You are on page 1of 15

Cellular and Molecular Life Sciences

https://doi.org/10.1007/s00018-020-03506-x Cellular and Molecular Life Sciences

REVIEW

Mechanisms of larynx and vocal fold development and pathogenesis


Vlasta Lungova1 · Susan L. Thibeault1

Received: 4 December 2019 / Revised: 12 March 2020 / Accepted: 16 March 2020


© Springer Nature Switzerland AG 2020

Abstract
The larynx and vocal folds sit at the crossroad between digestive and respiratory tracts and fulfill multiple functions related
to breathing, protection and phonation. They develop at the head and trunk interface through a sequence of morphogenetic
events that require precise temporo-spatial coordination. We are beginning to understand some of the molecular and cel-
lular mechanisms that underlie critical processes such as specification of the laryngeal field, epithelial lamina formation and
recanalization as well as the development and differentiation of mesenchymal cell populations. Nevertheless, many gaps
remain in our knowledge, the filling of which is essential for understanding congenital laryngeal disorders and the evaluation
and treatment approaches in human patients. This review highlights recent advances in our understanding of the laryngeal
embryogenesis. Proposed genes and signaling pathways that are critical for the laryngeal development have a potential to be
harnessed in the field of regenerative medicine.

Keywords  Laryngeal · Vocal cords · Congenital · Embryology

Introduction linked diseases related to laryngeal development and/or


regeneration. The recent introduction and use of transgenic
Voice problems occur in between 3 and 9% of the population mice in voice research should significantly improve our
in the United States [78]. Although a majority of these prob- understanding of normal laryngeal cell biology and geneti-
lems arise due to vocal fold (VF) structural and neurologic cally linked disease processes.
abnormalities, phonotrauma and/or laryngeal inflammation Basic research into the mechanisms controlling laryngeal
[38], there is a clear segment of the population in which morphogenesis is important for a number of reasons. First,
voice disorders develop secondary to defects that occur dur- congenital laryngeal defects are potentially life-threatening
ing intrauterine development [11]. These congenital voice at birth, as they can be accompanied with airway obstruc-
disorders can be caused by a wide variety of abnormalities tion, and can cause problems in voicing across the lifespan
along the aerodigestive tract, including larynx and vocal [1, 99]. A better understanding of how these early devel-
folds (VF). Molecular and cellular mechanisms that con- opmental processes are regulated is necessary for under-
trol formation of laryngeal structures are beginning to be standing and evaluating functional prognosis in treatment
elucidated, but we are still far behind the research that has approaches. Second, abnormalities in formation of laryngeal
been done in upper airways or lungs [36, 61]. Frequently structures can seriously affect development and maturation
used animal models, such as felines, non-human primates, of other respiratory structures, such as lungs. Failure in VF
canines, pigs, rabbits or rats allow for the examination of recanalization in laryngeal webs/atresia results in polyhy-
laryngeal biology and testing of broad arrays of medical, dramnios lungs [48], while congenital laryngeal clefts may
surgical and behavioral interventions [3, 10, 12, 17, 30, 54, lead to chronic interstitial lung damage related to aspiration
77, 89]; however, these animal models have limitations for and gastroesophageal reflux [63]. Lastly, identifying genes
studying epithelio-mesenchymal interactions in genetically and signaling molecules that drive VF morphogenesis could
be utilized in VF tissue engineering to improve postnatal VF
tissue repair [6, 43, 46] and/or to create developmentally
* Susan L. Thibeault derived human VF mucosa for clinical and pharmacological
thibeault@surgery.wisc.edu
applications [53].
1
Department of Surgery, University of Wisconsin Madison,
5103 WIMR, 1111 Highland Ave, Madison, WI 53705, USA

13
Vol.:(0123456789)
V. Lungova, S. L. Thibeault

Here, we address current understanding of molecular for breathing. The CT muscle works as a VF tensor. Spe-
and cellular mechanisms that regulate different stages of the cific to humans are transverse and oblique arytenoid muscles
laryngeal morphogenesis, from specification of the laryn- located in the tracheoesophageal septum. They approximate
geal field to complete VF separation using transgenic mouse the VF along with LCA. Specific to rodents is a thin pair
models and correlate these findings with human data from of muscles called the superior cricoarytenoid (SCA). These
patients with congenital laryngeal malformations. We also muscles connect the dorsal surface of the AC with the CC
point to events which are underexplored and that deserve midline prominence. They assist in the VF adduction and
further attention. Proposed genes and signaling pathways likely serve the same role as the transverse and oblique inter-
that are critical for development of the larynx and VF have arytenoid muscles in humans [42, 88].
a potential to be harnessed in the field of VF regenerative The VF stretch horizontally across the larynx; they attach
medicine. to the TC ventrally and AC dorsally. Alike in humans, the
murine VF are composed of three distinct tissue types
(Fig. 1d–h). The outer layer is squamous non-keratinizing
Structure and function of the larynx stratified epithelium (EP), which transits into pseudostrati-
and vocal folds fied cranially, in the laryngeal vestibule, and caudally, in
the subglottic region. Squamous epithelial cells are adapted
The mammalian larynx fulfills multiple functions related to to withstand constant abrasions they experience during
breathing, protection and phonation starting with the first collisions and form a protective barrier against injury and
breath of a newborn. During breathing, the larynx main- environmental or systemic-based irritants [45]. Below the
tains a widely open air channel with abducted VF to con- epithelium is the lamina propria (LP) with fibroblasts and
trol inspiratory and expiratory airflow. Nevertheless, dur- extracellular matrix. This layer responds to the mechanical
ing swallowing VF approximate, the epiglottis closes the force applied by the air passing from the lungs. In humans,
entrance into the laryngeal cavity and the larynx acts as the LP gradually changes its density from the jelly-like
a sphincter to avoid aspiration. The VF are housed in the superficial layer to the stiffer layers containing elastic and
larynx and besides swallowing, they are involved in voice collagenous fibers and the muscle. The change in density
production in humans [59]. Whether the VF participate in from the superficial layer to the muscle allows the creation of
audible and ultrasonic vocalizations in mice is still debat- the mucosal wave that occurs during vibration. The presence
able [55, 74]. or absence of the mucosal wave determines VF vibratory
Current descriptive anatomic studies have revealed simi- behavior and, thus, influences the voice quality [59]. The
larities in the structure and function between human and thyroarytenoid (TA) muscle forms the deepest VF portion in
murine larynges and VF [57, 88, 89] (Fig. 1a–c). In both humans and mice [37, 51, 86]. The function of this muscle
species, the laryngeal framework is composed of four main is to shorten and relax the VF, which modifies VF elasticity
cartilages interconnected by ligaments and membranes, and and tension. While rodents use a different mechanism to gen-
moved by extrinsic and intrinsic laryngeal muscles. These erate ultrasonic and audible sound than humans [55], their
cartilages include the epiglottis (Epi), thyroid cartilage (TC), precise control of laryngeal activity and properties of the
cricoid cartilage (CC) and a pair of arytenoid cartilages (AC) VF are similar to humans and other mammals [73, 74, 97].
which rotate and rock on the CC to facilitate VF movement. Mice vocalizations can have a fundamental frequency
Specific to humans are pairs of cuneiform and corniculate range between 100 and 120,000 Hz; whereas, the range
cartilages. The corniculate cartilages are minor cartilaginous for human vocalizations is much reduced in comparison,
structures, which articulate with the apices of the AC and between 100 and 3000 Hz [55, 91]. In humans, fundamental
prolong them backward and medial. The cuneiform carti- frequency is determined by mass and tension of the VF and
lages are located within the aryepiglottic folds and act to air pressure during vibration. VF mass and tension are typi-
strengthen the VF. Specific to rodents is an alar cartilage cally regulated via muscle contraction and relaxation of the
(Ar) that is situated superior to the VF ventral attachment various laryngeal muscles (TA, PCA, PCA, CT). Certainly,
and is involved in ultrasonic vocalization [55, 74, 89]. changes in the constituents of the lamina propria can also
The intrinsic laryngeal muscles are skeletal voluntary change VF mass. The supralaryngeal vocal tract, consisting
muscles and are innervated by cranial nerves. The intrin- of both the oral and nasal airways serve as a time-varying
sic laryngeal muscles common to both species include the acoustic filter that suppresses the passage of sound energy
posterior cricoarytenoid muscles (PCA) located in the tra- at certain frequencies while allowing its passage at other
cheoesophageal septum, lateral cricoarytenoid (LCA) and frequencies. Formants are those frequencies at which local
cricothyroid muscles (CT), located on the lateral aspect of energy maxima are sustained by the supralaryngeal vocal
the larynx. LCA controls VF adduction for sound produc- tract and are determined, in part, by the overall shape, length
tion and/or swallowing, while PCA controls VF abduction and volume of the vocal tract [102]. Similar to humans, mice

