Thyroglobulin

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Thyroid Gland

3
CHAPTER

In the adult human, normal operation of a wide below the cricoid cartilage (Figure 3.1). The two
variety of physiological processes affecting vir- large lateral lobes that comprise the bulk of the
tually every organ system requires appropri- gland lie on either side of the trachea and are
ate amounts of thyroid hormone. Governing all connected by a thin isthmus. A third structure,
these processes, thyroid hormone acts as a mod- the pyramidal lobe, which may be a remnant of
ulator, or gain control, rather than an all-or-none the embryonic thyroglossal duct, is sometimes
signal that turns the process on or off. In the also seen as a finger-like projection extending
immature individual, thyroid hormone plays an headward from the isthmus. The thyroid gland
indispensable role in growth and development. in the normal human being weighs about 20 g
Its presence in optimal amounts at a critical time but is capable of enormous growth, sometimes
is an absolute requirement for normal develop- achieving a weight of several hundred grams
ment of the nervous system. In its role in growth when stimulated intensely over a long period
and development too, its presence seems to be of time. Such enlargement of the thyroid gland,
required for the normal unfolding of processes which may be grossly obvious, is called a goiter,
whose course it modulates but does not initiate. and is one of the most common manifestations
Because thyroid hormone affects virtually every of thyroid disease.
system in the body in this way, it is difficult to The thyroid gland receives its blood supply
give a simple, concise answer to the naive but through the inferior and superior thyroid arteries,
profound question: What does thyroid hormone which arise from the external carotid and subcla-
do? The response of most endocrinologists would vian arteries. Relative to its weight, the thyroid
be couched in terms of consequences of hormone gland receives a greater flow of blood than most
excess or deficiency. Indeed, deranged function other tissues of the body. Venous drainage is
of the thyroid gland is among the most prevalent through the paired superior, middle, and inferior
of endocrine diseases and may affect as many as thyroid veins into the internal jugular and innom-
4 to 5% of the population in the United States. inate veins. The gland is also endowed with a rich
In regions of the world where the trace element lymphatic system that may play an important
iodine is scarce, the incidence of deranged thy- role in delivery of hormone to the general circula-
roid function may be even higher. tion. The thyroid gland also has an abundant sup-
ply of sympathetic and parasympathetic nerves.
Some studies suggest that sympathetic stimula-
MORPHOLOGY tion or infusion of epinephrine or norepinephrine
may increase secretion of thyroid hormone,
The human thyroid gland is located at the base but it is probably only of minor importance in
of the neck and wraps around the trachea just the overall regulation of thyroid function.

43

CH003-P373975.indd 43 6/18/2008 4:30:52 PM


Basic Medical Endocrinology

The functional unit of the thyroid gland is the folli- functional state of one lobule may differ widely from that
cle, which is composed of epithelial cells arranged as hol- of an adjacent lobule. Secretory cells of the thyroid gland
low vesicles of various shapes ranging in size from 0.02 to are derived embryologically and phylogenetically from two
0.3 mm in diameter; it is filled with a glycoprotein colloid sources. Follicular cells, which produce the classical thyroid
called thyroglobulin (Figure 3.2). There are about three mil- hormones, thyroxine and triiodothyronine, arise from endo-
lion follicles in the adult human thyroid gland. Epithelial derm of the primitive pharynx. Parafollicular, or C cells, are
cells lining each follicle may be cuboidal or columnar, located between the follicles and produce the polypeptide
depending on their functional state, with the height of hormone calcitonin, which is discussed in Chapter 8. These
the epithelium being greatest when its activity is highest. cells arise from the ultimobranchial body associated with
Each follicle is surrounded by a dense capillary network the fifth branchial arch.
separated from epithelial cells by a well-defined basement
membrane. Groups of densely packed follicles are bound
together by connective tissue septa to form lobules that
receive their blood supply from a single small artery. The THYROID HORMONES

The thyroid hormones are α-amino acid derivatives of tyro-


sine (Figure 3.3). The thyronine nucleus consists of two
benzene rings in ether linkage, with an alanine side chain
Thyroid cartilage in the para position on the inner or tyrosyl ring and a
Cricothyroid ligament hydroxyl group in the para position in the outer or phenolic
ring. Thyroxine was the first thyroid hormone to be isolated
Common carotid artery
Sternocleidomastoid and characterized. Its name derives from thyroid oxyindole,
muscle which describes the chemical structure erroneously assigned
Cricothyroid muscle to it when it was initially isolated in 1914.
Cricoid cartilage Triiodothyronine a considerably less abundant, but far
Thyroid gland more potent hormone than thyroxine in most assay systems,
Cupula of pleura was not discovered until 1953. Both hormone molecules are
Trachea exceptionally rich in iodine, which comprises more than half
of their molecular weight. Thyroxine contains four atoms of
FIGURE 3.1 Gross anatomy of the thyroid gland. (From Netter, F.H.
(1989) Atlas of Human Anatomy, 2nd Ed. Novartis Summit New Jersey, Icon
iodine and is abbreviated as T4; triiodothyronine, which has
Learning Systems, LLC, a subsidiary of MediMedia, Inc, Reprinted with three atoms of iodine, is abbreviated as T3.
permission from Icon Learning Systems, LLC, illustrated by Frank H. Netter,
MD. All rights reserved.)

I I
HO- -O- -C-C-COOH
NH2
I I
Thyroxine
3,5,3’,5’-tetraiodothyronine (T4)

I I
HO- -O- -C-C-COOH
NH2
I
Colloid 3,5,3’-triiodothyronine (T3)

I I
FIGURE 3.2 Low power photomicrograph of a rat thyroid gland. The HO- -O- -C-C-COOH
thyroid follicles shown in cross section are filled with uniformly staining NH2
colloid and are each composed of a single layer of epithelial cells (red arrow). I
Note that the follicles are not uniform in size or shape. The white arrow 3,3’,5’-triiodothyronine
points to a parafollicular cell and the green arrow points to a connective Reverse T3 (rT3)
tissue septum separating two lobules. (Courtesy of Dr. William Cooke,
Department of Cell Biology, University of Massachusetts Medical School.) FIGURE 3.3 Thyroid hormones.

