You are on page 1of 7

2926 Hyponatremia, for example, appears to be more common and is more ANATOMY AND DEVELOPMENT

likely to be due to infectious diseases such as cholera, shigellosis, and The thyroid (Greek thyreos, shield, plus eidos, form) consists of two lobes
other diarrheal disorders. In these circumstances, hyponatremia is connected by an isthmus. It is located anterior to the trachea between
probably due to gastrointestinal losses of salt and water (hypovolemia the cricoid cartilage and the suprasternal notch. The normal thyroid is
type II), but other abnormalities, including undefined infectious tox- 12–20 g in size, highly vascular, and soft in consistency. Four parathyroid
ins, also may contribute. The causes of DI are similar worldwide except glands, which produce parathyroid hormone (Chap. 410), are located
that malaria and venoms from snake or insect bites are much more posterior to each pole of the thyroid. The recurrent laryngeal nerves
common in some tropical climates. traverse the lateral borders of the thyroid gland and must be identified
during thyroid surgery to avoid injury and vocal cord paralysis.
■ FURTHER READING The thyroid gland develops from the floor of the primitive pharynx
Bichet DG: Regulation of thirst and vasopressin release. Annu Rev during the third week of gestation. The developing gland migrates
along the thyroglossal duct to reach its final location in the neck. This
PART 12

Physiol 81:359, 2019.


Bichet DG et al: GNAS: A new nephrogenic cause of inappropriate feature accounts for the rare ectopic location of thyroid tissue at the
antidiuresis. J Am Soc Nephrol 30:722, 2019. base of the tongue (lingual thyroid) as well as the occurrence of thy-
Fenske W et al: A copeptin based approach in the diagnosis of diabetes roglossal duct cysts along this developmental tract. Thyroid hormone
insipidus. N Engl J Med 379:428, 2018. synthesis begins at about 11 weeks’ gestation.
Endocrinology and Metabolism

Oiso Y et al: Clinical review: Treatment of neurohypophyseal diabetes Neural crest derivatives from the ultimobranchial body give rise to
insipidus. J Clin Endocrinol Metab 98:3958, 2013. thyroid medullary C cells that produce calcitonin, a calcium-lowering
Robertson GL: Vaptans for the treatment of hyponatremia. Nat Rev hormone. The C cells are interspersed throughout the thyroid gland,
Endocrinol 7:151, 2011. although their density is greatest in the juncture of the upper one-third
Robertson GL: Diabetes insipidus: Differential diagnosis and man- and lower two-thirds of the gland. Calcitonin plays a minimal role in
agement. Best Pract Res Clin Endocrinol Metab 30:205, 2016. calcium homeostasis in humans, but the C cells are important because
of their involvement in medullary thyroid cancer.
Thyroid gland development is orchestrated by the coordinated expres-
sion of several developmental transcription factors. Thyroid transcrip-
tion factor (TTF)-1, TTF-2, NKX2-1, and paired homeobox-8 (PAX-8)
are expressed selectively, but not exclusively, in the thyroid gland. In
combination, they dictate thyroid cell development and the induction
of thyroid-specific genes such as thyroglobulin (Tg), thyroid peroxidase

382 Thyroid
(TPO), the sodium iodide symporter (Na+/I–, NIS), and the thyroid-stim-
Gland ulating hormone (TSH) receptor (TSH-R). Mutations in these develop-
Physiology and Testing mental transcription factors or their downstream target genes are rare
causes of thyroid agenesis or dyshormonogenesis, although the causes of
J. Larry Jameson, Susan J. Mandel, most forms of congenital hypothyroidism remain unknown (see Chap.
Anthony P. Weetman 383, Table 383-1). Because congenital hypothyroidism occurs in ~1 in
4000 newborns, neonatal screening is now performed in most industri-
alized countries. Transplacental passage of maternal thyroid hormone
occurs before the fetal thyroid gland begins to function and provides
The thyroid gland produces two related hormones, thyroxine (T4) and significant hormone support to a fetus with congenital hypothyroidism.
triiodothyronine (T3) (Fig. 382-1). Acting through thyroid hormone Early thyroid hormone replacement in newborns with congenital hypo-
receptors (TR) α and β, these hormones play a critical role in cell dif- thyroidism prevents potentially severe developmental abnormalities.
ferentiation and organogenesis during development and help maintain The thyroid gland consists of numerous spherical follicles composed
thermogenic and metabolic homeostasis in the adult. Autoimmune of thyroid follicular cells that surround secreted colloid, a protein-
disorders of the thyroid gland can stimulate overproduction of thyroid aceous fluid containing large amounts of thyroglobulin, the protein
hormones (thyrotoxicosis) or cause glandular destruction and hormone precursor of thyroid hormones (Fig. 382-2). The thyroid follicular cells
deficiency (hypothyroidism). Benign nodules and various forms of are polarized—the basolateral surface is apposed to the bloodstream
thyroid cancer are relatively common and amenable to detection by and an apical surface faces the follicular lumen. Increased demand for
physical examination, ultrasound, and other imaging techniques. thyroid hormone is regulated by TSH, which binds to its receptor on
the basolateral surface of the follicular cells. This binding
leads to Tg reabsorption from the follicular lumen and
I I NH2 proteolysis within the cytoplasm, yielding thyroid hor-
HO
3'
O
3
CH2 CH COOH
mones for secretion into the bloodstream.
5' 5

I I
REGULATION OF THE THYROID
Thyroxine (T4)
AXIS
3,5,3',5'-Tetraiodothyronine TSH, secreted by the thyrotrope cells of the anterior pitu-
Deiodinase 1 or 2 Deiodinase 3>2 itary, plays a pivotal role in control of the thyroid axis and
(5'-Deiodination) (5-Deiodination) serves as the most useful physiologic marker of thyroid
hormone action. TSH is a 31-kDa hormone composed of
α and β subunits; the α subunit is common to the other
I I I I
NH2 NH2 glycoprotein hormones (luteinizing hormone, follicle-
HO O CH2 CH COOH HO O CH2 CH COOH
stimulating hormone, human chorionic gonadotropin
[hCG]), whereas the TSH β subunit is unique to TSH.
I I The extent and nature of carbohydrate modification are
Triiodothyronine (T3) Reverse T3 (rT3) modulated by thyrotropin-releasing hormone (TRH) and
3,5,3'-Triiodothyronine 3,3',5'-Triiodothyronine influence the biologic activity of the hormone.
FIGURE 382-1 Structures of thyroid hormones. Thyroxine (T4) contains four iodine atoms. The thyroid axis is a classic example of an endocrine
Deiodination leads to production of the potent hormone triiodothyronine (T3) or the inactive feedback loop (Chap. 377). Hypothalamic TRH stimulates
hormone reverse T3. pituitary production of TSH, which, in turn, stimulates
of Tg into the thyroid follicular cell allows proteolysis and the release 2927
Hypothalamus T3 T4 of newly synthesized T4 and T3.

