Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

RevIewS

The role of thyroglobulin in thyroid


hormonogenesis
Cintia E. Citterio   1,2, Héctor M. Targovnik1,2 and Peter Arvan   3*
Abstract | In humans, the thyroid hormones T3 and T4 are synthesized in the thyroid gland in
a process that crucially involves the iodoglycoprotein thyroglobulin. The overall structure of
thyroglobulin is conserved in all vertebrates. Upon thyroglobulin delivery from thyrocytes to the
follicular lumen of the thyroid gland via the secretory pathway , multiple tyrosine residues can
become iodinated to form mono-iodotyrosine (MIT) and/or di-iodotyrosine (DIT); however,
selective tyrosine residues lead to preferential formation of T4 and T3 at distinct sites.
T4 formation involves oxidative coupling between two DIT side chains, and de novo T3 formation
involves coupling between an MIT donor and a DIT acceptor. Thyroid hormone synthesis is
stimulated by TSH activating its receptor (TSHR), which upregulates the activity of many thyroid
gene products involved in hormonogenesis. Additionally , TSH regulates post-translational
changes in thyroglobulin that selectively enhance its capacity for T3 formation — this process is
important in iodide deficiency and in Graves disease. 167 different mutations, many of which are
newly discovered, are now known to exist in TG (encoding human thyroglobulin) that can lead to
defective thyroid hormone synthesis, resulting in congenital hypothyroidism.

Metamorphosis
Many biological processes are regulated by thyroid Origins of thyroid hormonogenesis
An abrupt developmental hormones in vertebrates and some invertebrates 1,2 In humans, the primary, original source of thy-
change in the shape or form including metamorphosis (in echinoderms3–5 and in roid hormones is the iodoglycoprotein thyroglobu-
of an animal. some chordates, including teleosts, and many amphib- lin (Supplementary Fig. 1), which is the most highly
ians6) and control of seasonality7, as well as growth, expressed protein in the thyroid gland. Interestingly,
development, heart rate and thermogenesis in mam- thyroid-hormone-receptor-mediated bioactivity has
mals8. In many of these processes, thyroid hormones been traced back in evolution to aquatic life forms that
primarily regulate the gene expression controlling oxi- precede the first appearance of the thyroglobulin (TG)
dative metabolism9. In vertebrates, thyroid hormones gene15. Specifically, in both sea urchin16,17 and amphi-
are members of the family of peptide hormones, and oxus18, many orthologues of genes involved in thyroid
in this case, a protein precursor generates an amino hormone synthesis are already present19–22, although
1
Universidad de Buenos Aires,
Facultad de Farmacia y
acid-derived bioactive product of ~0.8 kDa. In humans, these species lack a gene encoding a vertebrate-like
Bioquímica, Departamento de once secreted into the bloodstream, T4 is long lived thyroglobulin. Amphioxus, which does not have
Microbiología, Inmunología y (t1/2 = ~7 days), whereas T3, which exhibits far more a thyroid follicular structure, accumulates iodide
Biotecnología/Cátedra de potent hormone action, is short lived (t1/2 ≤ 12 hours)10. and synthesizes thyroid hormone in the pharyngeal
Genética, Buenos Aires,
Through the activation of thyroid hormone recep- endostyle 18; this organ has functional equivalence to
Argentina.
tors, T3 modulates gene expression to regulate devel- the vertebrate thyroid gland23, including the expres-
2
CONICET-Universidad de
Buenos Aires, Instituto de
opment and many of the phenotypes (growth, heart sion of transcription termination factor 1 (TTF1),
Inmunología, Genética y rate and thermogenesis) described above 11,12. In a homeobox protein NKX2.1 and paired box protein
Metabolismo (INIGEM), negative-feedback loop, thyroid hormone levels PAX8, which are required for thyroid specification24.
Buenos Aires, Argentina. undergo continuous surveillance by central hypotha- Biosynthesis of a thyroid-hormone-containing pro-
3
Division of Metabolism, lamic control over pituitary secretion of TSH, which tein with a sedimentation coefficient of 17S–19S has
Endocrinology & Diabetes, activates thyroidal TSH receptors (TSHRs) to stimulate been reported in the amphioxus endostyle25; however,
University of Michigan
Medical School, Ann Arbor,
thyroid hormone synthesis13,14. genome database searches reveal no clear TG homo-
MI, USA. In this Review, we highlight the role of thyroglob- logue. Nevertheless, amphioxus genes encoding pro-
*e-mail: parvan@umich.edu ulin in hormonogenesis within the thyroid gland and teins with thyroglobulin type 1 modules and other
https://doi.org/10.1038/ consider the topic from evolutionary, biochemical, proteins bearing a putative thyroglobulin-cholinesteras
s41574-019-0184-8 ­molecular, cellular and physiological aspects. e-like (ChEL) domain (thyroglobulin domains are

Nature Reviews | Endocrinology


Reviews

Key points The availability of H2O2 at the apical membrane of


thyrocytes enables thyroid peroxidase (TPO) to catalyse
• The first definitive evidence of a complete TG gene appears with the development the oxidation of iodide39, which is necessary for protein
of the vertebrates, and once appearing in evolution, the entire structure of iodination in the follicular lumen. Most of the pro-
thyroglobulin, as well as its ability to be secreted, has been retained thereafter. tein substrate for iodination is provided by secreted
• The synthesis of T3 and T4 within thyroglobulin involves oxidative coupling between thyroglobulin, the concentration of which is 100–400 mg
iodinated tyrosine residues on thyroglobulin. per mL in the follicular lumen40. The H2O2-generating
• The main T3-forming site within thyroglobulin couples a mono-iodotyrosine donor at system at the apical surface involves the activity of
the antepenultimate residue of one monomer with a di-iodotyrosine acceptor in the dual-function oxidase 2 (DUOX2) and DUOX1, which
same residue of the apposed monomer within a dimer.
both belong to the NADPH oxidase family. DUOX2 is
• Post-translational modifications of thyroglobulin include phosphorylation for which more efficient in the production of H2O2 and more highly
the secretory pathway kinase FAM20C has been implicated.
expressed than DUOX1 (ref.41). The DUOX maturation
• TSH stimulation of thyrocytes promotes post-translational modifications that can factors, DUOXA2 and DUOXA1, facilitate the intra-
alter thyroglobulin structure in a way that favours T3 formation upon iodination,
cellular delivery of DUOX2 and DUOX1 to the plasma
whereas defects in TSH-mediated stimulation result in thyroglobulin with diminished
capacity to form T3.
membrane and regulate the activity of these enzymes42.
Reactive iodide in the follicular lumen preferen-
• 167 TG mutations exist that can cause congenital hypothyroidism; although the
disease is usually inherited as an autosomal recessive trait, patients with congenital
tially reacts with tyrosine residues that are closest to
hypothyroidism bearing monoallelic mutations of TG have recently been reported. the apical plasma membrane; this is an area enriched in
newly secreted thyroglobulin protein that is also highly
accessible for endocytic internalization. The process of
described in detail below) have been reported 19. A synthesis and release of thyroid hormone from newly
best guess of the ancestral amphioxus TG-related synthesized thyroglobulin has been termed ‘first come,
gene (Bf_123169) predicts an encoded protein of first served’43,44. Both singly iodinated tyrosine residues
~2,400 amino acids; it lacks the ChEL domain (needed (mono-iodotyrosine (MIT)) and doubly iodinated tyro­
for thyroglobulin function), but an independently sine residues (di-iodotyrosine (DIT)) are formed during
expressed ChEL domain could potentially function in a iodination of thyroglobulin. In addition, some thyroglob-
thyroglobulin-like heteromeric complex. ulin dimers (17S; with each monomer ~330 kDa) can
The complete TG gene probably first appeared undergo a crosslinking side reaction to generate cova-
through intragenic duplication and gene fusion lent dimers (19S; ~660 kDa) as well as tetramers (27S)
events26. The first definitive evidence of a complete and even higher-order covalent complexes that together
TG gene appears with the development of the verte- can constitute up to 30% of the total thyroglobulin in the
brates27 (Fig. 1a,b), and once appearing, the entire struc- follicular lumen45,46. These intermolecular covalent link-
ture encoded by TG seems to have been conserved ages include disulfide bonds, 3-3ʹ-dityrosine bridges and
throughout all vertebrates thereafter27,28. The earliest γ-glutamyl-lysine bridges40,45,47,48; however, non-iodinated
vertebrates bearing TG that have been studied to date thyroglobulin dimers have no i­ ntermolecular crosslinks
are lamprey larvae, which show an exocrine secretion between the monomer partners47.
of iodoprotein-containing thyroid hormones29 from Thyroglobulin iodination is favoured at specific
the pharyngeal endostyle before metamorphic transi- sites on the protein, which are thought to be based
tion into adult lamprey, which have a true endocrine largely on tyrosine residue exposure at the surface of
thyroid gland29,30. When comparing the thyroglobulin the thyroglobulin tertiary and/or quaternary structure
protein from lamprey to zebrafish to Xenopus to human, and might additionally be affected by the sequence of
although there are variations, the overall modular struc- immediately flanking amino acids. Although the ability
tures including the most critical disulfide-bond-forming of thyroglobulin to be iodinated is not unique, the effi-
cysteine residues, as well as the regional structure of ciency of thyroid hormone synthesis in thyroglobulin,
­thyroglobulin, are all retained27,31 (Fig. 1b). even under low-iodide conditions, is remarkable. At the
time of iodination, selective DIT and MIT residues of
Thyroglobulin in hormonogenesis thyroglobulin undergo a coupling reaction for de novo
The role of iodide. Iodide is initially absorbed in the formation of thyroid hormones within the thyroglob-
gastrointestinal tract and enters the systemic circulation; ulin polypeptide backbone. Classic studies report that
upon reaching the thyroid gland, iodide traverses the TPO shows no marked specificity over lactoperoxidase
basolateral plasma membrane and enters the cytoplasm or myeloperoxidase (all three enzymes are able to cat-
of thyroid follicular epithelial cells (hereafter referred to alyse iodination) in driving hormonogenic coupling
as thyrocytes) via the activity of the sodium–iodide sym- within thyroglobulin49. Rather, T3 and T4 formation is
porter (NIS; encoded by SLC5A5)32. Thyroidal uptake based upon the native conformation of the thyroglobu-
of iodide is increased by iodide efflux across the apical lin substrate, which has been evolutionarily selected for
Endostyle membrane into the follicular lumen, which is mediated efficient thyroid hormone synthesis50 as well as iodide
A longitudinal, ciliated, by the activities of pendrin33, anoctamin 1 (ref.34) and storage. Under conditions of a normal iodide supply,
grooved organ located on the ClC5 (refs35,36) (Fig. 2). The effect of NIS enables thyro- thyroidal thyroglobulin contains, on average, 2.5 res-
ventral wall of the pharynx of cytes to concentrate iodide roughly 30–60-fold within idues of T4 and 0.7 residues of T3 per thyroglobulin
chordates that has functional
equivalence to the vertebrate
the cytosol of thyrocytes37,38, and the activity of apical dimer51, but thyroid hormone-rich and hormone-poor
thyroid gland, among other transporters and iodination machinery further increases thyroglobulin molecules coexist, in variable proportions,
functions. overall iodide sequestration in the thyroid gland. within thyroid follicles.

www.nature.com/nrendo
Reviews

a Region I Region II Region III

NH2 Linker Hinge ChEL domain COOH


T4
T3
b
Human

Y5 Y130 Y1291 Y2554 Y2747

Mouse

Y5 Y130 Y1290 Y2552 Y2744

Domestic cows

Y5 Y130 Y1291 Y2555 Y2748

Frog

Y5 Y130 Y1287 Y2559 Y2750

Zebrafish

Y5 Y125 Y2505 Y2707

Lamprey

Y5 Y131 Y1339 Y2604

Fig. 1 | regional structure and primary hormonogenic sites found within thyroglobulin. a | In vertebrates, the regions of
monomeric thyroglobulin are composed of cysteine-rich repeats within the regions I, II, III and the carboxy-terminal
cholinesterase-like (ChEL) domain. Thyroglobulin contains ~70 tyrosine residues; however, only a few tyrosine residues are
capable of synthesizing thyroid hormones. T4 is primarily formed at a conserved site near the amino terminus (red), and T3 is
mainly synthesized at conserved sites in the carboxy terminus (blue). A typical thyroid hormone distribution for a thyroglobulin
dimer is 2.5 residues of T4 and 0.7 residues of T3. b | The localizations of hormonogenic tyrosine residues are shown in human
(Homo sapiens), mouse (Mus musculus), domestic cows (Bos taurus), frog (Xenopus tropicalis), zebrafish (Danio rerio) and
lamprey (Petromyzon marinus) thyroglobulins. Acceptor T4 and acceptor T3 hormonogenic tyrosine residues are indicated
with light red squares and light blue squares, respectively. Dashed lines indicate missing thyroglobulin sequence in lamprey.