44

CH003-P373975.indd 44 6/18/2008 4:30:54 PM


Thyroid Gland 3
Biosynthesis symporter, can block the uptake of iodide. This property can
be exploited for diagnostic or therapeutic purposes.
Several aspects of the production of thyroid hormone are
unusual: Thyroglobulin synthesis
Thyroglobulin is the other major component needed for syn-
1 Thyroid hormones contain large amounts of iodine.
thesis of thyroxine and triiodothyronine. Thyroglobulin is
Biosynthesis of active hormone requires adequate
the matrix for thyroid hormone synthesis and is the form in
amounts of this scarce element. This need is met by an
which hormone is stored in the gland. It is a large glycopro-
efficient energy-dependent transport mechanism that
tein that forms a stable dimer with a molecular mass of about
allows thyroid cells to take up and concentrate iodide.
660,000 daltons. Like other secretory proteins, thyroglobulin
The thyroid gland is also the principal site of storage of
is synthesized on ribosomes, glycosylated in the cisternae of
this rare dietary constituent.
the endoplasmic reticulum, translocated to the Golgi appa-
2 Thyroid hormones are partially synthesized extracellularly ratus, and packaged in secretory vesicles, which discharge it
at the luminal surface of follicular cells and stored in an from the apical surface into the lumen. Because thyroglobulin
extracellular compartment, the follicular lumen.
3 The hormone therefore is doubly secreted, in that the
DIT DIT
precursor molecule, thyroglobulin, is released from apical Follicular
DIT
surfaces of follicular cells into the follicular lumen, only DIT MIT Lumen
DIT MIT
DIT
to be taken up again by follicular cells and degraded TG H2O2 T4 T4
to release T4 and T3, which are then secreted into the
blood from the basal surfaces of follicular cells.
P
TO TPO
4 Thyroxine, the major secretory product, is not the
DIT
biologically active form of the hormone, but must be
MIT
transformed to T3 at extrathyroidal sites.
T4
TG
Biosynthesis of thyroid hormones can be considered Vescicle Endosome
as the sum of several discrete processes (Figure 3.4), all of
which depend upon the products of three genes that are
expressed predominantly, if not exclusively, in thyroid folli-
E.R.
cle cells: the sodium iodide symporter (NIS), thyroglobulin, Golgi MIT
PF
and thyroid peroxidase. DIT T4
MIT
Nucleus DIT
Iodine trapping Lysosome

Under normal circumstances iodide is about 25 to 50 times T4


Iⴚ ITDI
more concentrated in the cytosol of thyroid follicular cells
3Na⫹
than in blood plasma, and during periods of active stimula- NIS ATP
tion, it may be as high as 250 times that of plasma. Iodine T4
Iⴚ 2Na⫹ 2K⫹
is accumulated against a steep concentration gradient by the
action of an electrogenic “iodide pump” located in the baso- FIGURE 3.4 Thyroid hormone biosynthesis and secretion. Iodide
lateral membranes. The pump is actually a sodium iodide (I⫺) is transported into the thyroid follicular cell by the sodium/iodide
symporter (NIS) in the basal membrane and diffuses passively into the
symporter that couples the transfer of two ions of sodium lumen through the iodide channel called pendrin (P). Thyroglobulin
with each ion of iodide. Iodide is thus transported against its (TG) is synthesized by microsomes on the rough endoplasmic reticulum
concentration driven by the favorable electrochemical gradi- (ER), processed in the cisternae of the ER and the Golgi where it is
packaged into secretory granules and released into the follicular lumen.
ent for sodium. Energy is expended by the sodium potas- In the presence of hydrogen peroxide (H2O2) produced in the luminal
sium ATPase (the sodium pump), which then extrudes three membrane by thyroid oxidase (TO), the thyroid peroxidase (TPO) oxidizes
ions of sodium in exchange for two ions of potassium to iodide, which reacts with tyrosine residues in TG in the follicular lumen to
produce monoiodotyrosyl (MIT) and diiodotyrosyl (DIT) within the peptide
maintain the electrochemical gradient for sodium. Outward
chain. The TPO reaction also catalyzes the coupling of iodotyrosines to
diffusion of potassium maintains the membrane potential. form thyroxine (T4) and some triiodothyronine (T3, not shown) within
Like other transporters, the sodium iodide symporter has TG. Secretion of T4 begins with phagocytosis of TG, fusion of TG-laden
a finite capacity and can be saturated. Consequently, other endosomes with lysosomes and proteolytic digestion to peptide fragments
(PF), MIT, DIT, and T4. T4 is released from the cell at the basal membrane.
anions, such as perchlorate, pertechnetate, and thiocyanate, MIT and DIT are deiodinated by iodotyrosine deiodinase (ITDI) and
which compete for binding sites on the sodium iodide recycled.

