Peran Hormon Thyroid Pada Craniofacial

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

REvIEWS

Role of thyroid hormones in


craniofacial development
Victoria D. Leitch 1,2
, J. H. Duncan Bassett 1
* and Graham R. Williams 1
*
Abstract | The development of the craniofacial skeleton relies on complex temporospatial
organization of diverse cell types by key signalling molecules. Even minor disruptions to these
processes can result in deleterious consequences for the structure and function of the skull.
Thyroid hormone deficiency causes delayed craniofacial and tooth development, dysplastic
facial features and delayed development of the ossicles in the middle ear. Thyroid hormone
excess, by contrast, accelerates development of the skull and, in severe cases, might lead to
craniosynostosis with neurological sequelae and facial hypoplasia. The pathogenesis of these
important abnormalities remains poorly understood and underinvestigated. The orchestration
of craniofacial development and regulation of suture and synchondrosis growth is dependent
on several critical signalling pathways. The underlying mechanisms by which these key pathways
regulate craniofacial growth and maturation are largely unclear, but studies of single-​gene
disorders resulting in craniofacial malformations have identified a number of critical signalling
molecules and receptors. The craniofacial consequences resulting from gain-​of-function and
loss-​of-function mutations affecting insulin-​like growth factor 1, fibroblast growth factor
receptor and WNT signalling are similar to the effects of altered thyroid status and mutations
affecting thyroid hormone action, suggesting that these critical pathways interact in the
regulation of craniofacial development.

Craniosynostosis
The development of the craniofacial skeleton is complex involved. We then consider the physiological control of
Premature fusion of the fibrous and relies on the temporospatial organization of diverse thyroid status by the hypothalamic–pituitary–thyroid
calvarial sutures. cell types by key signalling molecules. Even minor dis- (HPT) axis and the regulation of thyroid hormone
ruptions to these intricate and integrated processes can (3,5,3′-l-​triiodothyronine, T3) action in bone. In subse-
Facial hypoplasia
Reduced growth of features in
result in deleterious consequences for the structure and quent sections, we review the consequences of thyroid
the midface, which results in an function of the skull. hormone deficiency and excess and the effect of genetic
abnormal facial appearance. The mechanisms of thyroid hormone action in bone disorders that alter T3 action on the development of the
and cartilage, and the consequences of thyroid hor- craniofacial skeleton in mouse models and human dis-
mone deficiency and excess on the skeleton, have been ease. Finally, we discuss current understanding of the
thoroughly reviewed1. Nevertheless, and even though cellular and molecular mechanisms of T3 action in bone
the skull is exquisitely sensitive to changes in thyroid and cartilage in the context of craniofacial development.
1
Molecular Endocrinology hormone availability, the effects of thyroid hormones
Laboratory, Department of on craniofacial development have not been considered Craniofacial development
Metabolism, Digestion and systematically. Thyroid hormone deficiency causes Development of the craniofacial skeleton begins by
Reproduction, Imperial
College London, London, UK.
delayed craniofacial and tooth development, dysplastic 4 weeks of gestation in humans, with the formation
facial features and delayed development of the ossicles of distinct condensations of mesoderm-​derived and
2
Royal Melbourne Institute
of Technology (RMIT) Centre in the middle ear. By contrast, high levels of thyroid neural crest-​derived mesenchyme, and continues into
for Additive Manufacturing, hormone accelerate development of the skull and can adulthood2,3. The skull is uniquely formed by a com-
RMIT University, Melbourne, lead to craniosynostosis , neurological sequelae and plex integration of bones that have diverse origins and
VIC, Australia. facial hypoplasia in severe cases. The pathogenesis of develop from both intramembranous and endochondral
*e-​mail: d.bassett@ these important abnormalities is poorly understood and ossification (Fig. 1a; Table 1).
imperial.ac.uk;
underinvestigated.
graham.williams@
imperial.ac.uk In this Review, we first discuss the processes of Intramembranous ossification. Intramembranous ossifi-
https://doi.org/10.1038/ craniofacial bone formation via intramembranous and cation is the osteoblast-​driven process that forms the vis-
s41574-019-0304-5 endochondral ossification and the signalling pathways cerocranium and cranial vault, together with the scapula,

Nature Reviews | Endocrinology


Reviews

Key points and posterior regions of the supraorbital ridge, respec-


tively, to form the primordia of the frontal and parietal
• Thyroid hormone deficiency during development results in delayed intramembranous bones16,17 (Fig. 1d). The frontal and parietal bone pri-
and endochondral ossification in the skull, which manifests as patent or persistent mordial cells begin to express bone-​specific markers at
fontanelles, patent sutures, delayed dentition, wormian bones and deafness. E10.5–12.5. Mineralization of these rudiments begins
• Thyroid hormone excess during development results in advanced intramembranous at E14.0–14.5 via intramembranous ossification17–20.
and endochondral ossification in the skull, which manifests as premature fusion of Formation of the occipital bone during embryogene-
the calvarial sutures and cranial base synchondroses.
sis involves five distinct rudiments: four of these develop
• These characteristic craniofacial malformations indicate that thyroid hormones have a via endochondral ossification within the cranial base,
pivotal role in development and growth of the craniofacial skeleton and demonstrate
whereas the interparietal section develops by intramem-
that the skull is exquisitely sensitive to changes in thyroid status.
branous ossification to form part of the cranial vault3.
• Thyroid hormone-​induced changes in fibroblast growth factor receptor, insulin-​like
Like the occipital bone, the temporal bone comprises
growth factor 1 and WNT signalling in osteoblasts and chondrocytes indicate
that these pathways are intricately involved in the regulation of craniofacial
five components that include the bones of the inner ear.
development by T3. The squamous and tympanic sections of the temporal
bone form from neural crest cells and develop by intra­
membranous ossification, while the remainder form via
ilium and lateral two-​thirds of the clavicle4,5. The process endochondral ossification2. The interparietal occipital
begins with aggregation of mesenchymal cells to form bone is largely mesodermal in origin, with additional
dense, tightly packed condensations. Cell-​surface hepa- areas of neural crest-​derived tissue18. The interparietal
ran sulfate proteoglycans (HSPGs), neural cell adhesion occipital bone develops from two, or sometimes three,
molecule, N-​cadherin, other proteoglycans, hyaladher- ossification centres that fuse together to form a single
ins, versican and tenascin all contribute to the cell–cell bone, with the midline of the two ossification centres
adhesions that develop during mesenchyme conden- housing the neural crest-​derived cells18,21. Occasionally,
sation6–8. At the centre of these condensations, mes- extra centres of intramembranous ossification form
enchymal cells differentiate directly to pre-​osteoblasts in the calvaria and eventually become wormian bones3.
and then osteoblasts that express runt-​related tran- The reason for wormian bone formation is unknown,
scription factor 2 (RUNX2) and secrete type I collagen but it has been postulated to correct or compensate for
(COL1A1) and other extracellular matrix proteins, increased suture or fontanelle width in conditions in
including osteocalcin, bone morphogenetic protein 2 which development of the skull is delayed22. Continued
(BMP2), transforming growth factor-​β (TGFβ) and growth of calvarial bones occurs via proliferation of
insulin-​like growth factor 1 (IGF1)9. Calcium phos- osteoprogenitor cells at the osteogenic front. When the
phate is then deposited on the extracellular matrix to osteogenic fronts meet, they either merge and ossify
form mineralized bone2,6,10 (Fig. 1b). the margin, or form a suture3.

Endochondral ossification. Some bones of the cranial Sutures. Sutures comprise two osteogenic fronts and a
base, the temporal bones and the bones of the middle region of fibrous mesenchyme encased by the pericra-
ear develop by endochondral ossification. This process nium and dura mater (Fig. 1b). Importantly, sutures con-
also occurs in the long bones and throughout the rest tain cells at various stages of differentiation, including
of the skeleton. During endochondral ossification, cells osteoprogenitors, osteoblasts, osteoclasts, fibroblasts and
at the centre of mesenchyme condensations differentiate osteocytes3. At birth, the calvarium has paired coronal
into chondrocytes, which secrete a cartilaginous extra- and lambdoid sutures, a sagittal suture and a metopic
cellular matrix rich in type II collagen (COL2A1) that suture, together with an anterior and posterior fontanelle
forms an anlage, or template, for the developing bone. where the sutures intersect. Like the craniofacial bones,
These chondrocytes undergo proliferation, maturation the mesenchymal cells contained within the suture
and hypertrophic differentiation, ultimately secreting derive from either the neural crest or mesoderm; these
type X collagen (COL10A1), alkaline phosphatase (ALP) developmental origins influence the pathogenesis of
and angiogenic factors, including vascular endothelial suture disorders such as the craniosynostosis syndromes.
growth factor (VEGF)11,12. Hypertrophic chondrocytes The sutures normally remain patent (that is, open and
might apoptose or transdifferentiate to become osteo- capable of growth) during neonatal and early develop-
blasts, and additional osteoblast and osteoclast progen- ment to enable rapid expansion of the brain during the
itor cells arrive via newly formed blood vessels13–15. The postnatal period. The fontanelles are the first regions to
cartilage anlage mineralizes, forming a template for bone fuse and ossify, with closure of the posterior and ante-
formation and, finally, remaining cartilage is resorbed rior fontanelles in humans completed by 3 months and
by osteoclasts to leave mineralized bone matrix (Fig. 1c). 2 years of age, respectively. The age at which calvarial
suture fusion occurs, however, is variable, although it
Intramembranous cranial vault. The cranial vault com- is generally agreed that the metopic suture fuses dur-
Calvaria prises paired frontal and parietal bones, the interpari­ ing childhood in humans, whereas the coronal, sagittal
The upper part of the skull that etal section of the occipital bone and the squamous and lambdoid sutures remain patent into adulthood3,16.
surrounds the brain. sections of the temporal bones (Fig. 1a). Development of Similarly, the posterior frontal suture in mice, which
Wormian bones
the cranial vault begins between embryonic day (E) 8.0 corresponds to the metopic suture in humans, fuses by
Additional bones that form and E9.5 in mice, with migration of neural crest and postnatal day (P) 40, whereas the sagittal, coronal and
between sutures in the skull. mesodermal cells from the mid-​hindbrain to the anterior lambdoid sutures are still patent at P250 (ref.23).

www.nature.com/nrendo
Reviews

a Metopic suture Ethmoid bone Fronto-


ethmoidal
Frontal synchondrosis
bone
Spheno-frontal
Frontal synchondrosis
Frontal bone
bone Sphenoid Spheno-
Parietal bone Coronal bone
suture occipital
synchondrosis
Anterior-
intraoccipital
Temporal synchondrosis
bone Parietal
bone Sagittal Posterior-
suture intraoccipital
synchondrosis
Occipital
Occipital bone bone
Lambdoid
suture Occipital
bone

Suture – intramembranous ossification Synchondroses – endochondral ossification


Osteoblast HZ PZ RZ PZ HZ
Osteoprogenitor
Mesenchymal
cell

Osteocyte Osteoid

Chondrocyte Osteoblast

Mesoderm Neural crest


Mixed mesoderm and neural crest

Fig. 1 | craniofacial development. a | Schematic representation of the skull showing the bones of the cranial vault and
cranial base together with the associated sutures and synchondroses. Intramembranous ossification in the sutures of
the cranial vault and endochondral ossification in the synchondroses of the cranial base are also shown. Purple cells
represent osteoblasts and the blue cells are chondrocytes at different stages of maturation. b | Mesodermal and neural
crest origin of the bones of the skull. HZ, hypertrophic zone; PZ, proliferative zone; RZ, resting zone.

Endochondral cranial base. The cranial base com- By E16 in mice, the 14 cartilage plates of the cranial base
prises the basioccipital, sphenoid and ethmoid bones, have fused to form one large cartilaginous structure,
together with part of the temporal bone3 (Fig.  1a) . the chondrocranium, which covers the entire base of the
These bones derive from mesenchymal condensations skull24,27,28. Ossification centres form in the chondrocra-
at the anterior (neural crest-​derived) and posterior nium and undergo endochondral ossification to form
(mesoderm-​derived) regions of the cranial base24 (Fig. 1d). the bones of the skull base. Between these bones, areas
Chondrocyte differentiation occurs at the centre of the of cartilage persist. Like the sutures between the intra­
condensations and the newly differentiated chondro- membranous bones, these regions, the synchondroses,
cytes secrete extracellular matrix rich in COL2A1 and are critical for continued growth during postnatal matu­
proteoglycans to form the cartilage template11,12,25,26. ration. Along with the cranial base, the ossicles of the