Thyroglobulin trafficking. Follicular thyroglobulin is A large cohort of both cathepsin proteases and plasma
re-internalized into thyrocytes via endocytosis44, which glutamate carboxypeptidase57–59,67,68 is thought to contrib-
promotes thyroglobulin proteolysis that liberates thy- ute to the ultimate lysosomal degradation of thyroglobulin,
roid hormones from the polypeptide backbone, leading which leads to the liberation of thyroid hormones26,31,69,70.
to thyroid hormone secretion via the basolateral plasma The monocarboxylate transporter 8 (MCT8), which is
membrane into the bloodstream (Fig. 2). TSH stimulates expressed in multiple cell types, can facilitate the efflux
endocytic uptake and lysosomal degradation of thy- of thyroid hormones from the thyroid gland as well as
roglobulin52,53, and the rate of thyroid hormone produc- their influx into various cells and tissues. In mice, MCT10
tion correlates with endocytic transfer to lysosomes54,55. shows an overlapping expression with MCT8 in organs
During TSH-stimulated endocytosis, some digestive such as the liver, kidney and thyroid and is particularly
proteolytic enzymes are apically regurgitated from thy- important in facilitating thyroid hormone transport when
rocytes, which enables the predigestion of thyroglobu- MCT8 is missing71. In addition, l-type amino acid trans-
lin before completion of endocytic internalization and porters LAT1 and/or LAT2 can function as secondary thy-
lysosomal delivery40,52,56–59. In addition, some intact thy- roid hormone transporters72–74. Upregulation of lysosomal
roglobulin might directly enter the bloodstream by misdi- thyroglobulin processing and MCT8-mediated thyroid
rected secretion, by leakage from disrupted follicles or by hormone transport are physiologically coordinated for the
transcytosis from the follicular lumen to the basolateral release and delivery of thyroid hormones to the body71,75.
surface of thyrocytes43,44,60,61. Furthermore, some endo- The lysosomal proteolysis of thyroglobulin also yields a
cytically internalized thyroglobulin can potentially be large supply of uncoupled iodotyrosines. MIT and DIT
apically recycled back to the thyroid follicular lumen62–66. can be released into the bloodstream but are substantially

Nature Reviews | Endocrinology


Reviews

Follicular lumen Oxidation of I–


I+ Iodination
I

of tyrosines Coupling
DUOX
Anoctamin 1 system
T4
PDS ClC5 H2O2 Thyroglobulin T3
dimer Apical membrane
TPO

Golgi Iodide recycled


MIT
apparatus
Thyroglobulin
post-translational
modifications

IYD1
ER
DIT
Lysosome

Nucleus

TG gene

NIS Na+/K+-ATPase
Gαq Gαs
PLC AC
Basolateral membrane TSH TSHR
MCT8
Blood vessel

Fig. 2 | Overview of de novo thyroid hormone biosynthesis. Thyroid follicles, which are the functional unit for thyroid
hormone biosynthesis, compose a monolayer of polarized thyrocytes (otherwise known as thyroid follicular epithelial
cells) with the apical surface contacting the follicular lumen and the basolateral membrane facing the bloodstream.
Thyroglobulin is the predominant protein expressed by the thyrocytes and is the primary original source of thyroid
hormones. Hormonogenesis is stimulated by interaction of TSH with its receptor (TSHR), and the central steps involve
the coordination of iodide (I−), H2O2, thyroid peroxidase (TPO) and thyroglobulin. I− traverses the basolateral plasma
membrane of thyrocytes via the activity of the sodium–iodide symporter (NIS) and effluxes across the apical membrane
into the follicular lumen as mediated by the activities of pendrin (PDS), anoctamin 1 and ClC5. A source of H2O2 is
provided by the dual-function oxidase (DUOX) system. In the presence of I− and H2O2, TPO catalyses the iodination of
thyroglobulin tyrosine residues, beginning with the molecules that are closest to the apical plasma membrane. At the
time of iodination, selective di-iodotyrosine (DIT) and mono-iodotyrosine (MIT) residues of thyroglobulin can undergo
the coupling reaction to initiate formation of T4 and T3. Follicular thyroglobulin is re-internalized into the thyrocytes via
endocytosis, which promotes the thyroglobulin proteolysis that liberates thyroid hormones from the polypeptide
backbone, leading to thyroid hormone secretion into the bloodstream via transporters such as MCT8 at the basolateral
plasma membrane. The I− contained within uncoupled MIT and DIT is recycled by iodotyrosine dehalogenase 1 (IYD1).
AC, adenylyl cyclase; ER , endoplasmic reticulum; PLC, phospholipase C.

deiodinated by iodotyrosine dehalogenase 1 (also known the thyroid hormonogenic coupling reaction, is one of the
as IYD1) before they escape from the thyroid gland76–78; least-understood aspects of the biochemistry of thyroid
this process allows for intrathyroidal iodide recycling, hormone synthesis. Evidence suggests that coupling
which is essential to the maintenance of thyroid hormone might proceed either through a free radical interme-
synthesis under conditions of low dietary iodide77. The net diate or through ionic oxidation79. Regardless of the
result of thyroglobulin iodination, coupling, endocytosis, mechanism, hormonogenic coupling always involves
proteolysis and thyroid hormone transport is that 100% two iodotyrosine residues, one of which (referred to as
of the body’s supply of T4 derives from de novo thyroid the donor residue) loses its iodophenolic ring to become
Thyroid hormonogenic
coupling reaction hormone biosynthesis (Fig. 2). dehydroalanine. Either DIT or MIT could be a donor
A post-translational residue, but to generate bioactive thyroid hormone, the
modification resulting in Biochemistry of T4 and T3 synthesis acceptor residue must be DIT (Fig. 3b). After coupling,
the physical transfer of a As mentioned above, H2O2 enables TPO to catalyse the acceptor residue thereby ultimately bears a two-
mono-iodotyrosine
or di-iodotyrosine donor to a
the oxidation of iodide, and reactive iodide can react ring amino acid side chain with the outer ring derived
di-iodotyrosine acceptor within with tyrosine residues on thyroglobulin to generate either from a DIT donor (destined to become T4) or a
a protein. DIT and MIT (Fig. 3a,b). The next step in the process, MIT donor (­ destined to become T3), and this two-ring

www.nature.com/nrendo
Reviews

a b Thyroglobulin-T3 Thyroglobulin-T4
H2N H2N
NH NH
COOH COOH
NH
O NH O NH
Thyroglobulin-Tyr DIT MIT DIT DIT
I O I O
O
TPO– Fe O I OH I OH I
I–
I OH I OH
H2O OH
I–
H COOH H COOH
TPO– Fe O H2N N H2N N
O NH O NH
H2O2
I I
O O
OH–
O O
TPO– Fe I I I

I OH I OH
H2N H2N
NH NH
COOH COOH
O O

I I

I O I O

I I I
OH OH

c T3 NH2
DIO1, DIO2 ORD DIO1, DIO3 IRD
OH
I I O

HO O I
NH2
T4 NH2 T2 OH
OH I I O
I I O
HO O
HO O I
rT3 NH2
I
OH
DIO1, DIO3 IRD I I O DIO1, DIO2 ORD

HO O

Fig. 3 | Physiological enzymatic reactions involved in thyroid hormone synthesis. a | Thyroid peroxidase (TPO)
catalyses the iodination that leads to the coupling of mono-iodotyrosine (MIT) and di-iodotyrosine (DIT) within
thyroglobulin. b | Coupling during de novo thyroid hormone synthesis entails the transfer of an iodophenoxyl ring from a
donor MIT or DIT residue to an acceptor DIT residue within thyroglobulin, yielding T3 or T4, respectively. c | Additionally ,
type 1 and type 2 iodothyronine deiodinases (DIO1 and DIO2) provide outer ring deiodinase (ORD) activity on the T4
molecule to produce T3. Thyroid hormone inactivation occurs by inner ring deiodination (IRD) catalysed primarily by DIO3
(and secondarily by DIO1). rT3, 3, 3′, 5′-tri-iodothyronine; T2, 3, 3′-tri-iodothyronine.

product is still embedded within the thyroglobulin also might contribute importantly to local T3 generation
­polypeptide chain (Fig. 3b). in tissues including the brain, pituitary gland, thyroid
In healthy rats, 55% of circulating T3 appears to gland and brown adipose tissue86. Both DIO1 and DIO2
be derived directly from the thyroid gland80, whereas catalyse deiodination of the outer phenolic ring of the
in healthy humans, only ~21% of daily T3 produc- T4 molecule, which results in an outer ring that emu-
tion is ordinarily derived from thyroidal secretion81. lates the one donated by MIT in the coupling reaction
Importantly, T3 can be generated from T4 in the cytosol described above, to produce T3. By contrast, thyroid hor-
of cells in selected peripheral tissues (such as kidney and mone inactivation — which is also of great physiological
liver), as well as in thyrocytes, by the actions of type 1 significance — occurs by deiodination of the iodothyro-
iodothyronine deiodinase (DIO1) and type 2 iodothyro- nine inner ring, which is catalysed primarily by DIO3,
nine deiodinase (DIO2)82–85 (Fig. 3c). Importantly, DIO1 the third member of the deiodinase group85.
contributes primarily to circulating plasma levels of T3 Thyroidal T3 formed de novo within the thyroglobu-
(ref.84), whereas DIO2 contributes to plasma T3 levels and lin protein (Fig. 3b) is released from the thyroid gland in a

Nature Reviews | Endocrinology


Reviews

manner similar to the release of T4 described above. This Thyroglobulin protein structure
process does not involve the deiodination of T4 within The newly synthesized thyroglobulin precursor protein
thyroglobulin84,87. Furthermore, hormone-containing contains an ~19-residue signal peptide (Supplementary
thyroglobulin is localized within the thyroid follicular Fig. 1), which is immediately removed upon successful
lumen as well as in membrane-bound endocytic and delivery of thyroglobulin into the endoplasmic reticu-
lyso­somal vesicles88, which are sites inaccessible to DIO1 lum (ER)98. Thyroglobulin is synthesized as a mono­
and DIO2 — the catalytic sites of these enzymes topo­ meric protein with a 12S sedimentation coefficient
logically face the cytosol85. Therefore, thyroglobulin (~330 kDa). The multidomain monomeric protein has
itself is not a substrate for deiodination89, and the extent distinct modules: four thyroglobulin type 1 repeats, a
of thyroidal release of T3 derived from thyroglobulin is linker segment, six additional type 1 repeats, a hinge
largely dependent upon the pre-existing T3 content of segment, three thyroglobulin type 2 repeats, a final
thyroglobulin, which is formed during its iodination. type 1 repeat, five thyroglobulin type 3 repeats and a
Notably, animals with whole-body double knockout of carboxy-terminal ChEL domain of ~570 residues, fol-
DIO1 and DIO2 deiodinases exhibit completely normal lowed by a thyroglobulin short unique tail sequence of ~32
circulating T3 levels, which derive entirely from thyroidal residues27,31,70 (Supplementary Fig. 1). Thyroglobulin
secretion84. Moreover, such animals possess as much or modules are cysteine-rich repeat domains bearing
more T3-containing thyroglobulin — the synthesis of intradomain disulfide bonds99. Type 1 modules contain
which is regulated exclusively by the hypothalamus– the central CWCV sequence and are subdivided into
pituitary–thyroid axis — as that found in the thyroid type 1A (containing six cysteine residues) and type 1B
glands of control animals87. (containing four cysteine residues, in the 9th and 11th
repeats)100. Type 2 modules have two cysteine residues
TG gene expression within a short span of 14–17 residues99. Type 3 mod-
Specific transcription factors such as the paired-domain ules are subdivided into type 3A (with eight cysteine
protein PAX8, the homeodomain protein NKX2.1, the ­residues) and type 3B (with six cysteine residues)99.
forkhead-domain protein E1 (FOXE1), homeobox- Overall, the monomeric structure of thyroglobulin
containing transcription factor NKX2.5, the haemato- is divided into four regions (Supplementary Fig. 1):
poietically expressed homeobox HEX (also known as region I contains the first 10 of the 11 thyroglobulin
PRH) and transcriptional co-regulators are all involved type 1 modules, including both the linker and hinge seg-
in the differentiation programme for human thyroid ments (Fig. 1a); region II contains the type 2 repeats and
follicular cells90,91, including transcription of the TG the embedded 11th type 1 repeat; region III contains the
gene26,91. Among these transcription factors, PAX8 type 3 repeats; and region IV contains the ChEL domain
and NKX2.1 are stimulated by the transcriptional plus the short unique tail sequence26 (Supplementary
co-activator with PDZ-binding motif (TAZ; also known Fig. 1). The carboxy-terminal ChEL portion of thy-
as WWTR1), resulting in their biochemical association roglobulin is a member of the classic α-hydrolase and
and synergistic mode of action, which has a prominent β-hydrolase fold family, which is best represented by
role in thyroid cell differentiation and thyroid gene tran- acetylcholines­terase (with a central β-sheet flanked on
scription91. By contrast, HEX acts as a repressor of the both sides by α-helices)101, to which the thyroglobulin
TG promoter92,93. TSH action on its receptor enables pos- ChEL domain exhibits 47% residue similarity102,103.
itive induction of thyroid-specific genes including TG91, Although the 3D structure of thyroglobulin has not
largely through the modulation of intracellular cAMP been elucidated, strong circumstantial evidence exists
levels13,94, which can stimulate TG enhancer function95. for interactions between different regions. The ChEL
The human TG gene (a ~270 kb single-copy gene domain is implicated in the conformational matura-
localized on chromosome 8q24.22) is expressed at very tion and export of newly synthesized thyroglobulin
high levels in thyrocytes26, which is a distinguishing fea- through the thyroid secretory pathway to the follicular
ture of the thyroid gland. The TG gene is organized into lumen98,104,105. Interestingly, when engineered to bear a
48 exons with sizes in the range of 63–1,101 nucleotides signal peptide for delivery into the ER, regions II and III
and containing introns that range in size, including one (expressed together) and ChEL both behave as fully
of 64 kb (refs31,96) that encodes the human SRC-like folded proteins, which pass the conformational require-
adaptor protein (SLAP)97. TG (human thyroglobulin, ments for ER quality control to function as efficient and
NCBI Reference Sequence: NG_015832.1) encodes independently successful secretory proteins106. However,
an mRNA containing a 41-nucleotide 5ʹ-untranslated contiguous thyroglobulin regions I, II and III are not
region followed by a single open reading frame of competent for intracellular transport and require a sep-
8,307 bases and 105 bp of a 3ʹ-untranslated sequence. arately expressed secretory ChEL to enable complete
In human thyroid tissues, thyroglobulin mRNA is folding and export through the secretory pathway106.
very heterogeneous with 12 predicted transcript vari- After a period of monomer folding, thyroglobulin
Thyroglobulin short unique
tail sequence ants (NCBI Reference Sequences: XM_017013797.1, self-associates noncovalently into a homodimer in the
Refers to the final ~32 XM_005251042.4, XM_017013796.1, XM_017013800.1, ER (~660 kDa)107. Moreover, thyroglobulin can fur-
residues of the thyroglobulin XM_017013799.1, XM_005251040.4, XM_017013798.1, ther multimerize in the follicular lumen, as described
sequence containing the XM_017013795.1, XM_005251038.4, XM_017013793.1, above45,46,108. Although the intermonomer contact sur-
antepenultimate tyrosine
residue that can form T3 and is
XM_017013794.1 and XM_006716622.3) in addition to face might not be limited to ChEL, this domain has been
conserved throughout all the full-length thyroglobulin mRNA (NCBI Reference found to be both necessary and sufficient for dimeriza-
vertebrates. Sequence: NM_003235.4). tion. Each ChEL monomer contributes two α-helices to