45

CH003-P373975.indd 45 6/18/2008 4:30:58 PM


Basic Medical Endocrinology

secretion into the lumen is coupled with its synthesis, follicu- with the rest remaining as MIT and DIT. After coupling is
lar cells do not have the extensive accumulation of secretory complete, each thyroglobulin molecule normally contains
granules characteristic of protein-secreting cells. Iodination to one to three molecules of T4. T3 is considerably scarcer, with
form mature thyroglobulin does not take place until after the one molecule being present in only 20 to 30% of thyroglob-
thyroglobulin is discharged into the lumen. ulin molecules. T3 may be formed either by deiodination of
T4 or by coupling one residue of DIT to one of MIT.
Incorporation of iodine Exactly how coupling is achieved is not known. One
possible mechanism involves joining two iodotyrosine resi-
Iodide that enters at the basolateral surfaces of the fol-
dues that are in close proximity to each other on either
licular cell must be delivered to the follicular lumen where
two separate strands of thyroglobulin or adjacent folds of
hormone biosynthesis takes place. Iodide diffuses through-
the same strand. Free radicals formed by the action of thy-
out the follicular cell and exits from the apical membrane
roperoxidase react to form the ether linkage at the heart of
by way of a sodium-independent iodide transporter called
the thyronine nucleus, leaving behind in one of the peptide
pendrin, which is also expressed in brain and kidney. In
chains the serine or alanine residue that was once attached
order for iodide to be incorporated into tyrosine residues
to the phenyl group that now comprises the outer ring of
in thyroglobulin, it must first be converted to some higher
T4 (Figure 3.5). An alternative mechanism involves cou-
oxidized state. This step is catalyzed by the thyroid-specific
pling a free diiodophenylpyruvate (deaminated DIT) with a
thyroperoxidase in the presence of hydrogen peroxide, whose
molecule of DIT in peptide linkage within the thyroglobu-
formation may be rate-limiting. Hydrogen peroxide is
lin molecule by a similar reaction sequence. Regardless of
generated by the catalytic activity of a calcium-dependent
which model proves correct, it is sufficient to recognize the
NADPH oxidase that is present in the brush border.
central importance of thyroperoxidase for formation of the
Thyroperoxidase is the key enzyme in thyroid hormone for-
thyronine nucleus as well as iodination of tyrosine residues.
mation and is thought to catalyze the iodination and cou-
In addition, the mature hormone is formed while in peptide
pling reactions described next in addition to the activation
linkage within the thyroglobulin molecule, and remains a
of iodide. Thyroperoxidase spans the brush border mem-
part of that large storage molecule until lysosomal enzymes
brane on the apical surface of follicular cells and is oriented
set it free during the secretory process.
with its catalytic domain facing the follicular lumen.
Addition of iodine molecules to tyrosine residues in
thyroglobulin is called organification. Thyroglobulin is iodin- Hormone storage
ated at the apical surface of follicular cells as it is extruded
into the follicular lumen. Iodide acceptor sites in thyroglob- The thyroid is unique among endocrine glands in that it stores
ulin are in sufficient excess over the availability of iodide that its product extracellularly as large precursor molecules in fol-
no free iodide accumulates in the follicular lumen. Although licular lumens. In the normal individual, approximately 30%
posttranslational conformational changes orchestrated by of the mass of the thyroid gland is thyroglobulin, which cor-
endoplasmic reticular proteins organize the configuration of responds to about two to three months’ supply of hormone.
thyroglobulin to increase its ability to be iodinated, iodina- Mature thyroglobulin is a high molecular weight molecule,
tion and hormone formation do not appear to be particularly probably a dimer of the thyroglobulin precursor peptide, and
efficient. Tyrosine is not especially abundant in thyroglobu- contains about 10% carbohydrate and about 0.5% iodine. The
lin and comprises only about 1 in 40 residues of the peptide tyrosine residues that are situated just a few amino acids away
chain. Only about 10% of the 132 tyrosine residues in each from the C and N terminals are the principal sites of iodothy-
thyroglobulin dimer appear to be in positions favorable for ronine formation. MIT and DIT at other sites in thyroglobu-
iodination. The initial products formed are monoiodotyrosine lin comprise an important reservoir for iodine and constitute
(MIT) and diiodotyrosine (DIT), and they remain in peptide about 90% of the total pool of iodine in the body.
linkage within the thyroglobulin molecules. Normally more
DIT is formed than MIT, but when iodine is scarce there is
less iodination and the ratio of MIT to DIT is reversed. Secretion

Thyroglobulin stored within follicular lumens is separated


Coupling from extracellular fluid and the capillary endothelium by a
The final stage of thyroxine biosynthesis is the coupling of virtually impenetrable layer of follicular cells. In order for
two molecules of DIT to form T4 within the peptide chain. secretion to occur, thyroglobulin must be brought back into
This reaction also is catalyzed by thyroperoxidase. Only follicular cells by a process of endocytosis. Upon acute stim-
about 20% of iodinated tyrosine residues undergo coupling, ulation with TSH, long strands of protoplasm (pseudopodia)

46

CH003-P373975.indd 46 6/18/2008 4:30:59 PM


Thyroid Gland 3
Thyroglobulin A

NH CO
COOH
CH
I
CH2
HO I
I
HO
CH2
I
CH
CO NH NH2

TPO H2O2

NH CO
COOH
CH
I
CH2
HO * I
I
*O
CH2 B
I
CH
CO NH NH2

Rearrangement

NH CO
COOH
CH
CH3
I I
HO O
CH2
I I
CH
CO NH NH2

FIGURE 3.5 Hypothetical coupling scheme for intramolecular formation


of T4 based on model reaction with purified thyroid peroxidase. (Modified
from Taurog, A. (2000) In Braverman, L.E. and Utiger, R.D., eds. Werner FIGURE 3.6 Scanning electron micrographs of the luminal microvilli
and Ingbar’s The Thyroid, 8th ed., p. 71. Lippincott Williams and Wilkins, of dog thyroid follicular cells. A. TSH secretion suppressed by feeding
Philadelphia.) thyroid hormone; magnification 36,000x. B. At 1 hr after TSH; magnification
16,500x. (From Balasse, P.D., Rodesch, F.R., Neve, P.E. et al. (1972)
Observations en microscopie a balayage de la surface apicale y de
reach out from the apical surfaces of follicular cells to sur- cellules folliculares thyroidiennes chez le chien. C. R. Acad. Sci. [D] (Paris),
274: 2332.)
round chunks of thyroglobulin, which are taken up in endo-
cytic vesicles (Figure 3.6). In chronic situations uptake is
probably less dramatic than shown in Figure 3.6, but nev- are released into the bloodstream, in a ratio of about 20:1,
ertheless requires an ongoing endocytic process. The endo- perhaps by a process of simple diffusion down a concentra-
cytic vesicles migrate toward the basal portion of the cells tion gradient.
and fuse with lysosomes, which simultaneously migrate Monoiodotyrosine and DIT cannot be utilized for
from the basal to the apical region of the cells to meet synthesis of thyroglobulin and are rapidly deiodinated by
the incoming endocytic vesicles. As fused lyso-endosomes a specific microsomal deiodinase. Virtually all the iodide
migrate toward the basement membrane, thyroglobulin released from iodotyrosines is recycled into thyroglobu-
is degraded to peptide fragments and free amino acids, lin. Deiodination of iodotyrosine provides about twice as
including T4, T3, MIT, and DIT. Of these, only T4 and T3 much iodide for hormone synthesis as the iodide pump and