13
Mechanisms of larynx and vocal fold development and pathogenesis

Fig. 1  Morphology of the murine larynx and vocal folds. a–f Sche- in the panel of e shows the detailed morphology of the right vocal
matic illustrations of the murine laryngeal cartilages and muscles. fold (f). g, h Hematoxylin–eosin staining showing morphology of the
Sagittal section through the murine larynx showing the position of murine vocal folds in coronal (g) and transverse section (h). Brack-
the VF (in yellow) and epiglottis, thyroid cartilage, alar cartilage, eted regions in the panel of g, h show detailed morphology of the
arytenoid cartilages and cricoid cartilage (a). Frontal view of the right vocal fold in coronal and transversal sections, respectively. Scale
murine larynx showing the attachment of the cricothyroid muscle bar 500 µm. A anterior, Ar alar cartilage, AC arytenoid cartilage, CC
to the thyroid and cricoid cartilages (b). Sagittal section through the cricoid cartilage, CT cricothyroid muscle, D dorsal, Ep epithelium,
murine larynx showing the attachment of the thyroarytenoid muscle, Epi epiglottis, LP lamina propria, P posterior, PCA posterior cricoar-
lateral cricoarytenoid muscle, superior and posterior cricoarytenoid ytenoid muscle, SCA superior cricoarytenoid muscle, TA thyroaryt-
muscles (c). Transverse sections of the murine VF, the cranial (ante- enoid muscle, TC thyroid cartilage, V ventral, VF vocal fold
rior) section (d) and caudal (posterior) section (e). Bracketed region

typically use their larynx to create sound, albeit differently, model [74]. In this model, a ventral pouch, which is posi-
such that USV is possible. Theories into the mechanism(s) tioned downstream of the VF and surrounded by cartilagi-
for the creation of ultrasonic vocalization (USV) have been nous structures, has part of its entrance edge supported by
various, with the most likely mechanism being the edge-tone alar cartilage or “alar edge”. Glottal airflow passes over the

13
V. Lungova, S. L. Thibeault

alar edge of the ventral pouch. Contraction of the alar edge microanatomy is well adapted to withstand the high mechan-
via contraction of the TA muscles results in a shorter dis- ical stresses and forces they experience during the vibratory
tance between the edges of the ventral pouch and a smaller cycle and collisions. Further looking into the developmental
ventral pouch lumen. During voicing, undulating airflow origin of laryngeal structures is important to understand the
interacts with the alar edge of the ventral pouch allowing relationship between these structures, their unique properties
for resonance within pouch, similar to a Helmholtz reso- and etiology of defects in laryngeal and VF development.
nator. Fundamental frequency is dependent upon glottal
airflow velocity and glottis to alar edge distance. Increases
in fundamental frequency are perceived when airflow Gene fate mapping of the laryngeal and VF
velocity increases and/or when the glottis to alar edge dis- structures
tance decreases, such that the volume in the ventral pouch
decreases concomitantly. The TA muscle indirectly regulates Only recently comprehensive descriptions of developmental
airflow through the glottis via VF adduction and it has been origin of mammalian laryngeal and VF tissue have become
hypothesized in its role in regulating glottis to alar edge dis- available using mouse transgenic models [52, 86]. These
tance and regulating ventral pouch size. studies revised and extended previous contradictory inves-
Overall, the larynx and VF in mice and humans are well tigations based on descriptive studies and lineage-tracing
suited to carry out their function. The finely orchestrated experiments in birds [24, 40, 65, 66] or amphibians [71].
action of intrinsic laryngeal muscles attached to the laryn- Precursors for laryngeal and VF structures come from
geal framework facilitates VF movement necessary for multiple sources in developing embryos (Fig. 2A). First,
breathing, swallowing and vocalization. Moreover, the VF laryngeal structures develop in close proximity with the

Fig. 2  Developmental origin of laryngeal and vocal fold structures. ate lamina of the thyroid cartilage in the caudal-most region of the
a Schematic illustration of major cell populations that contribute thyroid cartilage (in red). d Schematic illustration showing that neural
to the laryngeal and vocal fold structures including anterior foregut crest-derived cells expressing Wnt1Cre+/−mGFP contribute to for-
endoderm (in blue), lateral mesoderm (in red) and neural crest cells mation of the loose connective tissue in the ventral lamina propria,
(in yellow). b Schematic illustration showing that anterior foregut nervous tissue and thyroid cartilage, except for its medial caudal-most
endoderm-derived cells expressing ShhCre+/−TdTom Red give rise portion (in yellow). AC arytenoid cartilage, AFE anterior foregut
primarily to the laryngeal and VF epithelium (in blue). c Schematic endoderm, CC cricoid cartilage, D dorsal, EP vocal fold epithelium,
illustration showing that cells of the lateral mesoderm-derived cells LM lateral mesoderm, NCC neural crest cell, TA thyroarytenoid mus-
expressing Mesp1Cre+/−TdTom Red give rise to the cricoid cartilage, cle, TC thyroid cartilage, V ventral
arytenoid cartilages, intrinsic laryngeal muscles, and the intermedi-

13
Mechanisms of larynx and vocal fold development and pathogenesis

endodermal foregut tube, similar to other respiratory struc- expression of the skeletal muscle-specific genes, such as
tures. Endodermal cells express sonic hedgehog (Shh) gene Myf5 [67]. Mechanisms that specify and guide migration
which is required for normal patterning of respiratory, diges- of myogenic precursors into the laryngeal region as well as
tive and genitourinary organs and enables to genetically their further differentiation into intrinsic laryngeal muscles
label and follow the fate of endodermal cells [32]. Second, have not yet been fully elucidated.
the endodermal foregut tube is enveloped laterally and ven- Mapping of descendants of NCC using Wnt1Cre+/−mGFP
trally with the lateral mesoderm that expresses mesoderm reporter mice [86] has shown the neural crest cells are a
posterior BHLH transcription factor 1 (Mesp1). Mesp1 gene pluripotent population of cells that represent precursors for
plays a key role in the rostro-caudal patterning of somitic connective and nerve tissues (Fig. 2d). NCC contribute to
mesoderm and labels mesoderm-derived tissues [79]. Third, the formation of the loose connective tissue in the ventral
the lateral mesoderm becomes partially displaced by sec- LP and the TC, except for the medial caudal-most portion
ondary population of mesenchymal neural crest cells (NCC) of the TC [86]. Wnt1Cre+/−mGFP-positive cells have also
that migrate to the site of laryngeal development from the been found within laryngeal muscles and were identified
hindbrain region. The NCC transiently express protoonco- either as neurons by acetylated tubulin immunofluorescent
gen wingless-type MMTV integration site family, member staining or fascia separating the muscles [86]. Similar to
1 (Wnt-1) gene that is used to visualize descendant of the myogenic precursors, NCC move en masse along branchial
NCC [14]. arches into the target site and mix with the mesoderm-
Lineage-tracing experiments using ShhCre+/−TdTomato derived structures during craniofacial morphogenesis [68].
reporter mice [52] revealed that endodermal cells primar- Recent experiments using Fuz−/− mutants have shown that
ily give rise to the laryngeal and VF epithelium (Fig. 2b). laryngeal NCC become specified prior to leaving the hind-
These endodermal cells are formed during gastrulation [29] brain region. Whole animal deletion of Fuz gene results in
and are initially located ventral to the future postotic hind- excessive accumulation of NCC in the developing larynx
brain region. Interestingly, the ShhCre lineage signal also which leads to severe malformations of laryngeal cartilages
extends into adjacent mesenchyme suggesting that some Shh and muscles and the absence of glottis. However, conditional
expressing cells may delaminate into VF lamina propria. deletion of the Fuz gene in Wnt1Cre+/−FuzFlox/− mutants does
These cells intermix with Sox9 expressing chondrocytes or not affect laryngeal morphogenesis [86]. Further experi-
surround MF-20 expressing myoblasts [52]. The function of ments are needed to elucidate the specification and migra-
these cells, as well as their contribution to differentiation of tory pattern of laryngeal NCC as well as their integration
mesenchymal structures remains to be investigated. into mesoderm-derived tissues, such as laryngeal cartilages,
Lateral mesoderm is a source of the AC and CC, and or nerve tissue.
intrinsic laryngeal muscles including TA muscle as deter- In summary, lineage-tracing data have revealed two
mined by Mesp1 Cre+/−TdTomato reporter lineage trac- important aspects of laryngeal morphogenesis that deserve
ing [86] (Fig. 2c). Further investigation of the origin of further investigations. First, the intrinsic laryngeal muscles
intrinsic laryngeal muscles has revealed that mamma- originate from different mesodermal cell populations and
lian intrinsic laryngeal muscles originate from the cra- develop via distinct genetic programs than typical skeletal
nial (unsegmented) mesoderm based on the robust Isl1 limb muscles. This supports the notion that intrinsic laryn-
expression, similar to the head muscles, and not from the geal muscles represent specialized muscle groups closely
somitic (segmented) Pax3-positive mesoderm as clas- related to the extraocular and head muscles that significantly
sic skeletal limb muscles do [86]. A close relationship differ from typical skeletal muscle structure and function
between intrinsic laryngeal and extraocular head muscles [57]. This could have an impact on voice therapy, as sev-
has been confirmed by independent experiments using eral voice therapy approaches suggested in the literature
animal models and human tissues. They demonstrate that are based on principles of exercise physiology in classic
intrinsic laryngeal muscles share many structural features skeletal muscles, as discussed elsewhere [88].Second, the
with extraocular muscles and stand apart from typical limb larynx develops at the “head (neural crest-derived)–trunk
skeletal muscles in terms of their muscles fiber size, myo- (mesoderm-derived)” interface and cells from both sides of
sin heavy chain isomorphs, innervation patterns, regenera- this interface move into the site where the larynx develop-
tive capacity and response to diseases [30, 57, 70, 81, 82, ments. This requires precise temporo-spatial coordination to
88]. Myogenic cells enter the branchial arches and migrate achieve an integrated outcome. The complexity of develop-
into the site of laryngeal development along with NCC, ment, therefore, renders the laryngeal apparatus suscepti-
which is reflected in their innervation by the cranial nerves ble to perturbation and, subsequently, the pathogenesis of
(superior and inferior branches of the vagal nerve) [28, congenital anomalies. In the next part of this review, we
67]. Myogenic precursors are committed to the myogenic will focus on developmental events that are critical for
lineage prior to leaving the somites, as evidenced by their