www.nature.com/nrendo
Reviews

a four-helix bundle that is used for tail-to-tail homo­ owing to misfolding. The asparagine (N)-linked oli-
dimerization as occurs in the related protein acetylcho- gosaccharides on thyroglobulin include both simple
linesterase and many of its evolutionary homologues109. N-acetylglucosamine plus multiple mannose residues,
In thyroglobulin, homodimerization thereby brings which are characteristic of ER processing128, as well as
the contiguous short unique tail sequences from two complex biantennary (Fig. 4b) or triantennary oligo-
­adjoining monomers into close proximity109,110. saccharides with trimmed mannose residues but con-
taining Golgi-added galactose, fucose and sialic acid129.
Post-translational processing Thyroglobulin contains multiple N-linked glycosylation
During its trafficking through the intracellular secre- acceptor sites; of the 20 potential sites in human thy-
tory pathway, thyroglobulin undergoes substantial roglobulin (Supplementary Fig. 1), 16 are actually used26,
post-translational processing as a noncovalent dimer whereas other species might have as few as ten N-linked
before its delivery to the follicular lumen for iodination oligosaccharides per monomer118. TSH is known to reg-
and hormonogenesis. Several of these post-translational ulate thyroidal glycosylation, including the stimulation
modifications might have structural consequences of oligosaccharyl transferase activity112 and the upreg-
that could impact thyroglobulin hormonogenic ulation of N-acetylglucosaminyltransferase 1 (ref.59), as
potential47,59,111–117. well as increased maturation of thyroglobulin N-linked
oligosaccharides from high-mannose to the complex
Catalysed disulfide bond formation. Each thyroglob- type111,126 including increased galactose113 and decreased
ulin monomer has the capacity to form up to 60 2,6-bound sialic acid114.
intramolecular disulfide bonds118. Cysteine residues Human thyroglobulin is also known to contain sul-
present in the repeating thyroglobulin modules are fated O-linked glycosaminoglycans130. The O-linked
covalently bound by intradomain disulfide bonds99. oligosaccharides contain d-galactosamine attached
A conserved pattern of Cys1–Cys2, Cys3–Cys4 and to serine and threonine via an O-glycosidic linkage or
Cys5–Cys6 disulfides are thought to form in type 1A contain d-glucuronic acid N-acetyl-d-galactosamine
repeats100, and type 1B, type 2 and type 3 repeats also sulfate disaccharide plus chondroitin 6-sulfate, which
include intradomain disulfide bonding. Last, the six are attached to the polypeptide chain through a
cysteine residues of the thyroglobulin ChEL domain are d-galactosyl-d-xylosyl-serine linkage124. Interestingly, it
thought to form three intrachain disulfide bonds, which has been reported that the presence of a single chondroi-
occur at the same positions within the related protein tin 6-sulfate on Ser2730 of native human thyroglobulin
acetylcholinesterase119. enhances its hormone-forming ability127. The mod-
The formation of disulfide pairs in thyroglobu- ifications on each glycosylation site show variability
lin is hypothesized to be catalysed by endogenous among the population of thyroglobulin molecules, as
ER oxido­reductases120,121, and folding intermediates identified in porcine thyroglobulin by liquid chroma-
of thyroglobulin bearing mixed disulfide bonds with tography and tandem mass spectrometry131. Although
ER oxidoreductases can occasionally be detected120,121. of potentially great physiological importance, the role of
The ER oxidoreductases implicated in thyroglobulin TSH-regulated changes in the patterns and efficiency
folding include ERp57, protein disulfide-isomerase of Golgi-based O-glycosylation of thyroglobulin, and
(PDI), ERp72 and P5 (Fig. 4a). Thyroglobulin is such a large their effects on thyroid hormone synthesis, remains
protein that the same monomer can be simultaneously poorly understood.
bound by more than one ER oxidoreductase, each pre-
sumably located at its own preferred binding site to facil- Phosphorylation of thyroglobulin. Thyroglobulin
itate thyroglobulin conformational maturation121,122. In undergoes phosphorylation, some of which is included
addition, the isomerase function of ER oxidoreductases within carbohydrate (50%), serine (30%) and tyrosine
such as ERp57, in conjunction with glycan-dependent (20%)117,132. Approximately 75–85% of the thyroglobu-
calnexin (CNX) and/or calreticulin (CRT) chaperone lin phosphotyrosine and phosphoserine residues have
binding, can help to reshuffle non-native disulfides and been recovered in tryptic or cyanogen-bromide-derived
to form new disulfide bonds121,123 (Fig. 4a). peptides comprising <10% of the thyroglobulin protein
and nearly devoid of carbohydrate, which suggests
Glycosylation of thyroglobulin. Approximately 10% that these residues are not randomly located along the
of the molecular mass of thyroglobulin is composed of thyroglobulin molecule117. Phosphorylation is a Golgi
carbohydrate124. Thyroglobulin glycosylation is thought modification that occurs during thyroglobulin trans-
to influence multiple important functions such as pro- port through the secretory (and possibly endocytic)
tein folding and trafficking125, thyroglobulin immuno- pathway133 and is thought to improve the efficiency of T3
reactivity125, iodination and hormone synthesis126,127. formation87. This fact is of potential physiological impor-
Only a few individual thyroglobulin glycosylation sites tance because increased de novo T3 formation within
are conserved between human, Xenopus, zebrafish and thyroglobulin is known to occur when TSHR activity
lamprey27; however, the presence of multiple N-linked is stimulated87. FAM20C is a novel secretory pathway
De novo T3 formation and/or O-linked glycans added during thyroglobulin (casein) serine kinase that catalyses the phosphoryla-
Refers to the synthesis of T3 synthesis and intracellular transport is considered to tion of secreted proteins bearing S-X-E motifs (where
within the thyroglobulin
polypeptide backbone that
be an essential structural feature. In vivo and in vitro X is any amino acid134; Fig. 4c). Interestingly, FAM20C
occurs as a result of the studies show that unglycosylated thyroglobulin loses the shows a TSH dose-dependent increase in mRNA expres-
coupling of iodotyrosines. ability to synthesize thyroid hormones126, presumably sion level in the PCCL3 thyrocyte cell line87. Moreover,

Nature Reviews | Endocrinology


Reviews

a
HS ERp57
CNX/CRT
SH

HS
SH

S-S
PDI ERp57
HS CNX/CRT
S-S
SH

S-
S-S
ERp72

S
HS SH

S-
S-

S
S
S-S
S-S

S-S
SH
P5 SH
SH

HS

b c
ER Polypeptide Cell
membrane
Golgi
Asn-X-Ser/Thr

Polypeptide ----S-X-E--
FAM20C FAM20C
Polypeptide

Asn-X-Ser/Thr
ADP ATP
Polypeptide
Mannose trimming
Golgi
Further mannose
trimming
----S-X-E-- ----S-X-E--
Polypeptide P P

Asn-X-Ser/Thr

Polypeptide

Glucose N-acetylglucosamine Galactose


Mannose Sialic acid Fucose

d Region I Region II Region III


5
NH2 Linker Hinge ChEL domain COOH
Y

PAPS
Sulfotransferase
PAP
5

Y SO3H

www.nature.com/nrendo
Reviews

◀ Fig. 4 | Thyroglobulin post-translational modifications. a | Early thyroglobulin folding for iodination and hormonogenesis of thyroglobulin
is required for thyroid hormonogenesis. Disulfide bond formation in thyroglobulin is (Asp-Tyr or Glu-Tyr) is also favoured for tyrosine sulfa-
catalysed simultaneously by more than one endoplasmic reticulum (ER) oxidoreductase, tion141, which strongly suggests that these are competing
including ER resident protein 57 (ERp57), protein disulfide-isomerase (PDI), ERp72 and reactions. Interestingly, in porcine thyroglobulin, some
P5. Isomerase function of ER oxidoreductase ERp57 in conjunction with glycan-
of the Tyr5 residues (the most prevalent site of T4 forma-
dependent calnexin (CNX) and calreticulin (CRT) chaperone binding can help to form
new disulfide bonds and to reshuffle non-native disulfides. b | In the ER , asparagine-
tion) were found to be sulfated115,142 (Fig. 4d). Conceivably,
linked glycans on thyroglobulin are considered simple N-acetylglucosamine plus the sulfate group could be removed before thyroglobulin
high-mannose oligosaccharides, with terminal glucose residues that are removed, iodination and coupling143, offering a number of spec-
which is characteristic of ER processing. In the Golgi, thyroglobulin contains complex ulative alternatives for possible regulation of de novo
oligosaccharides that have trimmed mannose residues but contain Golgi-added hormonogenesis within thyroglobulin.
galactose, fucose and sialic acid in biantennary structures like that shown here or in
triantennary glycan structures (not shown). TSH receptor stimulation favours the Iodination of tyrosine residues within thyroglobu-
maturation of asparagine-linked oligosaccharides from high-mannose type to the lin. The iodine content of human thyroglobulin varies
complex type. c | FAM20C catalyses the phosphorylation of secreted proteins within largely with iodide intake, which can be in the region
S-X-E motifs (where X is any amino acid). Human thyroglobulin phosphoserine-2721
of 0.05–1.1% (w:w), that is, 2.5–55 atoms of iodine per
(S, in red) is a FAM20C canonical phosphorylation site, confirmed by mass spectrometry ,
and is conserved between human, mouse and rat. A segment of the thyroglobulin mol of thyroglobulin126. Although thyroglobulin has
carboxy-terminal cholinesterase-like (ChEL) domain and short unique tail sequence is ~70 tyrosine residues, classic studies have shown that,
shown within the boxed area. Importantly , phosphorylation is thought to improve the under physiological conditions, only 16 sites are iodin­
efficiency of T3 formation in thyroglobulin. d | Sulfation and hormonogenesis are ated in human thyroid glands, and 13–15 sites are
hypothesized to be competitive reactions, given that Tyr5 of porcine thyroglobulin has iodinated in thyroglobulin derived from rat thyroid
been found to be sulfated. 3'-Phosphoadenosine 5'-phosphosulfate (PAPS) is used by glands144. By contrast, a 2011 analysis of unpurified
sulfotransferases in the Golgi complex as a sulfate source for protein sulfation. thyroglobulin from mouse thyroid by shotgun mass
spectrometry revealed as many as 37 iodinated tyro­
small interfering RNA (siRNA)-mediated knockdown of sine residues145. As only a few positions are involved in
FAM20C significantly decreased the ability of secreted hormonogenesis (maximally approximately five donor–
thyroglobulin to undergo de novo T3 formation87. In acceptor iodotyrosine pairs), these findings emphasize
addition, dephosphorylation of secreted thyroglobulin that thyroglobulin non-hormonogenic tyrosine residues
partially inhibited its de novo T3 formation87. have an underappreciated iodide storage function that
Of the potential phosphorylation sites that are both might have played a critical role in thyroid physiology
predicted casein kinase and canonical FAM20C targets, during the evolution of land-dwelling vertebrates145.
the serine residue at position 2721 of human thyroglob- Nevertheless, studies have shown that the initial iodina-
ulin has been shown by mass spectrometry to actually tion of thyroglobulin tends to occur at preferred tyrosine
be a phosphoserine residue135 (Fig. 4c). This site is specif- residues that are involved in hormonogenesis89, driven
ically conserved between human, rat and mouse and is by the native 3D structure of thyroglobulin, which offers
thereby implicated as a potential regulator of de novo T3 tyrosine residue exposure as well as structural proxim-
formation within thyroglobulin. ity of donor and acceptor iodotyrosines. Residues that
flank the tyrosine residue may also influence iodina-
Sulfation of thyroglobulin. Sulfation is a late post- tion and hormonogenesis. The Asp-Tyr or Glu-Tyr
translational modification step that takes place in the motif has been described as a ‘consensus sequence’ in
trans-Golgi network and contributes to the negative that it appears to be associated with many (but not all)
charge of the thyroglobulin molecule. Sulfation involves a hormonogenic tyrosine residues145,146. In many species,
complex enzymatic mechanism that is dependent on the Ser-Tyr-Ser or Thr-Tyr-Ser occurs at the T3 synthesis
production of 3ʹ-phosphoadenosine 5ʹ-phosphosulfate site within the carboxy-terminal region of thyroglobu-
(PAPS) in the cytosol and its subsequent transport into lin89,145,147, and Glu-X-Tyr is commonly associated with
the trans-Golgi compartment, where it is used as a sub- non-hormonogenic iodotyrosines89,145,147.
strate by protein sulfotransferases136. The sulfate residues
of thyroglobulin are largely present in complex carbo- Thyroglobulin hormonogenic sites
hydrates, such as chondroitin sulfate chains127,137; the In human thyroglobulin, three main hormonogenic
average sulfate content in purified thyroidal thyroglob- acceptor sites have been identified — Tyr5 (site A),
ulin is ~1.5 mol sulfate per mol complex oligosaccharide Tyr2554 (site B) and Tyr2747 (site C) plus the minor
units115,130. Evidence suggests that human thyroglobulin site Tyr1291 (site D) (Fig. 1b) — some studies describe
derived from thyroid nodules has an increased incorpora- Tyr685 as a fifth site31,70,147,148. At human Tyr972, both
tion of [35S]sulfate into its oligosaccharide units compared MIT and DIT were recovered147, although this could vary
with that derived from healthy thyroid tissue138,139. between species145.
Interestingly, it has been reported that TSH generally
downregulates sulfate incorporation into thyroglobulin Site A. In all vertebrate species examined, the most
Thyroglobulin and specifically downregulates thyroglobulin tyrosine important T4-forming site is at Tyr5 (refs144,149) (site A),
non-hormonogenic tyrosine sulfation115. TSH could modulate thyroglobulin sulfa- with Tyr130 (human numbering) serving as an impor-
residues tion by influencing the relative expression levels of sulfo­ tant outer ring donor for thyroxine formation at this
Refers to the tyrosines within
thyroglobulin that are not
transferases and glycosyltransferases111 or by altering site 149 (Fig.  1b) . In most species, Tyr5 accounts for
directly involved in thyroid the rate of passage of thyroglobulin through the Golgi 40–50% of all T4 present in thyroglobulin and 25% of T3.
hormone synthesis. complex140. In addition, the same consensus sequence Furthermore, the tyrosines at positions orthologous to