47

CH003-P373975.indd 47 6/18/2008 4:30:59 PM


Basic Medical Endocrinology

is therefore of great significance in hormone biosynthesis. Binding of TSH to the receptor results in activation
Patients who are genetically deficient in thyroidal tyrosine of both adenylyl cyclase through Gαs and phospholipase C
deiodinase readily suffer symptoms of iodine deficiency and through Gαq and leads to increases in both the cyclic AMP
excrete MIT and DIT in their urine. Normally, virtually no and diacylglycerol/IP3 second messenger pathways (see
MIT or DIT escape from the gland. Chapter 1). Activation of the cyclic AMP pathway appears
Synthesis of thyroglobulin and its export in vesicles to be the more important transduction mechanism, as all
into the follicular lumen is an ongoing process that takes the known effects of TSH can be duplicated by cyclic AMP.
place simultaneously with uptake of thyroglobulin in other Because TSH increases cyclic AMP production at much
vesicles moving in the opposite direction. These opposite lower concentrations than are needed to increase phospho-
processes, involving vesicles laden with thyroglobulin mov- lipid turnover, it is likely that IP3 and DAG are redundant
ing into and out of the cells, are somehow regulated so that mediators that reinforce the effects of cyclic AMP at times
under normal circumstances thyroglobulin neither accu- of intense stimulation, but it is also possible that these second
mulates in follicular cells nor are the lumens depleted. The messengers signal some unique responses. Increased turnover
physiological mechanisms for such traffic control are not yet of phospholipid is associated with release of arachidonic acid
understood. and the consequent increased production of prostaglandins
that also follows TSH stimulation of the thyroid.
In addition to regulating all aspects of hormone bio-
synthesis and secretion, TSH increases blood flow to the
CONTROL OF THYROID FUNCTION thyroid. With prolonged stimulation TSH also increases
the height of the follicular epithelium (hypertrophy), and
Effects of thyroid-stimulating hormone can stimulate division of follicular cells (hyperplasia).
Stimulation of thyroid follicular cells by TSH is a good
Although the thyroid gland can carry out all the steps of example of a pleiotropic effect of a hormone in which there
hormone biosynthesis, storage, and secretion in the absence are multiple separate but complementary actions that sum-
of any external signals, autonomous function is far too slug- mate to produce an overall response. Each step of hormone
gish to meet bodily needs for thyroid hormone. The princi- biosynthesis, storage, and secretion appears to be directly
pal regulator of thyroid function is the thyroid-stimulating stimulated by a cyclic AMP-dependent process that is
hormone (TSH), which is secreted by thyrotropes in the accelerated independently of the preceding or follow-
pituitary gland (see Chapter 2). It may be recalled that TSH ing steps in the pathway. Thus even when increased iodide
consists of two glycosylated peptide subunits including the transport is blocked with a drug that specifically affects the
same α-subunit that is also found in FSH, and LH. The iodide pump, TSH nevertheless accelerates the remaining
β-subunit is the part of the hormone that confers thyroid- steps in the synthetic and secretory process. Similarly, when
specific stimulating activity, but free β-subunits are inactive, iodination of tyrosine is blocked by a drug specific for the
and stimulate the thyroid only when linked to α-subunits organification process, TSH still stimulates iodide transport
in a complex three-dimensional configuration. and thyroglobulin synthesis.
Thyroid-stimulating hormone binds to a single class of Most of the responses to TSH depend upon activation
heptiahelical G-protein-coupled receptors (see Chapter 1) of protein kinase A and the resultant phosphorylation of
in the basolateral surface membranes of thyroid follicular proteins including transcription factors such as CREB (cyclic
cells. The TSH receptor is the product of a single gene, but AMP response element binding protein; see Chapter 1).
it is comprised of two subunits held together by a disulfide TSH increases expression of genes for the sodium iodide
bond. It appears that after the molecule has been prop- symporter, thyroglobulin, thyroid oxidase, and thyroid
erly folded and its disulfide bonds formed, a loop of about peroxidase. These effects are exerted through cooperative
50 amino acids is excised proteolytically from the extracellular interactions of TSH activated nuclear proteins with thyroid-
portion of the receptor. The α-subunit includes about 300 specific transcription factors whose expression is also
residues at the amino terminus and contains most of the enhanced by TSH. TSH appears to increase blood flow by
TSH binding surfaces. The β-subunit contains the seven activating the gene for the inducible form of nitric oxide
membrane-spanning alpha helices and the short carboxyl synthase, which increases production of the potent vasodila-
terminal tail in the cytoplasm. Reduction of the disulfide tor, nitric oxide, and by inducing expression of paracrine fac-
bond may lead to release of the α-subunit into the extra- tors that promote capillary growth (angiogenesis). Precisely
cellular fluid, and may have important implications for the how TSH increases thyroid growth is not understood, but it
development of antibodies to the TSH receptor and thyroid is apparent that synthesis and secretion of a variety of local
disease (see later). growth factors is induced.

48

CH003-P373975.indd 48 6/18/2008 4:31:02 PM


Thyroid Gland 3
Effects of the thyroid-stimulating follicular cell to the lumen. Thus thyroglobulin may have
immunoglobulins significant effects in regulating its own synthesis and may
temper the stimulatory effects of TSH, which remains the
Overproduction of thyroid hormone, hyperthyroidism, which primary and most important regulator of thyroid function.
is also known as Graves’ disease, usually is accompanied
by extremely low concentrations of TSH in blood plasma,
yet the thyroid gland gives every indication of being under
intense stimulation. This paradox was resolved when it was
found that blood plasma of affected individuals contains a THYROID HORMONES IN BLOOD
substance that stimulates the thyroid gland to produce and
secrete thyroid hormone. This substance is an immunoglob- More than 99.8% of thyroid hormone circulating in blood
ulin secreted by lymphocytes and is almost certainly an is bound firmly to three plasma proteins, thyroxine-binding
antibody to the TSH receptor. Thyroid-stimulating immu- globulin (TBG), transthyretin (TTR), and albumin. Of these,
noglobulin (TSI) can be found in the serum of virtually all TBG is quantitatively the most important and accounts for
patients with Graves’ disease, suggesting an autoimmune more than 70% of the total protein-bound hormone (both
etiology to this disorder. T4 and T3). About 10 to 15% of circulating T4 and 10% of
It is interesting to note that when reacting with the circulating T3 are bound to TTR and nearly equal amounts
TSH receptor, antibodies trigger the same sequence of are bound to albumin. TBG carries the bulk of the hormone
responses that are produced when TSH interacts with the even though its concentration in plasma is only 6% that of
receptor. This fact indicates that all the information needed TTR and less than 0.1% that of albumin because its affinity
to produce the characteristic cellular response to TSH for both T4 and T3 is so much higher than that of the other
resides in the receptor rather than the hormone. The role of proteins. All three thyroid hormone-binding proteins bind
the hormone therefore must be limited to activation of the T4 at least 10 times more avidly than T3, so that the free,
receptor. Similar effects also have been seen with antibodies unbound concentrations of T3 and T4 are nearly equal. All
to receptors for other hormones. three binding proteins are large enough to escape filtration
by the kidney so that virtually no thyroid hormone appears
in urine, and little crosses the capillary endothelium. The
Autoregulation of thyroid hormone less than 1% of hormone present in free solution is in equi-
synthesis librium with bound hormone and is the only hormone that
can escape from capillaries to produce biological activity or
Although production of thyroid hormones is severely be acted on by tissue enzymes.
impaired when too little iodide is available, iodide uptake The total amount of thyroid hormone bound to plasma
and hormone biosynthesis are blocked temporarily when the proteins represents about three times as much hormone as
concentration of iodide in blood plasma becomes too high. is secreted and degraded in the course of a single day. Thus
This effect of iodide has been exploited clinically to produce plasma proteins provide a substantial reservoir of extrathyroi-
short-term suppression of thyroid hormone secretion. This dal hormone. We therefore should not expect acute increases
inhibitory effect of iodide apparently depends on its being or decreases in the rate of secretion of thyroid hormones to
incorporated into some organic molecule and is thought bring about large or rapid changes in circulating concentra-
to represent an autoregulatory phenomenon that protects tions of thyroid hormones. For example, if the rate of thy-
against overproduction of thyroxine. Blockade of thyroid roxine secretion were doubled for one day, we could expect
hormone production is short-lived, and the gland eventually its concentration in blood to increase by no more than 30%,
“escapes” from the inhibitory effects of iodide by mechanisms even if there were no accompanying increase in the rate of
that include downregulation of the sodium iodide symporter. hormone degradation. A tenfold increase in the rate of secre-
Biosynthetic activity of the thyroid gland may also tion lasting for 60 minutes would give only a 12% increase in
be regulated by thyroglobulin that accumulates in the fol- total circulating thyroxine, and if thyroxine secretion stopped
licular lumen. Acting through a receptor on the apical sur- completely for one hour, its concentration would decrease
face of follicular cells, thyroglobulin decreases expression of by only 1%. Furthermore, because the binding capacity of
thyroid-specific transcription factors and thereby downreg- plasma proteins for thyroid hormones is far from saturated,
ulates transcription of the genes for thyroglobulin, the thy- an even massive increase in secretion rate would have lit-
roid peroxidase, the sodium iodide symporter and the TSH tle effect on the concentration of hormone that is unbound.
receptor. Further effects of thyroglobulin include increased These considerations seem to rule out changes in thyroid hor-
transcription of pendrin, which delivers iodide from the mone secretion as effectors of minute-to-minute regulation