13
V. Lungova, S. L. Thibeault

understanding the laryngeal and VF morphogenesis and can intrinsic laryngeal muscle formation and initiation of
lead to laryngeal congenital malformations. stratification in the VF epithelium [52]. Laryngeal carti-
lages and muscles complete their development prior to VF
separation. Soon after VF separation (E17.5–18.5 in mice
Overview of laryngeal and vocal fold and week 12 in humans), the intrinsic laryngeal muscles
morphogenesis approximate the VF to close the glottis or pull them apart
to enable bidirectional flow of amniotic fluid [48]. After
The first signs of laryngeal morphogenesis appear dur- delivery, amniotic fluid is replaced with air as a newborn
ing early embryonic stages, embryonic (E) day 9.5 in takes its first breath. In humans, the presence of laryngeal
mice, which corresponds to week 4 (Carnegie stage 11) cartilages as well as detection of the VF motion serves
in humans, with a formation of a sagittal slit at the level as orientation points for sonographic examination of the
of the fourth pharyngeal pouch, labeled as the laryngotra- developing human fetal larynx as early as 11 weeks’ ges-
cheal groove (LG) [35, 80, 100] (Fig. 3a–c). There has tation to evaluate obstructive pathological lesions [48].
been much controversy over the interpretation of this ana- Inability to identify these anatomical elements along with
tomic location. In the past, some authors believed that the the marked polyhydramnios and echogenic lungs indicate
LG represented the primordium of the upper subglottic (or congenital upper airway anomalies such as laryngeal atre-
infraglottic) cavity [80]. The most recent re-examination sia or tracheoesophageal fistula [48].
of the early development of the mammalian larynx using In summary, the major morphogenetic events of the
computer generated three-dimensional (3D) reconstruc- laryngeal and VF embryogenesis include the apposition of
tion supports the notion that this opening represents the lateral walls of the primitive LPh and the EL formation that
primordium of the supraglottic cavity, called the primitive bring VF primordia together. Then, it is the separation of
laryngopharynx (LPh), which develops as the RD expands the larynx from the esophagus and the EL disintegration
and elongates [35]. As an anatomic landmark, the primi- once supporting mesenchymal structures develop and firmly
tive LPh appears at E10.5 in mouse models and Carnegie anchor growing VF. The process of the EL recanalization
stage 13 in humans [35, 51, 100]. It is located between prevents airway obstruction and is critical for further devel-
the fourth pharyngeal pouch, as its cranial border, and opment of other parts of the respiratory system, particularly
the cephalic portion of the respiratory diverticulum, as the lungs. Reciprocal epithelial–mesenchymal interactions
its caudal border. The VFs originate from the caudal LPh that regulate these morphogenetic events are poorly under-
segment; the cranial part of the LPh becomes the laryngeal stood as laryngeal development has received little attention
vestibule (LV) (supraglottis); while, the segment below the by experimental biologists.
prospective VFs, including the cephalic portion of the res-
piratory diverticulum (RD), becomes the subglottis (SG).
In the course of development (Fig. 3), the primitive Early stages of laryngeal morphogenesis—
LPh becomes bilaterally compressed due to the approxi- specification of the laryngeal and vocal fold
mation of the lateral walls toward the center of the lumen progenitors
(at E10.5 in mice and Carnegie stage 13 in humans). Soon
after, the closely juxtaposed lateral walls fuse together Signaling pathways that control early LPh
and form the epithelial lamina (EL) which temporarily remodeling and specification of VF epithelial
obliterates the ventral LPh lumen (at E11.5 in mice and progenitors
Carnegie stages 14–16 in humans). The EL is first reorgan-
ized to form the laryngeal part of the tracheoesophageal Early stages of laryngeal morphogenesis are associated
septum (E13.5), and then completely disintegrates to open with development of the respiratory diverticulum (RD) in
the laryngotracheal tube (E13.5–18.5 in mice; Carnegie the ventral foregut endoderm. Thus, signaling pathways
stages 19–23 up to week 12 in humans). The process of that drive respiratory progenitor identity are likely to act on
EL recanalization initiates with the formation of cavities specification of the laryngeal cell fate as well. RD embry-
such as the laryngeal cecum (ventrally) and pharyngoglot- ogenesis begins with a separation of the trachea from the
tic duct (dorsally) that extend into the primitive LPh from foregut; the lung bud appearing as an out pouching of the
cranial and caudal directions, respectively, and exert pres- RD at E9.5 in mice and approximately 28 days in humans
sure on the epithelial walls. By the gradual expansion of [36, 61]. This early histogenic RD remodeling requires a
these cavities, the laryngeal cecum unites with the phar- localized domain of Nkx2-1 expression in the ventral wall of
yngoglottic duct and VF move apart [51, 52]. In both spe- the anterior foregut endoderm and the presence of Shh [4].
cies, mice and humans, the process of EL disintegration Reducing levels of Nkx2-1 and/or Shh or disabling the ability
is concurrent with the laryngeal cartilage chondrification, of adjacent mesodermal cells to properly respond results in

13
Mechanisms of larynx and vocal fold development and pathogenesis

Fig. 3  Schematic illustration of larynx and vocal fold development. a, closely juxtaposed lateral walls fuse and create the EL. Differentia-
b Time scales of the laryngeal and VF development showing that the tion of mesenchymal structures initiates. At E13.5, the tracheoesoph-
human larynx and VF (a) pass through similar developmental stages ageal septum is formed and the EL begins its recanalization, due to
as the murine larynx and VF (b). These stages include the specifi- the expansion of the laryngeal cecum and pharyngoglottic duct that
cation of the laryngeal field, apposition of the lateral walls, EL for- exert pressure on the epithelial walls. During EL recanalization,
mation, EL recanalization and maturation of the VF epithelium and laryngeal cartilages and muscles continue to differentiate. At E18.5,
mesenchymal structures during the fetal period (in humans) and the VF are separated and mesenchymal structures are fully devel-
postnatally (in mice). Schematic illustrations demonstrate the murine oped to anchor the growing VF. EL epithelial lamina, Es esophagus,
larynx and VF development in more detail. The first image shows LB lung bud, LC laryngeal cecum, LG laryngotracheal groove, LPh
the position of the laryngotracheal groove at the level of the fourth primitive laryngopharynx, PA pharyngeal arch, PD pharyngoglottic
pharyngeal pouch at stage E 9.5. At E10.5, the primitive LPh initiates duct, PP pharyngeal pouch, RD respiratory diverticulum, Tr trachea,
its formation. It becomes bilaterally compressed due to the approxi- VF vocal fold
mation of the lateral walls to the center of the lumen. At E11.5, the

13
V. Lungova, S. L. Thibeault

defects of tracheoesophageal separation and tracheal chon- Deletion of Shh in Shh null mutants affects the apposition
drogenesis leading to tracheoesophageal atresia/fistula [50]. of the lateral walls; they do not approach each other and the
Recent studies have shown that Shh and Gli1, Gli2 and Gli3 primitive LPh remains open. This is consistent with the fact
are strongly expressed in the primitive LPh endoderm and that lack of Shh signaling may downregulate gene expression
surrounding mesenchyme, respectively [51, 86] (Fig. 4a). and proliferation of the adjacent mesenchymal target cells.

Fig. 4  Genes and signaling molecules participating in larynx and proliferation via Cyclin D1 expression and simultaneous differen-
vocal fold development. a Genes and signaling molecules that con- tiation of basal p63+ K8+ progenitors. In the lamina propria, laryn-
trol specification of the laryngeal field and early stages of primitive geal cartilages initiate differentiation into Sox9+ chondrocytes and
LPh remodeling. Shh is expressed in the LPh endoderm (red color) MF-20+ myoblasts. Other genes and signaling molecules acting on
and likely controls the juxtaposition of Nkx2-1 (green solid line) differentiation of laryngeal cartilages and muscles include R-spon-
and Sox2 (yellow solid line) expression domains in the LPh endo- din2, Eph/EphrinB2 signaling. c Genes and signaling molecules
derm and cell proliferation. In the lamina propria, Gli genes along that control EL recanalization and further differentiation of laryn-
with genes involved in the pharynx segmentation, such as RA, Tbx1, geal cartilages (dark gray), muscle (brown dashed lines) and nerves
β-Cat, Hox genes, Fgf signaling, contribute to initial differentiation (purple curvy lines). Proper stratification of VF basal progenitors has
of the mesenchymal cells and incorporation of migratory cell popula- been linked to β-Catenin (blue color) which controls conversion of
tions. (B) Genes and signaling molecules that control the fusion of p63+ cytokeratin (K) 8+ basal progenitors into functional basal p63+,
lateral walls and simultaneous differentiation of supportive mesen- K8− cells capable of stratification. AC arytenoid cartilage, CC cricoid
chymal structures—cartilages (dark gray) and muscles (brown dashed cartilage, EL epithelial lamina, LPh primitive laryngopharynx, TC
lines). β-Catenin (blue color) expressed in the EL likely controls cell thyroid cartilage

13
Mechanisms of larynx and vocal fold development and pathogenesis

Moreover, Shh deletion also alters Nkx2-1 and Sox2 expres- esophagus [13, 69]. In human patients, voice and laryngeal
sion in the primitive LPh endoderm, which may also disrupt defects are commonly associated with defects in Shh/Gli
approximation of lateral walls. signaling pathway [26]. Apart from defects described in Pal-
Besides Shh and a localized expression of Nkx2-1, the lister–Hall syndrome, bifid epiglottis and laryngeal clefts are
developing RD responds to Wnt signals emanating from found in patients with Oral–Facial–Digital syndromes and
adjacent mesoderm. Combined loss of Wnt2/2b in the ven- Bardet–Biedl syndrome [27, 34, 83, 85].
tral anterior foregut mesoderm leads to a complete loss of The pharyngeal arches and their segmental arrangement
Nkx2-1 lung endoderm and a failure to form the trachea represent another source of signaling for successful migra-
and branching lungs [31]. This phenotype is recapitulated tion and incorporation of NCC and myoblasts. Signaling
upon loss of beta-Catenin in the anterior foregut endoderm pathways that control caudal patterning of the pharyngeal
[32]. Specific to laryngeal development, high levels of Wnt apparatus, include—retinoic acid (RA), Tbx1, Hox genes,
signaling, as indicated by Axin2 in situ hybridization, have and Fgf signaling (Fig.  4a). RA is a morphogen that is
been reported in the primitive LPh endoderm and in adjacent important for the caudal pharyngeal pouch segmentation
mesenchyme during EL formation at E11.5 in mice [52] and formation of the ­3rd–6th pharyngeal arches [95]. Vita-
(Fig. 4b). Conditional deletion of beta-Catenin in the LPh min A-deficient rat embryos demonstrate laryngotracheal
endoderm disrupts the timing of EL formation. Nevertheless, cartilage malformation, incomplete separation of the glottis,
the approximation of lateral walls appear normal suggesting and/or laryngeal clefts [87]. Similarly, mouse mutants for
that Wnt signaling was not required for the juxtaposition of Raldh2 gene, the key enzyme responsible for the conversion
the lateral walls, but for their fusion and specification of VF of vitamin A (retinol) into retinoic acid, exhibit defects in
basal epithelial progenitors. Investigation of other signaling 3rd–6th pharyngeal arches formation, which also leads to
pathways, such as Bmp or Fgf signaling, may help to under- laryngeal cartilage malformations [64, 95].
stand epithelio-mesenchymal interactions that control these Besides a direct effect of RA on pharyngeal segmenta-
early morphogenetic events. tion, RA also mediates transcription of other genes such
as Tbx1 or Hox genes. Recent studies have shown that, in
Signaling pathways that control migratory cell a mouse model, decreased embryonic levels of RA down-
populations and differentiation of the mesenchymal regulate the expression of Tbx1, which results in pharyngeal
progenitors apparatus malformations and defects in cardiovascular and
craniofacial development resembling a DiGeorge syndrome
Early stages of LPh remodeling are also associated with phenotype [76]. The similar phenotype could be achieved by
intense migration of NCC and cranial myoblasts [28] and the mesenchymal inactivation of beta-Catenin which also
initial differentiation of mesenchymal progenitors in the LP suppresses the expression Tbx1 and its downstream signal-
(Fig. 4a). Excessive or insufficient cell migration as well as ing pathways [41]. These data confirmed that mesenchymal
inability of migratory cells to respond to positional clues Tbx1 is necessary for pharyngeal arch development [103]
may disrupt development of laryngeal cartilages, muscles and mutation in Tbx1 gene is closely associated with the
and laryngeal innervation. Signaling pathways known to etiology of DiGeorge syndrome. In patients, DiGeorge syn-
date that control migration and/or incorporation of laryngeal drome, or velo-cardio-facial syndrome, is linked with the
NCC and myoblasts in the LP include (1) Shh signaling via 22q11.2 microdeletion [98], which alters Tbx1 gene expres-
Gli proteins expressed in the LP mesenchyme and (2) signal- sion [16, 49, 62]. Besides congenital heart defects and crani-
ing pathways that control pharyngeal segmentation, as the ofacial malformations, these patients exhibit a wide range of
migratory cell populations enter the laryngeal region via the laryngeal abnormalities including laryngeal webs (the most
fourth and sixth pharyngeal (branchial) arches. frequent anomaly), subglottic stenosis, laryngomalacia and/
Recent studies have shown that Shh/Gli signaling is likely or atrophic VF [21, 25].
involved in regulation of migration and/or integration of In addition to regulation of Tbx1 expression, RA is a well-
laryngeal NCC in the LP. Deletion of Gli3 in Gli3−/− mutant known regulator of Hox gene activity through the presence
mice results in an excessive accumulation of NCC in the of RA response elements in the promoter region of many
ventral glottic compartment. This influences the attachment Hox genes [28, 58]. Analyses of Hox gene loss-of-function in
of the vocal ligaments and muscles as well as vocalization transgenic mice identified two Hox gene groups, Hoxa-3 and
[86]. Gli3 mutant mice can also display a more severe phe- Hoxa-5, which are involved in laryngeal and tracheal carti-
notype with a wide range of developmental abnormalities lage development. Deletion of Hoxa-3 in the caudal pharyn-
resembling Pallister–Hall syndrome [13]. Similar to human geal endoderm results in multiple skeletal defects, including
patients with Pallister–Hall syndrome, Gli3 mutant mice dysmorphogenesis of the CC and TC [56]. Similarly, loss-
showed an absence of the epiglottis and a laryngeal cleft of-function in Hoxa-5 gene in the mesenchyme surrounding
resulting in an abnormal connection between the larynx and

13
V. Lungova, S. L. Thibeault

the RD reveals a hyperplastic cricoid, disorganization of the experiments. In mice with a DiGeorge syndrome phenotype,
tracheal rings and hypoplasia of the pulmonary tree [5]. manipulation of beta-Catenin–Tbx1 signaling in the mesen-
Lastly, Fgf signaling also plays a key role in pharyngeal chyme led to mesenchymal defects and problems with the
pouch/arch formation and segmentation. Signals from the EL recanalization [41] suggesting that the EL disintegration
pharyngeal endoderm mediated via Fgf3 and Fgf8 interact depends on reciprocal interactions between epithelial and
with the Fgf receptors expressed in the NCC to facilitate the mesenchymal cells in the LP.
development of NCC-derived cartilages [28]. The role of
Fgf signaling in the laryngeal morphogenesis has not been
investigated yet. Differentiation of mesenchymal cells
in the lamina propria

Epithelial lamina formation EL recanalization is accompanied with simultaneous differ-


and recanalization entiation of mesenchyme into cartilages; muscles, fibroblasts
and neurofilaments that are major structural and functional
The next critical period of the laryngeal development that components of the larynx and VF (Fig. 4b, c). The inability
deserves further attention is related to the establishment of of mesenchyme to properly differentiate may lead to ana-
the EL and its recanalization. In a mouse model, the lateral tomic abnormalities of laryngeal cartilages and muscles and/
walls of the primitive LPh are brought together and fuse to or defective neuromuscular control that can affect the lumen
form the EL at E11.5 (Fig. 4b). The movement of the lateral patency and/or VF mobility.
walls towards the midline is likely caused by rapidly prolif-
erating epithelial and mesenchymal cells, which simultane- Differentiation of supportive cartilages
ously initiate differentiation into VF basal progenitors (epi-
thelial cells) and cartilages (mesenchymal cells) at the sites In a mouse model, differentiation of laryngeal cartilages ini-
of swellings [51, 80, 100]. The decrease in cell proliferation tiates with the formation of the EL, when an early marker
and/or failure in differentiation of epithelial or mesenchymal of cartilaginous structures SOX9 [23] appears at the site of
progenitors may disrupt EL formation and its consequent arytenoid swellings (prospective AC) and ventrally close to
recanalization. If EL recanalization does not occur until the tip of the EL [52]. This ventral region of SOX9+ cells
E18.5 in mice and the week 12 in humans, laryngeal webs gives rise to the intermediate lamina of the TC. The lateral
will result [35, 52, 80, 100]. laminae of the TC develop in a close proximity to the thyroid
Laryngeal webs are congenital defects associated with gland and gradually fuse with the intermediate lamina to
different degrees of failure of EL resorption during intrau- form the TC. The CC likely initiates its development later
terine development. They can cause acute, life-threatening along with the establishment of the tracheoesophageal sep-
respiratory complications in newborns, and chronic, lifelong tum (at E13.5 in mice). Failure in septum formation in beta-
problems with voicing [1, 33, 99]. Molecular mechanisms Catenin conditional mutants leads to the absence of the CC
that underline the establishment of the EL and its recanaliza- [52]. The subepithelial location of SOX9+ cells, next to the
tion are beginning to be elucidated. To date, the proper tim- EL expressing Shh, is likely important for Sox9 gene activa-
ing of the EL formation and recanalization has been linked tion. As previously reported, in the somitic mesoderm, Shh
to beta-Catenin function [41, 52]. Conditional deletion of signals from the notochord promote Bmp expression in the
beta-Catenin in the LPh endoderm delays the fusion of the adjacent mesenchyme to induce chondrogenesis and expres-
lateral walls, which leads to the absence of the tracheoe- sion of cartilage-specific genes such as Sox9 [101]. Simi-
sophageal septum and defective VF separation resembling larly, Sox9 expression could be promoted by Fgf signaling.
milder to severe cases of laryngeal webs [52]. The delay in Explants of airway structures including the larynx cultured
EL formation is more than likely caused by a decrease in in the exogenous FGF18 demonstrated hyperplastic growth
cell proliferation in epithelial and mesenchymal cells medi- of laryngeal and tracheal cartilages and increased expression
ated by the interaction between Cyclin D1, a downstream of Sox9 [22].
target of beta-Catenin, and cell cycle inhibitor p27kip and Sox9 has been identified as an early marker and impor-
by the defective differentiation of VF basal progenitors into tant regulator of cartilage differentiation in both mice and
functional p63 + cytokeratin (K)8-basal cells capable of humans [2, 8, 60]. Humans and transgenic mice with hap-
stratification. Moreover, downregulation of beta-Catenin in losufficiency of Sox9 expression have, among other skel-
the LPh endoderm had a secondary effect on differentia- etal defects, upper airway defects, secondary hypoplasia of
tion of mesenchymal cells in the LP, which could also act laryngeal and tracheal cartilages [23]. Similarly, in mice,
on EL recanalization [52]. Involvement of mesenchymal conditional deletion of Sox9 using Wnt1Cre+/− results in
structures in EL recanalization has been confirmed in other specific loss of the TC [60]. Interestingly, migration and

13
Mechanisms of larynx and vocal fold development and pathogenesis

localization of Wnt1Cre+/− Sox9Flox/− NCC demonstrate that explanation is that laryngeal muscles evolve from individual
NCC which successfully reach their laryngeal destination mesenchymal masses. In a mouse model, double staining for
lose their chondrogenic potential. During chondrogenesis, distribution of SOX9+ chondrocytes and MF-20+ myoblasts
Sox9 is co-expressed with Col2a1, the gene encoding type-II in developing LP supports the second theory of individual
collagen, a major cartilage matrix protein, which is a direct mesenchymal masses. The TA muscle initiates its differ-
regulatory target of Sox9 [8]. In humans and mice, a total entiation during the EL establishment at E11.5 along with
loss of type-II collagen function in null mutants results in the differentiation of AC and the TC, while dorsal intrinsic
severe skeletal and cartilaginous abnormalities but individu- laryngeal muscles develop two days later along with the tra-
als heterozygous for Col2al are relatively mildly affected cheoesophageal septum and CC [52].
[47, 75]. Simultaneous with differentiation of laryngeal carti-
Other signaling pathways that have been shown to regu- lages and muscles, axons invade the mesenchyme of the
late the development and differentiation of laryngeal carti- LP [72]. In a rat model of laryngeal muscle innervation,
lages include Wnt via R-spondin 2 and Eph/EphrinB2 sign- staining for neuronal class III β-tubulin confirmed the pres-
aling. Whole animal loss-of-function R-spondin 2 leads to ence of axons in the LP during the establishment of the EL,
laryngo-tracheal malformations [9]. In these animals, aryt- which correlates with the onset of myogenesis in the VF.
enoids fused to the ventral part of the cricoid, and the dorsal Nerve growth cones enter the larynx from the dorso-lateral
aspect of the cricoid was absent. Defective Eph/Ephrin B2 region in a time- and muscle-specific manner [72]. After
signaling affects the midline fusion of the CC in the dorsal complete EL recanalization (E21 in a rat, which correlates
laryngotracheal wall producing posterior laryngeal clefts with E17.5–18.5 in a mouse), neuromuscular junctions are
[63]. Patients with laryngeal clefts suffer from dysphagia, present with a single axon innervating multiple motor end
with possible aspiration, and often have associated neuro- plates arranged in a central band within the muscle. In verte-
logical disorders [15, 63], which corresponds to the role of brate embryogenesis, axon guidance is highly conserved and
Ephrins in the septation events [20] as well as in the axon controlled via extracellular chemoattractants such as netrins,
guidance in the central and peripheral nervous system [19, Slits, semaphorins, ephrins [19] and nerve growth factors
63]. (NGF) [94]. Delay or errors in the development of the neuro-
Immaturity of neuromuscular control along with possi- logical control of the laryngeal musculature may lead to the
ble defects in chondrification may also contribute to laryn- collapse of laryngeal tissues as described in laryngomalacia
gomalacia [44]. Laryngomalacia is the most common laryn- [44] or defects in VF mobility, known as VF paralysis [39].
geal anomaly in infants [7], which can occur as an isolated This type of disorder can be either unilateral or bilateral and
anomaly or as a part of malformation complexes. Patients is associated with neonatal stridor and airway obstruction.
with laryngomalacia exhibit omega shaped epiglottis, redun- Approximately half of all cases of bilateral VF paralysis
dant bulky arytenoids and inward collapse of the arytenoid are congenital followed by birth trauma or idiopathic [18].
folds, causing inspiratory stridor and airway obstruction In humans, novel chromosomal translocation between chro-
[90]. Molecular data are available for syndromic type of mosomes 5 and 14 has been identified in familial congenital
laryngomalacia, in DiGeorge or Bardet–Biedl syndromes, bilateral VF paralysis [39]. There are currently no molecu-
as these syndromes are associated with malacic bifid epiglot- lar data in animal models that elucidate the temporo-spatial
tis [16, 21, 85, 92]. relationship between myogenic progenitors and ingrowing
peripheral recurrent nerve to validate human data and pro-
Differentiation of muscles and nerves pose other mechanisms responsible for defects in laryngeal
myogenesis and neuromuscular control.
Along with laryngeal cartilages, mesenchymal cells in the
LP differentiate into intrinsic laryngeal muscles. These Differentiation of fibroblasts in the LP
muscle cells do not show overt signs of muscle organiza- and extracellular matrix deposition
tion, such as alignment of primary myocytes, until adja-
cent cartilaginous structures have begun to condense [52]. Considerably less is known about the development and dif-
There are two theories to explain the genesis of laryngeal ferentiation of the fibroblasts in the LP and proteins they
muscles [84]. The sphincter theory states that the larynx is secrete into the extracellular matrix (ECM), such as collagen
surrounded throughout its initial stages of development by and elastin. Abnormalities in production of these proteins
two mesenchymal zones or sphincters, one outside and one can negatively influence VF biomechanics and voice pro-
inside the larynx. The outer zone gives rise to inferior phar- duction [96, 97]. In humans, haplodeficiency in the elastin
yngeal constrictor muscles, the CT and the TC; while, the gene expression is associated with Supravalvular aortic ste-
inner zone gives rise to the TA, lateral and dorsal intrinsic nosis and Williams syndrome. These patients have abnor-
laryngeal muscles, and remaining cartilages. The alternate mal, rough, and hoarse voices as compared to normals, and

13
V. Lungova, S. L. Thibeault

have greater instability of fundamental frequency during closely associated with respiratory, cardiac or craniofa-
phonation [96]. Molecular data of patients with Williams cial development. The unique morphogenetic event, which
syndrome revealed deletion of the critical region at the chro- seems to be critical for the VF development, is the estab-
mosome 7q11.23 which includes the elastin gene suggesting lishment of the EL. Why the lateral walls of the primitive
that abnormal VF elastin production may account for hoarse LPh temporarily fuse and obliterate the ventral lumen is
voice [93]. Future research regarding the molecular mecha- still a mystery. At the moment, we can only guess that EL
nisms of ECM stratification and remodeling is necessary to formation may be required for symmetric development of
understand VF tissue biomechanics to generate strategies the VF and supportive mesenchymal structures in the LP
for its restoration. to ensure that the VF meet midline and close the glottis
All genes and signaling pathways known so far that par- after they fully separate. EL formation likely starts with
ticipate in the larynx and VF morphogenesis are summarized signals from the epithelium, but how these signals are
in the Table 1. translated into mesenchymal cells is not known. Similarly,
involvement of mesenchymal structures in EL formation
and recanalization remains to be investigated.
Summary and future directions Determination of the contribution of other cell types in
laryngeal and VF embryogenesis is also very important.
Despite recent investigations, we are only beginning to These cell types include: (1) VF fibroblasts in the LP for
understand the cellular and molecular mechanisms of the studying molecular basis of ECM compartmentalization
larynx and VF development. As we have seen, the larynx with different biomechanical properties, (2) invading axons
is composed of many closely related interdependent tissues for investigation of mechanisms of recurrent nerve innerva-
and cell types—epithelial cells, fibroblasts, and different tion and (3) endothelial cells for studying the development
kinds of ECM, cartilages, muscle tissue, and nerves. To of VF vasculature. Besides supplying tissues with nutrients
move further in our understanding of the mechanisms of and oxygen, blood brings immune cells into the VF region.
the laryngeal embryogenesis, we need to know much more Further research is needed to examine the contribution of
about the transcription factors and signaling pathways that immune cells to laryngeal and VF morphogenesis.
mediate the reciprocal interactions between these progeni- From the clinical perspective, further investigations into
tors. So far, the focus has been on the specification of the candidate genes and signaling molecules responsible for
VF epithelium and the early stages of the primitive LPh defective phenotypes described here may allow for improved
remodeling. Particularly, because these early morphoge- genetic testing, better prenatal counseling, and potentially
netic events occur during periods of organogenesis are better insight into disease pathophysiology and mechanisms

Table 1  Summary of genes and signaling pathways that are known to control murine larynx and vocal fold development
Stages Duration Characteristic events Genes and signaling molecules

Specification of E9.5–E10.5 Formation of the primitive LPh Shh, Gli1, Gli2, Gli3, Nkx2-1, Sox2, FoxA2,
laryngeal and VF Apposition of lateral walls Fuz, Myf5, RA, Tbx1, Hox genes, Fgf, Wnt/
progenitors beta-Catenin
Migration of NCCs and myoblasts into the
laryngeal region
EL formation E11.5–E12.5 Fusion of lateral walls Wnt/beta-Catenin, Sox2, Nkx2-1, FoxA2, p63,
Differentiation of VF basal epithelial pro- K8, Cyclin D1, R-spondin 2, Bmp, Sox9,
genitors Fgf18, Col2A1, Ephrin B2/Eph, MF-20
Initiation of differentiation of cartilages and
muscles
EL recanalization E13.5–E18.5 Formation of the tracheoesophageal septum Foxa2, Wnt/beta-Catenin, Sox2, Sox9, MF-20,
VF separation Formation of the laryngeal cecum and phar- Col2a1, p63, K8, K5, K14
yngoglottic duct
Initiation of stratification of VF epithelium
EL recanalization
Differentiation of laryngeal cartilages, intrin-
sic laryngeal muscles and neurons
Maturation Birth to postnatal stages Stratification of the VF epithelium p63, K5, K14, Sox2, Sox9, MF-20
·Differentiation and stratification of the
lamina propria

13
Mechanisms of larynx and vocal fold development and pathogenesis

of congenital laryngeal disorders. Similarly, candidate genes the mouse Tbx1 gene. Genomics 43(3):267–277. https​://doi.
and signaling pathways active during VF development may org/10.1006/geno.1997.4829
17. Connor NP et al (2002) Neuromuscular junction changes in
allow for improved therapies for postnatal VF repair and aged rat thyroarytenoid muscle. Ann Otol Rhinol Laryngol
regeneration for both the LP and epithelium. 111(7):579–586. https​://doi.org/10.1177/00034​89402​11100​703
18. Daya H et al (2000) Pediatric vocal fold paralysis: a long-
Acknowledgements  This work was supported by grants NIH NIDCD term retrospective study. Arch Otolaryngol Head Neck Surg
R01 DC004336 and R01 DC012773. 126(1):21–25. https​://doi.org/10.1001/archo​tol.126.1.21
19. Dickson BJ (2002) Molecular mechanisms of axon guidance.
Science 298:1959–1964
20. Dravis C, Henkemeyer M (2011) Ephrin-B reverse signaling
References controls septation events at the embryonic midline through
separate tyrosine phosphorylation-independent signaling
1. Ahmad SM, Soliman AMS (2007) Congenital anomalies of avenues. Dev Biol 355(1):138–151. https​://doi.org/10.1016/j.
the larynx. Otolaryngol Clin N Am 40(1):177–191. https​://doi. ydbio​.2011.04.020
org/10.1016/j.otc.2006.10.004 21. Ebert B et al (2018) Congenital and iatrogenic laryngeal and
2. Akiyama H et  al (2002) The transcription factor Sox9 has vocal abnormalities in patients with 22q11.2 deletion. Int J
essential roles in successive steps of the chondrocyte differ- Pediatr Otorhinolaryngol 109(March):17–20. https​: //doi.
entiation pathway and is required for expression of Sox5 and org/10.1016/j.ijpor​l.2018.03.006
Sox6. Genes Dev 16(21):2813–2828. https​://doi.org/10.1101/ 22. Elluru RG, Thompson F, Reece A (2009) Fibroblast growth
gad.10178​02 factor 18 gives growth and directional cues to airway cartilage.
3. Andreatta RD et al (2002) Mucosal afferents mediate laryngeal Laryngoscope 119(6):1153–1165. https​: //doi.org/10.1002/
adductor responses in the cat. J Appl Physiol 93(5):1622–1629. lary.20157​
https​://doi.org/10.1152/jappl​physi​ol.00417​.2002 23. Elluru RG, Whitsett JA (2014) Potential role of Sox9 in pat-
4. Arsić D et al (2003) Differences in the levels of sonic hedge- terning tracheal cartilage ring formation in an embryonic
hog protein during early foregut development caused by expo- mouse model Ravindhra. Arch Otolaryngol Head Neck Surg
sure to Adriamycin give clues to the role of the Shh gene in 130(6):732–736. https​://doi.org/10.1038/jid.2014.371
oesophageal atresia. Pediatr Surg Int 19(6):463–466. https​:// 24. Evans DJR, Noden DM (2006) Spatial relations between avian
doi.org/10.1007/s0038​3-003-0959-8 craniofacial neural crest and paraxial mesoderm cells. Dev Dyn
5. Aubin J et al (1997) Early postnatal lethality in Hoxa-5 mutant 235(5):1310–1325. https​://doi.org/10.1002/dvdy.20663​
mice is attributable to respiratory tract defects. Dev Biol 25. Fokstuen S, Bottani A, Medeiros PF, Antonarakis SE, Stoll
192(2):432–445. https​://doi.org/10.1006/dbio.1997.8746 CSA (1997) Laryngeal atresia type III (glottic web) with
6. Ban MJ et al (2017) The efficacy of fibroblast growth fac- 22q11.2 microdeletion: report of three patients. Am J Med
tor for the treatment of chronic vocal fold scarring: from ani- Genet 70(2):130–133
mal model to clinical application. Clin Exp Otorhinolaryngol 26. França MM et al (2010) Novel heterozygous nonsense GLI2
10(4):349–356. https​://doi.org/10.21053​/ceo.2016.00941​ mutations in patients with hypopituitarism and ectopic pos-
7. Bedwell J, Zalzal G (2016) Laryngomalacia. Semin Pediatr terior pituitary lobe without holoprosencephaly. J Clin
Surg 15(1):119–122 Endocrinol Metab 95(11):384–391. https​://doi.org/10.1210/
8. Bell DM et al (1997) SOX9 directly regulates the type-II col- jc.2010-1050
lagen gene. Nat Genet 16(2):174–178. https​://doi.org/10.1038/ 27. Franco B, Thauvin-Robinet C (2016) Update on oral-facial-
ng069​7-174 digital syndromes (OFDS). Cilia BioMed Central 5(1):1–11.
9. Bell SM et al (2008) R-spondin 2 is required for normal laryn- https​://doi.org/10.1186/s1363​0-016-0034-4
geal-tracheal, lung and limb morphogenesis. Development 28. Frisdal A, Trainor PA (2014) Development and evolution of
135(6):1049–1058. https​://doi.org/10.1242/dev.01335​9 the pharyngeal apparatus. Wiley Interdiscip Rev Dev Biol
10. Bergrin M et al (2006) Three-dimensional compartmentaliza- 3(16):403–418. https​://doi.org/10.1038/jid.2014.371
tion of myosin heavy chain and myosin light chain isoforms 29. Garcia-Martinez V, Alvarez IS, Schoenwolf GC (1993) Loca-
in dog thyroarytenoid muscle. Am J Physiol Cell Physiol tions of the ectodermal and nonectodermal subdivisions of the
290(5):1446–1458. https​://doi.org/10.1152/ajpce​ll.00323​.2005 epiblast at stages 3 and 4 of avian gastrulation and neurulation.
11. Bhattacharyya N (2015) The prevalence of pediatric voice J Exp Zool. https​://doi.org/10.1002/jez.14026​70409​
and swallowing problems in the United States. Laryngoscope 30. Goding GS, Al-Sharif KI, McLoon LK (2005) Myonuclear
125(3):746–750. https​://doi.org/10.1002/lary.24931​ addition to uninjured laryngeal myofibers in adult rabbits.
12. Bless DM et al (2004) Growth factor therapy for vocal fold scar- Ann Otol Rhinol Laryngol 114(7):552–557. https ​ : //doi.
ring in a canine model. Ann Otol Rhinol Laryngol 113(10):777– org/10.1177/00034​89405​11400​711
785. https​://doi.org/10.1177/00034​89404​11301​002 31. Goss AM et al (2009) ‘Wnt2/2b and beta-catenin signaling are
13. Bose J (2002) Pallister–Hall syndrome phenotype in mice necessary and sufficient to specify lung progenitors in the fore-
mutant for Gli3. Hum Mol Genet 11(9):1129–1135. https​:// gut. Dev Cell 17(2):290–298. https​://doi.org/10.1016/j.devce​
doi.org/10.1093/hmg/11.9.1129 l.2009.06.005
14. Chai Y et al (2000) Fate of the mammalian cranial neural crest 32. Harris-Johnson KS et al (2009) beta-Catenin promotes res-
during tooth and mandibular morphogenesis. Development piratory progenitor identity in mouse foregut. Proc Natl Acad
127(8):1671–1679 Sci USA 106(38):16287–16292. https​: //doi.org/10.1073/
15. Chiang T et al (2014) ‘Surgical management of type I and II pnas.09022​74106​
laryngeal cleft in the pediatric population. Int J Pediatr Otorhi- 33. Hartnick CJ, Cotton RT (2000) Congenital laryngeal anomalies
nolaryngol 78(12):2244–2249. https​://doi.org/10.1016/j.ijpor​ laryngeal atresia, stenosis, webs, and clefts. Otolaryngol Clin
l.2014.10.023 N Am 33:6. https​://doi.org/10.1016/S0030​-6665
16. Chieffo C et  al (1997) Isolation and characterization of a 34. Hayes LL et al (2008) Laryngeal and tracheal anomalies in an
gene from the DiGeorge chromosomal region homologous to infant with oral-facial-digital syndrome type VI (Váradi-Papp):