Nature Reviews | Endocrinology


Reviews

a Region I Region II Region III

Y5 Y130

NH2 Linker Hinge ChEL domain COOH

I I
O

I I
Coupling OH

Y2747
b

I I
O
Dimerization OH
I

Coupling

Fig. 5 | Mechanisms of de novo hormonogenesis at conserved sites on thyroglobulin. a | Formation of T4 at the


evolutionarily conserved Tyr5 site within thyroglobulin involves the di-iodotyrosine (DIT) acceptor undergoing
intramonomeric coupling with a DIT donor residue. b | Formation of T3 at the evolutionarily conserved antepenultimate
tyrosine residue of thyroglobulin involves intermolecular coupling of donor mono-iodotyrosine (MIT) and acceptor DIT
from apposed monomers within the thyroglobulin dimer. Type 1 repeats, light blue boxes; type II repeats, yellow boxes;
type III repeats, darker blue boxes; carboxy-terminal cholinesterase-like (ChEL) domain, pale yellow rectangle; zone of
dimerization, pink double trapezoid.

human thyroglobulin Tyr5 and Tyr130 are conserved within thyroglobulin26. Interestingly, this conserved res-
in mammals and other vertebrates (Fig.  1b), and in idue abundantly forms both MIT (making it a possible
these species, they are also thought to serve as main donor residue in coupling; Fig. 3a) and DIT (as expected
sites of T4 synthesis27,144–146,150–152. A purified human thy­ for an acceptor residue)147,152. Proteolytic cleavage of thy-
roglobulin polypeptide fragment from Asn1 to Met171 roglobulin at Lys2742 excises a carboxy-terminal SYSK
(which maintains most of its 3D conformation as tetrapeptide that is too small to be captured by mass
demonstrated by antibody epitope mapping153) is itself spectrometry, causing it to escape detection145 and thus
capable of forming T4 upon iodination at Tyr5, with interfering with definitive identification of this puta-
the loss of the Tyr130 phenolic side chain150, which tive T3-containing fragment. A study recently reported
demonstrates the potential for intramolecular coup­ that upon iodination, the side chain from the antepe-
ling of these residues at the amino-terminal portion of nultimate residue of mouse thyroglobulin (MIT2744)
thyroglobulin — a conclusion that has been independently closely interacts with and donates to the same residue
supported by several groups and methodologies149,154 (DIT2744) of the apposed monomer within the thy-
(Figs 1b,5). In mouse thyroglobulin, Tyr239 has been roglobulin dimer to form T3 (ref.110) (Figs. 1b,5). This crit-
suggested as an alternative potential iodotyrosyl donor ical site C falls within the short unique tail sequence that
residue to the hormonogenic Tyr5 (ref.145). immediately follows the thyroglobulin ChEL domain.
The development of a simple and specific method to
Site B. Iodination of human thyroglobulin at Tyr2554 detect T3 formation upon in vitro iodination of recom-
(site B) is conserved between humans, mammals and binant thyroglobulin made it possible, for the first time,
other vertebrates 27,145,152. However, different views to detect the mechanism of de novo T3 hormonogenesis
exist about the utilization of site B as a hormonogenic at site C110. For example, studies demonstrated that the
donor152 or acceptor145, which might depend upon the secretory ChEL region (lacking regions I, II and III),
species and conditions (such as iodide concentration) which is sufficient to form a secreted homodimer109, is
or the technology used for analysis. able to form close contact between the antepenultimate
tyrosine side chain and the side chain of the same residue
Site C. The antepenultimate thyroglobulin tyrosine res- in an adjacent secretory ChEL (or that of an adjacent
idue has been conserved throughout evolution for 500 full-length thyroglobulin)110. Furthermore, secretory
million years27,145,152 (Tyr2747 according to human num- ChEL maintains the potential to synthesize T3 at site C110.
bering; also known as site C) and is the major T3-forming Indeed, an artificially engineered mutation that allows
site26,110 (Fig. 1b), accounting for ~50% of T3 production for covalent (disulfide) ‘stapling’ of the apposed unique

www.nature.com/nrendo
Reviews

optimize hormone production in special situations, such


Cytoplasm
↑ Expression of TG as in iodide deficiency. However, a more likely hypo­
Nucleus ↑ Thyroglobulin endocytosis and thesis is that the tyrosine residues that are less well con-
degradation served in their precise location between species might
↑ T3 formed within thyroglobulin function primarily as non-hormonogenic sites needed
Gene expression for iodine storage.

Regulation of thyroid hormonogenesis


CREB ELK1 TSH action on its receptor stimulates thyroglobulin
endocytosis from the apical membrane of thyrocytes
for hormonogenesis (Fig. 2) as well as the expression and
activities of critical gene products involved in thyroid
hormone formation. In addition, TSH can stimulate
ERK1/ERK2 NF-KB thyroidal DIO1 and DIO2 deiodinase activities, which
CREB Ca2+ can further increase T3 production86,156 (Fig. 3b). TSH
MEK1/MEK2 binds to its G protein-coupled receptor TSHR on the
PKA RAP1 RAF PKC basolateral membrane of thyrocytes and mainly activates
the guanine nucleotide-binding protein Gαs to stimulate
cAMP DAG Ins(1,4,5)P3 cAMP synthesis157 (Fig. 6). Activation of the cAMP cas-
cade stimulates the expression of thyroglobulin, TPO,
Gαq NIS and TSHR158,159. By contrast, there is no significant
ATP Gαs PtdIns(4,5)P2
PLC induction of the H2O2-generating DUOX system by TSH
AC or forskolin (a chemical activator of adenylyl cyclase)
TSHR in human thyrocytes in primary culture160. However,
Basolateral human DUOX2 promoter activity can be positively
membrane induced by TSH and forskolin at high doses in PCCL3
TSH (rat) thyrocytes161. Through a protein kinase A (PKA)-
dependent pathway, TSH also regulates the insertion of
pendrin into the plasma membrane of thyrocytes162.
Fig. 6 | TSH regulation of thyroid hormone biosynthesis in thyroglobulin. TSH binds to An additional effect of TSHR activation occurs via
its G protein-coupled receptor (TSHR) on the basolateral membrane of thyrocytes, which the Gαq–Gα11-mediated phospholipase C (PLC)–diacyl­
primarily activates guanine nucleotide-binding protein Gαs to catalyse cAMP synthesis glycerol–calcium pathway, which increases production
(this is the major TSH-activated signalling pathway) and the Gαq–Gα11-mediated signalling of H2O2 by the DUOX–DUOXA system163 to enhance
pathway.  TSHR stimulation induces the expression of critical genes involved in thyroid TPO activity164. Mice deficient in Gαq–Gα11-mediated sig-
hormone formation, including TG, TPO, NIS and TSHR, and promotes thyroglobulin nalling show impairment of thyroid hormone secretion
endocytosis and degradation for hormone release as well as both dual-function oxidase 2
in response to TSH; however, these mice can still carry
(DUOX2) and type 1 and type 2 iodothyronine deiodinase (DIO1 and DIO2) activities to
further increase thyroid hormone production. AC, adenylyl cyclase; CREB, cAMP response out Gαs-mediated thyroid hormone synthesis, which still
element binding; DAG, diacylglycerol; ELK1, ETS transcription factor ; Ins(1,4,5)P3, seems to be the major TSH-activated signalling path-
inositol-1,4,5-triphosphate; NF-κB, nuclear factor-κB; PKA , protein kinase A ; PLC, way165. Thyrocyte-specific deletion of Gαs in adult mice
phospholipase C; PtdIns(4,5)P2, phosphatidylinositol 4,5-bisphosphate. rapidly leads to impaired thyrocyte function and hypo-
thyroidism with a substantial compensatory increase
in serum TSH166. Interestingly, in vitro studies utiliz-
tail sequences ten residues away from the antepenulti- ing a specific TSHR agonist that activates Gαs showed
mate T3 site enables increased contact between these that thyroglobulin secreted from PCCL3 rat thyrocytes
tail sequences and significantly enhances T3 formation was capable of increased T3 synthesis upon iodination
at site C110. Thus, the unique tail sequence of thyroglob- under standard conditions87,167. Moreover, incubation
ulin, which contains site C, has essentially become incor- of PCCL3 cells in the presence of dibutyryl cAMP also
porated into the conserved ChEL domain during TG resulted in the secretion of thyroglobulin with enhanced
evolution27: ChEL provides homodimerization, which capability for de novo T3 formation. These findings
enhances MIT–DIT coupling at the ­antepenultimate are consistent with TSH binding to TSHR and acting
tyrosine residues110 (Figs 1b,5). through stimulatory G proteins and cAMP production
to induce modifications of thyroglobulin that alter its
Minor hormonogenic sites. The ‘minor’ hormonogenic structure such as to enhance its intrinsic ability to form
site D has been reported at position Tyr1291 (human T3 upon iodination87.
numbering), which is conserved in bovine thyroglob- Notably, thyroglobulin is an intrinsic negative-
ulin as well as in other mammals145,147. Studies utilizing feedback regulator that can limit the effects of
a fragment of bovine thyroglobulin encompassing resi- TSH108,168,169, affecting TSHR-mediated cAMP–PKA
dues 1218–1591 suggested position 1375 to be a donor and PLC–PKC signalling pathways and NKX2.1 and
for position 1291 (ref.155). Additionally, more minor PAX8 expression170 and also potentially exerting effects
hormonogenic sites have been found that are poorly on thyroid cell proliferation171. Various metabolites have
conserved between human, Xenopus, zebrafish and also been reported to inhibit thyrocyte function172,173,
lamprey27. Minor sites such as these might be used to the most important of which appears to be iodide itself.

Nature Reviews | Endocrinology


Reviews

Iodide negatively influences the TSH response, and high mutations, 89 missense mutations, 5 insertions or dupli-
doses of iodide can be used to block TSH-driven thyroid cations, 15 deletions and 1 imperfect DNA inversion89,183
hormone synthesis174. (Supplementary Table 1). Interestingly, ~30% of these
human TG mutations have been described within the
Graves disease and iodide deficiency. As discussed past 2 years. TG mutations have also been described in
above, TSHR stimulation influences several important Afrikaner cattle (p.R697*)184, Dutch goats (p.Y296*)185,
thyroglobulin post-translational modifications that cog/cog mice (p.L2263P)186, rdw/rdw rats (p.G2300R)187
could increase de novo T3 formation87. Two common and Wistar Hannover GALAS rats (c.749-1 G>T)188.
thyroid conditions — Graves disease (with an annual The animal models provide a heterogeneous preclin-
incidence of ~30 cases per 100,000 people) and iodide ical spectrum of congenital hypothyroidism. Because
deficiency (affecting ~2 billion people worldwide) — of the autosomal recessive inheritance, patients who
cause hyperthyroidism and hypothyroidism, respec- present clinically with hypothyroidism are generally
tively175; however, both conditions share the feature of either homozygous or compound heterozygous; how-
hyperstimulation of thyroidal TSHRs. TSHR hyperac- ever, a few monoallelic variants have been described.
tivation can also occur as a consequence of activating Conceivably, the apparent absence of a second muta-
mutations in the TSHR, a rare condition that has been tion in human patients could be explained by technical
described primarily in patients of European ances- limitations of direct TG sequencing (neither micro­
try176–179. A relative increase in T3 synthesis is observed in deletions involving one or several exons nor muta-
each of these disorders148,176–178,180,181. Interestingly, incu- tions in distant regulatory or intronic regions of the
bation of PCCL3 cells with TSHR-stimulating immuno- TG gene can be excluded). Moreover, in the setting of TG
globulin G (IgG), or unpurified sera from some patients haploinsufficiency, patients might develop clinical disease
with Graves disease, induces increased T3 formation by the undetected presence of a mutation in a second
upon in vitro iodination of secreted thyroglobulin87. gene that also disrupts thyroid homeostasis. With this
Thus far, the molecular contributions to the relative in mind, several monoallelic TG mutations have been
increase in T3 synthesis remain incompletely defined, found in individuals with mild congenital hypothyroid-
but the data suggest that de novo T3 formation within ism189. Moreover, a report exists of a patient with severe
thyroglobulin is one of the important contributors87,148. hypothyroidism who harbours pathogenic heterozy-
Thyroglobulin secreted from thyrocytes with genetic gous variants of both TG and TPO genes, suggesting
deletion of TSHRs consistently exhibited decreased that some patients might develop hypothyroidism as a
de novo T3 formation compared with T3 synthesized in consequence of oligogenicity190.
the thyroglobulin secreted from thyrocytes with stim- The p.C1058R and p.C1977S mutations are the most
ulated TSHRs87. By contrast, there is no demonstrable frequently identified TG mutations in the Japanese
evidence that TSHR stimulation enhances the efficiency population, whereas p.R277* is the most frequently
of de novo T4 formation per thyroglobulin molecule87,182, identified TG mutation found in white individuals in
despite the fact that overall T4 production clearly rises in families from Brazil, Argentina, Spain and France26,191
patients with Graves disease. (Supplementary Table 1). The structural consequence
of the p.R277* mutation is the expression of a truncated
Genetic disorders caused by TG mutation protein, which has a complete loss of the central and
Hormonogenesis defects caused by TG mutations are carboxy-terminal hormonogenic domains and, conse-
inherited as an autosomal recessive trait, causing con- quently, a limited ability to generate thyroid hormone.
genital hypothyroidism, which has an estimated global Theoretically, however, the p.R277* thyroglobulin poly-
incidence of approximately 1 in 100,000 newborn peptide, if iodinated, should retain its ability for T4 and
babies183. Some patients present with thyroid enlarge- T3 synthesis because it still harbours both the acceptor
ment shortly after birth, others later in life, and in a few Tyr5 and the donor Tyr130. More important than any
individuals no goitre develops. Primary hypothyroidism limitation in the ability of thyroglobulin to generate
and thyroid gland enlargement depend on the severity of active thyroid hormones, most mutant thyroglobulin
the TG defect, the amount of dietary iodide intake and proteins are misfolded, which causes their retention
patient adherence to thyroid hormone replacement ther- in the ER with premature degradation, as observed in
apy. Untreated severe congenital hypothyroidism can preclinical (animal) models192. Nevertheless, limited
result in irreversible mental delay and short stature175. amounts of mutated thyroglobulin molecules might
The biochemical profile of patients with homozygous reach the follicular lumen, enabling iodination with the
or compound heterozygous TG mutations is charac­ potential synthesis of thyroid hormones, as observed in
terized by low or absent serum levels of thyroglobulin, many patients with congenital hypothyroid goitre and
high serum TSH, normal perchlorate discharge test defective thyroglobulin.
(indicating a normal iodide organification mechanism), Remarkably, patients have been identified with
Haploinsufficiency low serum levels of circulating T4 and variable levels of compound heterozygous TG mutations encoding
A condition that arises when circulating T3 (usually resulting in a relatively increased p.R277*/p.C1981W-p.P2183R or p.C1245R/p.G2356R
there is a complete loss of T3:T4 ratio)183. The first described human mutation (Supplementary Table 1), who exhibited an absolute
function of one copy of a gene, causing a TG defect associated with congenital hypo- increase in plasma levels of T3 (refs193,194). Bioinformatic
and the remaining functional
copy of the gene is not
thyroidism was c.275-3 C>G (Supplementary Table 1). studies of thyroglobulin p.C1981W-p.P2183R predict
adequate to preserve normal In 2019, 167 human TG gene mutations have been that the protein could be efficiently secreted193, and
function in a diploid organism. identified to date: 21 splice site mutations, 36 nonsense functional studies of recombinant thyroglobulin p.C175*

www.nature.com/nrendo
Reviews

Monoallelic mutations
demonstrate its secretion, leaving open the possibility the T3 formation sites within thyroglobulin that are
Changes in the genetic that thyroglobulin p.R277* might also be secreted191. used in Graves disease, as well as to understand how
sequence in one of two Possibly the increase in plasma T3 might be due to TSHR-stimulated post-translational processing impacts
homologous alleles (paternal elevated DIO2 activity194 in the thyroid gland; alterna- thyroglobulin structure to enhance its hormonogenic
or maternal) of a single gene.
tively, certain mutations in TG might provoke structural capability. For this, new technologies including X-ray
changes that trigger the appearance of cryptic sites of crystallography of purified thyroglobulin regions or
T3 formation in thyroglobulin that become favoured by cryo-electron microscopy, combined with other tools
TSH hyperstimulation. that have the capability of single-molecule resolution,
Finally, a few patients with TG mutations do not might advance the field. Using these technologies in
develop goitre. For example, a goitre has not been conjunction with large-scale synthesis of recombinant
reported in patients homozygous for p.G2300D195, which thyroglobulin domains could enable direct visualiza-
occurs in the position equivalent to the p.G2298R muta- tion of the proximity between donor and acceptor hor-
tion responsible for the phenotype in rdw/rdw dwarf rats monogenic iodotyrosine residues. Furthermore, such
that develop a hypoplastic thyroid gland187. Similar to technology might conceivably help in the analysis of the
goitrous congenital hypothyroidism, misfolded thy- rare compound heterozygous genotypes of patients who
roglobulin mutants responsible for nongoitrous hypo- actually exhibit a selective elevation of plasma T3 levels.
thyroidism in rdw/rdw dwarf rats196 are linked to an Finally, unusual diseases have been identified, such
increased ER stress response197, but importantly, the as cystinosis199 and Fabry disease200, in which hypothy-
Tgrdw/rdw rats exhibit proteotoxic thyrocyte cell death198, roidism is a common yet poorly understood feature and
which appears to explain the lack of goitre. in which pathogenic intracellular thyroglobulin traf-
ficking has been implicated. As personalized medicine
Conclusions continues to expand, unexpected new gene products
Recognizing the importance of thyroglobulin in thy- might come to light that, in some capacity, modulate or
roid hormone synthesis could provoke new studies contribute to thyroid hormonogenesis. As such, further
investigating ancestral pre-vertebrates that synthesize study is needed (utilizing next-generation sequencing
thyroid hormones despite the absence of a complete platforms) of the interesting and rare patients with con-
vertebrate-style TG gene. The possibility exists that in genital hypothyroidism who have monoallelic mutations
the endostyle of pre-vertebrates, thyroglobulin regions of TG. Such patients might ultimately highlight some of
expressed as separate gene products might potentially the most physiologically important genetic interactions
account for thyroid hormone synthesis. Alternatively, regulating the role of TG in thyroid hormone synthe-
unrelated gene products, whose identification remains sis. As a purely hypothetical example, genes regulating
to be determined, might serve as substrates for iodina- autophagy might allow thyroglobulin entrapped in the
tion and thyroid hormone synthesis, although their hor- ER to be brought to an autophagosomal compartment
monogenic potential would presumably be less efficient in which it might conceivably encounter molecules of
than that of thyroglobulin, which has been evolutionarily the iodination machinery, bypassing hormonogenesis at
selected for this function. the follicular lumen. Of course, 167 different human TG
Additionally, we now recognize that de novo T3 for- mutations exist that are linked to hypothyroidism, and
mation within thyroglobulin is an important contributor synthetic interactions with other defective genes might
to enhanced T3 production in pathological states during vary for different mutant TG alleles, altering disease
which TSHRs are hyperstimulated87,148. To understand severity. Clearly, there is still much to learn.
the molecular mechanisms underlying the increased T3
production, future studies will need to further clarify Published online xx xx xxxx

1. Tovo-Neto, A., da Silva Rodrigues, M., Habibi, H. R. & metabolism: crosstalk between nuclear receptors. 17. Kinjo, S., Kiyomoto, M., Yamamoto, T., Ikeo, K. &
Nóbrega, R. H. Thyroid hormone actions on male Int. J. Mol. Sci. 19, 1–20 (2018). Yaguchi, S. HpBase: a genome database of a sea
reproductive system of teleost fish. Gen. Comp. 10. Nicoloff, J. T., Low, J. C., Dussault, J. H. & Fisher, D. A. urchin, Hemicentrotus pulcherrimus. Dev. Growth
Endocrinol. 265, 230–236 (2018). Simultaneous measurement of thyroxine and Differ. 60, 174–182 (2018).
2. Saito, M., Yamasu, K. & Suyemitsu, T. Binding triiodothyronine peripheral turnover kinetics in man. 18. Paris, M. & Laudet, V. The history of a developmental
properties of thyroxine to nuclear extract from sea J. Clin. Invest. 51, 473–483 (1972). stage: metamorphosis in chordates. Genesis 46,
urchin larvae. Zoolog. Sci. 29, 79–82 (2012). 11. Kalderon, B., Hertz, R. & Bar-Tana, J. Effect of thyroid 657–672 (2008).
3. Hodin, J. Expanding networks: Signaling components hormone treatment on redox and phosphate 19. Paris, M., Brunet, F., Markov, G., Schubert, M. &
in and a hypothesis for the evolution of metamorphosis. potentials in rat liver. Endocrinology 131, 400–407 Laudet, V. The amphioxus genome enlightens the
Integr. Comp. Biol. 46, 719–742 (2006). (1992). evolution of the thyroid hormone signaling pathway.
4. Heyland, A., Reitzel, A. M. & Hodin, J. Thyroid 12. Venditti, P. & Di Meo, S. Thyroid hormone-induced Dev. Genes Evol. 218, 667–680 (2008).
hormones determine developmental mode in sand oxidative stress. Cell. Mol. Life Sci. 63, 414–434 20. Holland, L. Z. et al. The amphioxus genome
dollars (Echinodermata: Echinoidea). Evol. Dev. 6, (2006). illuminates vertebrate origins and cephalochordate
382–392 (2004). 13. Nagayama, Y. et al. Regulation of thyroid peroxidase biology. Genome Res. 18, 1100–1111 (2008).
5. Saito, M. et al. Induction of metamorphosis in the and thyroglobulin gene expression by thyrotropin in 21. Putnam, N. H. et al. The amphioxus genome and the
sand dollar Peronella japonica by thyroid hormones. cultured human thyroid cells. J. Clin. Endocrinol. evolution of the chordate karyotype. Nature 453, 1–7
Dev. Growth Differ. 40, 307–312 (1998). Metab. 68, 1155–1159 (1989). (2008).
6. Laudet, V. The origins and evolution of vertebrate 14. Kang, H. S. et al. GLIS3 is indispensable for TSH/ 22. Paris, M. et al. Active metabolism of thyroid hormone
metamorphosis. Curr. Biol. 21, R726–R737 (2011). TSHR-dependent thyroid hormone biosynthesis and during metamorphosis of amphioxus. Integr. Comp.
7. Dardente, H., Hazlerigg, D. G. & Ebling, F. J. P. Thyroid follicular cell proliferation. J. Clin. Invest. 127, Biol. 50, 63–74 (2010).
hormone and seasonal rhythmicity. Front. Endocrinol. 4326–4337 (2017). 23. Ogasawara, M. & Satoh, N. Isolation and
5, 1–11 (2014). 15. Köhrle, J. in Thyroid Hormone Nuclear Receptor characterization of endostyle-specific genes in the
8. Mullur, R., Liu, Y.-Y. & Brent, G. A. Thyroid hormone Methods and Protocols (eds Plateroti, M. & Samarut, J.) Ascidian Ciona intestinalis. Biol. Bull. 195, 60–69
regulation of metabolism. Physiol. Rev. 94, 355–382 85–104 (Springer New York, 2018). (1998).
(2014). 16. Cary, G. A., Cameron, A. R. & Hinman, V. F. 24. Kimura, S. Thyroid-specific enhancer-binding protein:
9. Kouidhi, S. & Clerget-Froidevaux, M.-S. Integrating EchinoBase: tools for echinoderm genome analyses. role in thyroid function and organogenesis.
thyroid hormone signaling in hypothalamic control of Methods Mol. Biol. 1757, 349–369 (2018). Trends Endocrinol. Metab. 7, 247–252 (1996).