49

CH003-P373975.indd 49 6/18/2008 4:31:02 PM


Basic Medical Endocrinology

of any homeostatic process. On the other hand, because so Observations in human subjects confirm that T3 actu-
much of the circulating hormone is bound to plasma bind- ally is formed extrathyroidally and can account for most of
ing proteins, we might expect that the total amount of T4 the biological activity of the thyroid gland. Thyroidectomized
and T3 in the circulation would be affected significantly by subjects given pure T4 in physiological amounts have nor-
decreases in the concentration of plasma-binding proteins, mal amounts of T3 in their circulation. Furthermore, the
as might occur with liver or kidney disease. rate of metabolism of T3 in normal subjects is such that
about 30 μg of T3 is replaced daily, even though the thyroid
gland secretes only 5 μg each day. Thus nearly 85% of the T3
that turns over each day must be formed by deiodination of
METABOLISM OF THYROID HORMONES T4 in extrathyroidal tissues. This extrathyroidal formation of
T3 consumes about 35% of the T4 secreted each day. The
remainder is degraded to inactive metabolites.
Because T4 is bound much more tightly by plasma proteins
Extrathyroidal metabolism of T4 centers around
than T3, a greater fraction of T3 is free to diffuse out of
selective and sequential removal of iodine from the thyro-
the vascular compartment and into cells where it can pro-
nine nucleus catalyzed by three different enzymes called
duce its biological effects or be degraded. Consequently, it
iodothyronine deiodinases (Figure 3.8). These enzymes are
is not surprising that the half-time for disappearance of an
seleno-proteins and share the unusual feature of having the
administered dose of 125I-labeled T3 is only one-sixth of
rare amino acid seleno-cysteine in their active sites. The
that for T4, or that the lag time needed to observe effects
type-I deiodinase is expressed mainly in the liver and kidney,
of T3 is considerably shorter than that needed for T4.
but is also found in the central nervous system, the ante-
However, because of the binding proteins, both T4 and T3
rior pituitary gland, and the thyroid gland itself. The type I
have unusually long half-lives in plasma, measured in days
deiodinase is a membrane-bound enzyme with its catalytic
rather than seconds or minutes (Figure 3.7). It is notewor-
domain oriented to face the cytoplasm. T4 gains access to
thy that the half-lives of T3 and T4 are increased with thy-
the enzyme after crossing the plasma membrane facilitated
roid deficiency and shortened with hyperthyroidism.
mainly by monocarboxylate transporter 8, which is present in
Although the main secretory product of the thyroid
most cells and which also transports T3. After deiodination,
gland and the major form of thyroid hormone present in
the newly produced T3 readily escapes into the circulation
the circulating plasma reservoir is T4, it is primarily T3 and
and accounts for about 80% of the T3 in blood. The type
not T4 that binds to thyroid hormone receptors (see later).
I deiodinase can remove an iodine molecule either from
In fact, T4 can be considered to be a prohormone that
the outer (phenolic) ring of T4, or from the inner (tyrosyl)
serves as the precursor for extrathyroidal formation of T3.
ring. Iodines in the phenolic ring are designated 3⬘ and 5⬘;
iodines in the inner ring are designated simply 3 and 5.
The 3 and 5 positions on either ring are chemically equiv-
10
alent, but there are profound functional consequences of
% Injected radioactivity/L serum

T4 Half-life ⫽ 6.2 days removing an iodine from the inner or outer rings of thy-
roxine. Removing an iodine from the outer ring produces
3⬘,3,5 triiodothyronine, usually designated as T3, and con-
verts thyroxine to the form that binds to thyroid hormone
1.0 receptors. Removal of an iodine from the inner ring produces
3⬘,5⬘,3 triiodothyronine, which is called reverse T3 (rT3).
T3 Half-life ⫽ 1.0 day
Reverse T3 cannot bind to thyroid hormone receptors and
can only be further deiodinated. Hepatic expression of the
type I deiodinase is under negative feedback control by T3,
0.1 and is therefore most abundant when T3 levels are low.
0 1 2 3 4 5 6 7 8 9 10 11 The type II deiodinase resides in the endoplasmic
Days after I.V. injection of radioactive reticulum in many extrahepatic tissues, but is absent from
T3 or T4
the liver. It accounts for most of the extrathyroidal produc-
FIGURE 3.7 Rate of loss of serum radioactivity after injection of labeled tion of T3 in normal humans. Its presence in the brain and
thyroxine or triiodothyronine into human subjects. (Plotted from data
pituitary gland is thought to account for about half of the
of Nicoloff, J.D., Low, J. C., Dussault, J.H. et al. (1972) Simultaneous
measurement of thyroxine and triiodothyronine peripheral turnover T3 available to bind to receptors in these tissues. T3 and T4
kinetics in man. J. Clin. Invest. 51: 473.) are taken up by facilitated diffusion driven by blood levels