TSH-R Iodine Metabolism and Transport Iodide uptake is a critical
Basal
NIS
first step in thyroid hormone synthesis. Ingested iodine is bound to
I- serum proteins, particularly albumin. Unbound iodine is excreted in
the urine. The thyroid gland extracts iodine from the circulation in
I- a highly efficient manner. For example, 10–25% of radioactive tracer
(e.g., 123I) is taken up by the normal thyroid gland over 24 h in an
TRH cAMP iodine-replete state; this value can rise to 70–90% in Graves’ disease.

CHAPTER 382 Thyroid Gland Physiology and Testing


+ – Iodide uptake is mediated by NIS, which is expressed at the basolateral
membrane of thyroid follicular cells. NIS is most highly expressed in
Tg
Pituitary Apical the thyroid gland, but low levels are present in the salivary glands, lac-
tating breast, and placenta. The iodide transport mechanism is highly
DIT
TPO regulated, allowing adaptation to variations in dietary supply. Low
Follicular
cell
iodine levels increase the amount of NIS and stimulate uptake, whereas
Tg-MIT
Tg + I- high iodine levels suppress NIS expression and uptake. The selective
TSH expression of NIS in the thyroid allows isotopic scanning, treatment
Co

up
ling Iodination
+ of hyperthyroidism, and ablation of thyroid cancer with radioisotopes
of iodine, without significant effects on other organs. Mutation of the
NIS gene is a rare cause of congenital hypothyroidism, underscoring its
Thyroid
Thyroid follicle
importance in thyroid hormone synthesis. Another iodine transporter,
pendrin, is located on the apical surface of thyroid cells and mediates
iodine efflux into the lumen. Mutation of the pendrin gene causes Pen-
T4 T3 dred syndrome, a disorder characterized by defective organification of
iodine, goiter, and sensorineural deafness.
Iodine deficiency is prevalent in many mountainous regions and in
Peripheral
central Africa, central South America, and northern Asia (Fig. 382-3).
actions Europe remains mildly iodine-deficient, and health surveys indicate
that iodine intake has been falling in the United States and Australia.
FIGURE 382-2 Regulation of thyroid hormone synthesis. Left. Thyroid hormones The World Health Organization (WHO) estimates that about 2 billion
T4 and T3 feed back to inhibit hypothalamic production of thyrotropin-releasing
hormone (TRH) and pituitary production of thyroid-stimulating hormone (TSH). people are iodine-deficient, based on urinary excretion data. In areas
TSH stimulates thyroid gland production of T4 and T3. Right. Thyroid follicles are of relative iodine deficiency, there is an increased prevalence of goiter
formed by thyroid epithelial cells surrounding proteinaceous colloid, which contains and, when deficiency is severe, hypothyroidism and cretinism. Cretin-
thyroglobulin. Follicular cells, which are polarized, synthesize thyroglobulin and ism is characterized by intellectual disability and growth retardation
carry out thyroid hormone biosynthesis (see text for details). DIT, diiodotyrosine; and occurs when children who live in iodine-deficient regions are not
MIT, monoiodotyrosine; NIS, sodium iodide symporter; Tg, thyroglobulin; TPO, treated with iodine or thyroid hormone to restore normal thyroid hor-
thyroid peroxidase; TSH-R, thyroid-stimulating hormone receptor.
mone levels during early life. These children are often born to mothers
thyroid hormone synthesis and secretion. Thyroid hormones act via with iodine deficiency, and it is likely that maternal thyroid hormone
negative feedback predominantly through thyroid hormone receptor deficiency worsens the condition. Concomitant selenium deficiency
β2 (TRβ2) to inhibit TRH and TSH production (Fig. 382-2). The “set may also contribute to the neurologic manifestations of cretinism.
point” in this axis is established by TSH. TRH is the major positive reg- Iodine supplementation of salt, bread, and other food substances has
ulator of TSH synthesis and secretion. Peak TSH secretion occurs ~15 markedly reduced the prevalence of cretinism. Unfortunately, however,
min after administration of exogenous TRH. Dopamine, glucocorti- iodine deficiency remains the most common cause of preventable intel-
coids, and somatostatin suppress TSH but are not of major physiologic lectual disability, often because of societal resistance to food additives
importance except when these agents are administered in pharmaco- or the cost of supplementation. In addition to overt cretinism, mild
logic doses. Reduced levels of thyroid hormone increase basal TSH iodine deficiency can lead to subtle reduction of IQ. Oversupply of
production and enhance TRH-mediated stimulation of TSH. High thy- iodine, through supplements or foods enriched in iodine (e.g., shell-
roid hormone levels rapidly and directly suppress TSH gene expression fish, kelp), is associated with an increased incidence of autoimmune
and inhibit TRH stimulation of TSH secretion, indicating that thyroid thyroid disease. The Recommended Dietary Allowance (RDA) is
hormones are the dominant regulator of TSH production. Like other 220 μg iodine per day for pregnant women and 290 μg iodine per day
pituitary hormones, TSH is released in a pulsatile manner and exhib- for breastfeeding women. Because the effects of iodine deficiency are
its a diurnal rhythm; its highest levels occur at night. However, these most severe in pregnant women and their babies, the American Thy-
TSH excursions are modest in comparison to those of other pituitary roid Association has recommended that all pregnant and breastfeeding
hormones, in part, because TSH has a relatively long plasma half-life women in the United States and Canada take a prenatal multivitamin
(50 min). Consequently, single measurements of TSH are adequate for containing 150 μg iodine per day. Urinary iodine is >100 μg/L in
assessing its circulating level. TSH is measured using immunoradio- iodine-sufficient populations.
metric assays that are highly sensitive and specific. These assays readily
distinguish between normal and suppressed TSH values; thus, TSH can Organification, Coupling, Storage, and Release After iodide
be used for the diagnosis of primary hyperthyroidism (low TSH) or enters the thyroid, it is trapped and transported to the apical mem-
primary hypothyroidism (high TSH). brane of thyroid follicular cells, where it is oxidized in an organifica-
tion reaction that involves TPO and hydrogen peroxide produced by
THYROID HORMONE SYNTHESIS, dual oxidase (DUOX) and DUOX maturation factor (DUOXA). The
METABOLISM, AND ACTION reactive iodine atom is added to specific tyrosyl residues within Tg, a
large (660 kDa) dimeric protein that consists of 2769 amino acids. The
■ THYROID HORMONE SYNTHESIS iodotyrosines in Tg are then coupled via an ether linkage in a reaction
Thyroid hormones are derived from Tg, a large iodinated glycoprotein. that is also catalyzed by TPO. Either T4 or T3 can be produced by this
After secretion into the thyroid follicle, Tg is iodinated on tyrosine reaction, depending on the number of iodine atoms present in the
residues that are subsequently coupled via an ether linkage. Reuptake iodotyrosines. After coupling, Tg is taken back into the thyroid cell,
2928 Global scorecard of iodine nutrition in 2021
Iodine intake in the general population assessed by median urinary iodine concentration (mUIC) in school-age children (SAC)a
Studies conducted in 2005–2020
PART 12
Endocrinology and Metabolism