Nature Reviews | Endocrinology


Reviews

25. Monaco, F., Dominici, R., Andreoli, M., Pirro, R. D. E. 44. Marinò, M. et al. Role of thyroglobulin endocytic Biochem. Biophys. Res. Commun. 268, 42–46
& Roche, J. Thyroid hormone formation in pathways in the control of thyroid hormone release. (2000).
thyroglobulin synthesized in the Amphioxus Am. J. Physiol. Cell Physiol. 279, C1295–C1306 66. Mezghrani, A. et al. Protein-disulfide Isomerase (PDI)
(Branchiostoma lanceolatum pallas). Comp. Biochem. (2000). in FRTL5 Cells. J. Biol. Chem. 275, 1920–1929
Physiol. B 70, 341–343 (1981). 45. Delom, F., Mallet, B., Carayon, P. & Lejeune, P. Role of (2000).
This paper demonstrates, within the amphioxus extracellular molecular chaperones in the folding of 67. Friedrichs, B. et al. Thyroid functions of mouse
endostyle, the synthesis of thyroid hormone in a oxidized proteins. Refolding of colloidal thyroglobulin cathepsins B, K, and L. J. Clin. Invest. 111,
large protein that has properties similar to those by protein disulfide isomerase and immunoglobulin 1733–1745 (2003).
of thyroglobulin, despite complete genome heavy chain-binding protein. J. Biol. Chem. 276, 68. Oda, K. et al. Follicular thyroglobulin induces
sequencing subsequently confirming the absence 21337–21342 (2001). cathepsin H expression and activity in thyrocytes.
of a vertebrate-style TG gene in this organism. 46. Berndorfer, U., Wilms, H. & Herzog, V. Multimerization Biochem. Biophys. Res. Commun. 483, 541–546
26. Targovnik, H. M. in Werner & Ingbar’s The Thyroid: of thyroglobulin (TG) during extracellular storage: (2017).
A Fundamental and Clinical Text (eds Braverman, L. E. isolation of highly cross-linked TG from human thyroids. 69. Kopp, P. in Werner & Ingbar’s The Thyroid:
& Cooper, D.) 74–92 (Lippincott Williams & Wilkins, J. Clin. Endocrinol. Metab. 81, 1918–1926 (1996). A Fundamental and Clinical Text (eds Braverman, L. E.
2012). 47. Leonardi, A. et al. Presence of dityrosine bridges in & Cooper, D.) 48–74 (Lippincott Williams & Wilkins,
This comprehensive review describes the thyroglobulin and their relationship with iodination. 2012).
structure and function of thyroglobulin and TG Biochem. Biophys. Res. Commun. 202, 38–43 70. van de Graaf, S. a et al. Up to date with human
and discusses mutations that cause congenital (1994). thyroglobulin. J. Endocrinol. 170, 307–321 (2001).
hypothyroidism. 48. Baudry, N. et al. Dityrosine bridge formation and 71. Müller, J. et al. Tissue-specific alterations in thyroid
27. Holzer, G. et al. Thyroglobulin represents a novel thyroid hormone synthesis are tightly linked and are hormone homeostasis in combined Mct10 and Mct8
molecular architecture of vertebrates. J. Biol. Chem. both dependent on N-glycans. FEBS Lett. 396, deficiency. Endocrinology 155, 315–325 (2018).
291, 16553–16566 (2016). 223–226 (1996). 72. Krause, G. & Hinz, K. M. Thyroid hormone transport
This paper confirms the presence of TG in lamprey 49. Taurog, A., Dorris, M. L. & Lamas, L. Comparison of across L-type amino acid transporters: What can
and Xenopus and indicates functional conservation lactoperoxidase- and thyroid peroxidase-catalyzed molecular modelling tell us? Mol. Cell. Endocrinol.
in thyroid hormone synthesis throughout all iodination and coupling. Endocrinology 94, 458, 68–75 (2017).
vertebrates. 1286–1294 (1974). 73. Hinz, K. M. et al. Structural insights into thyroid
28. Smith, J. J. et al. Sequencing of the sea lamprey 50. Lamas, L. & Taurog, A. The importance of hormone transport mechanisms of the L-type amino
(Petromyzon marinus) genome provides insights into thyroglobulin structure in thyroid peroxidase- acid transporter 2. Mol. Endocrinol. 29, 933–942
vertebrate evolution. Nat. Genet. 45, 415–421 catalyzed conversion of diiodotyrosine to thyroxine. (2015).
(2013). Endocrinology 100, 1129–1136 (1977). 74. Mayerl, S. et al. Thyroid hormone transporters MCT8
29. Youson, J. H. Is lamprey metamorphosis regulated This paper shows that the native structure of and OATP1C1 control skeletal muscle regeneration.
by thyroid hormones? Am. Zool. 37, 441–460 thyroglobulin is critical to the efficiency of the Stem Cell Rep. 10, 1959–1974 (2018).
(1997). hormonogenic coupling reaction. 75. Weber, J. et al. Interdependence of thyroglobulin
30. Kluge, B., Renault, N. & Rohr, K. B. Anatomical 51. Dunn, J. T. & Dunn, A. D. Update on intrathyroidal processing and thyroid hormone export in the mouse
and molecular reinvestigation of lamprey endostyle iodine metabolism. Thyroid 11, 407–414 (2001). thyroid gland. Eur. J. Cell Biol. 96, 440–456 (2017).
development provides new insight into thyroid 52. Brix, K. & Lemansky, P. H. V. Evidence for This paper suggests that lysosomal degradation of
gland evolution. Dev. Genes Evol. 215, 32–40 extracellularly acting cathepsins mediating thyroid thyroglobulin and thyroid hormone transporter
(2005). hormone liberation in thyroid epithelial cells. proteins in the thyroid gland are physiologically
31. Malthiéry, Y. & Lissitzky, S. Primary structure of Endocrinology 137, 1963–1974 (1996). regulated.
human thyroglobulin deduced from the sequence of 53. Rubio, I. G. S. & Medeiros-Neto, G. Mutations of the 76. Gnidehou, S. et al. Cloning and characterization of a
its 8448-base complementary DNA. Eur. J. Biochem. thyroglobulin gene and its relevance to thyroid novel isoform of iodotyrosine dehalogenase 1
165, 491–498 (1987). disorders. Curr. Opin. Endocrinol. Diabetes. Obes. 16, (DEHAL1) DEHAL1C from human thyroid: comparisons
This paper determines the complete cDNA 373–378 (2009). with DEHAL1 and DEHAL1B. Thyroid 16, 715–724
nucleotide sequence of thyroglobulin, thereby 54. Mascia, A. et al. Rab7 regulates CDH1 endocytosis, (2006).
establishing that the region-specific domain circular dorsal ruffles genesis, and thyroglobulin 77. Gnidehou, S. et al. Iodotyrosine dehalogenase 1
structure of human thyroglobulin is comparable to internalization in a thyroid cell line. J. Cell. Physiol. (DEHAL1) is a transmembrane protein involved in the
that in other vertebrate species. 231, 1695–1708 (2016). recycling of iodide close to the thyroglobulin iodination
32. Matos, L. P. L. et al. Regulation of thyroid 55. Croizet-Berger, K., Daumerie, C., Couvreur, M., site. FASEB J. 18, 1574–1576 (2004).
sodium-iodide symporter in different stages of goiter: Courtoy, P. J. & van den Hove, M.-F. The endocytic 78. Renko, K. et al. A nonradioactive DEHAL assay for
possible involvement of reactive oxygen species. catalysts, Rab5a and Rab7, are tandem regulators of testing substrates, inhibitors, and monitoring
Clin. Exp. Pharmacol. Physiol. 12, 3218–3221 thyroid hormone production. Proc. Natl Acad. Sci. endogenous activity. Endocrinology 157, 4516–4525
(2017). USA 99, 8277–8282 (2002). (2018).
33. Benvenga, S. & Guarneri, F. Homology of pendrin, 56. Brix, K., Linke, M., Tepel, C. & Herzog, V. Cysteine 79. Gavaret, J., Cahnmann, H. J. & Nunez, J. Thyroid
sodium-iodide symporter and apical iodide proteinases mediate extracellular prohormone hormone synthesis in thyroglobulin. J. Biol. Chem.
transporter. Front. Biosci. 23, 1864–1873 (2018). processing in the thyroid. Biol. Chem. 382, 717–725 256, 9167–9173 (1981).
34. Silveira, J. C. & Kopp, P. A. Pendrin and anoctamin as (2001). 80. Chanoine, J. P. et al. The thyroid gland is a major
mediators of apical iodide efflux in thyroid cells. 57. Jordans, S. et al. Monitoring compartment-specific source of circulating T3 in the rat. J. Clin. Invest. 91,
Curr. Opin. Endocrinol. Diabetes Obes. 22, 374–380 substrate cleavage by cathepsins B, K, L, and S at 2709–2713 (1993).
(2015). physiological pH and redox conditions. BMC Biochem. 81. Larsen, P. R. Thyroidal triiodothyronine and thyroxine
35. Fong, P. Thyroid iodide efflux: a team effort? J. Physiol. 10, 23 (2009). in Graves’ disease: correlation with presurgical
589, 5929–5939 (2011). 58. Linke, M., Jordans, S., Mach, L., Herzog, V. & Brix, K. treatment, thyroid status, and iodine content. J. Clin.
36. Van Den Hove, M. F. et al. The loss of the chloride Thyroid stimulating hormone upregulates secretion of Endocrinol. Metab. 41, 1098–1104 (1975).
channel, ClC-5, delays apical iodide efflux and induces cathepsin B from thyroid epithelial cells. Biol. Chem. 82. Schneider, M. J. et al. Targeted disruption of the type 2
a euthyroid goiter in the mouse thyroid gland. 383, 773–784 (2002). selenodeiodinase gene (DIO2) results in a phenotype
Endocrinology 147, 1287–1296 (2006). 59. Suban, D. et al. Cathepsin C and plasma glutamate of pituitary resistance to T4. Mol. Endocrinol. 15,
37. Portulano, C., Paroder-Belenitsky, M. & Carrasco, N. carboxypeptidase secreted from Fischer rat thyroid 2137–2148 (2001).
The Na+/I- symporter (NIS): mechanism and medical cells liberate thyroxin from the N-terminus of 83. Schneider, M. J. et al. Targeted disruption of the type 1
impact. Endocr. Rev. 35, 106–149 (2014). thyroglobulin. Biochimie 94, 719–726 (2012). selenodeiodinase gene (Dio1) results in marked changes
38. Rhoden, K. J., Cianchetta, S., Duchi, S. & Romeo, G. 60. Lisi, S. et al. Preferential megalin-mediated in thyroid hormone economy in mice. Endocrinology
Fluorescence quantitation of thyrocyte iodide transcytosis of low-hormonogenic thyroglobulin: 147, 580–589 (2006).
accumulation with the yellow fluorescent protein a control mechanism for thyroid hormone release. 84. Galton, V. A., Schneider, M. J., Clark, A. S. & St.
variant YFP-H148Q/I152L. Anal. Biochem. 373, Proc. Natl Acad. Sci. USA 100, 14858–14863 Germain, D. L. Life without thyroxine to
239–246 (2008). (2003). 3,5,3′-triiodothyronine conversion: Studies in mice
39. Belforte, F. S. et al. Kinetic characterization of human 61. Marino, M., Zheng, G. & Mccluskey, R. T. Megalin devoid of the 5′-deiodinases. Endocrinology 150,
thyroperoxidase. Normal and pathological enzyme (gp330) is an endocytic receptor for thyroglobulin on 2957–2963 (2009).
expression in Baculovirus System: a molecular model cultured fisher rat thyroid cells. J. Biol. Chem. 274, This paper reveals normal serum T3 levels in mice
of functional expression. Mol. Cell. Endocrinol. 404, 12898–12904 (1999). with whole-body double knockout of DIO1 and
9–15 (2015). 62. Miquelis, R. et al. The N-acetylglucosamine-specific DIO2 deiodinases, indicating robust T3 formation
40. Saber-Lichtenberg, Y. et al. Covalent cross-linking of receptor of the thyroid. Binding characteristics, partial in the absence of deiodination of T4.
secreted bovine thyroglobulin by transglutaminase. characterization, and potential role. J. Biol. Chem. 85. Bianco, A. C. & Larsen, P. R. Cellular and structural
FASEB J. 14, 1005–1014 (2000). 262, 15291–15298 (1987). biology of the deiodinases. Thyroid 15, 777–786
41. Muzza, M. & Fugazzola, L. Disorders of H2O2 63. Miquelis, R. et al. Intracellular routing of (2005).
generation. Best Pract. Res. Clin. Endocrinol. Metab. GLcNAc-bearing molecules in thyrocytes: selective 86. Lavado-Autric, R. et al. Deiodinase activities in
31, 225–240 (2017). recycling through the Golgi Apparatus Raymond. thyroids and tissues of iodine-deficient female rats.
42. De Deken, X., Corvilain, B., Dumont, J. E. & Miot, F. J. Cell Biol. 123, 1695–1706 (1993). Endocrinology 154, 529–536 (2013).
Roles of DUOX-mediated hydrogen peroxide in 64. Pacifico, F., Liguoro, D., Acquaviva, R., Formisano, S. 87. Citterio, C. E. et al. De novo triiodothyronine
metabolism, host defense, and signaling. Antioxid. & Consiglio, E. Thyroglobulin binding and TSH formation from thyrocytes activated by Thyroid
Redox Signal. 20, 2776–2793 (2014). regulation of the RHL-1 subunit of the Stimulating Hormone. J. Biol. Chem. 292,
43. Lisi, S. et al. Defective thyroglobulin storage in asialoglycoprotein receptor in rat thyroid. Biochimie 15434–15444 (2017).
LDL receptor-associated protein-deficient mice. 81, 493–496 (1999). This paper demonstrates increased de novo T3
Am. J. Physiol. Cell Physiol. 290, C1160–C1167 65. Montuori, N. et al. The rat asialoglycoprotein receptor formation in thyroglobulin secreted from
(2006). binds the amino-terminal domain of thyroglobulin. TSH-stimulated thyrocytes and suggests that

www.nature.com/nrendo
Reviews

thyroglobulin phosphorylation may contribute to 110. Citterio, C. E., Morishita, Y., Dakka, N., Veluswamy, B. 130. Spiro, R. G. & Bhoyroo, V. D. Occurrence of sulfate
this effect. & Arvan, P. Relationship between the dimerization of in the asparagine-linked complex carbohydrate units
88. Sellitti, D. F. & Suzuki, K. Intrinsic regulation of thyroid thyroglobulin and its ability to form triiodothyronine. of thyroglobulin. Identification and localization of
function by thyroglobulin. Thyroid 24, 625–638 J. Biol. Chem. 293, 4860–4869 (2018). galactose 3-sulfate and N-acetylglucosamine 6-sulfate
(2014). This paper confirms that the major T3-forming site residues in the human and calf proteins. J. Biol. Chem.
89. Di Jeso, B. & Arvan, P. Thyroglobulin from molecular of thyroglobulin involves MIT–DIT coupling of the 263, 14351–14358 (1988).
and cellular biology to clinical endocrinology. antepenultimate residues of the two thyroglobulin 131. Zhao, J., Song, E., Zhu, R. & Mechref, Y. Parallel data
Endocr. Rev. 37, 2–36 (2016). monomers within the thyroglobulin dimer. acquisition of in-source fragmented glycopeptides to
90. Dentice, M., Cordeddu, V., Rosica, A. & Macchia, P. E. 111. Desruisseau, S., Franc, J. L., Gruffat, D. & Chabaud, O. sequence the glycosylation sites of proteins.
Missense mutation in the transcription factor Glycosylation of thyroglobulin secreted by porcine cells Electrophoresis 37, 1420–1430 (2016).
NKX2–5: a novel molecular event in the pathogenesis cultured in chamber system: thyrotropin controls the 132. Tassi, V., Liguoro, D., Consiglio, E. & Acquaviva, A.
of thyroid dysgenesis. J. Clin. Endocrinol. Metab. 91, number of oligosaccharides and their anionic residues. in Advances in Post-Translational Modifications of
1428–1433 (2005). Endocrinology 134, 1676–1684 (1994). Proteins and Aging (eds Zappia, V., Galletti, P., Porta, R.
91. Ma, R., Morshed, S. A., Latif, R. & Davies, T. F. TAZ 112. Desruisseau, S., Valette, A., Franc, J. L. & Chabaud, O. & Wold, F.) 541–549 (Springer US, 1988).
induction directs differentiation of thyroid follicular Thyrotropin controls dolichol-linked sugar pools and 133. Wen, G., Ringseis, R. & Eder, K. Endoplasmic reticulum
cells from human embryonic stem cells. Thyroid 27, oligosaccharyltransferase activity in thyroid cells. stress inhibits expression of genes involved in thyroid
292–299 (2017). Mol. Cell. Endocrinol. 122, 223–228 (1996). hormone synthesis and their key transcriptional
92. Lacroix, L. et al. HEX, PAX-8 and TTF-1 gene 113. Di Jeso, B. et al. Modulation of the carbohydrate regulators in FRTL-5 thyrocytes. PLOS ONE 12,
expression in human thyroid tissues: a comparative moiety of thyroglobulin by thyrotropin and calcium in e0187561 (2017).
analysis with other genes involved in iodide fisher rat thyroid line-5. J. Biol. Chem. 267, 134. Tagliabracci, V. S. et al. Secreted kinase
metabolism. Clin. Endocrinol. 64, 398–404 (2006). 1938–1944 (1992). phosphorylated extracellular proteins that regulate
93. Pellizzari, L. et al. Expression and function of the 114. Grollman, E. F., Saji, M., Shimura, Y., Lau, J. T. & biomineralization. Science 336, 1150–1153 (2012).
homeodomain-containing protein Hex in thyroid cells. Ashwell, G. Thyrotropin regulation of sialic acid 135. Mertins, P. et al. Proteogenomics connects somatic
Nucleic Acids Res. 28, 2503–2511 (2000). expression in rat thyroid cells. J. Biol. Chem. 268, mutations to signalling in breast cancer. Nature 534,
94. Berg, V., Vassart, G. & Christophe, D. Identification of 3604–3609 (1993). 55–62 (2016).
a thyroid-specific and cAMP-responsive enhancer in 115. Nlend, M. C., Cauvi, D., Venot, N., Desruisseau, S. & 136. Cauvi, D., Venot, N., Nlend, M.-C. & Chabaud, O. M.
the upstream sequences of the human thyroglobulin Chabaud, O. Thyrotropin regulates tyrosine sulfation Thyrotropin and iodide regulate sulfate concentration
promoter. Biochim. Biophys. Acta 1307, 35–38 of thyroglobulin. Eur. J. Endocrinol. 141, 61–69 in thyroid cells. Relationship to thyroglobulin sulfation.
(1996). (1999). Can. J. Physiol. Pharmacol. 81, 1131–1138 (2003).
95. Berg, V., Vassart, G. & Christophe, D. A zinc-dependent 116. Haeberli, A., Kneubuehl, F. & Studer, H. Changes in 137. Schneider, A. B., Mccurdy, A., Chang, T., Dudlak, D.
DNA-binding activity co-operates with cAMP-responsive the polypeptide assembly of guinea pig thyroglobulin & Magner, J. Metabolic labeling of human thyroglobulin
-element-binding protein to activate the human induced by thyrotropin-regulated thyroid activity. with [35S]sulfate: incorporation into chondroitin 6-sulfate
thyroglobulin enhancer. Biochem. J. 323, 349–357 Endocrinology 109, 523–529 (1981). and endoglycosidase-F-susceptible carbohydrate units.
(1997). 117. Consiglio, E. et al. Characterization of phosphate Endocrinology 122, 2428–2435 (1988).
96. Baas, F., van Ommen, G. J., Bikker, H., Arnberg, A. C. residues on thyroglobulin. J. Biol. Chem. 262, 138. Schneider, A. B. & Dudlak, D. Chondroitin chain and
& de Vijlder, J. J. The human thyroglobulin gene is 10304–10314 (1987). complex carbohydrate chains of human thyroglobulin:
over 300 kb long and contains introns of up to 64 kb. 118. Yang, S., Pollock, H. G. & Rawitch, A. B. Glycosylation studies in normal and neoplastic thyroid tissue.
Nucleic Acids Res. 14, 5171–5186 (1986). in human thyroglobulin: location of the N-linked Endocrinology 124, 356–362 (1989).
97. Meijerink, P. H. et al. The gene for the human Src-like oligosaccharide units and comparison with bovine 139. Sakurai, S., Fogelfeld, L., Ries, A. & Schneider, A. B.
adaptor protein (hSLAP) is located within the 64-kb thyroglobulin 1. Arch. Biochem. Biophys. 327, 61–70 Anionic complex-carbohydrate units of human
intron of the thyroglobulin gene. Eur. J. Biochem. 254, (1996). thyroglobulin. Endocrinology 127, 2056–2063
297–303 (1998). 119. MacPhee-Quigley, K., Vedvick, T. S., Taylor, P. (1990).
98. Lee, J. & Arvan, P. Repeat motif-containing regions & Taylor, S. S. Profile of the disulfide bonds in 140. Chambard, M. et al. Thyrotrophin regulation of apical
within thyroglobulin. J. Biol. Chem. 286, 26327–26333 acetylcholinesterase. J. Biol. Chem. 261, and basal exocytosis of thyroglobulin by porcine
(2011). 13565–13570 (1986). thyroid monolayers. J. Mol. Endocrinol. 4, 193–199
99. Veneziani, B. M., Giallauria, F. & Gentile, F. The 120. Di Jeso, B. et al. Mixed-disulfide folding (1990).
disulfide bond pattern between fragments obtained by intermediates between thyroglobulin and 141. Nlend, M. C., Cauvi, D., Venot, N. & Chabaud, O.
the limited proteolysis of bovine thyroglobulin. endoplasmic reticulum resident oxidoreductases Sulfated tyrosines of thyroglobulin are involved in
Biochimie 81, 517–525 (1999). ERp57 and protein disulfide isomerase. Mol. Cell. thyroid hormone synthesis. Biochem. Biophys. Res.
100. Molina, F., Bouanani, M., Pau, B. & Granier, C. Biol. 25, 9793–9805 (2005). Commun. 262, 193–197 (1999).
Characterization of the type-1 repeat from 121. Di Jeso, B. et al. Transient covalent interactions of 142. Venot, N., Nlend, M. C., Cauvi, D. & Chabaud, O.
thyroglobulin, a cysteine-rich module found in proteins newly synthesized thyroglobulin with oxidoreductases The hormonogenic tyrosine 5 of porcine thyroglobulin
from different families. Eur. J. Biochem. 240, 125–133 of the endoplasmic reticulum. J. Biol. Chem. 289, is sulfated. Biochem. Biophys. Res. Commun. 298,
(1996). 11488–11496 (2014). 193–197 (2002).
101. Cousin, X. et al. The alpha/beta fold family of proteins This paper identifies several distinct chaperone 143. Nlend, M. C., Cauvi, D. M., Venot, N. & Chabaud, O.
database and the cholinesterase gene server ESTHER. and oxidoreductase partners that engage newly Role of sulfated tyrosines of thyroglobulin in thyroid
Nucleic Acids Res. 25, 143–146 (1997). synthesized thyroglobulin during its early folding in hormonosynthesis. Endocrinology 146, 4834–4843
102. Targovnik, H. M., Citterio, C. E. & Rivolta, C. M. the ER. (2005).
Thyroglobulin gene mutations in congenital 122. Di Jeso, B. et al. Folding of thyroglobulin in the 144. Palumbo, G., Gentile, F., Condorelli, G. L. & Salvatore, G.
hypothyroidism. Horm. Res. Paediatr. 75, 311–321 calnexin/calreticulin pathway and its alteration by loss The earliest site of iodination in thyroglobulin is residue
(2011). of Ca2+ from the endoplasmic reticulum. Biochem. J. number 5. J. Biol. Chem. 265, 8887–8892 (1990).
103. De Jaco, A., Dubi, N., Camp, S. & Taylor, P. Congenital 370, 449–458 (2003). This paper shows that thyroglobulin Tyr5 becomes
hypothyroidism mutations affect common folding and 123. Jessop, C. E. et al. ERp57 is essential for efficient iodinated before other tyrosines, suggesting its
trafficking in the α/β-hydrolase fold proteins. FEBS J. folding of glycoproteins sharing common structural exposure on the surface of the 3D molecular
279, 4293–4305 (2012). domains. EMBO J. 26, 28–40 (2007). structure of thyroglobulin.
104. Lee, J., Di jeso, B. & Arvan, P. The cholinesterase-like 124. Vali, M., Rose, N. R. & Caturegli, P. Thyroglobulin as 145. Dedieu, A., Gaillard, J.-C., Pourcher, T., Darrouzet, E.
domain of thyroglobulin functions as an intramolecular autoantigen: structure-function relationships. & Armengaud, J. Revisiting iodination sites in
chaperone. J. Clin. Invest. 118, 2950–2958 (2008). Rev. Endocr. Metab. Disord. 1, 69–77 (2000). thyroglobulin with an organ-oriented shotgun strategy.
This paper demonstrates that the thyroglobulin 125. Xavier, A. C. W., Maciel, R. M. B., Vieira, J. G. H., J. Biol. Chem. 286, 259–269 (2011).
ChEL domain can function independently to assist Dias-da-Silva, M. R. & Martins, J. R. M. Insights into 146. Dunn, J. T. & Dunn, A. D. The importance of
in the folding stability, intracellular transport and the posttranslational structural heterogeneity of thyroglobulin structure for thyroid hormone
secretion of the upstream regions of thyroglobulin. thyroglobulin and its role in the development, biosynthesis. Biochimie 81, 505–509 (1999).
105. Park, Y.-N. & Arvan, P. The acetylcholinesterase diagnosis, and management of benign and malignant This comprehensive review summarizes the main
homology region is essential for normal conformational thyroid diseases. Arch. Endocrinol. Metab. 60, 66–75 hormonogenic sites of thyroglobulin that are
maturation and secretion of thyroglobulin. J. Biol. (2016). involved in T4 and T3 formation.
Chem. 279, 17085–17089 (2004). 126. Mallet, B., Lejeune, P., Baudry, N., Niccoli, P. & 147. Lamas, L., Anderson, P., Fox, J. W. & Dunn, J. T.
106. Lee, J., Di Jeso, B. & Arvan, P. Maturation of Carayon, P. N-glycans modulate in vivo and in vitro Consensus sequences for early iodination and
thyroglobulin protein region I. J. Biol. Chem. 286, thyroid hormone synthesis. J. Biol. Chem. 270, hormonogenesis in human thyroglobulin. J. Biol.
33045–33052 (2011). 29881–29888 (1995). Chem. 264, 13541–13545 (1989).
107. Kim, P. S. & Arvant, P. Folding and assembly of newly 127. Conte, M. et al. A single chondroitin 6-sulfate 148. Izumi, M. & Larsen, P. R. Triiodothyronine, thyroxine,
synthesized thyroglobulin occurs in a pre-Golgi oligosaccharide unit at Ser-2730 of human and iodine in purified thyroglobulin from patients with
compartment. J. Biol. Chem. 266, 12412–12418 thyroglobulin enhances hormone formation and limits Graves’ disease. J. Clin. Invest. 59, 1105–1112 (1977).
(1991). proteolytic accessibility at the carboxyl terminus. 149. Dunn, A. D., Corsi, C. M., Myers, H. E. & Dunn, J. T.
108. Suzuki, K. et al. Thyroglobulin autoregulation of Potential insights into thyroid homeostasis and Tyrosine 130 is an important outer ring donor for
thyroid-specific gene expression and follicular autoimmunity. J. Biol. Chem. 281, 22200–22211 thyroxine formation in thyroglobulin. J. Biol. Chem.
function. Rev. Endocr. Metab. Disord. 1, 217–224 (2006). 273, 25223–25229 (1998).
(2000). 128. Tsuji, T., Yamam, Irimura, T. & Osawa, T. Structure This paper provides independent evidence in
109. Lee, J., Wang, X., Di Jeso, B. & Arvan, P. The of carbohydrate unit A of porcine thyroglobulin. support of earlier work suggesting that DIT at
cholinesterase-like domain, essential in thyroglobulin Biochem. J. 195, 691–699 (1981). residue 130 of thyroglobulin is the donor to the
trafficking for thyroid hormone synthesis, is required 129. Yamamoto, K., Tsuji, T., Irimura, T. & Osawa, T. acceptor DIT at residue 5.
for protein dimerization. J. Biol. Chem. 284, The structure of carbohydrate unit B of porcine 150. Marriq, C., Lejeune, P. J., Venot, N. & Vinet, L.
12752–12761 (2009). thyroglobulin. Biochem. J. 195, 701–713 (1981). Hormone formation in the isolated fragment 1–171 of

Nature Reviews | Endocrinology


Reviews

human thyroglobulin involves the couple tyrosine 5 169. Carvalho, D. P. & Dupuy, C. Thyroid hormone 188. Sato, A. et al. A novel mutation in the thyroglobulin
and tyrosine 130. Mol. Cell. Endocrinol. 81, 155–164 biosynthesis and release. Mol. Cell. Endocrinol. 458, gene that causes goiter and dwarfism in Wistar
(1991). 6–15 (2017). Hannover GALAS rats. Mutat. Res. 762, 17–23
151. Ohmiya, Y., Hayashi, H. & Kondo, T. K. Y. Location of 170. Huang, H., Shi, Y., Liang, B., Cai, H. & Cai, Q. (2014).
dehydroalanine bovine thyroglobulin residues in the Iodinated TG in thyroid follicular lumen regulates 189. Vigone, M. C., Capalbo, D., Weber, G. & Salerno, M.
amino acid sequence of bovine thyroglobulin. J. Biol. TTF-1 and PAX8 expression via TSH/TSHR signaling Mild hypothyroidism in childhood: who, when and how
Chem. 265, 9066–9071 (1990). pathway. J. Cell. Biochem. 118, 3444–3451 (2017). should be treated? J. Endocr. Soc. 2, (1024–1039
152. Cetrangolo, G. P. et al. Hormonogenic donor Tyr2522 171. Noguchi, Y. et al. Thyroglobulin (Tg) induces thyroid (2018).
of bovine thyroglobulin. Insight into preferential T3 cell growth in a concentration-specific manner by a 190. Nicholas, A. K. et al. Comprehensive screening of eight
formation at thyroglobulin carboxyl terminus at low mechanism other than thyrotropin / cAMP stimulation. known causative genes in congenital hypothyroidism
iodination level. Biochem. Biophys. Res. Commun. Biochem. Biophys. Res. Commun. 391, 890–894 with gland-in-situ. J. Clin. Endocrinol. Metab. 101,
450, 488–493 (2014). (2010). 4521–4531 (2016).
This paper presents strong mass spectrometry 172. Padron, A. S. et al. Administration of 191. Peteiro-Gonzalez, D. et al. New insights into
evidence for efficient mono-iodination and 3,5-diiodothyronine (3,5-T2) causes central thyroglobulin pathophysiology revealed by the study
di-iodination of the antepenultimate residue that hypothyroidism and stimulates thyroid-sensitive of a family with congenital goiter. J. Clin. Endocrinol.
is engaged in T3 synthesis in thyroglobulin. tissues. J. Endocrinol. 221, 415–427 (2014). Metab. 95, 3522–3526 (2010).
153. Mallet, B. et al. Tyrosine iodination and iodotyrosyl 173. Schanze, N. et al. 3-iodothyronamine decreases 192. Kim, P. S. & Arvan, P. Endocrinopathies in the family
coupling of the N-terminal thyroid hormone forming expression of genes involved in iodide metabolism in of endoplasmic reticulum (ER) storage diseases:
site of human thyroglobulin modulate its binding to mouse thyroids and inhibits iodide uptake in PCCL3 disorders of chaperones. Endocr. Rev. 19, 173–202
auto- and monoclonal antibodies. Mol. Cell. thyrocytes. Thyroid 27, 11–22 (2017). (1998).
Endocrinol. 88, 89–95 (1992). 174. Ferreira, A. C. F. et al. Rapid regulation of thyroid 193. Citterio, C. E. et al. New insights into thyroglobulin
154. den Hartog, M. T., Sijmons, C. C., Bakker, O., sodium-iodide symporter activity by thyrotrophin and gene: molecular analysis of seven novel mutations
Ris-Stalpers, C. & de Vijlder, J. J. Importance of the iodine. J. Endocrinol. 184, 69–76 (2005). associated with goiter and hypothyroidism. Mol. Cell.
content and localization of tyrosine residues for 175. Woeber, K. A. Iodine and thyroid disease. Med. Clin. Endocrinol. 365, 277–291 (2013).
thyroxine formation within the N-terminal part of North Am. 75, 169–178 (1991). 194. Kanou, Y. et al. Thyroglobulin gene mutations
human thyroglobulin. Eur. J. Endocrinol. 132, 176. Führer, D. et al. Autosomal dominant nonautoimmune producing defective intracellular transport of
611–617 (1995). hyperthyroidism. Clinical features-diagnosis-therapy. thyroglobulin are associated with increased thyroidal
155. Gentile, F., Ferranti, P., Mamone, G., Malorni, A. & Exp. Clin. Endocrinol. Diabetes 106, 10–15 (1998). type 2 iodothyronine deiodinase activity. J. Clin.
Salvatore, G. Identification of hormonogenic tyrosines 177. Mai, V. & Burch, H. A. Stepwise approach to the Endocrinol. Metab. 92, 1451–1457 (2007).
in fragment 1218–1591 of bovine thyroglobulin by evaluation and treatment of subclinical 195. Hishinuma, A. et al. Haplotype analysis reveals
mass spectrometry. Biochemistry 272, 639–646 hyperthyroidism. Endocr. Pract. 18, 772–780 founder effects of thyroglobulin gene mutations
(1997). (2012). C1058R and C1977S in Japan. J. Clin. Endocrinol.
156. Marsili, A., Zavacki, A. M., Harney, J. W. & Larsen, P. R. 178. Toccafondi, R. et al. Effects of TSH on cAMP levels and Metab. 91, 3100–3104 (2006).
Physiological role and regulation of iodothyronine thyroid hormone release in human thyroid 196. Umezu, M., Kagabu, S., Jiang, J. & Sato, E. Evaluation
deiodinases: a 2011 update. J. Endocrinol. Invest. 34, ‘autonomous’ nodules: relationship with iodothyronine and characterization of congenital hypothyroidism in
395–407 (2011). and iodine content in thyroglobulin. Clin. Endocrinol. rdw dwarf rats. Lab. Anim. Sci. 48, 496–501 (1998).
157. Laugwitz, K. L. et al. The human thyrotropin receptor: 17, 537–546 (1982). 197. Baryshev, M. et al. Unfolded protein response is
A heptahelical receptor capable of stimulating 179. Kosugi, S., Hai, N., Okamoto, H., Sugawa, H. & Mori, T. involved in the pathology of human congenital
members of all four G protein families. Proc. Natl A novel activating mutation in the thyrotropin receptor hypothyroid goiter and rat non-goitrous congenital
Acad. Sci. USA 93, 116–120 (1996). gene in an autonomously functioning thyroid nodule hypothyroidism. J. Mol. Endocrinol. 32, 903–920
158. Maenhaut, C., Brabant, G., Vassart, G. & Dumont, J. E. developed by a Japanese patient. Eur. J. Endocrinol. (2004).
In vitro and in vivo regulation of thyrotropin receptor 143, 471–477 (2000). 198. Menon, S. et al. Oxidoreductase interactions include a
mRNA levels in dog and human thyroid cells. J. Biol. 180. Patel, Y. C., Pharoah, P. P. O. D., Hornabrook, R. W. role for ERp72 engagement with mutant thyroglobulin
Chem. 267, 3000–3007 (1992). & Hetzel, B. S. Serum triiodothyronine, thyroxine and from the rdw/rdw rat dwarf. J. Biol. Chem. 282,
159. Dohán, O. et al. The sodium/iodide symporter (NIS): thyroid-stimulating hormone in endemic goiter: 6183–6191 (2007).
characterization, regulation, and medical significance. a comparison of goitrous and nongoitrous subjects in 199. Gaide Chevronnay, H. P. et al. A mouse model suggests
Endocr. Rev. 24, 48–77 (2003). New Guinea. J. Clin. Endocrinol. Metab. 37, 783–789 two mechanisms for thyroid alterations in infantile
160. Raad, H., Eskalli, Z., Corvilain, B., Miot, F. & (1973). cystinosis: Decreased thyroglobulin synthesis due to
De Deken, X. Thyroid hydrogen peroxide production 181. Vagenakis, A. G. et al. Studies of serum triiodothyronine, endoplasmic reticulum stress/unfolded protein
is enhanced by the Th2 cytokines, IL-4 and IL-13, thyroxine and thyrotropin concentrations in endemic response and impaired lysosomal processing.
through increased expression of the dual oxidase goiter in Greece. J. Clin. Endocrinol. Metab. 37, Endocrinology 156, 2349–2364 (2015).
2and its maturation factorDUOXA2. Free Radic. 485–488 (1973). 200. Taguchi, A. et al. A symptomatic Fabry disease mouse
Biol. Med. 56, 216–225 (2013). 182. Gilbert, J. A. et al. Monoclonal pathogenic antibodies model generated by inducing globotriaosylceramide
161. Cardoso-Weide, L. C. et al. DuOx2 promoter to the thyroid-stimulating hormone receptor in synthesis. Biochem. J. 456, 373–383 (2013).
regulation by hormones, transcriptional factors and Graves’ disease with potent thyroid-stimulating
the coactivator TAZ. Eur. Thyroid J. 4, 6–13 (2015). activity but differential blocking activity activate Acknowledgements
162. Pesce, L. et al. TSH regulates pendrin membrane multiple signaling pathways. J. Immunol. 176, The authors acknowledge the support of NIH grant R01
abundance and enhances iodide efflux in thyroid cells. 5084–5092 (2006). DK40344.
Endocrinology 153, 512–521 (2012). 183. Targovnik, H. M., Citterio, C. E. & Rivolta, C. M.
163. Dupuy, C. et al. Mechanism of hydrogen peroxide Iodide handling disorders (NIS, TPO, TG, IYD). Best Author contributions
formation catalyzed by NADPH oxidase in thyroid Pract. Res. Clin. Endocrinol. Metab. 31, 195–212 C.E.C., H.M.T. and P.A. researched data for the article and
plasma membrane. J. Biol. Chem. 266, 3739–3743 (2017). made a substantial contribution to discussion of content.
(1991). 184. Ricketts, M. H., Simons, M. J., Mercken, L. & Dong, Q. C.E.C. and P.A. wrote, reviewed and/or edited the manuscript
164. Song, Y. et al. Association of duoxes with thyroid A nonsense mutation causes hereditary goitre in the before submission.
peroxidase and its regulation in thyrocytes. J. Clin. Afrikander cattle and unmasks alternative splicing of
Endocrinol. Metab. 95, 375–382 (2010). thyroglobulin transcripts. Proc. Natl Acad. Sci. USA Competing interests
165. Kero, J. et al. Thyrocyte-specific Gq / G11 deficiency 84, 3181–3184 (1987). The authors declare no competing interests.
impairs thyroid function and prevents goiter 185. Veenboer, G. J. & de Vijlder, J. J. Molecular basis of
development. J. Clin. Invest. 117, 2399–2407 the thyroglobulin synthesis defect in Dutch goats. Publisher’s note
(2007). Endocrinology 132, 377–381 (1993). Springer Nature remains neutral with regard to jurisdictional
166. Patyra, K. et al. Partial thyrocyte-specific G a s 186. Kim, P. S. et al. A single amino acid change in the claims in published maps and institutional affiliations.
deficiency leads to rapid-onset hypothyroidism, acetylcholinesterase-like domain of thyroglobulin
hyperplasia, and papillary thyroid carcinoma – like causes congenital goiter with hypothyroidism in the Reviewer information
lesions in mice. FASEB J. 32, 6239–6251 (2018). cog/cog mouse: a model of human endoplasmic Nature Reviews Endocrinology thanks X. DeDeken, J.
167. Latif, R. et al. New small molecule agonists to the reticulum storage diseases. Proc. Natl Acad. Sci. USA Lado-Abeal and other anonymous reviewer(s) for their
thyrotropin receptor. Thyroid 25, 51–62 (2015). 95, 9909–9913 (1998). contribution to the peer review of this work.
168. Luo, Y. et al. A novel role for flotillin-containing lipid 187. Kim, P. S. et al. A missense mutation G2320R in the
rafts in negative-feedback regulation of thyroid-specific thyroglobulin gene causes non-goitrous congenital Supplementary information
gene expression by thyroglobulin. Thyroid 26, primary hypothyroidism in the WIC-rdw rat. Supplementary information is available for this paper at
1630–1639 (2016). Mol. Endocrinol. 14, 1944–1953 (2000). https://doi.org/10.1038/s41574-019-0184-8.

www.nature.com/nrendo

You might also like