50

CH003-P373975.indd 50 6/18/2008 4:31:03 PM


Thyroid Gland 3
of free hormone. However, circulating levels of T3 normally the inner ring (Figure 3.8). Reverse T3 is also a favored sub-
remain fairly constant, whereas tissue demands for hor- strate for the type I deiodinase, and although it is formed at a
mone vary with changing physiological demands. The type similar rate as T3, it is degraded much faster than T3. Some
II deiodinase provides an alternative means for satisfying rT3 escapes into the bloodstream where it is avidly bound to
needs for T3 through intracellular deiodination of T4, inde- TBG and TTR.
pendently of the availability of the circulating hormone. As All three deiodinases can catalyze the oxidative removal
might be anticipated, the abundance of type II deodinase is of iodine from partially deiodinated hormone metabolites,
not constant; its expression is highest when blood concen- and through their joint actions the thyronine nucleus can be
trations of T4 are low. Both its synthesis and degradation completely stripped of iodine. The liberated iodide is then
are regulated. The enzyme turns over rapidly with a half- available for uptake by the thyroid and is recycled into hor-
life of only about 20 minutes, compared to many hours for mone. A quantitatively less important route for degradation
the type I enzyme. It is inactivated by ubiquitylation and of thyroid hormones includes shortening of the alanine side
degraded in proteasomes. However, ubiquitylation is revers- chain to produce tetraiodothyroacetic acid (Tetrac) and its
ible, and the type II enzyme can be rescued by activation of subsequent deiodination products. Thyroid hormones also are
de-ubiquitinating enzymes. Synthesis of the type II deiod- conjugated with glucuronic acid and excreted intact in the
ibnase also is regulated by other hormones and growth fac- bile. Bacteria in the intestine can split the glucuronide bond,
tors; in particular, by hormones that act through the cyclic and some of the thyroxine liberated can be taken up from the
AMP second messenger system (see Chapter 1). These intestine and returned to the general circulation. This cycle of
characteristics support the idea that this enzyme may pro- excretion in bile and absorption from the intestine is called
vide T3 to meet local demands. the enterohepatic circulation, and may be of importance in
The type III deiodinase removes an iodine from the maintaining normal thyroid economy when thyroid function
tyrosyl ring of T4 or T3, and hence its function is solely deg- is marginal or dietary iodide is scarce. Thyroxine is one of the
radative. It is widely expressed by many tissues throughout few naturally occurring hormones that is sufficiently resistant
the body. Reverse T3 is produced by both the type I and to intestinal and hepatic destruction that therapeutic doses
type III deiodinases and may be further deiodinated by the can readily be given by mouth.
type III deiodinase by removal of the second iodide from

I I
HO- -O- -C-C-COOH
NH2
I I

Deiodinase Type I Deiodinase Type I


Deiodinase Type III Deiodinase Type II

I I I I
HO- -O- -C-C-COOH HO- -O- -C-C-COOH
NH2 NH2
I I
3,3’,5’-triiodothyronine; reverse T3; rT3 3,5,3’-triiodothyronine; T3;

3’,5’-diiodothyronine 3’,3’-diiodothyronine (T2) 3,5-diiodothyronine


FIGURE 3.8 Metabolism of thyroxine. About 90% of
thyroxine is metabolized by sequential deiodination catalyzed
by deiodinases (types I, II, and III); the first step removes an
iodine from either the phenolic or tyrosyl ring producing
3’-monoiodothyronine 3-monoiodothyronine an active (T3) or an inactive (rT3) compound. Subsequent
deiodinations continue until all the iodine is recovered from
the thyronine nucleus. Green arrows designate deiodination
of the phenolic ring and red arrows indicate deiodination of
Thyronine the tyrosyl ring. Less than 10% of thyroxine is metabolized by
shortening the alanine side chain prior to deiodination.

51

CH003-P373975.indd 51 6/18/2008 4:31:03 PM


Basic Medical Endocrinology

PHYSIOLOGICAL EFFECTS OF THYROID neural development, but direct effects of maternal thyroid
HORMONES deficiency on the fetal brain have not been established.
However, failure of thyroid gland development in babies
Growth and maturation born to mothers with normal thyroid function have normal
brain development if properly treated with thyroid hor-
Skeletal system mones after birth. Maturation of the nervous system during
One of the most striking effects of thyroid hormones is the perinatal period has an absolute dependence on thyroid
on bodily growth (see Chapter 10). Although fetal growth hormone. During this critical period thyroid hormone must
appears to be independent of the thyroid, growth of the be present for normal development of the brain. In rats
neonate and attainment of normal adult stature require made hypothyroid at birth, cerebral and cerebellar growth
optimal amounts of thyroid hormone. Because stature or and nerve myelination are severely delayed. Overall size of
height is determined by the length of the skeleton, we might the brain is reduced along with its vascularity, particularly
anticipate an effect of T3 on growth of bones that elongate at the capillary level. The decrease in size may be partially
by expansion of cartilage in the epiphyseal plates followed accounted for by a decrease in axonal density and dendritic
by replacement of cartilage with bone (see Chapter 11). branching.
Cartilage cells at all stages of development express receptors Thyroid hormone deficiency also leads to specific
for thyroid hormone and the type 2 deiodinase. Their pro- defects in cell migration and differentiation. In human
gression from undifferentiated precursors, through mature infants the absence or deficiency of thyroid hormone during
and hypertrophic stages are all influenced by T3, which also this period is catastrophic and results in permanent, irrevers-
stimulates secretion of extracellular matrix proteins. In addi- ible mental retardation even if large doses of hormone are
tion to these presumably direct actions, thyroid hormones given later in childhood (Figure 3.9). If replacement ther-
appear to act permissively or synergistically with growth apy is instituted early in postnatal life, however, the tragic
hormone, insulin-like growth factor I (see Chapter 11), and consequences of neonatal hypothyroidism can be averted.
other growth factors that promote bone formation. They also Mandatory neonatal screening for hypothyroidism therefore
promote bone growth indirectly by actions on the pituitary has been instituted throughout the United States and many
gland and hypothalamus. Thyroid hormone is required for other countries. Precisely what thyroid hormones do during
normal growth hormone synthesis and secretion. the critical period, how they do it, and why the opportunity
Skeletal maturation is distinct from skeletal growth. for intervention is so brief are subjects of active research.
Maturation of bone results in the ossification and eventual Effects of T3 and T4 on the central nervous system
fusion of the cartilaginous growth plates, which occurs with are not limited to the perinatal period of life. In the adult,
sufficient predictability in normal development that indi- hyperthyroidism produces hyperexcitability, irritability, rest-
viduals can be assigned a specific “ bone age” from radiologi- lessness, and exaggerated responses to environmental stimuli.
cal examination of ossification centers. Thyroid hormones Emotional instability that can lead to full-blown psychosis
profoundly affect skeletal maturation. Bone age is retarded may also occur. Conversely, decreased thyroid hormone results
relative to chronological age in children who are deficient in in listlessness, lack of energy, slowness of speech, decreased
thyroid hormone and is advanced prematurely in hyperthy- sensory capacity, impaired memory, and somnolence. Mental
roid children. Uncorrected deficiency of thyroid hormone capacity is dulled, and psychosis (myxedema madness) may
during childhood results in retardation of growth and mal- occur. Conduction velocity in peripheral nerves is slowed and
formation of facial bones characteristic of juvenile hypothy- reflex time increased in hypothyroid individuals. The under-
roidism or cretinism. lying mechanisms for these changes are not understood.