Iodine intake
Insufficient Adequateb Excess
mUIC mUIC mUIC
<100 µg/L 100–299 µg/L ≥300 µg/L

National data 18 105 9

Sub-national data 3 13 4

No recent data 42

FIGURE 382-3 Worldwide iodine nutrition. aIn population monitoring of iodine status using urinary iodine concentration (UIC), school-age children (SAC) serve as a proxy
for the general population; therefore, preference has been given to studies carried out in SAC. The UIC data have been selected for each country in the following order of
priority: data from the most recent known nationally representative survey carried out between 2005 and 2020 in (i) SAC, (ii) SAC and adolescents, (iii) adolescents, (iv) women
of reproductive age, (v) other adults (excluding pregnant or lactating women), and (vi) other eligible populations. In the absence of recent national surveys, subnational data
were used in the same order of priority. Subnational UIC surveys are commonly carried out to provide a rapid assessment of population iodine status, but due to a lack of
sampling rigor, they may over- or underestimate the iodine status at the national level and should be interpreted with caution. bAdequate iodine intake in school-age children
corresponds to median UIC values in the range 100-299 μg/L, and includes categories previously referred to as “Adequate” (100-199 μg/L) and “More than adequate”
(200-299 μg/L). (Reproduced with permission from The Iodine Global Network. Global scorecard of iodine nutrition in 2021 in the general population based on data in school-age
children (SAC). IGN: Ottawa, Canada. 2021.)

where it is processed in lysosomes to release T4 and T3. Uncoupled the receptor. They mimic the conformational changes induced by TSH
mono- and diiodotyrosines (MIT, DIT) can be deiodinated by the binding or the interactions of thyroid-stimulating immunoglobulins
enzyme dehalogenase, thereby recycling any iodide that is not con- (TSIs) in Graves’ disease. Activating TSH-R mutations also occur as
verted into thyroid hormones. somatic events, leading to clonal selection and expansion of the affected
Disorders of thyroid hormone synthesis are rare causes of congenital thyroid follicular cell and autonomously functioning thyroid nodules.
hypothyroidism (Chap. 383). The vast majority of these disorders are
due to recessive mutations in TPO or Tg, but defects have also been Other Factors That Influence Hormone Synthesis and
identified in the TSH-R, NIS, pendrin, hydrogen peroxide generation, Release Although TSH is the dominant hormonal regulator of
and dehalogenase, as well as genes involved in thyroid gland devel- thyroid gland growth and function, a variety of growth factors, most
opment. In the case of biosynthetic defects, the gland is incapable of produced locally in the thyroid gland, also influence thyroid hormone
synthesizing adequate amounts of hormone, leading to increased TSH synthesis. These include insulin-like growth factor 1 (IGF-1), epider-
and a large goiter. mal growth factor, transforming growth factor β (TGF-β), endothelins,
and various cytokines. The quantitative roles of these factors are not
TSH Action TSH regulates thyroid gland function through the well understood, but they are important in selected disease states.
TSH-R, a seven-transmembrane G protein–coupled receptor (GPCR). In acromegaly, for example, increased levels of growth hormone and
The TSH-R is coupled to the α subunit of stimulatory G protein (GSα), IGF-1 are associated with goiter and predisposition to multinodular
which activates adenylyl cyclase, leading to increased production of goiter (MNG). Certain cytokines and interleukins (ILs) produced in
cyclic adenosine monophosphate (cAMP). TSH also stimulates phos- association with autoimmune thyroid disease induce thyroid growth,
phatidylinositol turnover by activating phospholipase C. Recessive whereas others lead to apoptosis. Iodine deficiency increases thyroid
loss-of-function TSH-R mutations cause thyroid hypoplasia and con- blood flow and upregulates the NIS, stimulating more efficient iodine
genital hypothyroidism. Dominant gain-of-function mutations cause uptake. Excess iodide transiently inhibits thyroid iodide organification,
sporadic or familial hyperthyroidism that is characterized by goiter, a phenomenon known as the Wolff-Chaikoff effect. In individuals with
thyroid cell hyperplasia, and autonomous function (Chap. 384). Most a normal thyroid, the gland escapes from this inhibitory effect and
of these activating mutations occur in the transmembrane domain of iodide organification resumes; the suppressive action of high iodide
may persist, however, in patients with underlying autoimmune thyroid TABLE 382-1 Characteristics of Circulating T4 and T3 2929
disease.
HORMONE PROPERTY T4 T3
Serum concentrations    
THYROID FUNCTION IN PREGNANCY
Five factors alter thyroid function in pregnancy: (1) the transient Total hormone 8 μg/dL 0.14 μg/dL
increase in hCG during the first trimester, which weakly stimulates the Fraction of total hormone in the 0.02% 0.3%
TSH-R; (2) the estrogen-induced rise in TBG during the first trimester, unbound form
which is sustained during pregnancy; (3) alterations in the immune Unbound (free) hormone 21 × 10–12M 6 × 10–12M
system, leading to the onset, exacerbation, or amelioration of an under- Serum half-life 7d 2d
lying autoimmune thyroid disease; (4) increased thyroid hormone Fraction directly from the thyroid 100% 20%