Nature Reviews | Endocrinology


Reviews

middle ear (malleus, incus and stapes) and parts of the cranial base abnormalities37, which might also be accom-
mandible also develop by endochondral ossification29,30. panied by serious comorbidities, including the Chiari
malformation at the base of the brain38.
Synchondroses. Chondrocytes align within synchon-
droses to form structures that are analogous to the epi- Key signalling pathways
physeal growth plates in long bones. The organization of The orchestration of craniofacial development and
proliferating and differentiating chondrocytes in these regulation of suture and synchondrosis growth is depen­
regions drives orientation and the anterior–posterior dent on several critical signalling pathways (Fig. 2). The
elongation of the cranial base28 (Fig. 1c). The three syn- underlying mechanisms by which these key pathways
chondroses responsible for the majority of the growth regulate craniofacial growth and maturation are largely
of the cranial base are the spheno-​occipital, spheno-​ unclear, but in single gene disorders resulting in cranio­
frontal and fronto-​ethmoidal. There are contradictory facial malformations, a number of critical signalling
reports in the literature regarding the age at which the molecules and receptors have been identified.
synchondroses fuse, although the consensus indicates
that the spheno-​frontal and fronto-​ethmoidal synchon- Fibroblast growth factor signalling. Mutations in the
droses start to fuse around 7 years of age3. By contrast, genes that encode fibroblast growth factor (FGF) recep-
the spheno-​occipital synchondrosis remains patent up to tors (FGFRs) cause over 20 craniofacial syndromes,
between 14 and 25 years of age31,32. In mice, the spheno-​ the most common of which are FGFR2-related cranio-
occipital synchondroses start to fuse at approximately synostoses39. Humans express 18 FGF ligands and four
P28 and the process is completed by P84 (ref.33). FGFRs, with each ligand binding to one or more of the
receptors. Specific FGFs can act as either endocrine
Abnormal craniofacial development or paracrine factors binding to FGFRs in a complex
Both genetic and environmental factors influence the combined with heparin and/or heparan sulfates and
processes of craniofacial formation and can cause serious β-​Klotho that results in an FGF–FGFR co-receptor with
deleterious effects to the developing and growing cranio- increased β-​glucuronidase activity40. FGFRs are tyro­
facial skeleton. Premature fusion of the calvarial sutures, sine kinase receptors that span the cell membrane; after
craniosynostosis, is one of the most common malfor- binding FGFs a cascade of intracellular signalling events
mations, occurring in approximately 1 in 2,500 births16. is initiated, including activation of the phosphoinositide
Fusion can occur in one or multiple sutures and results 3-kinase (PI3K), signal transducer and activator of tran-
in compensatory growth in other areas of the skull. scription protein (STAT) and mitogen-​activated protein
This premature fusion can also result in complications, kinase (MAPK) pathways40.
including raised intracranial pressure with neurological The FGFRs are highly homologous, with three of the
and cognitive developmental delay16. Delayed growth four isoforms (FGFR1, FGFR2 and FGFR3) expressed
at the suture manifests as large or persistent fontanelles in the skull. Of the 18 FGF ligands in humans, six have
and is a feature of a number of disorders, including been identified in the skull. FGF2, FGF4 and FGF9 are
cleidocranial dysplasia, achondroplasia, Down syndrome expressed in the suture mesenchyme, FGF8 and FGF18
and rickets34. are expressed in osteoblasts lining the bones of the cal-
Premature fusion of the synchondroses can change varia and FGF10 is expressed in the palate during early
the angle of the cranial base, which might cause retrogn­ embryogenesis41–43. FGF2, FGF4 and FGF8 can bind all
athia, malocclusion, midface hypoplasia or cleft palate35,36. three receptor isoforms expressed in the skull, FGF9
The incidence of these malformations has not been and FGF18 can only bind to FGFR2 and FGFR3, and
thoroughly investigated, but patients with syndromic FGF10 binds FGFR1 and FGFR2 (ref.44).
craniosynostosis are at increased risk of accompanying FGFR1 is expressed in the suture mesenchyme, in
osteoblasts at the osteogenic fronts and in chondrocytes
Table 1 | Timing of key craniofacial events in humans and mice in the cranial base. FGFR1 gain-​of-function mutations
lead to Pfeiffer (OMIM 101600) and Jackson–Weiss
developmental stage or event Age in humans Age in mice (OMIM 123150) syndromes, which are both charac-
Neural crest migration 21 days pc E8.0 terized by premature fusion of the calvarial sutures
Mesodermal and neural crest mesenchymal cell 28 days pc E12.5 and, in some cases, shortening of the cranial base35,41.
condensations Fgfr1 gain-​of-function mutations in mice also result in
Deciduous tooth buds begin to form 40 days pc E13.0 craniosynostosis of both the calvaria and cranial base,
via upregulation of Runx2 (ref.45). Conversely, loss-​
Ossification centre of mandible body appears 6–7 weeks pc E14.0
of-function mutations in FGFR1 are rare, but can result
Ossification centres of frontal, parietal, occipital 8 weeks pc E14.0 in hypomineralization of calvarial bones, micrognathia
(interparietal) and nasal bones appear
and dental agenesis46,47.
Formation of the chondrocranium begins 40 days pc E16.0 FGFR2 is expressed in proliferating osteoprogen-
Metopic suture fusion (posterior frontal in mouse) 2 years P10 itors, differentiating osteoblasts and chondrocytes at
Dental eruption (first molar) 6–7 years P20 the cranial base. FGFR2 gain-​of-function mutations
cause a variety of craniosynostosis syndromes including
Spheno-​occipital synchondrosis fusion 12–14 years P28 Crouzon (OMIM 123500), Pfeiffer (OMIM 101600),
Coronal, lambdoid and sagittal suture fusion Adulthood After P250 Apert (OMIM 101200) and Jackson–Weiss (OMIM
E, embryonic day; P, postnatal day; pc, post coitum. 123150) syndromes. Gain-​of-function mutations in

www.nature.com/nrendo
Reviews

Mesenchymal condensation Fig. 2 | Key signalling pathways in craniofacial


Mesenchymal cell
development. FGF–FGFR, IGF1–IGF1R, BMP and WNT
signalling pathways regulate key stages in chondrocyte and
osteoblast specification, proliferation and differentiation.
Green boxes indicate stages where the listed growth
factors promote the process and red boxes indicate stages
• SOX5 • RUNX2 where they prevent it. BMP, bone morphogenetic protein;
WNT • SOX6 • TWIST1 BMP
FGF, fibroblast growth factor; FGFR, fibroblast growth
• SOX9 • MSX2
factor receptor; IGF1R, insulin-​like growth factor 1
• WNT receptor; MSX2, msh homeobox 2; RUNX2, RUNX family
• FGFR2 transcription factor 2; SOX, SRY-​box transcription factor;
• IGF1R
TWIST1, Twist family bHLH transcription factor 1; WNT,
Osteoprogenitor WNT family member.

RUNX2 in hypomineralization of calvarial bones, micrognathia


and dental agenesis46,47.
Chondroprogenitor
• WNT1 FGFR3 is expressed in the cranial base and at low
• WNT4 levels in osteoblasts at the osteogenic front of calvar-
• WNT16
ial sutures42. FGFR3 gain-​of-function mutations cause
Crouzon syndrome (OMIM 612247), Muenke syndrome
• WNT (OMIM 602849) and achondroplasia (OMIM 100800).
FGFR2 • FGFR1
FGFR3 IGF1R • IGF1R Features of these syndromes include craniosynosto-
sis, hydrocephaly, hearing loss and frontal bossing
Osteoblast (protuberance of the frontal bones)49–51. Fgfr3 gain-of-
function mice have craniosynostosis and midface hypo-
plasia owing to decreased chondrocyte proliferation and
maturation52.
• FGF8
• FGF18 Ultimately, FGFR2 stimulates the proliferation of
Proliferating
osteoblast progenitor cells, while FGFR1 promotes
chondrocyte differentiation and FGFR3 regulates chondrocyte pro-
liferation and differentiation. The reason for the allelic
heterogeneity of the FGFR-​b ased craniosynostosis
• IGF1R
FGFR3
• WNT
syndromes is unknown but might be a consequence
of the activation of convergent downstream signalling
pathways in response to ligand binding. The actions of
the receptor are dependent on receptor dimerization,
autophosphorylation of the kinase domain and bind-
ing of specific adaptor proteins for each of the down-
stream pathways53. Thus the cellular, and consequently
Hypertrophic Osteocyte clinical, outcome of mutations can differ depending on
chondrocyte which functional domain within the FGFR contains
the mutation.

IGF signalling. The IGF signalling pathway includes


• IGF1 • WNT6 two ligands (IGF1 and IGF2) and three receptors (IGF1
• IGF2 • WNT7A receptor (IGF1R), insulin receptor and IGF2 receptor
• WNT1 • WNT7B
• WNT3A (IGF2R)). IGF1 is the predominant functional ligand
Surrounding tissue during the postnatal period, when it also mediates some
of the actions downstream of growth hormone signal-
ling, whereas IGF2 acts mainly during intra-​uterine
FGFR1 FGFR2 FGFR3 IGF1R development. Both ligands bind with high affinity to
IGF1R, but only IGF2 displays high-​affinity binding
for IGF2R and the insulin receptor. IGF1R is a tyrosine
kinase receptor; after ligand binding, IGF1R activates
Fgfr2 in mice also lead to ectopic cartilage formation the PI3K, MAPK and extracellular signal-​related kinase
in the calvarial sutures and increased amounts of carti- (ERK) pathways. By contrast, IGF2R is a signalling
lage in the cranial base48. Although they display fusion antagonist that regulates the actions of IGF2 by seques-
of the coronal suture, Fgfr2-overexpressing mice also tering the ligand, preventing its binding to and activation
have characteristic defects in the sagittal suture, where of IGFR1 signalling54.
increased FGFR2 signalling leads to an accumulation of IGF1 and IGF2 are expressed in the dura mater that
pre-​osteoblastic cells with reduced numbers of differenti- surrounds the suture mesenchyme at the time of calvar-
ated osteoblasts and decreased ossification. Like FGFR1, ial suture fusion. In patients with craniosynostosis, IGF1
loss-​of-function mutations in FGFR2 are rare, but result is upregulated in the fused and fusing suture tissue55.

Nature Reviews | Endocrinology


Reviews

Administration of recombinant IGF1 to rat cranial sutures regulates the timing of synchondrosis development and
also induces fusion56,57. Homozygous loss-of-function maturation during growth at the skull base.
mutations in IGF1 (OMIM 608747) cause microcephaly,
delayed bone age and deafness, and heterozygous loss- WNT. The WNT family of signalling molecules contains
of-function mutations in IGF2 (OMIM 616489) results 19 proteins, which bind Frizzled receptors and the LDL
in macrocephaly, frontal bossing, micro­gnathia, tria­ receptor-​related protein 5 (LRP5) and LRP6 co-​receptors
ngular face, low-set ears and delayed bone age58,59. In at the cell surface. WNT signalling is either dependent on
osteo­blasts, IGF1 regulates DNA binding of the master (canonical signalling) or independent of (non-​canonical
transcription factor RUNX2 via the PI3K and ERK1/2 signalling) the transcription factor β-​catenin. The exact
pathways60. Thus, IGF1 regulates expression of RUNX2 mode of action of the WNT ligands has not been deter-
target genes, inducing upregulation of Ocn and bone mined, but in most tissues signalling occurs locally via
sialoprotein (Bsp)61. In chondrocytes, IGF1 drives pro- paracrine actions between adjacent cells83. Evidence sug-
duction of COL2A1, aggrecan and SOX9 (refs62,63). The gests that the WNT protein remains tethered to the cell
consequences to the cranial base in Igf1−/− mice have membrane and acts at its site of expression84. Calvarial
not been reported, but the growth plates of long bones pre-​osteoblasts express WNT1, WNT4 and WNT16, and
of these mice have impaired hypertrophic chondrocyte surrounding tissues produce WNT1, WNT3a, WNT6,
differentiation64. The craniofacial phenotype in Igf2−/− WNT7a and WNT7b85,86. WNT signalling is important
mice has also not been reported, although decreased at all stages of bone development and growth. Its initial
BMD throughout the appendicular skeleton and cranial role is as the fate-​determinant of mesenchymal con-
dysmorphology indicates that a defect in growth of the densations in the craniofacial region. The presence or
skull is present65. absence of β-​catenin dictates whether the condensa-
IGFR1 is expressed in both chondrocytes and osteo­ tion will become intramembranous or endochondral,
blasts. Heterozygous loss-​of-function mutations in demonstrating that β-​catenin is a key molecular switch
IGFR1 (OMIM 270450) cause intrauterine growth retar- that distinguishes ossification fate87.
dation, microcephaly, short stature, delayed bone age Mutations in WNT ligands and their receptors result
and increased serum levels of IGF66–68. Both Igf1−/− and in a variety of systemic bone diseases, but those with
Igfr1−/− mice recapitulate this phenotype, with decreased specific malformations affecting the skull include tetra-​
ossification in the calvaria and viscerocranium69,70. amelia syndrome (WNT3; OMIM 273395), Robinow
Overall, the IGF–IGFR signalling pathway pro- syndrome (WNT5A; OMIM 180700), tooth agenesis
motes intramembranous and endochondral ossification and hypodontia (WNT10A; OMIM 150400, 257980;
in the skull, acting via PI3K and MAPK–ERK signal- WNT10B; OMIM 617073; LRP6; OMIM 616724) and
ling pathways to stimulate osteoblast and chondrocyte Van Buchem disease (LRP5; OMIM 607636). During
differentiation. bone growth, reduced WNT signalling caused by loss-​
of-function mutations in the WNT co-​receptor LRP5
RUNX2. RUNX2 is a transcription factor expressed in causes osteoporosis–pseudoglioma syndrome (OPPG),
osteoprogenitor cells in response to TGFβ and BMP2 which is associated with reduced calvarial thickness88.
signalling via the SMAD, p38 and ERK1/2 pathways71,72, Furthermore, reduced WNT signalling caused by LRP6
where it drives osteoblast differentiation and expres- deficiency in Lrp6−/− mice causes midface hypoplasia and
sion of Ocn, Osteopontin (Opn), Bsp, Alp and COL1A1 cleft palate89. By contrast, gain-​of-function mutations in
(ref.73). It is indispensable for osteoblast formation, as LRP5 result in increased WNT signalling and cause the
shown by the lack of osteoblasts and bone formation LRP5 high bone mass phenotype (LRP5–HBM) that is
in Runx2 knockout mice74,75. RUNX2 also controls cell associated with craniosynostosis and is further character-
proliferation in pre-​hypertrophic chondrocytes and is ized by increased calvarial thickness, foramen magnum
expressed in terminally differentiated hypertrophic stenosis and thickening of the mandible90,91. These pheno­
chondrocytes76,77. RUNX2 regulates expression of Fgfr1, types result from WNT-​regulated actions in both osteo-
Fgfr2 and Fgfr3 in the calvaria to enhance pre-​osteoblast blasts and chondrocytes. In osteoblasts, canonical WNT
proliferation, as well as the expression of Ihh and the signalling induces Alp and Runx2 expression in response
hypertrophic chondrocyte-​specific marker Col10a1 to BMP2 (refs92,93). In chondrocytes, canonical WNT
(refs77–80). Expression of Runx2 is also tightly regulated, signalling inhibits cell differentiation by downregulating
with MSX2 and TWIST1 downregulating its expression Sox9 and drives chondrocyte hypertrophy87,94.
in the calvarial sutures to prevent osteoblast differentia- Thus, WNT signalling promotes osteoblast pro-
tion and maintain suture patency81,82. Heterozygous loss-​ liferation and differentiation, stimulates chondrocyte
of-function mutations of RUNX2 result in cleidocranial hypertrophy and determines the intramembranous or
dysplasia (OMIM 119600), a condition characterized by endochondral ossification developmental fate of cranial
delayed closure of the anterior fontanelle and metopic mesenchyme.
suture, decreased intramembranous ossification in the
cranial vault, abnormal dental development and clavi­ Thyroid hormones
cular hypoplasia. These abnormalities are recapitulated The HPT axis. Thyroid hormones are essential homeo-
in Runx2 knockout mice74,75. static regulators of numerous developmental and meta­
Overall, RUNX2 has critical roles in both intramem- bolic processes. Circulating levels of the prohormone T4
branous and endochondral ossification. RUNX2 therefore (also known as thyroxine) and the active hormone T3 are
controls the timing of suture formation and closure and tightly controlled by the HPT axis, which forms a classic

www.nature.com/nrendo
Reviews

a b T4
T4
Hypothalamus T4 T3 T4
T3 T4 T4
TRH
MCT8, MCT10 MCT8, LAT1
or LAT1 or LAT2

T3
T3
T4
T4 T4 T3
Pituitary T3 T4 T4 T4
gland
T3 DIO3 DIO2 DIO3
TSH
T2 T3 T2 rT3
T3 rT3
T3