Autonomic nervous system


Central nervous system
The importance of the thyroid hormones for normal devel- Interactions between thyroid hormones and the autonomic
opment of the nervous system is well established. Thyroid nervous system, particularly the sympathetic branch, are
hormones and their receptors are present early in the devel- important throughout life. Increased secretion of thyroid
opment of the fetal brain, well before the fetal thyroid hormone exaggerates many of the responses that are medi-
gland becomes functional. T4 and T3 present in the fetal ated by the neurotransmitters norepinephrine and epineph-
brain at this time probably arise in the mother and cross rine released from sympathetic neurons and the adrenal
the placenta to the fetus, although the placenta is relatively medulla (see Chapter 4). In fact, many symptoms of hyper-
impermeable to T4 and T3. Some evidence suggests that thyroidism, including tachycardia (rapid heart rate) and
maternal hypothyroidism may lead to deficiencies in postnatal increased cardiac output, resemble increased activity of the

52

CH003-P373975.indd 52 6/18/2008 4:31:04 PM


Thyroid Gland 3
8 Thyroid result of increased rates of release of calcium from the sarco-
treatment Normal plasm reticulum and its reuptake. Opposite effects are seen
started
7 in hypothyroidism. Thyroid hormones upregulate expres-
sion of the genes for the calcium channel (ryanodine recep-
6 tor) through which calcium is released during contraction,
Bone age
and the sarcoplasmic reticular calcium ATPase (SERCa2)
by which it is retrieved during relaxation. In addition, T3
Developmental age

5
upregulates expression of the genes for the subunits of the
sodium/potassium ATPase and the potassium channel pro-
4
Height age teins that restore the membrane potential.
Cardiac output is increased in hyperthyroidism and
3
decreased in thyroid deficiency. Hyperthyroidism lowers
peripheral resistance by promoting relaxation of arteriolar
2
smooth muscle and by increasing venous tone. As a result,
Mental age
venous return is increased producing increased ventricular
1 filling (preload). The decrease in peripheral resistance also
decreases resistance to ventricular emptying (afterload). These
effects may be indirect, but when combined with the direct
3 4 5 6 7 8 9 effects on cardiac myocytes and enhanced sympathetic actions
Chronological age in years they have profound implications for cardiovascular function.
FIGURE 3.9 Effects of thyroid therapy on growth and development of
a child with no functional thyroid tissue. Daily treatment with thyroid
hormone began at 4.5 years of age (green arrow). Bone age rapidly Metabolism
returned toward normal, and the rate of growth (height age) paralleled the
normal curve. Mental development, however, remained infantile. (From
Wilkins, L. (1965) The Diagnosis and Treatment of Endocrine Disorders in
Oxidative metabolism and thermogenesis
Childhood and Adolescence. Charles C Thomas, Springfield, Illinois.) More than a century has passed since it was recognized that
the thyroid gland exerts profound effects on oxidative metab-
olism in humans. The so-called basal metabolic rate (BMR),
sympathetic nervous system. Thyroid hormones increase the which is a measure of oxygen consumption under defined
abundance of receptors for epinephrine and norepinephrine resting conditions, is highly sensitive to thyroid status. A
(β-adrenergic receptors) in the myocardium and some other decrease in oxygen consumption results from a deficiency of
tissues. Thyroid hormones may also increase expression of thyroid hormones, and excessive thyroid hormone increases
the stimulatory G-protein (Gαs) associated with adrenergic BMR. Oxygen consumption in all tissues except brain, tes-
receptors and downregulate the inhibitory G-protein (Gαi). tis, and spleen is sensitive to the thyroid status and increases
Either of these effects results in greater production of cyclic in response to thyroid hormone (Figure 3.10). Even though
AMP (see Chapter 1). Furthermore, through the agency of the dose of thyroid hormone given to hypothyroid animals
cyclic AMP, sympathetic stimulation activates the type II in the experiment shown in Figure 3.10 was large, there was
deiodinase, which accelerates local conversion of T4 to T3. a delay of many hours before effects were observable. In fact,
Because thyroid hormones exaggerate a variety of responses the rate of oxygen consumption in the whole animal did
mediated by β-adrenergic receptors, pharmacological block- not reach its maximum until four days after a single dose of
ade of these receptors is useful for reducing some of the hormone. The underlying mechanisms for increased oxygen
symptoms of hyperthyroidism. Conversely, the diverse func- consumption are incompletely understood.
tions of the sympathetic nervous system are compromised Oxygen consumption ultimately reflects activity of
in hypothyroid states. mitochondria and is coupled with formation of high-energy
bonds in ATP. In general, oxygen consumption is propor-
Cardiovascular system tional to energy utilization, but conversion of energy stored
in metabolic fuels to formation of ATP is not perfect, and
In addition to the indirect effects produced through inter- some energy is lost as heat. Thus oxygen consumption and
action with the sympathetic nervous system, thyroid hor- heat production are intimately related, and the effects of thy-
mones directly influence several aspects of cardiovascular roid hormones on oxygen consumption seem to be byprod-
function. Both the contraction and relaxation phases of the ucts of their essential role in calorigenesis. Although thyroid
cardiac cycle are accelerated in the hyperthyroid heart as a hormones have many functions in all vertebrates, their effects