CHAPTER 382 Thyroid Gland Physiology and Testing


metabolism by the placental type III deiodinase; and (5) increased Production rate, including peripheral 90 μg/d 32 μg/d
urinary iodide excretion, which can cause impaired thyroid hormone conversion
production in areas of marginal iodine sufficiency. Women with a Intracellular hormone fraction ~20% ~70%
precarious iodine intake (<50 μg/d) are most at risk of developing a
Relative metabolic potency 0.3 1
goiter during pregnancy or giving birth to an infant with a goiter and
hypothyroidism. The World Health Organization recommends a daily Receptor binding 10–10M 10–11M
iodine intake of 250 μg during pregnancy and lactation, and prenatal
vitamins should contain 150 μg per tablet. tightly bound than T4, the fraction of unbound T3 is greater than
The rise in circulating hCG levels during the first trimester is unbound T4, but there is less unbound T3 in the circulation because it
accompanied by a reciprocal fall in TSH that persists into the middle is produced in smaller amounts and cleared more rapidly than T4. The
of pregnancy. This reflects the weak binding of hCG, which is present unbound or “free” concentrations of the hormones are ~2 × 10–11 M for
at very high levels, to the TSH-R. Rare individuals have variant TSH-R T4 and ~6 × 10–12 M for T3, which roughly correspond to the thyroid
sequences that enhance hCG binding and TSH-R activation. hCG- hormone receptor–binding constants for these hormones (see below).
induced changes in thyroid function can result in transient gestational The unbound hormone is thought to be biologically available to tissues.
hyperthyroidism that may be associated with hyperemesis gravidarum, The homeostatic mechanisms that regulate the thyroid axis are directed
a condition characterized by severe nausea and vomiting and risk of toward maintenance of normal concentrations of unbound hormones.
volume depletion. However, since the hyperthyroidism is not causal,
antithyroid drugs are not indicated unless concomitant Graves’ disease Abnormalities of Thyroid Hormone–Binding Proteins
is suspected. Parenteral fluid replacement usually suffices until the A number of inherited and acquired abnormalities affect thyroid
condition resolves. hormone–binding proteins. X-linked TBG deficiency is associated
Normative values for most thyroid function tests differ during with very low levels of total T4 and T3. However, because unbound
pregnancy and, if available, trimester-specific reference ranges should hormone levels are normal, patients are euthyroid and TSH levels are
be used when diagnosing thyroid dysfunction during pregnancy. TSH normal. It is important to recognize this disorder to avoid efforts to
levels decrease at the end of the first trimester and then rise as gesta- normalize total T4 levels, because this leads to thyrotoxicosis and is
tion progresses so that the nonpregnant reference ranges can be used futile because of rapid hormone clearance in the absence of TBG. TBG
from mid-gestation to delivery. Total T4 and T3 levels are ~1.5× higher levels are elevated by estrogen, which increases sialylation and delays
throughout pregnancy, but the free T4, which is the same or slightly TBG clearance. Consequently, in women who are pregnant or taking
higher at the end of the 1st trimester, then progressively decreases so estrogen-containing contraceptives, elevated TBG increases total T4
that third-trimester values in healthy pregnancies are often below the and T3 levels; however, unbound T4 and T3 levels are normal. These fea-
nonpregnant lower reference cutoff. tures are part of the explanation for why women with hypothyroidism
During pregnancy, subclinical hypothyroidism occurs in 2% of require increased amounts of l-thyroxine replacement as TBG levels
women, but overt hypothyroidism is present in only 1 in 500. Prospec- are increased by pregnancy or estrogen treatment. Mutations in TBG,
tive randomized controlled trials have not shown a benefit for univer- TTR, and albumin may increase the binding affinity for T4 and/or T3
sal thyroid disease screening in pregnancy. Targeted TSH testing for and cause disorders known as euthyroid hyperthyroxinemia or familial
hypothyroidism is recommended for women planning a pregnancy if dysalbuminemic hyperthyroxinemia (FDH) (Table 382-2). These disor-
they have a strong family history of autoimmune thyroid disease, other ders result in increased total T4 and/or T3, but unbound hormone levels
autoimmune disorders (e.g., type 1 diabetes), infertility, prior preterm are normal. The familial nature of the disorders, and the fact that TSH
delivery or recurrent miscarriage, or signs or symptoms of thyroid levels are normal rather than suppressed, should suggest this diagnosis.
disease, or are older than 30 years. Thyroid hormone requirements Unbound hormone levels (ideally measured by dialysis) are normal
are increased by up to 45% during pregnancy in levothyroxine-treated in FDH. The diagnosis can be confirmed by using tests that measure
hypothyroid women. the affinities of radiolabeled hormone binding to specific transport
proteins or by performing DNA sequence analyses of the abnormal
■ THYROID HORMONE TRANSPORT transport protein genes.
AND METABOLISM Certain medications, such as salicylates and salsalate, can displace
thyroid hormones from circulating binding proteins. Although these
Serum-Binding Proteins T4 is secreted from the thyroid gland drugs transiently perturb the thyroid axis by increasing free thyroid
in about twentyfold excess over T3 (Table 382-1). Both hormones hormone levels, TSH is suppressed until a new steady state is reached,
are bound to plasma proteins, including thyroxine-binding globulin thereby restoring euthyroidism. Circulating factors associated with
(TBG), transthyretin (TTR, formerly known as thyroxine-binding pre- acute illness may also displace thyroid hormone from binding proteins
albumin [TBPA]), and albumin. The plasma-binding proteins increase (Chap. 384).
the pool of circulating hormone, delay hormone clearance, and may
modulate hormone delivery to selected tissue sites. The concentration Deiodinases T4 may be thought of as a precursor for the more
of TBG is relatively low (1–2 mg/dL), but because of its high affinity for potent T3. T4 is converted to T3 by the deiodinase enzymes (Fig. 382-1).
thyroid hormones (T4 > T3), it carries ~80% of the bound hormones. Type I deiodinase, which is located primarily in thyroid, liver, and kid-
Albumin has relatively low affinity for thyroid hormones but has a high neys, has a relatively low affinity for T4. Type II deiodinase has a higher
plasma concentration (~3.5 g/dL), and it binds up to 10% of T4 and affinity for T4 and is found primarily in the pituitary gland, brain,
30% of T3. TTR carries ~10% of T4 but little T3. brown fat, and thyroid gland. Expression of type II deiodinase allows it
When the effects of the various binding proteins are combined, to regulate T3 concentrations locally, a property that may be important
~99.98% of T4 and 99.7% of T3 are protein-bound. Because T3 is less in the context of levothyroxine (T4) replacement. Type II deiodinase
2930
TABLE 382-2 Conditions Associated with Euthyroid Hyperthyroxinemia with retinoic acid X receptors (RXRs)
(Chap. 377). The activated receptor
DISORDER CAUSE TRANSMISSION CHARACTERISTICS
can either stimulate gene transcription
Familial dysalbuminemic Albumin mutations, AD Increased T4 (e.g., myosin heavy chain α) or inhibit
hyperthyroxinemia (FDH) usually R218H Normal unbound T4 transcription (e.g., TSH β-subunit
Rarely increased T3 gene), depending on the nature of the
TBG       regulatory elements in the target gene.
Familial excess Increased TBG production XL Increased total T4, T3 Thyroid hormones (T3 and T4)
Normal unbound T4, T3 bind with similar affinities to TRα
Acquired excess Medications (estrogen), Acquired Increased total T4, T3 and TRβ. However, structural differ-
pregnancy, cirrhosis, Normal unbound T4, T3
ences in the ligand-binding domains
hepatitis provide the potential for developing
PART 12

Transthyretina       receptor-selective agonists or antag-


Excess Islet tumors Acquired Usually normal T4, T3
onists, and these are under investi-
gation. T3 is bound with 10–15 times
Mutations Increased affinity for T4 AD Increased total T4, T3
or T3
greater affinity than T4, which explains
Normal unbound T4, T3
Endocrinology and Metabolism

its increased potency. Although T4 is


Medications: propranolol, Decreased T4 → T3 Acquired Increased T4 produced in excess of T3, receptors are
ipodate, iopanoic acid, conversion Decreased T3 occupied mainly by T3, reflecting T4
amiodarone
Normal or increased TSH → T3 conversion by peripheral tissues,
Resistance to thyroid Thyroid hormone receptor AD Increased unbound T4, T3 T3 bioavailability in the plasma, and
hormone (RTH) β mutations Normal or increased TSH the greater affinity of receptors for T3.
Some patients clinically thyrotoxic After binding to TRs, thyroid hormone
induces conformational changes in the
a
Also known as thyroxine-binding prealbumin (TBPA). receptors that modify its interactions
Abbreviations: AD, autosomal dominant; TBG, thyroxine-binding globulin; TSH, thyroid-stimulating hormone; XL, X-linked. with accessory transcription factors.
Importantly, in the absence of thyroid
hormone binding, the aporeceptors
is also regulated by thyroid hormone; hypothyroidism induces the bind to co-repressor proteins that inhibit gene transcription. Hormone
enzyme, resulting in enhanced T4 → T3 conversion in tissues such binding dissociates the co-repressors and allows the recruitment of
as brain and pituitary. T4 → T3 conversion is impaired by fasting, co-activators that enhance transcription. The discovery of TR interac-
systemic illness or acute trauma, oral contrast agents, and a variety of tions with co-repressors explains the fact that TR silences gene expres-
medications (e.g., propylthiouracil, propranolol, amiodarone, gluco- sion in the absence of hormone binding. Consequently, hormone
corticoids). Type III deiodinase inactivates T4 and T3 and is the most deficiency has a profound effect on gene expression because it causes
important source of reverse T3 (rT3), including in the sick euthyroid gene repression as well as loss of hormone-induced stimulation. This
syndrome. This enzyme is expressed in the human placenta but is not concept has been corroborated by the finding that targeted deletion
active in healthy individuals. In the sick euthyroid syndrome, especially of the TR genes in mice has a less pronounced phenotypic effect than
with hypoperfusion, the type III deiodinase is activated in muscle and hormone deficiency.
liver. Massive hemangiomas and other tumors that express type III
deiodinase are a rare cause of consumptive hypothyroidism. Thyroid Hormone Resistance Resistance to thyroid hormone
(RTH) is an autosomal dominant disorder characterized by elevated
■ THYROID HORMONE ACTION thyroid hormone levels and inappropriately normal or elevated TSH.
Thyroid Hormone Transport Circulating thyroid hormones
enter cells by passive diffusion and via specific transporters such as
the monocarboxylate 8 transporter (MCT8), MCT10, and organic
anion-transporting polypeptide 1C1. Mutations in the MCT8 gene T3 Nucleus
have been identified in patients with X-linked psychomotor retardation
and thyroid function abnormalities (low T4, high T3, and high TSH). T4
After entering cells, thyroid hormones act primarily through nuclear T3 CoR
receptors, although they also have nongenomic actions through stim- 1
2
ulating mitochondrial enzymatic responses and may act directly on
blood vessels and the heart through integrin receptors. T3
CoA 3
Nuclear Thyroid Hormone Receptors Thyroid hormones bind RXR TR
with high affinity to nuclear TRs α and β. Both TRα and TRβ are Cytoplasm
expressed in most tissues, but their relative expression levels vary TRE Gene
among organs; TRα is particularly abundant in brain, kidneys, gonads,
4
muscle, and heart, whereas TRβ expression is relatively high in the
pituitary and liver. Both receptors are variably spliced to form unique Gene expression
isoforms. The TRβ2 isoform, which has a unique amino terminus, is
selectively expressed in the hypothalamus and pituitary, where it plays
a role in feedback control of the thyroid axis (see above). The TRα2
isoform contains a unique carboxy terminus that precludes thyroid FIGURE 382-4 Mechanism of thyroid hormone receptor action. The thyroid
hormone binding; it may function to inhibit the action of other TR hormone receptor (TR) and retinoid X receptor (RXR) form heterodimers that bind
isoforms. specifically to thyroid hormone response elements (TRE) in the promoter regions
The TRs contain a central DNA-binding domain and a C-terminal of target genes. In the absence of hormone, TR binds co-repressor (CoR) proteins
that silence gene expression. The numbers refer to a series of ordered reactions
ligand-binding domain. They bind to specific DNA sequences, termed that occur in response to thyroid hormone: (1) T4 or T3 enters the nucleus; (2) T3
thyroid response elements (TREs), in target genes (Fig. 382-4). The binding dissociates CoR from TR; (3) co-activators (CoA) are recruited to the T3-
receptors bind as homodimers or, more commonly, as heterodimers bound receptor; and (4) gene expression is altered.
Individuals with RTH do not, in general, exhibit signs and symptoms ■ LABORATORY EVALUATION 2931
that are typical of hypothyroidism because hormone resistance is par-
tial and is compensated by increased levels of thyroid hormone. The Measurement of Thyroid Hormones The enhanced sensitivity
clinical features of RTH can include goiter, attention deficit disorder, and specificity of TSH assays have greatly improved laboratory assess-
mild reduction in IQ, delayed skeletal maturation, tachycardia, and ment of thyroid function. Because TSH levels change dynamically in
impaired metabolic responses to thyroid hormone. response to alterations of T4 and T3, a logical approach to thyroid testing
Classical forms of RTH are caused by mutations in the TRβ gene. is to first determine whether TSH is suppressed, normal, or elevated.
These mutations, located in restricted regions of the ligand-binding With rare exceptions (see below), a normal TSH level excludes a primary
domain, cause loss of receptor function. However, because the mutant abnormality of thyroid function. This strategy depends on the use of
receptors retain the capacity to dimerize with RXRs, bind to DNA, immunochemiluminometric assays (ICMAs) for TSH that are sensitive
enough to discriminate between the lower limit of the reference interval

CHAPTER 382 Thyroid Gland Physiology and Testing


and recruit co-repressor proteins, they function as antagonists of the
remaining normal TRβ and TRα receptors. This property, referred to as and the suppressed values that occur with thyrotoxicosis. Extremely
“dominant negative” activity, explains the autosomal dominant mode sensitive assays can detect TSH levels ≤0.004 mIU/L, but, for practical
of transmission. The diagnosis is suspected when unbound thyroid purposes, assays sensitive to ≤0.1 mIU/L are sufficient. The widespread
hormone levels are increased without suppression of TSH. Similar availability of the TSH ICMA has rendered the TRH stimulation test
hormonal abnormalities are found in other affected family members, obsolete, because the failure of TSH to rise after an intravenous bolus
although the TRβ mutation arises de novo in ~20% of patients. DNA of 200–400 μg TRH has the same implications as a suppressed basal
sequence analysis of the TRβ gene provides a definitive diagnosis. RTH TSH measured by ICMA. Because the antibodies used in the ICMA are
must be distinguished from other causes of euthyroid hyperthyroxine- biotinylated, biotin supplements, including biotin in multivitamins, can
mia (e.g., FDH) and inappropriate secretion of TSH by TSH-secreting interfere with TSH measurement, resulting in falsely low TSH values
pituitary adenomas (Chap. 380). In most patients, no treatment is and falsely high T4 or T3 levels. Therefore, patients should be advised
indicated; the importance of making the diagnosis is to avoid inappro- to stop taking biotin for at least 2 days prior to thyroid function testing.
priate treatment of mistaken hyperthyroidism and to provide genetic The finding of an abnormal TSH level must be followed by measure-
counseling. ments of circulating thyroid hormone levels to confirm the diagnosis of
A distinct form of RTH is caused by mutations in the TRα gene. hyperthyroidism (suppressed TSH) or hypothyroidism (elevated TSH).
Affected patients have many clinical features of congenital hypothy- Automated immunoassays are widely available for serum total T4 and
roidism including growth retardation, skeletal dysplasia, and severe total T3. T4 and T3 are highly protein-bound, and numerous factors
constipation. In contrast to RTH caused by mutations in TRβ, thyroid (illness, medications, genetic factors) can influence protein binding. It
function tests include normal TSH, low or normal T4, and normal is useful, therefore, to measure the free, or unbound, hormone levels,
or elevated T3 levels. These distinct clinical and laboratory features which correspond to the biologically available hormone pool. Two direct
underscore the different tissue distribution and functional roles of TRβ methods are used to measure unbound thyroid hormones: (1) unbound
and TRα. Thyroxine treatment appears to alleviate some of the clinical thyroid hormone competition with radiolabeled T4 (or an analogue)
manifestations of patients with RTH caused by TRα mutations. for binding to a solid-phase antibody, and (2) physical separation of
the unbound hormone fraction by ultracentrifugation or equilibrium
■ PHYSICAL EXAMINATION dialysis. Although early unbound hormone immunoassays suffered
In addition to the examination of the thyroid itself, the physical exami- from artifacts, newer assays correlate well with the results of the more
nation should include a search for signs of abnormal thyroid function technically demanding and expensive physical separation methods. An
and the extrathyroidal features of ophthalmopathy and dermopathy indirect method that is now less commonly used to estimate unbound
(Chap. 384). Examination of the neck begins by inspecting the seated thyroid hormone levels is to calculate the free T3 or free T4 index from
patient from the front and side and noting any surgical scars, obvious the total T4 or T3 concentration and the thyroid hormone binding ratio
masses, or distended veins. The thyroid can be palpated with both (THBR). The latter is derived from the T3-resin uptake test, which deter-
hands from behind or while facing the patient, using the thumbs to mines the distribution of radiolabeled T3 between an absorbent resin
palpate each lobe. It is best to use a combination of these methods, and the unoccupied thyroid hormone–binding proteins in the sample.
especially when nodules are small. The patient’s neck should be slightly The binding of the labeled T3 to the resin is increased when there is
flexed to relax the neck muscles. After locating the cricoid cartilage, reduced unoccupied protein binding sites (e.g., TBG deficiency) or
the isthmus, which is attached to the lower one-third of the thyroid increased total thyroid hormone in the sample; it is decreased under the
lobes, can be identified and then followed laterally to locate either opposite circumstances. The product of THBR and total T3 or T4 pro-
lobe (normally, the right lobe is slightly larger than the left). By asking vides the free T3 or T4 index. In effect, the index corrects for anomalous
the patient to swallow sips of water, thyroid consistency can be better total hormone values caused by variations in hormone-protein binding.
appreciated as the gland moves beneath the examiner’s fingers. Total thyroid hormone levels are elevated when TBG is increased
Features to be noted include thyroid size, consistency, nodularity, due to estrogens (pregnancy, oral contraceptives, hormone therapy,
and any tenderness or fixation. An estimate of thyroid size (normally tamoxifen, selective estrogen receptor modulators, inflammatory liver
12–20 g) should be made, and a drawing is often the best way to record disease) and decreased when TBG binding is reduced (androgens,
findings. Ultrasound imaging provides the most accurate measurement nephrotic syndrome). Genetic disorders and acute illness can also
of thyroid volume and nodularity and is useful for assessment of goiter cause abnormalities in thyroid hormone–binding proteins, and various
prevalence in iodine-deficient regions. However, ultrasound is not drugs (phenytoin, carbamazepine, salicylates, and nonsteroidal anti-
indicated if the thyroid physical examination is normal. The size, loca- inflammatory drugs [NSAIDs]) can interfere with thyroid hormone
tion, and consistency of any nodules should also be defined. A bruit binding. Because unbound thyroid hormone levels are normal and the
or thrill over the gland, located over the insertion of the superior and patient is euthyroid in all of these circumstances, assays that measure
inferior thyroid arteries (supero- or inferolaterally), indicates increased unbound hormone are preferable to those for total thyroid hormones.
vascularity, associated with turbulent rather than laminar blood flow, For most purposes, the unbound T4 level is sufficient to confirm
as occurs in hyperthyroidism. If the lower borders of the thyroid lobes thyrotoxicosis, but 2–5% of patients have only an elevated T3 level
are not clearly felt, a goiter may be retrosternal. Large retrosternal (T3 toxicosis). Thus, unbound T3 levels should be measured in patients
goiters can cause venous distention over the neck and difficulty breath- with a suppressed TSH but normal unbound T4 levels.
ing, especially when the arms are raised (Pemberton’s sign). With any There are several clinical conditions in which the use of TSH as a
central mass above the thyroid, the tongue should be extended, as screening test may be misleading, particularly without simultaneous
thyroglossal cysts then move upward. The thyroid examination is not unbound T4 determinations. Any severe nonthyroidal illness can cause
complete without assessment for lymphadenopathy in the supraclavic- abnormal TSH levels. Although hypothyroidism is the most common
ular and cervical regions of the neck. cause of an elevated TSH level, rare causes include a TSH-secreting
2932 pituitary tumor (Chap. 380), thyroid hormone resistance, and assay is subnormal to determine if functioning thyroid nodules are pres-
artifact. Conversely, a suppressed TSH level, particularly <0.01 mIU/L, ent. Functioning or “hot” nodules are almost never malignant, and
usually indicates thyrotoxicosis. However, subnormal TSH levels fine-needle aspiration (FNA) biopsy is not indicated. The vast majority
between 0.01 and 0.1 mIU/L may be seen during the first trimester of of thyroid nodules do not produce thyroid hormone (“cold” nodules),
pregnancy (due to hCG secretion), after treatment of hyperthyroidism and these are more likely to be malignant (~5–10%). Whole-body and
(because TSH can remain suppressed for several months), and in thyroid scanning is also used in the treatment and, now less frequently,
response to certain medications (e.g., high doses of glucocorticoids in the surveillance of thyroid cancer. After thyroidectomy for thyroid
or dopamine). TSH levels measured by immunoassay may also be cancer, the TSH level is raised by either using a thyroid hormone with-
suppressed in patients ingesting biotin supplements <18 h prior to a drawal protocol or recombinant human TSH injection (Chap. 385).
blood draw because the TSH capture antibodies are biotinylated and Administration of either 131I or 123I (in higher activities than used to
the exogenous biotin can interfere with the subsequent streptavidin image the thyroid gland alone) allows whole-body scanning (WBS)
capture. Importantly, secondary hypothyroidism, caused by hypo- to detect the thyroid remnant. WBS imaging is also performed after
PART 12

thalamic-pituitary disease, is associated with a variable (low to high- therapeutic administration of 131I, which confirms remnant ablation
normal) TSH level, which is inappropriate for the low T4 level. Thus, TSH and may reveal iodine-avid metastases.
should not be used as an isolated laboratory test to assess thyroid function Thyroid Ultrasound Ultrasonography is the most valuable tool
in patients with suspected or known hypothalamic or pituitary disease. for the diagnosis and evaluation of patients with nodular thyroid
Endocrinology and Metabolism

Tests for the end-organ effects of thyroid hormone excess or deple- disease (Chap. 385). Evidence-based guidelines recommend thyroid
tion, such as estimation of basal metabolic rate, tendon reflex relax- ultrasonography for all patients suspected of having thyroid nodules
ation rates, or serum cholesterol, are relatively insensitive and are not by either physical examination or another imaging study. Using 10- to
useful as clinical determinants of thyroid function. 12-MHz linear transducers, resolution and image quality are excellent,
Tests to Determine the Etiology of Thyroid Dysfunction allowing the characterization of nodules and cysts >3 mm. Sonographic
Autoimmune thyroid disease is detected most easily by measuring patterns that combine suspicious sonographic features are highly sug-
circulating antibodies against TPO and Tg. Because antibodies to Tg gestive of malignancy (e.g., hypoechoic solid nodules with infiltrative
alone are less common, it is reasonable to measure only TPO anti- borders and microcalcifications, >90% cancer risk), whereas other
bodies. About 5–15% of euthyroid women and up to 2% of euthyroid patterns correlate with a lower likelihood of cancer (isoechoic solid
men have thyroid antibodies; such individuals are at increased risk of nodules, 5–10% cancer risk). Some patterns suggest benignity (e.g.,
developing thyroid dysfunction. Almost all patients with autoimmune spongiform nodules, defined as those with multiple small internal cys-
hypothyroidism, and up to 80% of those with Graves’ disease, have tic areas, or simple cysts, <3% cancer risk) (see Chap. 385, Fig. 385-2).
TPO antibodies, usually at high levels. These patterns have been incorporated into validated risk stratification
TSIs are antibodies that stimulate the TSH-R in Graves’ disease. systems (RSSs) for sonographic imaging of thyroid nodules (American
They are most commonly measured by commercially available tracer College of Radiology [ACR] Thyroid Imaging Reporting and Data
displacement assays called TRAb (TSH receptor antibody) with the System [TI-RADS], American Thyroid Association, European Thyroid
assumption that elevated levels in the setting of clinical hyperthyroid- Association [EU-TIRADS] and others) (see Chap. 385, Fig. 385-1).
ism reflect stimulatory effects on the TSH receptor. A bioassay is less These systems are relatively concordant in the classification of thyroid
commonly used. Remission rates in patients with Graves’ disease after nodules; they differ in size cutoff recommendations for FNA. Not sur-
antithyroid drug cessation are higher with disappearance rather than prisingly, the RSSs with lower size cutoffs have higher sensitivity and
persistence of TRAb. Furthermore, the TRAb assay is used to predict lower specificity for thyroid cancer diagnosis than those with higher
both fetal and neonatal thyrotoxicosis caused by transplacental passage cutoffs. Nevertheless, all have been shown to reduce unnecessary FNAs
of high maternal levels of TRAb or TSI (>3× upper limit of normal) in by at least 45%, in part due to the recommendation not to perform
the last trimester of pregnancy. FNA of spongiform nodules.
Serum Tg levels are increased in all types of thyrotoxicosis except In addition to evaluating thyroid nodules, ultrasound is useful for
thyrotoxicosis factitia caused by self-administration of thyroid hor- monitoring nodule size and for the aspiration of nodules or cystic
mone. Tg levels are particularly increased in thyroiditis, reflecting lesions. Ultrasound-guided FNA biopsy of thyroid lesions lowers the
thyroid tissue destruction and release of Tg. The main role for Tg rate of inadequate sampling and decreases sample error, thereby reduc-
measurement, however, is in the follow-up of thyroid cancer patients. ing both the nondiagnostic and false-negative rates of FNA cytology.
After total thyroidectomy and radioablation, Tg levels should be Ultrasonography of the central and lateral cervical lymph node com-
<0.2 ng/mL in the absence of anti-Tg antibodies; measurable levels partments is indispensable in the evaluation of thyroid cancer patients,
indicate incomplete ablation or recurrent cancer. preoperatively and during follow-up. In addition, the ACR recom-
mends a survey of the cervical lymph nodes as part of every diagnostic
Radioiodine Uptake and Thyroid Scanning The thyroid gland thyroid sonographic examination.
selectively transports radioisotopes of iodine (123I, 125I, 131I) and 99mTc
pertechnetate, allowing thyroid imaging and quantitation of radioac- ■ FURTHER READING
tive tracer fractional uptake. Alexander EK et al: 2017 Guidelines of the American Thyroid Asso-
Nuclear imaging of Graves’ disease is characterized by an enlarged ciation for the diagnosis and management of thyroid disease during
gland and increased tracer uptake that is distributed homogeneously. pregnancy and postpartum. Thyroid 27:315, 2017.
Toxic adenomas appear as focal areas of increased uptake, with Braun D, Schweizer U: Thyroid hormone transport and transport-
suppressed tracer uptake in the remainder of the gland (reflecting ers. Vitam Horm 106:19, 2018.
suppressed TSH). In toxic MNG, the gland is enlarged—often with dis- Ortiga-Cavalho TM et al: Thyroid hormone receptors and resistance
torted architecture—and there are multiple areas of relatively increased to thyroid hormone disorders. Nat Rev Endocrinol 10:582, 2014.
(functioning nodules) or decreased tracer uptake (suppressed thyroid Rugge JB et al: Screening and treatment of thyroid dysfunction: An
parenchyma or nonfunctioning nodules). Subacute, viral, and postpar- evidence review for the U.S. Preventive Services Task Force. Ann
tum thyroiditis are associated with very low uptake because of follicular Intern Med 162:35, 2015.
cell damage and TSH suppression. Thyrotoxicosis factitia is also associ- Stoupa A et al: Update of thyroid developmental genes. Endocrinol
ated with low uptake because exogenous hormone suppresses TSH. In Metab Clin North Am 45:243, 2016.
addition, if there is excessive circulating exogenous iodine (e.g., from Tessler FN et al: ACR Thyroid Imaging Reporting and Data System
dietary sources of iodinated contrast dye), the radionuclide uptake is (TI-RADS): White paper of the ACR TI-RADS Committee. J Am
low even in the presence of increased thyroid hormone production. Coll Radiol 14:587 2017.
Thyroid scintigraphy is not used in the routine evaluation of patients Zimmermann MB, Boelaert K: Iodine deficiency and thyroid disor-
with thyroid nodules but should be performed if the serum TSH level ders. Lancet Diabetes Endocrinol 3:286, 2015.

You might also like