Thyroid Co-activator Co-activator


T3 T3
RXR TRα1 RXR TRα1
T4 T3

TRE TRE
Bone

Chondrocyte Osteoblast

Fig. 3 | Thyroid hormone action in chondrocytes and osteoblasts. a | The thyroid gland secretes T4 and T3, and
circulating levels are maintained by the hypothalamic–pituitary–thyroid (HPT) axis, which is a classic endocrine negative
feedback loop. b | Chondrocytes (blue) express the MCT8, MCT10 and LAT1 thyroid hormone transporters. Intracellular T3
supply is regulated by the activity of the inactivating enzyme DIO3, and the actions of T3 are mediated by TRα1. Osteoblasts
(purple) express the MCT8, LAT1 and LAT2 thyroid hormone transporters. Intracellular T3 supply is regulated by the relative
activities of the activating enzyme DIO2 and the inactivating enzyme DIO3, and the actions of T3 are mediated by TRα1.
DIO2, iodothyronine deiodinase type 2; DIO3, iodothyronine deiodinase type 3; LAT, L-​type amino acid transporter; MCT,
monocarboxylate transporter; rT3, reverse T3; RXR, retinoid X receptor; TRα1, thyroid hormone receptor α1; TRE, thyroid
response element; TRH, thyrotropin-​releasing hormone; TSH, thyroid-​stimulating hormone.

negative feedback loop. The axis originates in parvo­ The thyroid hormone transporters facilitate entry
cellular neurosecretory neurons of the hypothalamic of both T4 and T3, with each protein displaying differ-
paraventricular nucleus, which secrete thyrotropin-​ ing affinities for the two ligands. The prohormone T4
releasing hormone (TRH)95–97. TRH passes through the must be converted to the active hormone T3 in target
pituitary-​portal vasculature and binds to G protein-​ cells to mediate its physiological actions113,114. Activation
coupled TRH receptors on the surface of anterior pitui- of T4 to T3 occurs via removal of the 5′-iodine atom by
tary gland thyrotrophs, which stimulates synthesis and either iodothyronine deiodinase type 1 (DIO1) or DIO2
secretion of TSH98–101. In the pituitary, TSH then acts (refs114,115). A third deiodinase (DIO3) irreversibly inac-
via G protein-​coupled TSH receptors (TSHR) on thy- tivates T4 and T3 by removal of the 5-iodine atom, which
roid follicular cells to stimulate their growth and the converts them to reverse T3 (rT3) or T2, respectively116,117
synthesis and secretion of T3 and T4 (ref.102). Binding (Fig. 3b). Mitochondrial aminoadipate aminotransferase
of the active hormone T3 to nuclear thyroid hormone (AADAT) has been identified as an additional thyroid
receptors (TRs) in the hypothalamus and pituitary inhib- hormone-​inactivating enzyme. AADAT facilitates
its production and secretion of TRH and TSH, respec- removal of the alanine side chain of T4 and T3, forming
tively103,104 (Fig. 3a). The HPT axis set point maintains the TK4 and TK3, respectively106. Expression patterns of
physiological reciprocal relationship between circulating DIO1, DIO2 and DIO3 differ between tissues and stages
levels of T4, T3 and TSH and is genetically determined, of development, which suggests that these enzymes
with considerable variation between individuals105,106. have specific roles at different developmental stages and
Thyroid hormone entry into target cells is mediated in different tissues. For instance, DIO1 is not expressed in
by specific membrane transporters, including mono­ the skeleton, DIO2 is present in mature osteoblasts and
carboxylate transporters 8 and 10 (MCT8 and MCT10), DIO3 activity is present in chondrocytes, osteoblasts
L-type amino acid transporters 1 and 2 (LAT1 and and osteoclasts118. Thus, the action of thyroid hormone
LAT2), organic anion transporting polypeptide 1C1 in target tissues is dependent on the intracellular con-
(OATP1C1) and the solute carrier SLC17A4 (refs106–109) centration of T3, which is locally regulated by the relative
(Fig. 3b). Mct8, Mct10, Lat1 and Lat2 are expressed in expression of DIO2 and DIO3.
the skeleton, although currently only MCT8 is known to Ultimately, the cellular actions of thyroid hormone
be essential for thyroid hormone transport in bone110–112. are mediated by genomic (type 1) and non-​genomic

Nature Reviews | Endocrinology


Reviews

(types 2 and 3) actions of nuclear TRα and TRβ119,120. The skeletal consequences of abnormal thyroid hor-
The genomic actions are mediated by binding of TR (as a mone levels in bone and cartilage during development
homodimer, or heterodimer with retinoid X receptors and in adulthood have been reviewed comprehen-
(RXRs)) to thyroid response element sequences (TREs) sively1,143,144. Briefly, hypothyroidism causes delayed skel-
located within target gene promoters (Fig. 3b). In the etal development during childhood that is characterized
absence of T3, the TR complex is bound to DNA and by growth retardation, delayed tooth eruption and short
interacts with co-​repressor proteins that inhibit target stature owing to impaired endochondral ossification1.
gene transcription121. In the presence of T3, the TR Conversely, childhood thyrotoxicosis results in accel-
undergoes a conformational change, promoting release erated growth and advanced bone age but ultimately
of the co-​repressor, facilitating binding of a co-​activator results in short stature owing to premature fusion of the
protein and stimulating target gene transcription122–124. growth plates1. In adults, hypothyroidism inhibits bone
Non-​genomic actions involve phosphorylation of the TR turnover, which results in increased bone mass and
and might be mediated via indirect binding of the TR to mineralization. By contrast, hyperthyroidism stimu­
DNA or via second messenger signalling, including the lates increased bone turnover with a greater effect on
PI3K pathway125,126. bone resorption than formation, which results in bone
Like the deiodinases, the TRs display distinct tem- loss and osteoporosis1.
porospatial patterns of expression. TRα is the main
isoform expressed in bone, central nervous system and Effects of thyroid hormone deficiency
heart, while the liver expresses mainly TRβ1,127. Skeletal Congenital hypothyroidism. Congenital hypothyroidism
responses to T3 during bone development are medi- has a global incidence of between 1 in 1,400 and 1 in
ated by transcriptional activation of T3 target genes by 2,800 (ref.145) and results from thyroid gland dysgenesis,
genomic (type 1) actions of TRα119. defects of iodine organification, dyshormonogenesis, mater-
nal iodine insufficiency, maternal TSHR antibodies or
Thyroid hormone action in bone. Thyroid hormones are genetic mutations145. Genetic causes include mutations
essential for bone development and growth, and consid- of thyroid peroxidase (TPO), dual oxidase 2 (DUOX2),
erable progress has been made in defining the actions of paired box gene 8 (PAX8), TSHR and selenocysteine
T3 in the appendicular skeleton. In osteoblasts, T3 pro- insertion sequence binding protein 2 (SECISBP2)1,145–150.
motes proliferation, differentiation, nodule formation TPO (OMIM 606765) and DUOX2 (OMIM 606759)
and miner­a lization 128–130, acting via the FGF, IGF, mutations inhibit normal thyroid hormone synthesis,
MAPK and WNT signalling pathways104,131–133. In vivo, resulting in low circulating concentrations of T4 and T3
T3-induced proliferation and differentiation of osteoblasts and increased levels of TSH146,147. Mutations in PAX8
results in increased mineral apposition and bone forma- (OMIM 167415) cause thyroid gland dysgenesis, result-
tion during skeletal development134,135. The osteoblastic ing in low to normal circulating levels of T4 and mark-
response to thyroid hormone results in increased expres- edly increased levels of TSH148. Loss-​of-function TSHR
sion of several genes that have important roles in cranio­ mutations (OMIM 275200) result in either impaired
facial development, including Alp, Ocn, Osx, Igf1, Fgf1, binding of TSH to its receptor or reduced downstream
Fgf2, Fgf3, Fgfr1, Runx2 and Mmp9 (refs128,129,132,136,137). In signalling responses, which lead to high levels of TSH
chondrocytes, T3 inhibits cell proliferation and induces but low levels of T4 (ref.149). SECISBP2 loss-​of-function
differentiation and hypertrophy138–140, acting via the FGF mutations (OMIM 607693) prevent the insertion of
and WNT signalling pathways104,141. In vivo, the increased the amino acid selenocysteine into 25 different human
chondrocyte hypertrophy in response to T3 leads to the proteins, including the three deiodinase enzymes.
hypertrophic zone in the growth plate increasing in Selenocysteine is critical to the enzymatic activity of the
size, advanced development of secondary ossification deiodinases and SECISBP2 loss-​of-function mutations
centres and promotion of transdifferentiation to osteo- impair their ability to activate and inactivate thyroid
Mineral apposition blasts15,139. A number of T3 target genes in chondrocytes hormones150,151. Consistent with this finding, SECISBP2
The deposition of mineral-​
have been identified, including Col10a1, Alp, Fgfr1 and loss-​of-function mutations result in normal to high
containing bone matrix during
new bone formation. Fgfr3 (refs138–141). In osteoclasts, the evidence is less clear. levels of T4, low levels of T3 and high circulating levels
Although excess levels of thyroid hormone stimulate of TSH150.
Thyroid gland dysgenesis osteoclastic bone resorption117,118, there is conflicting The clinical craniofacial phenotypes for all forms of
Abnormal or evidence about whether this response is dependent on congenital hypothyroidism are similar and include a
underdevelopment of the
thyroid gland.
the presence of osteoblasts117,142. Thus, it is not known widened anterior fontanelle (50.3% incidence) and per-
whether T3 acts directly in osteoclasts, or whether the sistent posterior fontanelle (47.2% incidence)152. Other
Iodine organification thyroid hormone-​stimulated increase in osteoclast common craniofacial abnormalities include delayed
The incorporation of iodine activity results from an indirect mechanism following dental eruption, deafness, facial clefting, widened or per-
into the thyroglobulin protein
T3 action in another cell type. sistent sutures, supernumerary cranial bones (wormian
during the synthesis of
thyroid hormones by thyroid Overall, thyroid hormone promotes osteoblastic bone bones), microcephaly and micrognathia152–158. In severe,
follicular cells. formation and terminal hypertrophic differentiation in untreated hypothyroidism, the delay in fontanelle clo-
chondrocytes. These effects are demonstrated by the sure can persist into the teenage years and even adult-
Dyshormonogenesis increased bone formation and growth plate maturation hood159. Treatment of congenital hypothyroidism with
The inability to concentrate
iodine or synthesize thyroid
seen in the appendicular skeleton in hyperthyroidism levothyroxine replacement therapy is effective in pro-
hormones in thyroid follicular and the decreased bone formation and impaired growth moting longitudinal growth and dental eruption153; how-
cells. plate maturation characteristic of hypothyroidism1. ever, the predicted final height might not be achieved,

www.nature.com/nrendo
Reviews

particularly if the diagnosis of hypothyroidism is delayed Juvenile hypothyroidism. Primary hypothyroidism


and if treatment is not commenced immediately follow- in the perinatal, postnatal or childhood periods had a
ing diagnosis160. In the case of individuals with SECISBP2 reported incidence of approximately 69 in 100,000 in
mutations, levothyroxine can normalize TSH concentra- a community-​based study in Scotland, UK173. Juvenile
tions but, owing to impaired conversion of T4 to T3 by hypothyroidism results from autoimmune disease
DIO1 and DIO2, liothyronine treatment is necessary to (Hashimoto disease; OMIM 140300), thyroidectomy
achieve physiological euthyroidism and effectively treat following surgical treatment of malignancy or could
the consequences of hypothyroidism150,158,161. be idiopathic 173. Juvenile hypothyroidism presents
Pregnant rats treated with 2-mercapto-1-methyl imi- with growth retardation and delayed bone age, which
dazole or radioiodine give birth to hypothyroid offspring is frequently accompanied by persistent fontanelles
that model congenital hypothyroidism and display and wormian bones in the skull174,175. The age at which
characteristic abnormalities, including microcephaly, juvenile hypothyroidism first presents has a consider­
blunt snouts, abnormal facial features and deafness due able effect on the form and severity of these craniofacial
to abnormal ossicle morphology162,163. These animals manifestations. Fontanelle closure occurs by approxi­
respond well to levothyroxine treatment, recovering mately 24 months of age, and thus hypothyroidism
to achieve normal growth that is equivalent to their occurring after this age is not characterized by the com-
wild-​type counterparts164. Genetically modified mice mon complication of persistent fontanelles, which might
with congenital hypothyroidism also recapitulate the result in delayed diagnosis. Levothyroxine treatment is
craniofacial phenotype of the human condition1. Thus, effective in accelerating skeletal maturation; however,
Tshr−/− mice display thinning of the calvaria and enlar­ short stature can persist unless treatment is commenced
ged ossicles with delayed ossification165,166. Although immediately following early diagnosis175–177.
Secisbp2 −/− mice have been generated, their skele- The effects of juvenile hypothyroidism on craniofacial
tal phenotype has not yet been analysed167, whereas development have been studied in animal models follow-
Tpo-mutant, Pax8−/− and Hyt/Hyt mice with congen- ing thyroidectomy or radioiodine ablation. In juvenile
ital hypothyroidism have not been investigated for sheep, thyroidectomy results in delayed tooth eruption,
craniofacial abnormalities or defective skull devel- defects in the anterior maxilla, frontal and orbital bones
opment168,169. The molecular mechanisms that cause and a prominent jaw178,179. In rabbits, shortening of the
craniofacial abnormalities in congenital hypothyroid- facial bones occurs, which results in decreased length,
ism have also not been characterized, although delayed width and height of the face and cranium, and the devel-
calvarial ossification in hypothyroid rodents is accom­ opment of the mandible is retarded180. Hypothyroid
panied by decreased IGF1 signalling and osteocalcin juvenile rats have wide skulls, a decreased anterior–
production66,170. posterior cranial base length, sunken nasal bridge and an
Zebrafish are a useful model to study the effects of altered relationship between the position of the calvaria
congenital hypothyroidism in the craniofacial complex. and foramen magnum181. In starlings, juvenile hypo-
Fish that have undergone thyroid ablation have an over- thyroidism results in delayed formation of the palate
all decrease in bone mineralization and an increase in and patent cranial sutures182. In dogs, hypothyroidism
the amount of cartilage with malformation of the bones results in anterior–posterior shortening of the skull, with
of the skull171. In these fish, the frontal and parietal reduced bone growth in both the calvarial sutures and
bones fail to fuse, leaving a void in the apex of the skull synchondroses of the cranial base183.
similar to the enlarged fontanelle seen in humans and Overall, juvenile hypothyroidism provides extensive
mice with congenital hypothyroidism169. At the base evidence of a key role for thyroid hormones during post-
of the skull, the amount of cartilage in the synchon- natal craniofacial development that is conserved across
droses is increased and ossification and mineralization species. The craniofacial abnormalities are similar to
are decreased171. The bones of the Weberian apparatus those caused by congenital hypothyroidism, with defects
(analogous to the auditory ossicles in humans and mice) affecting both intramembranous and endochondral
are considerably malformed and disorganized, with ossification (Table 2).
abnormal secondary ossification centres and regions
of low mineralization171. Craniofacial development of THRA dominant-​negative mutations. Mutations in
flatfish is also reliant on thyroid hormone, with anti-​ THRA were first described in 2012 and cause resis­tance
thyroid treatment of Senegalese sole larvae resulting in to thyroid hormone-​α (RTHα), with 23 independent
inhibition of the normal asymmetrical migration of the mutations currently identified (OMIM 614450)184.
eye during thyroid hormone-​dependent metamorpho- Most mutations occur in the ligand-​binding domain of
sis172. This change is accompanied by abnormal ossi- TRα at the C-​terminus and result in a dominant-​negative
fication of the ethmoid bone and pseudomesial bone receptor that binds persistently to the co-​repressor
(a bone unique to flatfish) and decreased expression of NCoR, which prevents activation of T3 target gene tran-
osteonectin and matrix gla protein172. scription185,186. The severity of the RTHα syndrome is
Overall, the distinct craniofacial phenotype obser­ variable as the resulting phenotype correlates with the
ved in congenital hypothyroidism provides evidence of differing interaction affinities of individual mutant
important developmental roles for thyroid hormone dur- TRs for the transcriptional co-​repressor NCoR187. Thus,
ing the prenatal period that affect intramembran­ous ossi­ the presentation of patients with RTHα is variable, but the
fication of the cranial vault as well as endochondral clinical features are consistent with the consequences of
ossification of the facial bones and middle ear. severe congenital hypothyroidism and include impaired

Nature Reviews | Endocrinology


Reviews

Table 2 | craniofacial phenotypes resulting from thyroid hormone deficiency


causes craniofacial phenotype other abnormalities refs
Human
Congenital: cause unknown Persistent anterior fontanelle, large posterior Short stature, delayed bone age, absent 153,159,160

or dyshormonogenesis fontanelle, persistent metopic suture, delayed or delayed secondary ossification centre
dentition, microcephaly and micrognathia and intellectual disability
Juvenile: autoimmune Patent anterior fontanelle Short stature and delayed bone age 174

thyroiditis
Genetic: mutations in PAX8, Large and/or persistent posterior fontanelle, Short stature, delayed bone age, absent 155,156,158,161,183,

TSHR, SECISBP2 or THRA persistent anterior fontanelle, persistent sutures, or delayed secondary ossification centres, 184,188,191,193,194,234

microcephaly or macrocephaly with thickened intellectual disability, hearing loss and


calvaria in patients with THRA mutations, delayed macroglossia
dental eruption and wormian bones
Rodents
Congenital: maternal anti-​ Blunt snout, impaired cranial ossification and widened Decreased bone length, impaired 164,170,171

thyroid treatment and/or persistent sutures ossification and reduced bone strength
Juvenile: thyroidectomy, Reduced calvarial length, reduced cranial base length, Decreased bone length 178,180,182

radioiodine delayed dental eruption and shortened mandible


Genetic: mutations in Tshr, Thra Thinning of calvaria, large and/or persistent anterior Decreased bone length and bone mineral 165,195,196,198,199

fontanelle and wide and/or persistent sutures density and delayed ossification

growth, delayed bone age and cognitive deficits185. The nasal bridge, but normal head circumference191. Finally,
craniofacial malformations that result from RTHα are a further mutation (THRAN359Y) has been reported in a
also consistent but vary in severity according to the single patient and affects both TRα1 and TRα2 (ref.192).
potency of individual THRA mutations1,187,188. The mutant TRα1 has reduced binding affinity for
Truncating mutations, lacking all or part of the T3 and a moderate dominant-​negative effect but the
C-​terminal alpha helix of TRα, delete the domains criti­ consequences to TRα2 are unknown as the physiolo­
cal for co-​activator recruitment185. Additionally, the gical role of TRα2 has not been characterized192. The
deletion exposes the hydrophobic cleft that accom- THRAN359Y mutation is also accompanied by a micro­
modates NCoR185. Together, these changes result in a deletion and results in a unique phenotype that includes
strongly dominant-​negative TRα1 receptor. Examples micrognathia, macro­cephaly, persistent fontanelles,
of these truncating mutations are THRAE395X, THRAE403X absent clavicles and an abnormal rib cage192. The cause
and THRAC380fs387X (refs184,185,188). The phenotypic conse- of this fairly severe and unusual skeletal dysplasia
quences resulting from expression of potent dominant-​ remains unclear, but it is unlikely to result solely from
negative truncated receptors include macrocephaly, the dominant-​negative effects of the mutant TRα1.
delayed fusion of the cranial sutures, patent anterior Taking these results together, the consistent cranio­
fontanelle, wormian bones, delayed dentition and facial features associated with RTHα include macroce­
tooth eruption and thickening of the calvaria185,188–190. phaly, persistent fontanelles, delayed dentition, wormian
Missense mutations, such as THRAL274P, which do not bones and thickening of the calvaria. Owing to vary-
result in truncation of the receptor, might also have ing potencies of the individual THRA mutations, the
potent dominant-​negative activity188 and cause a severe effectiveness of treatment with thyroid hormone dif-
phenotype that includes facial dysmorphia, delayed fers depending on the mutation present. Some clinical
dentition, delayed anterior fontanelle closure, wormian features are improved following levothyroxine adminis-
bones during childhood and increased calvarial thickness tration, including constipation, energy levels185,193,194 and
during adolescence190. growth velocity195; however, effects on predicted final
Many missense mutations, however, result in milder height might be variable193,195.
phenotypes than those caused by THRAL274P because the A variety of mouse models with Thra mutations have
expressed mutant TRα1 exerts only moderate dominant-​ been generated, and all present with distinct craniofacial
negative activity. The point mutations in these instances phenotypes. TRα1PV/+ mice, which are euthyroid with
cause a morphological change at the T3 binding site of reduced T3 action, have normal-​sized skulls, but wid-
TRα1 that still allows ligand binding at a lower affinity ened and persistently patent cranial sutures and fonta-
than normal190. Examples include THRAR384H, THRAA263S, nelles with more porous frontal and parietal bones196,197.
THRAD211G, THRAA263V (refs188,190,191) and the resulting TRα1PV/+ mice also have enlarged ossicles in the middle
phenotypes comprise delayed dentition, persistence of ear that result from delayed ossification and are associ-
the anterior fontanelle, wormian bones, thickening ated with deafness166. TRα1R384C mutant mice are mildly
of the calvarial bones and macrocephaly during child- hypothyroid during early life and have a less severe
hood, with the macrocephaly and calvarial thickening resistance to T3 than TRα1PV/+ mice but also exhibit
persisting during adulthood188. For some missense delayed intramembranous ossification and delayed
mutations, for example THRAD211G, the phenotype is tooth eruption198,199. Furthermore, cell-​specific expres-
even milder and manifests only as a broad and flattened sion of a dominant-​negative TRα1L400R mutant receptor

www.nature.com/nrendo
Reviews

in chondrocytes results in malocclusion, shortening of accelerated tooth eruption and microcephaly213–215.


the snout and delayed mineralization of endochondral The cranial base is also affected, with decreased bone
ossification-​derived bones of the cranial base200. growth and reduced width and shortening and prema-
Overall, these data demonstrate a clear and important ture fusion of the synchondroses205,207,216. A mouse model
role for TRα1 in both intramembranous and endochon- of the human TSHRD633H mutation has been developed
dral ossification during craniofacial development that is that accurately recapitulates the features of the human
conserved in humans and mice. The phenotype in RTHα disease, but the detailed skeletal consequences have
shares features with congenital hypothyroidism and var- not yet been reported217. Similarly, genetically modi-
ies in severity depending on the potency of the THRA fied mice with the equivalent of the human TSHRV509A
dominant-​negative mutation. and TSHRI486F mutations have been generated but their
craniofacial phenotypes have not been reported218.
Effects of thyroid hormone excess In the zebrafish model of hyperthyroidism, an overall
Juvenile hyperthyroidism. Juvenile hyperthyroidism is increase in ossification in the craniofacial complex has
rare, although there are reports suggesting that the global been reported171. Specifically, the increased ossification
incidence is between 3 and 14 per 100,000 people and results in increased overlap at the junctions between the
that its incidence is increasing201,202. This condition can frontal and parietal bones, a reduction in cartilage and
be a consequence of Graves disease, genetic disorders or increased fusion rates in the synchondroses at the base
pituitary tumour (TSHoma), or an adverse effect of over- of the skull171. The bones of the Weberian apparatus in
treatment with levothyroxine. Genetic causes include hyperthyroid zebrafish are also malformed171.
gain-​of-function mutations in TSHR (OMIM 609152), The advanced intramembranous and endochondral
which result in constitutive activation of the recep- ossification that occurs in response to excess thyroid
tor and increased systemic thyroid hormone levels203, hormone during craniofacial development highlights the
and McCune–Albright syndrome (OMIM 174800), important role of T3 (Table 3). The contrast between
which results from mutations in GNAS, which encodes the phenotypes resulting from juvenile hypothyroidism
a component of the TSHR downstream intracellular and hyperthyroidism indicates that the skull is exquisitely
signalling cascade. sensitive to the level of available thyroid hormone.
The most common craniofacial manifestation of
juvenile hyperthyroidism is premature fusion of the THRB dominant-​negative mutations. Dominant-​
sutures, or craniosynostosis204–206. Fusion of the syn- negative mutations of THRB had an incidence of approx-
chondroses at the cranial base and advanced dental imately 1 in 40,000 in a study of Spanish patients, and
eruption have also been reported207,208. The incidence of they result in RTHβ (OMIM 188570)219. Mutations
craniosynostosis in those with childhood thyrotoxico- in THRB that lead to RTHβ are located mainly in the
sis is high, although the severity and number of sutures ligand-​binding domain of TRβ and result in reduced
affected varies between patients and is related to the time affinity for T3, RXR or co-​activator proteins that inter-
of disease onset204. Craniofacial manifestations of TSHR act with TR–RXR complexes at the TRE168,220–222. The
gain-​of-function mutations include frontal bossing, consequence is impaired negative feedback of the HPT
premature closure of the fontanelles and craniosynos- axis, resulting in increased circulating levels of T3 and T4
tosis203,209,210. Anti-​thyroid drugs might be effective in and normal or high levels of TSH1. Thus, individual T3
preventing further advancement in bone age or suture target tissues might display phenotypes of thyroid hor-
fusion206,210,211. However, if craniosynostosis has already mone deficiency or excess depending on whether they
occurred, surgical intervention might be required211,212. predominantly express and respond to TRα or TRβ,
Modelling juvenile hyperthyroidism in animals by respectively1. RTHβ is thus a complex condition that
administration of liothyronine or levothyroxine results causes a variety of phenotypes in different tissues, and
in a number of morphological abnormalities of the its heterogeneity is further confounded by treatment of
skull, including increased ossification in the sutures, individuals with anti-​thyroid drugs and thyroidectomy

Table 3 | craniofacial phenotypes resulting from thyroid hormone excess


causes craniofacial phenotype other abnormalities refs
Human
Juvenile: cause unknown Craniosynostosis, microcephaly, Advanced bone age, goitre, 205–207,211,

or Graves disease advanced dental age and hyperactivity and intellectual 212,235–237

malocclusion disability
Genetic: mutations Premature anterior fontanelle closure Advanced bone age, goitre, irritability, 209,210,220,

in TSHR or THRB and craniosynostosis intellectual disability and diarrhoea 238–241

Rodents
Juvenile: levothyroxine Advanced dentition, reduced Advanced skeletal development 174,216,217,

treatment suture width, premature suture and and increased bone size 218,238,239

synchondrosis fusion and microcephaly


Genetic: mutations Reduced fontanelle size and reduced Advanced bone age and advanced 226,234

in Thrb suture width growth plate maturation

Nature Reviews | Endocrinology


Reviews

followed by thyroid hormone replacement or use of the regulation of craniofacial development by thyroid
thyroid hormone analogues. hormones (Fig. 4).
Over 170 THRB mutations have been reported in
more than 3,000 patients from about 1,000 families223,224, Fibroblast growth factors. In osteoblasts, thyroid hor-
but the effects on the craniofacial skeleton have been mone excess in TRβ−/− and TRβPV/PV mice or following
incompletely investigated. However, frontal bossing, levothyroxine administration results in increased expres-
scaphocephaly and craniosynostosis have been reported sion of FGFR1, FGF1 and FGF2 (refs128,132,137,199,226,227).
and are mainly compatible with increased circulating Conversely, mouse models of thyroid hormone deficiency
concentrations of thyroid hormone in RTHβ acting (TRα0/0 mice, which lack all TRα transcripts expressed
via the normal TRα expressed in bone. Nevertheless, from the Thra locus, and TRα1R384C/+ and TRα1PV/+ mice,
in patients with potent dominant-​negative truncating which express dominant-​negative forms of the recep-
mutations affecting TRβ, delayed bone age has been tor) show decreased expression of Fgfr1 and Fgfr3 in
reported and, although no specific data are available, it osteoblasts196,225,226. These findings demonstrate a direct
is probable in such cases that the mutant TRβ protein relationship between thyroid hormone concentrations,
interferes with the normal actions of wild-​type TRα that FGF–FGFR signalling and intramembranous ossification.
promote bone maturation and growth. While it is known that FGF–FGFR signalling pro-
In mouse models, the consequences of Thrb muta- motes intramembranous bone formation in the calvaria,
tions are clearer than in humans. Thus, TRβ−/− and the mechanism by which thyroid hormone regulates
TRβPV/PV mice have elevated circulating levels of thy- this process is largely unknown227. In the case of FGFR1,
roid hormone owing to absence of TRβ or expression of T3 stimulation of Fgfr1 mRNA expression requires TRα,
dominant-​negative TRβ in the pituitary and hypothala- and the FGFR1 response to FGF2 is enhanced, with
mus, resulting in disruption of the HPT axis. TRβ−/− mice more rapid and increased autophosphorylation of
display accelerated calvarial bone growth and decreased FGFR1 and activation of the downstream MAPK path-
fontanelle size199,225, and TRβPV/PV mice exhibit prema- way132. Whether similar mechanisms result in activation
ture closure of the anterior fontanelle and coronal suture of FGFR2 signalling by T3 has not been determined.
with accelerated calvarial ossification226. Taken together, Nevertheless, T3 stimulation of FGFR1 and MAPK
studies in mice with Thrb deletion or mutation provide signalling in osteoblasts ultimately results in increased
strong in vivo evidence of a major role for TRα during expression of several genes involved in osteoblast
craniofacial development. differentiation, including ALP and OCN132,228,229.
In chondrocytes, expression of Fgfr1 and Fgfr3 is
Aetiology of the craniofacial phenotype increased in response to thyroid hormone by both gene­
Children with severe thyroid hormone deficiency and tic manipulation (TRβ−/− and TRβPV/PV mice) and treat-
excess present with characteristic craniofacial pheno- ment with liothyronine128. Mouse models of impaired
types (Supplementary Table 1). The recurring features T3 signalling in the skeleton (TRα0/0, TRα1R384C/+ and
of thyroid hormone deficiency in the skull are per- TRα1PV/+ mice) show decreased expression of both Fgfr1
sistent fontanelles and patent or wide sutures, while and Fgfr3 in chondrocytes196,225,226, indicating a direct
those of thyroid hormone excess are craniosynosto- relationship between thyroid hormone concentrations,
sis of the calvarial vault and cranial base sutures and FGFR signalling in chondrocytes and endochondral
synchondroses1. These severe craniofacial phenotypes ossification. Thus, FGFR3 is an important T3 target gene
indicate that, as in the peripheral skeleton, thyroid hor- in chondrocytes in vitro and in vivo. Increased levels of
mone is a critical regulator of both intramembranous FGFR3 in response to T3 result in enhanced activation
and endochondral ossification, and precise control of of MAPK signalling by FGF2 and FGF18 (refs134,141).
T3 action is essential for normal craniofacial growth By contrast, T3 inhibits activation of the STAT1 pathway
and development. mediated by FGF2 and FGF18 and has no effect on FGF9
Juvenile thyroid hormone deficiency results in a signalling132. Overall, T3 differentially regulates chon-
craniofacial phenotype that is similar to that caused drocyte FGFR3 signalling during chondrogenesis, with
by FGFR1, FGFR2, IGF1, IGFR1 and RUNX2 loss-​ the response dependent on the activating ligand and the
of-function mutations46,47,58,59,69,70. By contrast, juvenile downstream signalling pathway affected141.
thyroid hormone excess results in a craniofacial pheno- Furthermore, modulation of FGFR signalling by thy-
type similar to that caused by FGFR1, FGFR2 and LRP5 roid hormones might also involve indirect mechanisms
gain-​of-function mutations39,90. These observations mediated via HSPGs, which interact with FGF–FGFR–
suggest that calvarial intramembranous and endochon- co-​receptor complexes at the cell surface230. Increased
dral ossification are regulated by the actions of thyroid expression of HSPGs in proliferating chondrocytes in
hormone and these actions involve the FGF, IGF1 and growth plate cartilage has been observed in hypothyroid
WNT signalling pathways. mice, and T3 inhibited expression of HSPG core pro-
tein, polymerase and modification enzymes (encoded
Mechanism of action in the skull by Gpc6, Ext1 and Hs6st2) in chondrogenic ATDC5
The craniofacial similarities between gain-​of-function cells230. Thus, the role of T3 in endochondral ossifica-
and loss-​of-function mutations in IGF1, the FGFR tion involves additional interactions with key proteo-
genes, the WNT signalling pathway, patients with hyper­ glycan mediators of FGFR signalling, although such
thyroidism and hypothyroidism and animal models mechanisms have not been investigated in cranial base
of these disorders, suggest these pathways interact in synchondroses during craniofacial development.

www.nature.com/nrendo
Reviews

FGF signalling IGF signalling WNT signalling

a Chondrocyte
FGF2 IGF1 GH
FGF18 WNT
LRP5 or
FGFR3 ↓ HSPG IGF1R GHR LRP6 FZD

T3

β-catenin

↑ MAPK ↓ STAT1 ↑ PI3K–AKT ↑ STAT5

T3 T3

Co-activator Co-activator
T3 ↑ Fgfr3 T3
↑ Ghr ↑ Runx2
RXR TRα1 ↑ Fgfr1 RXR TRα1
↓ Ext1 ↑ Igf1 TCF/LEF
↓ Gpc6 ↑ Igf1r
↓ Hs6st2
TRE TRE TRE

b Osteoblast
FGF2 IGF1
WNT
FGFR1
↑ IGF1R
P

↑ MAPK

↑ ALP
↑ OCN ↑ MAPK ↑ AKT

T3 T3 T3

Co-activator Co-activator Co-activator


T3 T3 T3
RXR TRα1 ↑ Fgfr1 RXR TRα1 RXR TRα1
↑ Fgf1 ↑ Igf1
↑ Fgf2
TRE TRE TRE

Fig. 4 | Mechanism of thyroid hormone action in the skull. a | FGF, IGF and WNT signalling in chondrocytes (blue)
in the skull base. T3 induces FGFR expression and enhances MAPK signalling, induces GHR and IGF1R expression and
enhances PI3K–AKT and STAT5 signalling. T3 also enhances canonical WNT signalling and RUNX2 expression. b | FGF,
IGF and WNT signalling in skull vault osteoblasts (purple). T3 induces FGF and FGFR expression and enhances MAPK
signalling, induces IGF1 expression and enhances MAPK and AKT signalling and inhibits WNT signalling. AKT, protein
kinase B; ALP, alkaline phosphatase; Ext1, exostosin 1; FGF, fibroblast growth factor; FGFR, fibroblast growth factor
receptors; FZD, frizzled receptor; GH, growth hormone; GHR, growth hormone receptor; Gpc6, glypican 6; Hs6st2,
heparan sulfate 6-O-​sulfotransferase 2; HSPG, heparan sulfate proteoglycan; IGF1, insulin-​like growth factor 1;
IGF1R, insulin-​like growth factor 1 receptor; LEF, lymphoid enhancer-​binding factor; LRP, low-​density lipoprotein
receptor-​related protein; MAPK, mitogen-​activated protein kinase; OCN, osteocalcin; PI3K, phosphatidylinositol 3-
kinase; Runx2, RUNX family transcription factor 2; RXR, retinoid X receptor; STAT, signal transducer and activator
of transcription; TCF, transcription factor; TRα1, thyroid hormone receptor α1; TRE, thyroid response element; WNT,
WNT family member.

Nature Reviews | Endocrinology


Reviews

Insulin-​like growth factors. Increased expression of Igf1 Nevertheless, in  vitro studies using the Tcf/Lef
and Igf1r has been reported in osteoblasts and the cra- reporter system revealed that T3 activation of wild-​type
nial sutures in TRβ−/− and TRβPV/PV mice and following TRs actually results in inhibition of WNT signalling
thyroid hormone administration in wild-​type rats and in osteoblasts104, whilst in vivo studies demonstrated
mice128,216,226,231. IGF1 promotes osteoblast proliferation, changes in serum concentrations of the WNT inhibi-
differentiation and mineralization in accordance with tors dickkopf 1 and sclerostin in thyroid-​manipulated
the accelerated intramembranous ossification seen in mice rendered hyperthyroid by levothyroxine injection
animals exposed to increased T3 signalling. Stimulation or hypothyroid by addition of methimazole and sodium
of Igf1 expression by T3 in osteoblasts is mediated via perchlorate to drinking water134. Even so, studies indi-
TRα binding to a specific TRE located in the Igf1 pro- cate that manipulation of dickkopf 1 in mutant mice
moter133, and increased IGF1 results in activation of the does not notably affect skeletal responses to T3 in adult
AKT and MAPK pathways in osteoblasts132. The mecha­ mice232. In addition, T3 stimulates WNT signalling in
nism by which Igf1r expression is stimulated by T3 in chondrocytes, resulting in accumulation of β-catenin
osteoblasts has not yet been defined. and increased expression of Runx2 (refs104,233), find-
In chondrocytes, increased expression of growth ings that are consistent with the stimulatory effects of
hormone receptor (Ghr), Igf1 and Igf1r, and increased RUNX2 gain-of-function mutations during endochondral
activity of their downstream signalling pathways STAT5 ossification231.
and PI3K–AKT, have been reported in cranial base syn- Overall, it is evident that thyroid hormones exert
chondroses and growth plate chondrocytes in vitro in complex regulatory effects on WNT signalling in osteo-
response to T3 (refs216,231). Conversely, growth plate chon- blasts and chondrocytes that might affect the stability and
drocytes from TRα0/0 and TRα1PV/+ mice with impaired accumulation of β-​catenin and that could differ between
T3 signalling exhibit decreased expression of these cell types and during development and adulthood.
genes and their downstream signalling responses196,225.
Overall, analysis of the T3 response in TRα0/0, TRα1PV/+, Conclusions
TRβ−/− and TRβPV/PV mice is consistent with a model in Thyroid hormone deficiency results in delayed intra­
which local GH–GHR–IGF1–IGFR mediated STAT5 membranous and endochondral ossification in the skull,
and PI3K–AKT responses to T3 in chondrocytes are which manifests as patent or persistent fontanelles,
mediated via TRα. patent sutures, delayed dentition, wormian bones and
deafness. By contrast, thyroid hormone excess results
WNT signalling. The WNT signalling pathway, which in advanced intramembranous and endochondral
determines the intramembranous or endochondral ossification in the skull, which manifests as premature
fate of cranial mesenchyme during development, is an fusion of the calvarial sutures and cranial base synchon-
important target for thyroid hormones in osteoblasts and droses. These characteristic craniofacial malformations
chondrocytes. Investigation of the WNT–β-​catenin path- indicate that thyroid hormones have a pivotal role in
way in primary osteoblasts from wild-​type and ThrbPV/PV development and growth of the craniofacial skeleton,
mice revealed differential expression of 34 WNT path- and demonstrate that the skull is exquisitely sensitive to
way genes, 12 of which were activated in ThrbPV/PV osteo- changes in available T3. Furthermore, analysis of pheno­
blasts, including the Frizzled co-​receptor Fzd7 and cyclin types in TR-​mutant mice indicate that TRα mediates
D1 cell cycle regulator Ccnd1 (ref.104). Increased activity the major effects of thyroid hormones during skeletal
of the WNT pathway in ThrbPV/PV mice was demonstrated develop­ment, and the findings in mutant mice recapit-
by increased expression of Runx2 and decreased expres- ulate the craniofacial manifestations of thyroid disorders
sion of Rankl in trabecular bone and perichondrium in humans. In addition, changes induced by thyroid hor-
during endochondral ossification, whereas expression mone in FGFR, IGF1 and WNT signalling in osteoblasts
of the activating WNT ligand Wnt4 was decreased, thus and chondrocytes suggest that these pathways are intri-
revealing the complex nature of the WNT signalling cately involved in the regulation of craniofacial develop-
response to T3 in vivo104. In vitro studies revealed that ment by T3. An improved understanding of the role of
the dominant-​negative mutant TRβPV protein exhib- thyroid hormones during craniofacial development and
its a gain-​of-function activity that stabilizes β-catenin growth might result in novel therapeutic opportunities
concentrations in osteoblasts, suggesting a mechanism for patients with craniofacial disorders.
to explain the overall increased activation of the WNT
pathway in ThrbPV/PV mice. Published online xx xx xxxx

1. Bassett, J. H. D. & Williams, G. R. Role of thyroid and pathology. Exp. Cell Res. 325, 138–147 10. Hata, K., Takahata, Y., Murakami, T. & Nishimura, R.
hormones in skeletal development and bone (2014). Transcriptional network controlling endochondral
maintenance. Endocr. Rev. 37, 135–187 (2016). 6. Hall, B. K. & Miyake, T. All for one and one for all: ossification. J. Bone Metab. 24, 75–82 (2017).
2. Jin, S. W., Sim, K. B. & Kim, S. D. Development and condensations and the initiation of skeletal 11. Mackie, E. J., Ahmed, Y. A., Tatarczuch, L., Chen, K. S.
growth of the normal cranial vault: an embryological development. Bioessays 22, 138–147 (2000). & Mirams, M. Endochondral ossification: how
review. J. Korean Neurosurg. Soc. 59, 192–196 (2016). 7. DeLise, A. M., Fischer, L. & Tuan, R. S. Cellular cartilage is converted into bone in the developing
3. Sperber, G. H. & Sperber, S. M. Craniofacial interactions and signaling in cartilage development. skeleton. Int. J. Biochem. Cell Biol. 40, 46–62
Embyrogenetics and Development (PMPHUSA, Osteoarthritis Cartilage 8, 309–334 (2000). (2008).
2018). 8. Long, F. & Ornitz, D. M. Development of the 12. Mackie, E. J., Tatarczuch, L. & Mirams, M.
4. Moreira, C. A., Dempster, D. W. & Baron, R. endochondral skeleton. Cold Spring Harb. Perspect. The skeleton: a multi-​functional complex organ:
Anatomy and Ultrastructure of Bone – Histogenesis, Biol. 5, a008334 (2013). the growth plate chondrocyte and endochondral
Growth and Remodeling (MDText.com, 2019). 9. Garg, P. et al. Prospective review of mesenchymal ossification. J. Endocrinol. 211, 109–121 (2011).
5. Mishina, Y. & Snider, T. N. Neural crest cell signaling stem cells differentiation into osteoblasts. Orthop. 13. Park, R. W. et al. Dual pathways to endochondral
pathways critical to cranial bone development Surg. 9, 13–19 (2017). osteoblasts: a novel chondrocyte-​derived

www.nature.com/nrendo
Reviews

osteoprogenitor cell identified in hypertrophic 39. Neben, C. L. & Merrill, A. E. in Craniofacial cells in the presence or absence of TGF-​beta signaling.
cartilage. Biol. Open 4, 608–621 (2015). Development 1st edn Vol. 115 (ed. Chai, Y.) 493–542 J. Bone Miner. Res. 21, 626–636 (2006).
14. Houben, A. et al. β-​catenin activity in late hypertrophic (Elsevier, 2015). 63. Zhou, Q. et al. IGF-​I induces adipose derived
chondrocytes locally orchestrates osteoblastogenesis 40. Farrell, B. & Breeze, A. L. Structure, activation and mesenchymal cell chondrogenic differentiation in vitro
and osteoclastogenesis. Development 143, dysregulation of fibroblast growth factor receptor and enhances chondrogenesis in vivo. In Vitro Cell
3826–3838 (2016). kinases: perspectives for clinical targeting. Biochem. Dev. Biol. Anim. 52, 356–364 (2016).
15. Aghajanian, P., Xing, W., Cheng, S. & Mohan, S. Soc. Trans. 46, 1753–1770 (2018). 64. Wang, J., Zhou, J. & Bondy, C. A. Igf1 promotes
Epiphyseal bone formation occurs via thyroid 41. Ornitz, D. M. & Marie, P. J. FGF signaling pathways longitudinal bone growth by insulin-​like actions
hormone regulation of chondrocyte to osteoblast in endochondral and intramembranous bone augmenting chondrocyte hypertrophy. FASEB J.
transdifferentiation. Sci. Rep. 7, 10432 (2017). development and human genetic disease. Genes Dev. 13, 1985–1990 (1999).
16. Morriss-​Kay, G. M. & Wilkie, A. O. Growth of the 16, 1446–1465 (2002). 65. Hardouin, S. N., Guo, R., Romeo, P.-H., Nagy, A.
normal skull vault and its alteration in craniosynostosis: 42. Su, N., Jin, M. & Chen, L. Role of FGF/FGFR signaling & Aubin, J. E. Impaired mesenchymal stem cell
insights from human genetics and experimental in skeletal development and homeostasis learning differentiation and osteoclastogenesis in mice deficient
studies. J. Anat. 207, 637–653 (2005). from mouse models. Bone Res. 2, 14003 (2014). for Igf2-P2 transcripts. Development 138, 203–213
17. Yoshida, T., Vivatbutsiri, P., Morriss-​Kay, G. M., 43. Prochazkova, K., Prochazka, J., Marangoni, P. (2011).
Saga, Y. & Iseki, S. Cell lineage in mammalian & Klein, O. D. Bones, glands, ears and more: the 66. Abuzzahab, M. J. et al. IGF-​I receptor mutations
craniofacial mesenchyme. Mech. Dev. 125, 797–808 multiple roles of FGF10 in craniofacial development. resulting in intrauterine and postnatal growth
(2008). Front. Genet. 9, 542 (2018). retardation. N. Engl. J. Med. 349, 2211–2222
18. Jiang, X., Iseki, S., Maxson, R. E., Sucov, H. M. & 44. Helsten, T., Schwaederle, M. & Kurzrock, R. Fibroblast (2003).
Morriss-​Kay, G. M. Tissue origins and interactions in growth factor receptor signaling in hereditary and 67. Raile, K. et al. Clinical and functional characteristics
the mammalian skull vault. Dev. Biol. 241, 106–116 neoplastic disease: biological and clinical implications. of the human Arg59Ter insulin-​like growth factor I
(2002). Cancer Metastasis Rev. 34, 479–496 (2015). receptor (IGF1R) mutation: implications for a gene
19. Han, J. et al. Concerted action of Msx1 and Msx2 45. Zhou, Y. X. et al. A Pro250Arg substitution in mouse dosage effect of the human IGF1R. J. Clin. Endocrinol.
in regulating cranial neural crest cell differentiation Fgfr1 causes increased expression of Cbfa1 and Metab. 91, 2264–2271 (2006).
during frontal bone development. Mech. Dev. 124, premature fusion of calvarial sutures. Hum. Mol. 68. Fang, P. et al. Severe short stature caused by novel
729–745 (2007). Genet. 9, 2001–2008 (2000). compound heterozygous mutations of the insulin-​like
20. Ishii, M., Sun, J., Ting, M. C. & Maxson, R. E. 46. Merrill, A. E. et al. Bent bone dysplasia-​FGFR2 type, growth factor 1 receptor (IGF1R). J. Clin. Endocrinol.
in Craniofacial Development 1st edn Vol. 115 a distinct skeletal disorder, has deficient canonical FGF Metab. 97, E243–E247 (2012).
(ed. Chai, Y.) 131–156 (Elsevier, 2015). signaling. Am. J. Med. Genet. 90, 550–557 (2012). 69. Liu, J. P., Baker, J., Perkins, A. S., Robertson, E. J.
21. Pal, G. P., Tamankar, B. P., Routal, R. V. & Bhagwat, S. S. 47. Jarzabek, K., Wolczynski, S., Lesniewicz, R., Plessis, G. & Efstratiadis, A. Mice carrying null mutations of
The ossification of the membranous part of the & Kottler, M. L. Evidence that FGFR1 loss-​of-function the genes encoding insulin-​like growth factor I (Igf-1)
squamous occipital bone in man. J. Anat. 138, mutations may cause variable skeletal malformations and type 1 IGF receptor (Igf1r). Cell 75, 59–72
259–266 (1984). in patients with Kallmann syndrome. Adv. Med. Sci. (1993).
22. Bellary, S. S. et al. Wormian bones: a review. Clin. Anat. 57, 314–321 (2012). 70. Woitge, H. W. & Kream, B. E. Calvariae from fetal
26, 922–927 (2013). 48. Wang, Y. et al. Abnormalities in cartilage and bone mice with a disrupted Igf1 gene have reuced rates
23. Bradley, J. P., Levine, J. P., Roth, D. A., McCarthy, J. G. development in the Apert syndrome FGFR2(+/S252W) of collagen synthesis but maintain responsiveness to
& Longaker, M. T. Studies in cranial suture biology: IV. mouse. Development 132, 3537–3548 (2005). glucocorticoids. J. Bone Miner. Res. 15, 1956–1964
Temporal sequence of posterior frontal cranial suture 49. Pierre-​Kahn, A., Hirsch, J. F., Renier, D., Metzger, J. (2000).
fusion in the mouse. Plast. Reconstr. Surg. 98, & Maroteaux, P. Hydrocephalus and achondroplasia. 71. Huang, R. L., Yuan, Y., Tu, J., Zou, G. M. & Li, Q.
1039–1045 (1996). A study of 25 observations. Childs Brain 7, 205–219 Opposing TNF-​α/IL-1β- and BMP-2-activated MAPK
24. McBratney-​Owen, B., Iseki, S., Bamforth, S. D., (1980). signaling pathways converge on Runx2 to regulate
Olsen, B. R. & Morriss-​Kay, G. M. Developmental 50. Arnaud-​López, L., Fragoso, R., Mantilla-​Capacho, J. BMP-2-induced osteoblastic differentiation. Cell Death
and tissue origins of the mammalian cranial base. & Barros-​Núñez, P. Crouzon with acanthosis nigricans. Dis. 5, e1187 (2014).
Dev. Biol. 322, 121–132 (2008). Further delineation of the syndrome. Clin. Genet. 72, 72. Phimphilai, M., Zhao, Z., Boules, H., Roca, H.
25. Cheng, A., Hardingham, T. E. & Kimber, S. J. Generating 405–410 (2007). & Franceschi, R. T. BMP signaling is required for
cartilage repair from pluripotent stem cells. Tissue Eng. 51. Abdel-​Salam, G. M. H. et al. Muenke syndrome with RUNX2-dependent induction of the osteoblast
Part B Rev. 20, 257–266 (2014). pigmentary disorder and probable hemimegalencephaly: phenotype. J. Bone Miner. Res. 21, 637–646
26. Foster, N. C., Henstock, J. R., Reinwald, Y. & El Haj, A. J. an expansion of the phenotype. Am. J. Med. Genet. A (2006).
Dynamic 3D culture: Models of chondrogenesis 155A, 207–214 (2011). 73. Ducy, P., Zhang, R., Geoffroy, V., Ridall, A. L. &
and endochondral ossification. Birth Defects Res. 52. Lee, Y. C., Song, I. W., Pai, Y. J., Chen, S. D. & Karsenty, G. Osf2/Cbfa1: a transcriptional activator
C Embryo Today 105, 19–33 (2015). Chen, Y. T. Knock-​in human FGFR3 achondroplasia of osteoblast differentiation. Cell 89, 747–754
27. Thorogood, P. The developmental specification mutation as a mouse model for human skeletal (1997).
of the vertebrate skull. Development 103, 141–153 dysplasia. Sci. Rep. 7, 43220 (2017). 74. Komori, T. et al. Targeted disruption of Cbfa1 results
(1988). 53. Ornitz, D. M. & Itoh, N. The fibroblast growth factor in a complete lack of bone formation owing to
28. Wei, X., Hu, M., Mishina, Y. & Liu, F. Developmental signaling pathway. Wiley Interdiscip. Rev. Dev. Biol. 4, maturational arrest of osteoblasts. Cell 89, 755–764
regulation of the growth plate and cranial synchondrosis. 215–266 (2015). (1997).
J. Dent. Res. 95, 1221–1229 (2016). 54. Adamek, A. & Kasprzak, A. Insulin-​like growth factor 75. Otto, F. et al. Cbfa1, a candidate gene for cleidocranial
29. Mallo, M. Formation of the middle ear: recent progress (IGF) system in liver disease. Int. J. Mol. Sci. 19, dysplasia syndrome, is essential for osteoblast
on the development and molecular mechanisms. E1308 (2018). differentiation and bone development. Cell 89,
Dev. Biol. 231, 410–419 (2001). 55. Roth, D. A. et al. Immunolocalization of transforming 765–771 (1997).
30. Stocum, D. L. & Roberts, W. E. Part I: Development growth factor β1, β2, and β3 and insulin-​like growth 76. Enomoto, H. et al. Cbfa1 is a positive regulatory
and physiology of the temporomadibular joint. factor I in premature cranial suture fusion. Plast. factor in chondrocyte maturation. J. Biol. Chem. 275,
Curr. Osteoporos. Rep. 16, 360–368 (2018). Reconstr. Surg. 99, 300–309 (1997). 8695–8702 (2000).
31. Krishnan, K. & Kanchan, T. Evaluation of spheno-​ 56. Thaller, S. R., Hoyt, J., Tesluk, H. & Holmes, R. 77. Chen, H. et al. Runx2 regulates endochondral
occipital synchondrosis: a review of literature and The effect of insulin growth factor-1 on calvarial ossification through control of chondrocyte
considerations from forensic anthropologic point sutures in a Sprague-​Dawley rat. J. Craniofac. Surg. proliferation and differentiation. J. Bone Miner. Res.
of view. J. Forensic Dent. Sci. 5, 72–76 (2013). 4, 35–39 (1993). 29, 2653–2665 (2014).
32. Coll, G. et al. Human foramen magum area and 57. Bradley, J. P. et al. Increased IGF-​I and IGF-​II mRNA 78. Kawane, T. et al. Runx2 is required for the proliferation
posterior cranial fossa volume growth in relation and IGF-​I peptide in fusing rat cranial sutures suggest of osteoblast progenitors and induces proliferation
to cranial base synchondrosis closure in the course evidence for a paracrine role of insulin like growth by regulating Fgfr2 and Fgfr3. Sci. Rep. 8, 13551
of child development. Neurosurgery 79, 722–735 factors in suture fusion. Plast. Reconstr. Surg. 104, (2018).
(2016). 129–138 (1999). 79. Zheng, Q. et al. Type X collagen gene regulation
33. Vora, S. R., Camci, E. D. & Cox, T. C. Postnatal 58. Woods, K. A., Camacho-​Hubner, C., Savage, M. O. by Runx2 contributes directly to its hypertrophic
ontogeny of the cranial base and craniofacial skeleton & Clark, A. J. L. Intrauterine growth retardation and chondrocyte-​specific expression in vivo. J. Cell Biol.
in male C57BL/6J mice: a reference standard for postnatal growth failure associated with deletion of 162, 833–842 (2003).
quantitative analysis. Front. Physiol. 6, 417 (2016). the Insulin-​like growth factor i gene. N. Engl. J. Med. 80. Yan, J. et al. Smad4 deficiency impairs chondrocyte
34. Kiesler, J. & Ricer, R. The abnormal fontanel. Am. Fam. 335, 1363–1367 (1996). hypertrophy via the Runx2 transcription factor in
Physician 67, 2547–2552 (2003). 59. Begemann, M. et al. Paternally inherited IGF2 mouse skeletal development. J. Biol. Chem. 293,
35. Cendekiawan, T., Wong, R. W. K. & Rabie, A. B. M. mutation and growth restriction. N. Engl. J. Med. 9162–9175 (2018).
Relationships between cranial base synchondroses 373, 349–356 (2015). 81. Bialek, P. et al. A Twist code determines the onset
and craniofacial development: a review. Open Anat. J. 60. Qiao, M., Shapiro, P., Kumar, R. & Passaniti, A. of osteoblast differentiation. Dev. Cell 6, 423–435
2, 67–75 (2010). Insulin-​like growth factor-1 regulates endogenous (2004).
36. Goldstein, J. A. et al. Earlier evidence of shpeno-​ RUNX2 activity in endothelial cells through a 82. Shirakabe, K., Terasawa, K., Miyama, K., Shibuya, H. &
occipital synchondrosis fusion correlates with severity phosphatidylinositol 3-kinase/ERK-​dependent and Nishida, E. Regulation of the activity of the transcription
of midface hypoplasia in patients with syndromic Akt-​independent signaling pathway. J. Biol. Chem. factor Runx2 by two homeobox, Msx2 and Dlx5. Genes
craniosynostosis. Plast. Reconstr. Surg. 134, 279, 42709–42718 (2004). Cells 6, 851–856 (2001).
504–510 (2014). 61. Fujita, T. et al. Runx2 induces osteoblast and 83. Widelitz, R. Wnt signaling through canonical and non-​
37. Mazzaferro, D. M. et al. Incidence of cranial base chondrocyte differentiation and enhances their canonical pathways: recent progress. Growth Factors
suture fusion in infants with craniosynostosis. migration by coupling with PI3K-​Akt signaling. 23, 111–116 (2005).
Plast. Reconstr. Surg. 141, 559e–570e (2018). J. Cell Biol. 166, 85–95 (2004). 84. Nusse, R. & Clevers, H. Wnt/β-​Catenin signaling,
38. Tubbs, R. S. & Oakes, W. J. The Chiari Malformations 62. Longobardi, L. et al. Effect of IGF-​I in the disease, and emerging therapeutic modalities. Cell 169,
(Springer, 2013). chondrogenesis of bone marrow mesenchymal stem 985–999 (2017).