53

CH003-P373975.indd 53 6/18/2008 4:31:04 PM


Basic Medical Endocrinology

A of calcium from cytosol to the sarcoplasmic reticulum by


160 Animal calcium ATPases accounts for even more. Although most
Heart of the activity of the sodium/potassium ATPase resides in
150 Gastric mucosa brain and kidney whose rates of metabolism are minimally
Oxygen consumption as a percent

increased by T3, thyroid hormones are thought to promote


140
of hypothyroid control

entry of sodium into other cells and therefore increase the


Liver
work of the sodium/potassium ATPase. Both the expression
130 Smooth muscle
and activity of the sodium/potassium ATPase are decreased
Diaphragm
Kidney
in hypothyroid and increased in hyperthyroid subjects.
120
Skeletal muscle Similar changes are seen in calcium uptake and release from
110
the sarcoplasmic reticulum in both resting and working skel-
etal muscle. These changes in calcium dynamics may account
100 for the stiffness and delayed reflexes seen in hypothyroid
individuals. Additionally, ATP-consuming cycles that are
90 intrinsic components of carbohydrate, lipid, and protein
0 6 12 24 48 metabolism, are accelerated by thyroid hormones and also
Hours after thyroxine contribute to the increase in oxygen consumption.
Thyroid hormones decrease the efficiency of ATP for-
B
mation. Phosphorylation of ADP to form ATP is driven by
200
the energy of the proton gradient that is generated across
Animal
Oxygen consumption as a percent

the inner mitochondrial membrane by the electron transport


180
system. When ATP production is coupled optimally to oxy-
of hypothyroid control

160 gen consumption 1.5 to 2.5 molecules of ATP are formed


for each molecule of oxygen consumed. Leakage of protons
140 across the inner mitochondrial membrane partially dissipates
the gradient and results in fewer high energy phosphate
Spleen
120 bonds formed for each atom of oxygen converted to water.
Brain Energy that would otherwise be stored in phosphate bonds is
100
dissipated as heat. Leakage of protons is increased by uncou-
Testis
pling proteins (UCP) in the inner mitochondrial membrane.
80
N Tx-0 1 2 3 4 5 6 7 8
To date three proteins thought to have uncoupling activity
Days after thyroxine have been identified in mitochondrial membranes of various
tissues. All three appear to be upregulated by T3. Although
FIGURE 3.10 Effects of thyroxine on oxygen consumption by various
the physiological importance of UCP-1 is firmly established
tissues of thyroidectomized rats. Note in A the abscissa is in units of hours
and in B the units are days. (Redrawn from Barker, S.B. and Klitgaard, H.M. (see later) the physiological roles of UCP-2 and UCP-3
(1952) Metabolism of tissues excised from thyroxine-injected rats. Am. J. remain uncertain.
Physiol. 170: 81.) The calorigenic effects of thyroid hormones are essen-
tial for maintaining constancy of body temperature. Upon
exposure to a cold environment, heat production is increased
on oxygen consumption are seen only in those animals, birds, through at least two mechanisms: (1) shivering, which is a
and mammals that maintain a constant body temperature rapid increase in involuntary activity of skeletal muscle, and
(homeotherms). It is therefore not surprising that one of the (2) the so-called nonshivering thermogenesis seen in cold
classic signs of hypothyroidism is decreased tolerance to cold, acclimated individuals. As we have seen, the metabolic effects
whereas excessive heat production and sweating are seen in of T3 have a long lag time and hence increased production of
hyperthyroidism. Thyroid hormones increase oxygen con- T3 cannot be of much use for making rapid adjustments to
sumption and heat production by increasing the rate of ATP cold temperatures. The role of T3 in the shivering response is
breakdown and decreasing the efficiency of ATP synthesis. probably limited to maintenance of tissue sensitivity to sym-
Among the ATP-consuming processes that T3 appears pathetic stimulation. In this context, the importance of T3
to accelerate are the maintenance of cellular ionic gradients. derives from actions that were established before exposure to
The sodium/potassium ATPase maintains ionic integrity of cold temperature. Maintenance of sensitivity to sympathetic
all cells and has been estimated to account for as much as stimulation permits efficient mobilization of stored carbohy-
20% of total resting oxygen consumption, and the pumping drate and fat needed to fuel the shivering response and to

54

CH003-P373975.indd 54 6/18/2008 4:31:05 PM


Thyroid Gland 3
make circulatory adjustments for increased activity of skeletal 100
muscle. It may also be recalled that the sympathetic nervous
system regulates heat conservation by decreasing blood flow 10⫺10 M T3
80
through the skin. Piloerection in animals increases the thick- 10⫺8 M T3

Malic enzyme (U/mg)


ness of the insulating layer of fur. These responses are impor-
60
tant defenses in both acute and chronic exposure to cold.
Chronic nonshivering thermogenesis appears to
require increased production of T3, which acts in concert 40

with the sympathetic nervous system to increase heat pro-


duction and conservation. In humans and large animals thy- 20
roid hormone dependency increases in the activity of ion
pumps in muscle and provides the major source of increased 0
heat production. Brown fat is an important source of heat 100 300 500 1000
Glucose concentration (mg/dl)
in newborn humans and throughout life in small mammals.
This form of adipose tissue is especially rich in mitochondria, FIGURE 3.11 Effects of glucose and T3 on the induction of malic enzyme
which give it its unique brown color. Mitochondria in this (ME) in isolated hepatocyte cultures. Note that the amount of enzyme
present in tissues was increased by growing cells in higher and higher
tissue contain UCP 1, sometimes called thermogenin, which
concentrations of glucose. Blue bars show effects of glucose in the
allows them to oxidize relatively large amounts of fatty presence of a low (10⫺10 M) concentration of T3 . Red bars indicate that
acids, and produce heat. Although both T3 and the sympa- the effects of glucose were exaggerated when cells were grown in a high
thetic neurotransmitter, norepinephrine, can each upregulate concentration of T3 (10⫺8 M). (From Mariash, G.N. and Oppenheimer, J.H.
(1982) