
FGF10 during inner ear formation. Development 130:6329–6338. 99.
Bissonnette JP, Fekete DM. 1996. Standard atlas of the gross anatomy of the Hayashi T, Ray CA, Bermingham-McDonogh O. 2008b. Fgf20 is required
developing inner ear of the chicken. J Comp Neurol 368:620–630. for sensory epithelial specification in the developing cochlea. J Neurosci
Bok J, Bronner-Fraser M, Wu DK. 2005. Role of the hindbrain in 28:5991–5999.
dorsoventral but not anteroposterior axial specification of the inner ear. Huh SH, Jones J, Warchol ME, Ornitz DM. 2012. Differentiation of the
Development 132:2115–2124. lateral compartment of the cochlea requires a temporally restricted
Bok J, Chang W, Wu DK. 2007a. Patterning and morphogenesis of the FGF20 signal. Plos Biol 10:e1001231.
vertebrate inner ear. Int J Dev Biol 51:521–533. Jackler RK, Luxford WM, House WF. 1987. Congenital malformations of
Bok J, Dolson DK, Hill P, Ruther U, Epstein DJ, Wu DK. 2007b. Opposing the inner ear: a classification based on embryogenesis. Laryngoscope
gradients of Gli repressor and activators mediate Shh signaling along the 97:2–14.
dorsoventral axis of the inner ear. Development 134:1713–1722. Jacob J, Briscoe J. 2003. Gli proteins and the control of spinal-cord pattern-
Bok J, Raft S, Kong KA, Koo SK, Drager UC, Wu DK. 2011. Transient ing. EMBO Rep 4:761–765.
retinoic acid signaling confers anterior-posterior polarity to the inner ear. Jacques BE, Dabdoub A, Kelley MW. 2012. Fgf signaling regulates devel-
Proc Natl Acad Sci U S A 108:161–166. opment and transdifferentiation of hair cells and supporting cells in the
Brenski AC, Arjmand EM. 2003. Congenital Inner Ear Anomalies. In: basilar papilla. Hear Res 289:27–39.
Bluestone CD, Stool SE, Casselbrant ML et al., editors. Pediatric otolar- Jacques BE, Montcouquiol ME, Layman EM, Lewandoski M, Kelley MW.
yngology, Vol. 1. Philadelphia: SAUNDERS. p 441–455. 2007. Fgf8 induces pillar cell fate and regulates cellular patterning in the
Chang W, Brigande JV, Fekete DM, Wu DK. 2004. The development of mammalian cochlea. Development 134:3021–3029.
semicircular canals in the inner ear: role of FGFs in sensory cristae. Jeon SJ, Fujioka M, Kim SC, Edge AS. 2011. Notch signaling alters sensory
Development 131:4201–4211. or neuronal cell fate specification of inner ear stem cells. J Neurosci
Chang W, Lin Z, Kulessa H, Hebert J, Hogan BL, Wu DK. 2008. Bmp4 is 31:8351–8358.
essential for the formation of the vestibular apparatus that detects angular Kiernan AE, Ahituv N, Fuchs H et al. 2001. The Notch ligand Jagged1 is
head movements. Plos Genet 4:e1000050. required for inner ear sensory development. Proc Natl Acad Sci U S A
Chang W, Nunes FD, De Jesus-Escobar JM, Harland R, Wu DK. 1999. 98:3873–3878.
Ectopic noggin blocks sensory and nonsensory organ morphogenesis in Kobayashi Y, Nakamura H, Funahashi J. 2008. Epithelial-mesenchymal
the chicken inner ear. Dev Biol 216:369–381. transition as a possible mechanism of semicircular canal morphogenesis
Chang W, ten Dijke P, Wu DK. 2002. BMP pathways are involved in otic in chick inner ear. Tohoku J Exp Med 215:207–217.
capsule formation and epithelial-mesenchymal signaling in the develop- Kochhar A, Fischer SM, Kimberling WJ, Smith RJ. 2007. Branchio-oto-
ing chicken inner ear. Dev Biol 251:380–394. renal syndrome. Am J Med Genet A 143a:1671–1678.
Chrysostomou E, Gale JE, Daudet N. 2012. Delta-like 1 and lateral inhibi- Komada M. 2012. Sonic hedgehog signaling coordinates the proliferation
tion during hair cell formation in the chicken inner ear: evidence against and differentiation of neural stem/progenitor cells by regulating cell cycle
cis-inhibition. Development 139:3764–3774. kinetics during development of the neocortex. Congenit Anom Kyoto
Cole LK, Le Roux I, Nunes F, Laufer E, Lewis J, Wu DK. 2000. Sensory 52:72–77.
organ generation in the chicken inner ear: contributions of bone Ladher RK, Anakwe KU, Gurney AL, Schoenwolf GC, Francis-West PH.
morphogenetic protein 4, serrate1, and lunatic fringe. J Comp Neurol 2000. Identification of synergistic signals initiating inner ear develop-
424:509–520. ment. Science 290:1965–1967.

© 2014 Japanese Teratology Society


Inner ear development 25

Ladher RK, O’Neill P, Begbie J. 2010. From shared lineage to distinct Pirvola U, Ylikoski J, Trokovic R, Hebert JM, McConnell SK, Partanen J.
functions: the development of the inner ear and epibranchial placodes. 2002. FGFR1 is required for the development of the auditory sensory
Development 137:1777–1785. epithelium. Neuron 35:671–680.
Ladher RK, Wright TJ, Moon AM, Mansour SL, Schoenwolf GC. 2005. Propst EJ, Blaser S, Gordon KA, Harrison RV, Papsin BC. 2005. Temporal
FGF8 initiates inner ear induction in chick and mouse. Genes Dev bone findings on computed tomography imaging in branchio-oto-renal
19:603–613. syndrome. Laryngoscope 115:1855–1862.
Li X, Oghi KA, Zhang J et al. 2003. Eya protein phosphatase activity Puligilla C, Feng F, Ishikawa K et al. 2007. Disruption of fibroblast growth
regulates Six1-Dach-Eya transcriptional effects in mammalian factor receptor 3 signaling results in defects in cellular differentiation,
organogenesis. Nature 426:247–254. neuronal patterning, and hearing impairment. Dev Dyn 236:1905–1917.
Litsiou A, Hanson S, Streit A. 2005. A balance of FGF, BMP and WNT Radde-Gallwitz K, Pan L, Gan L, Lin X, Segil N, Chen P. 2004. Expression
signalling positions the future placode territory in the head. Development of Islet1 marks the sensory and neuronal lineages in the mammalian inner
132:4051–4062. ear. J Comp Neurol 477:412–421.
Magarinos M, Contreras J, Aburto MR, Varela-Nieto I. 2012. Early Rakowiecki S, Epstein DJ. 2013. Divergent roles for Wnt/beta-catenin
development of the vertebrate inner ear. Anat Rec (Hoboken) 295:1775– signaling in epithelial maintenance and breakdown during semicircular
1790. canal formation. Development 140:1730–1739.
Mansour SL, Li C, Urness LD. 2013. Genetic rescue of Muenke syndrome Ramsebner R, Ludwig M, Parzefall T et al. 2010. A FGF3 mutation asso-
model hearing loss reveals prolonged FGF-dependent plasticity in coch- ciated with differential inner ear malformation, microtia, and
lear supporting cell fates. Genes Dev 27:2320–2331. microdontia. Laryngoscope 120:359–364.
Mansour SL, Schoenwolf GC. 2005. Morphogenesis of the inner ear. In: Riccomagno MM, Martinu L, Mulheisen M, Wu DK, Epstein DJ. 2002.
Kelly M, Wu DK, Popper AN, Fay RR, editors. Development of the Inner Specification of the mammalian cochlea is dependent on Sonic hedgehog.
Ear. New York: Springer. p 43–84. Genes Dev 16:2365–2378.
Mansour SL, Twigg SRF, Freeland RM, Wall SA, Li C, Wilkie AOM. 2009. Salminen M, Meyer BI, Bober E, Gruss P. 2000. Netrin 1 is required for
Hearing loss in a mouse model of Muenke syndrome. Hum Mol Genet semicircular canal formation in the mouse inner ear. Development
18:43–50. 127:13–22.
Martin P, Swanson GJ. 1993. Descriptive and experimental analysis of the Satoh T, Fekete DM. 2005. Clonal analysis of the relationships between
epithelial remodellings that control semicircular canal formation in the mechanosensory cells and the neurons that innervate them in the chicken
developing mouse inner ear. Dev Biol 159:549–558. ear. Development 132:1687–1697.
Masuda S, Usui S, Matsunaga T. 2013. High prevalence of inner-ear and/or Senel E, Gulleroglu BN, Senel S. 2009. Additional temporal bone findings
internal auditory canal malformations in children with unilateral on computed tomography imaging in branchio-oto-renal syndrome.
sensorineural hearing loss. Int J Pediatr Otorhinolaryngol 77:228– Laryngoscope 119:832.
232. Sennaroglu L, Saatci I. 2002. A new classification for cochleovestibular
Montcouquiol M, Rachel RA, Lanford PJ, Copeland NG, Jenkins NA, malformations. Laryngoscope 112:2230–2241.
Kelley MW. 2003. Identification of Vangl2 and Scrb1 as planar polarity Steventon B, Mayor R, Streit A. 2012. Mutual repression between Gbx2 and
genes in mammals. Nature 423:173–177. Otx2 in sensory placodes reveals a general mechanism for ectodermal
Morsli H, Choo D, Ryan A, Johnson R, Wu DK. 1998. Development of the patterning. Dev Biol 367:55–65.
mouse inner ear and origin of its sensory organs. J Neurosci 18:3327– Stone JS, Shang JL, Tomarev S. 2003. Expression of Prox1 defines regions
3335. of the avian otocyst that give rise to sensory or neural cells. J Comp
Munnamalai V, Hayashi T, Bermingham-McDonogh O. 2012. Notch Neurol 460:487–502.
prosensory effects in the Mammalian cochlea are partially mediated by Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells from
Fgf20. J Neurosci 32:12876–12884. mouse embryonic and adult fibroblast cultures by defined factors. Cell
Neves J, Abello G, Petrovic J, Giraldez F. 2013. Patterning and cell fate in 126:663–676.
the inner ear: a case for Notch in the chicken embryo. Dev Growth Differ Takemoto T. 2013. Mechanism of cell fate choice between neural and
55:96–112. mesodermal development during early embryogenesis. Congenit Anom
Neves J, Kamaid A, Alsina B, Giraldez F. 2007. Differential expression of Kyoto 53:61–66.
Sox2 and Sox3 in neuronal and sensory progenitors of the developing Tarchini B, Jolicoeur C, Cayouette M. 2013. A molecular blueprint at the
inner ear of the chick. J Comp Neurol 503:487–500. apical surface establishes planar asymmetry in cochlear hair cells. Dev
Neves J, Parada C, Chamizo M, Giraldez F. 2011. Jagged 1 regulates the Cell 27:88–102.
restriction of Sox2 expression in the developing chicken inner ear: a Torres M, Giraldez F. 1998. The development of the vertebrate inner ear.
mechanism for sensory organ specification. Development 138:735–744. Mech Dev 71:5–21.
Neves J, Uchikawa M, Bigas A, Giraldez F. 2012. The prosensory function Urness LD, Paxton CN, Wang X, Schoenwolf GC, Mansour SL. 2010. FGF
of Sox2 in the chicken inner ear relies on the direct regulation of Atoh1. signaling regulates otic placode induction and refinement by controlling
PLoS ONE 7:e30871. both ectodermal target genes and hindbrain Wnt8a. Dev Biol 340:595–604.
Ohyama T, Basch ML, Mishina Y, Lyons KM, Segil N, Groves AK. 2010. Wang J, Mark S, Zhang X et al. 2005. Regulation of polarized extension and
BMP signaling is necessary for patterning the sensory and nonsensory planar cell polarity in the cochlea by the vertebrate PCP pathway. Nat
regions of the developing mammalian cochlea. J Neurosci 30:15044– Genet 37:980–985.
15051. White PM, Doetzlhofer A, Lee YS, Groves AK, Segil N. 2006. Mammalian
Ono K, Kita T, Sato S et al. 2014. FGFR1-Frs2/3 signalling maintains cochlear supporting cells can divide and trans-differentiate into hair cells.
sensory progenitors during inner ear hair cell formation. Plos Nature 441:984–987.
Genet:e1004118. Wright TJ, Mansour SL. 2003. Fgf3 and Fgf10 are required for mouse otic
Ozaki H, Nakamura K, Funahashi J et al. 2003. Six1 controls patterning of placode induction. Development 130:3379–3390.
the mouse otic vesicle. Development 131:551–562. Yamamoto N, Okano T, Ma X, Adelstein RS, Kelley MW. 2009. Myosin II
Pan N, Jahan I, Kersigo J et al. 2011. Conditional deletion of Atoh1 using regulates extension, growth and patterning in the mammalian cochlear
Pax2-Cre results in viable mice without differentiated cochlear hair cells duct. Development 136:1977–1986.
that have lost most of the organ of Corti. Hear Res 275:66–80. Yasuda M, Yamada S, Uwabe C, Shiota K, Yasuda Y. 2007. Three-
Pauley S, Lai E, Fritzsch B. 2006. Foxg1 is required for morphogenesis and dimensional analysis of inner ear development in human embryos. Anat
histogenesis of the mammalian inner ear. Dev Dyn 235:2470–2482. Sci Int 82:156–163.
Pera EM, Ikeda A, Eivers E, De Robertis EM. 2003. Integration of IGF, Zou D, Silvius D, Fritzsch B, Xu PX. 2004. Eya1 and Six1 are essential for
FGF, and anti-BMP signals via Smad1 phosphorylation in neural induc- early steps of sensory neurogenesis in mammalian cranial placodes.
tion. Genes Dev 17:3023–3028. Development 131:5561–5572.

© 2014 Japanese Teratology Society

You might also like