Nature Reviews | Endocrinology


Reviews

85. Parr, B. A., Shea, M. J., Vassileva, G. & McMahon, A. P. 110. Capelo, L. P., Beber, E. H., Fonseca, T. L. & male mice and their effects on bone mass, bone
Mouse Wnt genes exhibit discrete domains of Gouveia, C. H. A. The monocarboxylate transporter 8 turnover, and the Wnt inhibitors sclerostin and
expression in the early embryonic CNS and limb buds. and L-​type amino acid transporters 1 and 2 are dickkopf-1. Endocrinology 156, 3517–3527 (2015).
Development 119, 247–261 (1993). expressed in mouse skeletons and osteoblastic 135. Hu, Z. et al. Energy metabolism in the bone is
86. Kato, M. et al. Cbfa1-independent decrease in MC3T3-E1 cells. Thyroid 19, 171–180 (2009). associated with histomorphometric changes in rats
osteoblast proliferation, osteopenia, and persistent 111. Abe, S. et al. Monocarboxylate transporter 10 with hyperthyroidism. Cell. Physiol. Biochem. 46,
embryonic eye vascularization in mice deficient in Lrp5, functions as a thyroid hormone transporter in 1471–1482 (2018).
a Wnt coreceptor. J. Cell Biol. 157, 303–314 (2002). chondrocytes. Endocrinology 153, 4049–4058 136. Cheng, S., Xing, W., Pourteymoor, S. & Mohan, S.
87. Day, T. F., Guo, X., Garrett-​Beal, L. & Yang, Y. (2012). Effects of thyroxine (T4), 3,5,3’-triiodo-​L-thyronine (T3)
Wnt/β-​catenin signaling in mesenchymal progenitors 112. Leitch, V. D. et al. An essential physiological role for and their metabolites on osteoblast differentiation.
controls osteoblast and chondrocyte differentiation MCT8 in bone in male mice. Endocrinology 158, Calcif. Tissue Int. 99, 435–442 (2016).
during vertebrate skeletogenesis. Dev. Cell 8, 3055–3066 (2017). 137. O’Shea, P. J., Guigon, C. J., Williams, G. R. &
739–750 (2005). 113. Braverman, L. E., Ingbar, S. H. & Sterling, K. Cheng, S. Y. Regulation of fibroblast growth factor
88. Gong, Y. et al. LDL receptor-​related protein 5 (LRP5) Conversion of thyroxine (T4) to triiodothyronine (T3) receptor-1 (FGFR1) by thyroid hormone: identification
affects bone accrual and eye development. Cell 107, in athyreotic subjects. J. Clin. Invest. 49, 855–864 of a thyroid hormone response element in the murine
513–523 (2001). (1970). Fgfr1 promoter. Endocrinology 148, 5966–5976
89. Song, L. et al. Lrp6-mediated canonical Wnt signaling 114. Gereben, B. et al. Cellular and molecular basis of (2007).
is required for lip formation and fusion. Development deiodinase-​regulated thyroid hormone signaling. 138. Robson, H., Siebler, T., Stevens, D. A., Shalet, S. M.
136, 3161–3171 (2009). Endocr. Rev. 29, 898–938 (2008). & Williams, G. R. Thyroid hormone acts directly on
90. Roetzer, K. M. et al. Novel familial mutation of LRP5 115. Arrojo e Drigo, R. & Bianco, A. C. Type 2 deiodinase growth plate chondrocytes to promote hypertrophic
causing high bone mass: genetic analysis, clinical at the crossroads of thyroid hormone action. Int. J. differentiation and inhibit clonal expansion and cell
presentation, and characterization of bone matrix Biochem. Cell Biol. 43, 1432–1441 (2011). proliferation. Endocrinology 141, 3887–3897 (2000).
mineralization. Bone 107, 154–160 (2018). 116. St. Germain, D. L., Galton, V. A. & Hernandez, A. 139. Miura, M. et al. Thyroid hormones promote
91. Boyden, L. M. et al. High bone density due to a Minireview: defining the roles of the iodothyronine chondrocyte differentiation in mouse ATDC5 cells and
mutation in LDL-​receptor-related protein 5. N. Engl. deiodinases: current concepts and challenges. stimulate endochondral ossification in fetal mouse
J. Med. 346, 1513–1521 (2002). Endocrinology 150, 1097–1107 (2009). tibias through iodothyronine deiodinases in the growth
92. Rawadi, G., Vayssière, B., Dunn, F., Baron, R. 117. Kanatani, M. et al. Thyroid hormone stimulates plate. J. Bone Miner. Res. 17, 443–454 (2002).
& Roman-​Roman, S. BMP-2 controls alkaline osteoclast differentiation by a mechanism 140. Okubo, Y. & Reddi, A. H. Thyroxine downregulates
phosphatase expression and osteoblast mineralisation independent of RANKL-​RANK interaction. J. Cell. Sox9 and promotes chondrocyte hypertrophy.
by a Wnt autocrine loop. J. Bone Miner. Res. 18, Physiol. 201, 17–25 (2004). Biochem. Biophys. Res. Commun. 306, 186–190
1842–1853 (2003). 118. Williams, A. J. et al. Iodothyronine deiodinase enzyme (2003).
93. Gaur, T. et al. Canonical WNT signaling promotes activities in bone. Bone 43, 126–134 (2008). 141. Barnard, J. C. et al. Thyroid hormones regulate
osteogenesis by directly stimulating Runx2 gene 119. Hönes, G. S. et al. Noncanonical thyroid hormone fibroblast growth factor receptor signaling during
expression. J. Biol. Chem. 280, 33132–33140 (2005). signaling mediates cardiometabolic effects in vivo. chondrogenesis. Endocrinology 146, 5568–5580
94. Nagayama, M. et al. Wnt/beta-​catenin signaling Proc. Natl Acad. Sci. USA 114, E11323–E11332 (2005).
regulates cranial base development and growth. (2017). 142. Britto, J. M., Fenton, A. J., Holloway, W. R. &
J. Dent. Res. 87, 244–249 (2008). 120. Flamant, F. et al. Thyroid hormone signaling pathways: Nicholson, G. C. Osteoblasts mediate thyroid
95. Brownstein, M. J., Eskay, R. L. & Palkovits, M. time for a more precise nomenclature. Endocrinology hormone stimulation of osteoclastic bone resorption.
Thryotropin releasing hormone in the median eminence 158, 2052–2057 (2017). Endocrinology 134, 169–176 (1994).
is in processes of paraventricular nucleus neurons. 121. Astapova, I. et al. The nuclear receptor corepressor 143. Gouveia, C. H. A., Miranda-​Rodrigues, M.,
Neuropeptides 2, 197–201 (1982). (NCoR) controls thyroid hormone sensitivity and the Martins, G. M. & Neofiti-​Papi, B. Thyroid hormone
96. Segerson, T. P. et al. Localization of thyrotropin-​releasing set point of the hypothalamic-​pituitary-thyroid axis. and skeletal development. Vitam. Horm. 106,
hormone prohormone messenger ribonucleic acid in Mol. Endocrinol. 25, 212–224 (2011). 383–472 (2018).
rat brain by in situ hybridization. Endocrinology 121, 122. Leng, X., Tsai, S. Y., O’Malley, B. W. & Tsai, M. J. 144. Lindsey, R. C., Aghajanian, P. & Mohan, S.
98–107 (1987). Ligand-​dependent conformational changes in thyroid Thyroid hormone signaling in the development of the
97. Szarek, E., Cheah, P. S., Schwartz, J. & Thomas, P. hormone and retinoic acid receptors are potentially endochondral skeleton. Vitam. Horm. 106, 351–381
Molecular genetics of the developing neuroendocrine enhanced by heterodimerization with retinoic X (2018).
hypothalamus. Mol. Cell Endocrinol. 323, 115–123 receptor. J. Steroid Biochem. Mol. Biol. 46, 643–661 145. Wassner, A. J. & Brown, R. S. Congenital
(2010). (1993). hypothyroidism: recent advances. Curr. Opin.
98. Harris, A. R. et al. The physiological role of 123. Wagner, R. L. et al. A structural role for hormone in Endocrinol. Diabetes Obes. 22, 407–412 (2015).
thyrotropin-​releasing hormone in the regulation of the thyroid hormone receptor. Nature 378, 690–697 146. Bakker, B. et al. Two decades of screening for
thyroid-​stimulating hormone and prolactin secretion (1995). congenital hypothyroidism in the Netherlands: TPO
in the rat. J. Clin. Invest. 61, 441–448 (1978). 124. Fondell, J. D., Ge, H. & Roeder, R. G. Ligand induction gene mutations in total iodide organification defects
99. Steinfelder, H. J. et al. Thyrotropin-​releasing hormone of a transcriptionally active thyroid hormone receptor (an update). J. Endocrinol. Metab. 85, 3708–3712
regulation of human TSHB expression: role of the coactivator complex. Proc. Natl Acad. Sci. USA 93, (2000).
pituitary-​specific transcription factor (Pit-1/GHF-1) and 8329–8333 (1996). 147. Moreno, J. C. et al. Inactivating mutations in the
potential interaction with a thyroid hormone-​inhibitory 125. Hiroi, Y. et al. Rapid nongenomic actions of thyroid gene for thyroid oxidase 2 (THOX2) and congenital
element. Proc. Natl Acad. Sci. USA 88, 3130–3134 hormone. Proc. Natl Acad. Sci. USA 103, 14104–14109 hypothyroidism. N. Engl. J. Med. 347, 95–102
(1991). (2006). (2002).
100. Sun, Y., Lu, X. & Gershengorn, M. C. Thyrotropin-​ 126. Martin, N. P. et al. A rapid cytoplasmic mechanism for 148. Macchia, P. E. et al. PAX8 mutations associated
releasing hormone receptors – similarities and PI3 kinase regulation by the nuclear thyroid hormone with congenital hypothyroidism caused by thyroid
differences. J. Mol. Endocrinol. 30, 87–97 (2003). receptor, TRβ, and genetic evidence for its role in the dysgenesis. Nat. Genet. 19, 83–86 (1998).
101. Negi, C. S. Introduction to Endocrinology (PHI Learning, maturation of mouse hippocampal synapses in vivo. 149. Tao, Y. X. Inactivating mutations of G protein-​coupled
2009). Endocrinology 155, 3713–3724 (2014). receptors and diseases: structure-​function insights
102. Kopp, P. The TSH receptor and its role in thyroid 127. Forrest, D., Sjoberg, M. & Vennstrom, B. Contrasting and therapeutic implications. Phamacol. Ther. 111,
disease. Cell Mol. Life Sci. 58, 1301–1322 (2001). developmental and tissue-​specific expression of alpha 949–973 (2006).
103. Abel, E. D., Ahima, R. S., Boers, M. E., Elmquist, J. K. and beta thyroid hormone receptor genes. EMBO J. 9, 150. Dumitrescu, A. M. et al. Mutations in SECISBP2 result
& Wondisford, F. E. Critical role for thyroid hormone 1519–1528 (1990). in abnormal thyroid hormone metabolism. Nat. Genet.
receptor β2 in the regulation of paraventricular 128. Cray, J. J. Jr, Khaksarfard, K., Weinberg, S. M., 37, 1247–1252 (2005).
thyrotropin-​releasing hormone neurons. J. Clin. Invest. Elsalanty, M. & Yu, J. C. Effects of thyroxine exposure 151. Bubenik, J. L., Miniard, A. & Driscoll, D.
107, 1017-1023 (2001). on osteogenesis in mouse calvarial pre-​osteoblasts. Characterization of the UGA-​recoding and SECIS-​
104. O’Shea, P. J. et al. Advanced bone formation in mice PLoS One 8, e69067 (2013). binding activities of SECIS-​binding protein 2. RNA
with a dominant-​negative mutation in the thyroid 129. Park, S.-H. et al. Potential of L-​thyroxine to differentiate Biol. 11, 1402–1413 (2014).
hormone receptor β gene due to activation of osteoblast-​like cells via angiopoietin1. Biochem. 152. Smith, D. W. & Popich, G. Large fontanels in congenital
Wnt/β-​catenin protein signaling. J. Biol. Chem. 287, Biophys. Res. Commun. 478, 1409–1415 (2016). hypothyroidism: a potental clue toward earlier
17812–17822 (2012). 130. Kassem, M., Mosekilde, L. & Ericksen, E. F. Effects of recognition. J. Pediatr. 80, 753–756 (1972).
105. Taylor, P. N. et al. Whole-​genome sequence-​based triiodothyronine on DNA synthesis and differentiation 153. Loevy, H. T., Aduss, H. & Rosenthal, I. M. Tooth
analysis of thyroid function. Nat. Commun. 6, markers of normal human osteoblast-​like cells in vitro. eruption and craniofacial development in congenital
5681–5690 (2015). Biochem. Mol. Biol. Int. 30, 779–788 (1993). hypothyroidism: report of case. J. Am. Dent. Assoc.
106. Teumer, A. et al. Genome-​wide analyses identify 131. Ishisaki, A. et al. Activation of p38 mitogen-​activated 115, 429–431 (1987).
a role for SLC17A4 and AADAT in thyroid hormone protein kinase mediates thyroid hormone-​stimulated 154. Kaplan, S. B., Kemp, S. S. & Oh, K. S. Radiographic
regulation. Nat. Commun. 9, 4455 (2018). osteocalcin synthesis in osteoblasts. Mol. Cell. manifestations of congenital anomalies of the skull.
107. Friesema, E. C. et al. Effective cellular uptake and Endocrinol. 214, 189–195 (2004). Radiol. Clin. North Am. 29, 195–218 (1991).
efflux of thyroid hormone by human monocarboxylate 132. Stevens, D. A. et al. Thyroid hormone activates 155. Pezzuti, I. L., Lima, P. P. & Dias, V. M. Congenital
transporter 10. Mol. Endocrinol. 22, 1357–1369 fibroblast growth factor receptor-1 in bone. hypothyroidism: the clinical profile of affected
(2008). Mol. Endocrinol. 17, 1751–1766 (2003). newborns identified by the Newborn Screening
108. Visser, W. E., Friesema, E. C. & Visser, T. J. Minireview: 133. Xing, W. et al. Genetic evidence that thyroid hormone Program of the State of Minas Gerais, Brazil.
thyroid hormone transporters: the knowns and the is indispensable for prepubertal insulin-​like growth J. Pediatr. (Rio J) 85, 72–79 (2009).
unknowns. Mol. Endocrinol. 25, 1–14 (2011). factor-​I expression and bone acquisition in mice. 156. Bretones, P. et al. A familial case of congenital
109. Wirth, E. K., Schweizer, U. & Köhrle, J. Transport of J. Bone Miner. Res. 27, 1067–1079 (2012). hypothyroidism caused by a homozygous mutation
thyroid hormone in the brain. Front. Endocrinol. 5, 134. Tsourdi, E., Rijntjes, E., Kohrle, J., Hofbauer, L. C. & of the thyrotropin receptor gene. Thyroid 11, 977–980
98 (2014). Rauner, M. Hyperthyroidism and hypothyroidism in (2001).

www.nature.com/nrendo
Reviews

157. Gagné, N., Parma, J., Deal, C., Vassart, G. & Van Vliet, G. 181. Horn, G. & Ford, E. H. R. The effects of thyroid hypothyroidism. J. Plast. Reconstr. Aesthet. Surg.
Apparent congenital athyreosis contrasting with deficiency on the growth of the rat skull. J. Anat. 95, 66, e284–e286 (2013).
normal plasma thyroglobulin levels and associated with 131–136 (1961). 207. Riggs, W. Jr, Wilroy, R. S. Jr & Etteldorf, J. N. Neonatal
inactivating mutations in the thyrotropin receptor gene: 182. Dawson, A., McNaughton, F. J., Goldsmith, A. R. & hyperthyroidism with accelerated skeletal maturation,
are athyreosis and ectopic thyroid distinct entities? Degen, A. A. Ratite-​like neoteny induced by neonatal craniosynostosis, and brachydactyly. Radiology 105,
J. Clin. Endocrinol. Metab. 83, 1771–1775 (1998). thyroidectomy of European starlings, Sturnus vulgaris. 621–625 (1972).
158. Azevedo, M. F. et al. Selenoprotein-​related disease J. Zool. 232, 633–639 (1994). 208. Vucic, S. et al. Thyroid function during early life and
in a young girl caused by nonsense mutations in the 183. Dye, J. A. & Kinder, F. S. A prepotent factor in the dental development. J. Dent. Res. 96, 1020–1026
SBP2 gene. J. Clin. Endocrinol. Metab. 95, determination of skull shape. Am. J. Anat. 54, (2017).
4066–4071 (2010). 333–346 (1934). 209. Fuhrer, D., Wonerow, P., Willgerodt, H. & Paschke, R.
159. Huffmeier, U., Tietze, H. U. & Rauch, A. Severe skeletal 184. Sun, H. et al. New case of thyroid hormone resistance Identification of a new thyrotropin receptor germline
dysplasia caused by undiagnosed hypothyroidism. α caused by a mutation of THRA/TRα1. J. Endocr. Soc. mutation (Leu629Phe) in a family with neonatal
Eur. J. Med. Genet. 50, 209–215 (2007). 3, 665–669 (2019). onset of autosomal dominant nonautoimmune
160. Vakili, R. & Mazlouman, S. J. Dyshormonogenic 185. Bochukova, E. et al. A mutation in the thyroid hyperthyroidism. J. Clin. Endocrinol. Metab. 82,
hypothyroidism with normal neurological development, hormone receptor alpha gene. N. Engl. J. Med. 366, 4234–4238 (1997).
unexplained short stature and facial anomalies in three 243–249 (2012). 210. Kopp, P., Jameson, J. L. & Roe, T. F. Congenital
siblings. Clin. Dysmorphology 12, 21–27 (2003). 186. Fozzatti, L. et al. Nuclear receptor corepressor nonautoimmune hyperthyroidism in a nonidentical
161. Di Cosmo, C. et al. Clinical and molecular characterization (NCOR1) regulates in vivo actions of a mutated thyroid twin caused by a sporadic germline mutation in the
of a novel selenocysteine insertion sequence-​binding hormone receptor α. Proc. Natl Acad. Sci. USA 110, thyrotropin receptor gene. Thyroid 7, 765–770
protein 2 (SBP2) gene mutation (R128X). J. Clin. 7850–7855 (2013). (1997).
Endocrinol. Metab. 94, 4003–4009 (2009). 187. Markossian, S. et al. CRISPR/Cas9 editing of the mouse 211. Wilroy, R. S. J. & Etteldorf, J. N. Familial
162. Meyerhoff, W. L. Hypothyroidism and the ear: Thra gene produces models with variable resistance to hyperthyroidism including two siblings with neonatal
electrophysiological, morphological and chemical thyroid hormone. Thyroid 28, 139–150 (2018). Graves’ disease. J. Pediatr. 78, 625–632 (1971).
considerations. Laryngoscope 89, 1–25 (1979). 188. Demir, K. et al. Diverse genotypes and phenotypes 212. Menking, M. et al. Premature craniosynostosis
163. Gecgelen Cesur, M., Cesur, G., Ogrenim, M. & Alkan, A. of three novel thyroid hormone receptor-​α mutants. associated with hyperthyroidism in 4 children with
Do prenatal and postnatal hypothyroidism affect the J. Clin. Endocrinol. Metab. 101, 2945–2954 (2016). reference to 5 further cases in the literature.
craniofacial structure?: an experimental study. Angle 189. van Mullem, A. et al. Clinical phenotype and mutant Monatsschr. Kinderheilkd. 120, 106–110 (1972).
Orthod. 86, 983–990 (2016). TRα1. N. Engl. J. Med. 366, 1451–1453 (2012). 213. Karnofsky, D. & Cronkite, E. P. Effect of thyroxine on
164. Nanto-​Salonen, K., Glasscock, G. F. & Rosenfeld, R. G. 190. Moran, C. et al. Contrasting phenotypes in resistance eruption of teeth in newborn rats. Exp. Biol. Med. 40,
The effects of thyroid hormone on insulin-​like growth to thyroid hormone alpha correlate with divergent 568–570 (1939).
factor (IGF) and IGF-​binding protein (IGFBP) expression properties of thyroid hormone receptor α1 mutant 214. Hoath, S. B. & Pickens, W. L. Effect of thyroid hormone
in the neonatal rat: prolonged high expression of proteins. Thyroid 27, 973–982 (2017). and epidermal growth factor on tactile hair development
IGFBP-2 in methimazole-​induced congenital 191. van Gucht, A. L. et al. Resistance to thyroid hormone and craniofacial morphogenesis in the postnatal rat.
hypothyroidism. Endocrinology 129, 2563–2570 alpha in an 18-month old girl: clinical, therapeutic, J. Craniofac. Genet. Dev. Biol. 7, 161–167 (1987).
(1991). and molecular characteristics. Thyroid 26, 338–346 215. Akita, S., Nakamura, T., Hirano, A., Fujii, T. &
165. Abe, E. et al. TSH is a negative regulator of skeletal (2016). Yamashita, S. Thyroid hormone action on rat calvarial
remodelling. Cell 115, 151–162 (2003). 192. Espiard, S. et al. A novel mutation in THRA gene sutures. Thyroid 4, 99–106 (1994).
166. Cordas, E. A. et al. Thyroid hormone receptors control associated with an atypical phenotype of resistance 216. Durham, E. L. et al. Thyroxine exposure effects on the
developmental maturation of the middle ear and to thyroid hormone. J. Clin. Endocrinol. Metab. 100, cranial base. Calcif. Tissue Int. 101, 300–311 (2017).
the size of the ossicular bones. Endocrinology 153, 2841–2848 (2015). 217. Jaeschke, H. et al. Hyperthyroidism and papillary
1548–1560 (2012). 193. Van Mullem, A. et al. Clinical phenotype of a new type thyroid carcinoma in thyrotropin receptor D633H
167. Seeher, S. et al. Secisbp2 is essential for embryonic of thyroid hormone resistance caused by a mutation of mutant mice. Thyroid 28, 1372–1386 (2018).
development and enhances selenoprotein expression. the TRα1 receptor: consequences of LT4 treatment. 218. Neumann, S., Krohn, K., Chey, S. & Paschke, R.
Antioxid. Redox Signal. 21, 835–849 (2014). J. Clin. Endocrinol. Metab. 98, 3029–3038 (2013). Mutations in the mouse TSH receptor equivalent
168. Bassett, J. H. D. et al. A lack of thyroid hormones rather 194. Moran, C. et al. Resistance to thyroid hormone caused to human constitutively activating TSH receptor
than excess thyrotropin causes abnormal skeletal by a mutation in thyroid hormone receptor (TR)α1 and mutations also cause constitutive activity. Horm.
development in hypothyroidism. Mol. Endocrinol. 22, TRα2: clinical, biochemical, and genetic analyses of Metab. Res. 33, 263–269 (2001).
501–512 (2008). three related patients. Lancet Diabetes Endocrinol. 2, 219. Vela, A. et al. Thyroid hormone resistance from newborns
169. Endo, T. & Kobayashi, T. Excess TSH causes 619–626 (2014). to adults: a Spanish experience. J. Endocrinol. Invest. 42,
abnormal skeletal development in young mice with 195. Moran, C. et al. An adult female with resistance 941–949 (2019).
hypothyroidism via suppressive effects on the growth to thyroid hormone mediated by defective thyroid 220. Phillips, S. A. et al. Extreme thyroid hormone resistance
plate. Am. J. Physiol. Endocrinol. Metab. 305, hormone receptor α. J. Clin. Endocrinol. Metab. 98, in a patient with a novel truncated TR mutant. J. Clin.
E660–E666 (2013). 4254–4261 (2013). Endocrinol. Metab. 86, 5142–5147 (2001).
170. Takeuchi, H., Nakagawa, Y. & Igarashi, Y. Studies 196. O’Shea, P. J. et al. Contrasting skeletal phenotypes 221. Yoh, S. M., Chatterjee, V. K. & Privalsky, M. L.
on gene expression in calvaria and serum levels of in mice with an identical mutation targeted to thyroid Thyroid hormone resistance syndrome manifests as
insulin-​like growth factor-​I and bone Gla protein in hormone receptor α1 or β. Mol. Endocrinol. 19, an aberrant interaction between mutant T3 receptors
the methimazole-​induced congenital hypothyroid rat. 3045–3059 (2005). and transcriptional corepressors. Mol. Endocrinol. 11,
Endocr. J. 40, 351–362 (1993). 197. Bassett, J. H. D. et al. Thyroid hormone receptor α 470–480 (1997).
171. Keer, S. et al. Anatomical assessment of the adult mutation causes a severe and thyroxine-​resistant 222. Refetoff, S. & Dumitrescu, A. M. Syndromes of
skeleton of zebrafish reared under different thyroid skeletal dysplasia in female mice. Endocrinology 155, reduced sensitivity to thyroid hormone: genetic
hormone profiles. Anat. Rec. 302, 1754–1769 (2019). 3699–3712 (2014). defects in hormone receptors, cell transporters and
172. Campinho, M. A. et al. A thyroid hormone regulated 198. Tinnikov, A. et al. Retardation of post-​natal deiodination. Best Pract. Res. Clin. Endocrinol. Metab.
asymmetric responsive centre is correlated with eye development caused by a negatively acting thyroid 21, 277–305 (2007).
migration during flatfish metamorphosis. Sci. Rep. 8, hormone receptor α1. EMBO J. 21, 5079–5087 223. Weiss, R. E. & Refetoff, S. Resistance to thyroid
12267 (2018). (2002). hormone. Rev. Endocr. Metab. Disord. 1, 97–108
173. Hunter, I., Greene, S. A., MacDonald, T. M. 199. Bassett, J. H. D. et al. Thyroid status during skeletal (2000).
& Morris, A. D. Prevalence and aetiology of development determines adult bone structure and 224. Dumitrescu, A. M. & Refetoff, S. The syndromes of
hypothyroidism in the young. Arch. Dis. Child. 83, mineralization. Mol. Endocrinol. 21, 1893–1904 reduced sensitivity to thyroid hormone. Biochim.
207–210 (2000). (2007). Biophys. Acta 1830, 3987–4003 (2013).
174. Gutch, M. et al. Skeletal manifestations of juvenile 200. Desjardin, C. et al. Chondrocytes play a major role in 225. Bassett, J. H. D. et al. Thyroid hormone excess rather
hypothyroidism and the impact of treatment on the stimulation of bone growth by thyroid hormone. than thyrotropin deficiency induces osteoporosis in
skeletal system. Indian. J. Endocrinol. Metab. 17, Endocrinology 155, 3123–3135 (2014). hyperthyroidism. Mol. Endocrinol. 21, 1095–1107
S181–S183 (2013). 201. Leger, J. & Carel, J. C. Hyperthyroidism in childood: (2007).
175. Marzuillo, P. et al. Very early onset of autoimmune causes, when and how to treat. J. Clin. Res. Pediatr. 226. O’Shea, P. J. et al. A thyrotoxic skeletal phenotype of
thyroiditis in a toddler with severe hypothyroidism Endocrinol. 5, 50–56 (2013). advanced bone formation in mice with resistance to
presentation: a case report. Ital. J. Pediatr. 42, 61 202. Havgaard Kjær, R., Smedegård Andersen, M. thyroid hormone. Mol. Endocrinol. 17, 1410–1424
(2016). & Hansen, D. Increasing incidence of juvenile (2003).
176. Rivkees, S. A., Bode, H. H. & Crawford, J. D. Long-​term thyrotoxicosis in Denmark: a nationwide study, 227. Pepene, C. E. et al. Effects of triiodothyronine on the
growth in juvenile acquired hypothyroidism: the failure 1998-2012. Horm. Res. Paediatr. 84, 102–107 insulin-​like growth factor system in primary human
to achieve normal adult stature. N. Engl. J. Med. 318, (2015). osteoblastic cells in vitro. Bone 29, 540–546 (2001).
599–602 (1988). 203. Kopp, P. et al. Congenital hyperthyroidism caused by 228. Zhang, W. et al. Effects of insulin and insulin-​like
177. Boersma, B., Otten, B. J., Stoelinga, G. B. & Wit, J. M. a mutation in the thyrotropin-​receptor gene. N. Engl. growth factor 1 on osteoblast proliferation and
Catch-​up growth after prolonged hypothyroidism. J. Med. 322, 150–154 (1995). differentiation: differential signalling via Akt and ERK.
Eur. J. Pediatr. 155, 362–367 (1996). 204. Johnsonbaugh, R. E., Bryan, R. N., Hierlwimmer, U. R. Cell Biochem. Funct. 30, 297–302 (2012).
178. Liddell, H. S. The growth of the head in thyroidectomized & Georges, L. P. Premature craniosynostosis: a common 229. Rodriguez-​Carballo, E., Gámez, B. & Ventura, F.
sheep. Anat. Rec. 30, 327–332 (1925). complication of juvenile thyrotoxicosis. J. Pediatr. 93, p38 MAPK signaling in osteoblast differentiation.
179. Todd, T. W. & Wharton, R. E. The effect of thyroid 188–199 (1978). Front. Cell Dev. Biol. 4, 40 (2016).
deficiency upon skull growth in the sheep. Am. J. Anat. 205. Hirano, A., Akita, S. & Fujii, T. Craniofacial deformities 230. Bassett, J. H. D., Swinhoe, R., Chassande, O.,
55, 97–116 (1934). associated with juvenile hyperthyroidism. Cleft Palate Samarut, J. & Williams, G. R. Thyroid hormone
180. Hunter, M. W. & Sawin, P. B. The effects of Craniofac. J. 32, 328–333 (1995). regulates heparan sulfate proteoglycan expression
thyroidectomy on the skull of the domestic rabbit. 206. Higashino, T. & Hirabayashi, S. A secondary in the growth plate. Endocrinology 147, 295–305
Am. J. Anat. 71, 417–449 (1942). craniosynostosis associated with juvenile (2006).

Nature Reviews | Endocrinology


Reviews

231. Wang, L., Shao, Y. Y. & Ballock, R. T. Thyroid hormone-​ pressure due to premature fusion of skull sutures. Acknowledgements
mediated growth and differentiation of growth plate Arch. Dis. Child. 44, 252–257 (1969). J.H.D.B. and G.R.W. are funded by a Wellcome Trust Joint
chondrocytes involves IGF-1 modulation of beta-​catenin 237. Penfold, J. L. & Simpson, D. A. Premature Investigator Award (110141/Z/15/Z and 110140/Z/15/Z).
signaling. J. Bone Miner. Res. 25, 1138–1146 (2010). craniosynostosis - a complication of thyroid
232. Tsourdi, E. et al. The role of dickkopf-1 in thyroid replacement therapy. J. Pediatr. 86, 360–363 Author contributions
hormone-​induced changes of bone remodeling in male (1975). V.D.L., J.H.D.B. and G.R.W. researched data for the article,
mice. Endocrinology 160, 664–674 (2019). 238. Howie, R. N. et al. Effects of in utero thyroxine made substantial contributions to discussion of content of
233. Wang, L., Shao, Y. Y. & Ballock, R. T. Thyroid hormone exposure on murine cranial suture growth. PLoS One the article, wrote the article and reviewed and edited the
interacts with the Wnt/beta-​catenin signaling pathway 11, e0167805 (2016). manuscript before submission.
in the terminal differentiation of growth plate 239. Supornsilchai, V. et al. Expanding clinical spectrum of
chondrocytes. J. Bone Miner. Res. 22, 1988–1995 non-​autoimmune hyperthyroidism due to an activating Competing interests
(2007). germline mutation, p.M453T, in the thyrotropin The authors declare no competing interests.
234. Fryns, J. P. & Moerman, P. Unknown syndrome: receptor gene. Clin. Endocrinol. 70, 623–628
abnormal facies, hypothyroidism, and severe (2009). Publisher’s note
retardation: a second patient. J. Med. Genet. 25, 240. Gruters, A. et al. Severe congenital hyperthyroidism Springer Nature remains neutral with regard to jurisdictional
498–499 (1988). caused by a germ-​line neo mutation in the extracellular claims in published maps and institutional affiliations.
235. Ma, D. et al. A de novo 10.79 Mb interstitial deletion portion of the thyrotropin receptor. J. Clin. Endocrinol.
at 2q13q14.2 involving PAX8 causing hypothyroidism Metab. 83, 1431–1436 (1998). Supplementary information
and mullerian agenesis: a novel case report and 241. Chawla, R. et al. Squamosal suture craniosynostosis Supplementary information is available for this paper at
literature review. Mol. Cytogenet. 7, 85 (2014). due to hyperthyroidism caused by an activating TSH https://doi.org/10.1038/s41574-019-0304-5.
236. Robinson, D. C., Hall, R. & Munro, D. S. Graves’s receptor mutation (T632I). Thyroid 25, 1167–1172
disease, an unusual complication: raised intracranial (2015). © Springer Nature Limited 2020

www.nature.com/nrendo

You might also like