13
V. Lungova, S. L. Thibeault

report of a transitional type. Pediatr Radiol 38(9):994–998. to smoke exposure. Nat Commun. https​://doi.org/10.1038/s4146​
https​://doi.org/10.1007/s0024​7-008-0877-y 7-019-12069​-w
35. Henick DH (1993) Three-dimensional analysis of murine 54. Lüthe L, Häusler U, Jürgens U (2000) Neuronal activity in the
laryngeal development. Ann Otol Rhinol Laryngol Supplement medulla oblongata during vocalization. A single-unit recording
159:3–24 study in the squirrel monkey. Behav Brain Res 116(2):197–210.
36. Herriges M, Morrisey EE (2014) Lung development: orches- https​://doi.org/10.1016/S0166​-4328(00)00272​-2
trating the generation and regeneration of a complex organ. 55. Mahrt E et al (2016) Mice produce ultrasonic vocalizations by
Development (Cambridge) 141(3):502–513. https ​ : //doi. intra-laryngeal planar impinging jets. Curr Biol 26(19):R880–
org/10.1242/dev.09818​6 R881. https​://doi.org/10.1016/j.cub.2016.08.032
37. Hirano M (1974) Morphological structure of the vocal cord as a 56. Manley NR, Capecchi MR (1997) Hox group 3 paralogous
vibrator and its variations. Folia Phoniatrica Logopaed 26(2):89– genes act synergistically in the formation of somitic and neural
94. https​://doi.org/10.1159/00026​3771 crest-derived structures. Dev Biol 192(2):274–288. https​://doi.
38. Van Houtte E et al (2010) The prevalence of laryngeal pathology org/10.1006/dbio.1997.8765
in a treatment-seeking population with dysphonia. Laryngoscope 57. Marques MJ et al (2007) Intrinsic laryngeal muscles are spared
120(2):306–312. https​://doi.org/10.1002/lary.20696​ from myonecrosis in the mdx mouse model of Duchenne mus-
39. Hsu AK et al (2015) Familial congenital bilateral vocal fold cular dystrophy. Muscle Nerve 35(3):349–353. https​: //doi.
paralysis: a novel gene translocation. Int J Pediatr Otorhinolaryn- org/10.1002/mus.20697​
gol 79(3):323–327. https​://doi.org/10.1016/j.ijpor​l.2014.12.009 58. Marshall H, Morrison A, Studer M, Pöpperl HKR (1996) Reti-
40. Huang R et al (1997) The fate of the first avian somite. Anat noids and Hox genes. FASEB 10(9):969–978
Embryol 195(5):435–449. https​://doi.org/10.1007/s0042​90050​ 59. Miri AK (2014) Mechanical characterization of vocal fold tissue:
063 a review study. J Voice 28(6):657–667. https:​ //doi.org/10.1016/j.
41. Huh SH, Ornitz DM (2010) β-Catenin deficiency causes jvoic​e.2014.03.001
DiGeorge syndrome-like phenotypes through regulation of 60. Mori-Akiyama Y et al (2003) Sox9 is required for determination
Tbx1. Development 137(7):1137–1147. https​://doi.org/10.1242/ of the chondrogenic cell lineage in the cranial neural crest. Proc
dev.04553​4 Natl Acad Sci USA 100(16):9360–9365. https:​ //doi.org/10.1073/
42. Inagi K, Schultz E, Ford CN (1998) An anatomic study of the pnas.16312​88100​
rat larynx: establishing the rat model for neuromuscular func- 61. Morrisey EE, Hogan BLM (2010) ‘Preparing for the first breath:
tion. Otolaryngol Head Neck Surg 118(1):74–81. https​://doi. genetic and cellular mechanisms in lung development. Dev Cell
org/10.1016/S0194​-5998(98)70378​-X 18(1):8–23. https​://doi.org/10.1016/j.devce​l.2009.12.010
43. Kumai Y (2019) Pathophysiology of fibrosis in the vocal fold: 62. Morrow BE et al (2018) Molecular genetics of 22q11.2 deletion
current research, future treatment strategies, and obstacles to syndrome. Am J Med Genet Part A 176(10):2070–2081. https​://
restoring vocal fold pliability. Int J Mol Sci 20:10. https​://doi. doi.org/10.1002/ajmg.a.40504​
org/10.3390/ijms2​01025​51 63. Neilan RE et  al (2012) Characterization of the larynx in
44. Kusak B et al (2017) Types of laryngomalacia in children: inter- Ephrin-B2 knockout mice. Arch Otolaryngol Head Neck Surg
relationship between clinical course and comorbid conditions. 138(10):969. https​://doi.org/10.1001/archo​tol.2013.109
Eur Arch Otorhinolaryngol 274(3):1577–1583. https​: //doi. 64. Niederreither K et al (2003) The regional pattern of retinoic acid
org/10.1007/s0040​5-016-4334-5 synthesis by RALDH2 is essential for the development of poste-
45. Levendoski EE, Leydon C, Thibeault SL (2014) Vocal fold epi- rior pharyngeal arches and the enteric nervous system. Develop-
thelial barrier in health and injury: a research review. J Speech ment 130(11):2525–2534. https​://doi.org/10.1242/dev.00463​
Lang Hear Res 57(3):1679–1691. https​://doi.org/10.1044/2014 65. Noden DM (1983) The embryonic origins of avian cephalic and
46. Li L et al (2016) Tissue engineering-based therapeutic strategies cervical muscles and associated connective tissues. Am J Anat
for vocal fold repair and regeneration. Biomaterials 108:91–110. 168(3):257–276. https​://doi.org/10.1002/aja.10016​80302​
https​://doi.org/10.1016/j.bioma​teria​ls.2016.08.054 66. Noden DM (1986) Patterning of avian craniofacial muscles. Dev
47. Li S, Darwin JP (1995) Transgenic mice with targeted for col- Biol. https​://doi.org/10.1016/0012-1606(86)90138​-7
lagen 11 develop a skeleton with membranous and periosteal 67. Noden DM et al (1999) Differentiation of avian craniofacial
bone but no endochondral bone, pp 2821–2830 muscles: I. Patterns of early regulatory gene expression and
48. Liberty G et al (2013) The fetal larynx and pharynx: structure and myosin heavy chain synthesis. Dev Dyn 216(2):96–112. https​
development on two- and three-dimensional ultrasound. Ultra- ://doi.org/10.1002/(SICI)1097-0177(19991​0)216:2%3c96:AID-
sound Obstet Gynecol 42(2):140–148. https​://doi.org/10.1002/ DVDY2​%3e3.0.CO;2-6
uog.12358​ 68. Noden DM, Trainor PA (2005) Relations and interactions
49. Lipson AH et al (1991) Velocardiofacial (Shprintzen) syndrome: between cranial mesoderm and neural crest populations. J Anat
an important syndrome for the dysmorphologist to recognise. J 207:575–601
Med Genet 28(9):596–604. https:​ //doi.org/10.1136/jmg.28.9.596 69. Ondrey F, Griffith A, Van Waes C, Rudy S, Peters K, McCullagh
50. Litingtung Y et  al (1998) ‘Sonic hedgehog is essential to L, B. L. (2000) Asymptomatic laryngeal malformations are com-
foregut development. Nat Genet 20(1):58–61. https ​ : //doi. mon in patients with Pallister–Hall syndrome. Am J Med Genet
org/10.1038/1717 94(1):64–67
51. Lungova V et  al (2015) Ontogeny of the mouse vocal fold 70. Périé S et al (2000) Myosin heavy chain expression in human
epithelium. Dev Biol 399:2. https​://doi.org/10.1016/j.ydbio​ laryngeal muscle fibers: a biochemical study. Ann Otol Rhinol
.2014.12.037 Laryngol 109(2):216–220. https​://doi.org/10.1177/00034​89400​
52. Lungova V et al (2018) β-Catenin signaling is essential for mam- 10900​218
malian larynx recanalization and the establishment of vocal fold 71. Piekarski N, Olsson L (2007) Muscular derivatives of the cranial-
progenitor cells. Dev (Cambridge) 145:4. https:​ //doi.org/10.1242/ most somites revealed by long-term fate mapping in the Mexican
dev.15767​7 axolotl (Ambystoma mexicanum). Evol Dev 9(6):566–578. https​
53. Lungova V et al (2019) Human induced pluripotent stem cell- ://doi.org/10.1111/j.1525-142X.2007.00197​.x
derived vocal fold mucosa mimics development and responses

13
Mechanisms of larynx and vocal fold development and pathogenesis

72. Pitman MJ et al (2013) Embryologic innervation of the rat laryn- 89. Thomas LB et al (2009) Establishing a new animal model for the
geal musculature—A model for investigation of recurrent laryn- study of laryngeal biology and disease: an anatomic study of the
geal nerve reinnervation. Laryngoscope 123(12):3117–3126. mouse larynx. J Speech Lang Hear Res 52(3):802–811. https​://
https​://doi.org/10.1002/lary.24216​ doi.org/10.1044/1092-4388(2008/08-0087)
73. Riede T (2011) Subglottal pressure, tracheal airflow, and intrin- 90. Thompson DM (2007) Abnormal sensorimotor integrative func-
sic laryngeal muscle activity during rat ultrasound vocalization. tion of the larynx in congenital laryngomalacia: a new theory
J Neurophysiol 106(5):2580–2592. https​://doi.org/10.1152/ of etiology. Laryngoscope 117(6 SUPPL.):1–33. https​://doi.
jn.00478​.2011 org/10.1097/MLG.0b013​e3180​4a575​0
74. Riede T, Borgard HL, Pasch B (2017) Laryngeal airway recon- 91. Titze I, Riede T, Mau T (2016) Predicting achievable fundamen-
struction indicates that rodent ultrasonic vocalizations are pro- tal frequency ranges in vocalization across species. PLoS Com-
duced by an edge-tone mechanism. R Soc Open Sci 4:11. https​ put Biol 12(6):1–13. https​://doi.org/10.1371/journ​al.pcbi.10049​
://doi.org/10.1098/rsos.17097​6 07
75. Ritvaniemi P et al (1995) Identification of col2a1 gene muta- 92. Tsurumi H et al (2010) Bifid epiglottis: syndromic constituent
tions in patients with chondrodysplasias and familial osteoar- rather than isolated anomaly. Pediatr Int 52(5):723–728. https​://
thritis. Arthritis Rheum 38(7):999–1004. https:​ //doi.org/10.1002/ doi.org/10.1111/j.1442-200X.2010.03096​.x
art.17803​80717​ 93. Vaux KK et al (2003) Vocal cord abnormalities in Williams syn-
76. Roberts C et al (2005) Retinoic acid down-regulates Tbx1 expres- drome: a further manifestation of elastin deficiency. Am J Med
sion in vivo and in vitro. Dev Dyn 232(4):928–938. https​://doi. Genet 119A(3):302–304
org/10.1002/dvdy.20268​ 94. Vega-Cordova X et al (2010) Neurotrophin expression of laryn-
77. Rousseau B et al (2004) Characterization of chronic vocal fold geal muscles in response to recurrent laryngeal nerve transec-
scarringin a rabbit model. J Voice 18(1):116–124. https​://doi. tion. Laryngoscope 120(8):1591–1596. https​://doi.org/10.1002/
org/10.1016/j.jvoic​e.2003.06.001 lary.21026​
78. Roy N et al (2005) Voice disorders in the general population: 95. Vermot J et al (2003) Decreased embryonic retinoic acid syn-
prevalence, risk factors, and occupational impact. Laryngoscope thesis results in a DiGeorge syndrome phenotype in newborn
115(11):1988–1995. https:​ //doi.org/10.1097/01.mlg.000017​ 9174​ mice. Proc Natl Acad Sci USA 100(4):1763–1768. https​://doi.
.32345​.41 org/10.1073/pnas.04379​20100​
79. Saga Y et al (1999) MesP1 is expressed in the heart precursor 96. Watts CR, Awan SN, Marler JA (2008) ‘An investigation of voice
cells and required for the formation of a single heart tube. Devel- quality in individuals with inherited elastin gene abnormalities.
opment 126(15):3437–3447 Clin Linguist Phonet 22:3
80. Sanudo J, Domenech-Mateu JM (1990) The laryngeal pri- 97. Watts CR, Marler JA, Rousseau B (2011) Qualitative characteri-
mordium and epithelial lamina. A new interpretation. J Anat zation of elastic fiber distribution in the mouse vocal fold: further
171:207–222 development of an animal model. J Voice 25(1):1–6
81. Shinners MJ, Goding GS, McLoon LK (2006) Effect of recur- 98. Wong NS et al (2019) Patterns of dysphagia and airway protec-
rent laryngeal nerve section on the laryngeal muscles of adult tion in infants with 22q11.2-deletion syndrome. Laryngoscope.
rabbits. Otolaryngol Head Neck Surg 134(3):413–418. https​:// https​://doi.org/10.1002/lary.28317​
doi.org/10.1016/j.otohn​s.2005.11.037 99. Wyatt ME, Hartley BEJ (2005) Laryngotracheal reconstruction
82. Shiotani A, Westra WH, Flint PW (1999) Myosin heavy chain in congenital laryngeal webs and atresias. Otolaryngol Head
composition in human laryngeal muscles. Laryngoscope Neck Surg 132(2):232–238. https​://doi.org/10.1016/j.otohn​
109(9):1521–1524. https​://doi.org/10.1097/00005​537-19990​ s.2004.09.032
9000-00030​ 100. Zaw-Tun HA, Burdi AR (1985) Reexamination of the ori-
83. Silengo MC et al (1987) Oro-facial-digital syndrome II. Tran- gin and early development of the human larynx. Acta Anat
sitional type between the Mohr and the Majewski syndromes: 122(3):163–184
report of two new cases. Clin Genet 31(5):331–336. https​://doi. 101. Zeng L et al (2002) Shh establishes an Nkx3.2/Sox9 autoregula-
org/10.1111/j.1399-0004.1987.tb028​17.x tory loop that is maintained by BMP signals to induce somitic
84. Som PM, Curtin HD (2014) An updated and illustrated review of chondrogenesis. Genes Dev 16(15):1990–2005. https​://doi.
the complex embryology of the larynx and how laryngeal webs, org/10.1101/gad.10080​02
atresias, and stenoses develop. Neurographics 4(4):189–203. 102. Zhang Z (2016) Mechanics of human voice production and
https​://doi.org/10.3174/ng.41400​95 control. J Acoust Soc Ame 140(4):2614–2635. https​: //doi.
85. Stevens CA, Ledbetter JC (2005) Significance of bifid epiglot- org/10.1121/1.49645​09
tis. Am J Med Genet 134 A(4):447–449. https:​ //doi.org/10.1002/ 103. Zhang Z, Huynh T, Baldini A (2006) Mesodermal expression of
ajmg.a.30659​ Tbx1 is necessary and sufficient for pharyngeal arch and cardiac
86. Tabler JM et al (2017) Cilia-mediated hedgehog signaling con- outflow tract development. Development 133(18):3587–3595.
trols form and function in the mammalian larynx. eLife 6:1–26. https​://doi.org/10.1242/dev.02539​
https​://doi.org/10.7554/eLife​.19153​
87. Tateya I et  al (2007) Prenatal vitamin A deficiency causes Publisher’s Note Springer Nature remains neutral with regard to
laryngeal malformation in rats. Ann Otol Rhinol Laryngol jurisdictional claims in published maps and institutional affiliations.
116(10):785–792
88. Thomas LB et al (2008) Laryngeal muscles are spared in the
dystrophin deficient mdx mouse. J Speech Lang Hear Res
51(3):586–595. https​://doi.org/10.1044/1092-4388(2008/042)

13

You might also like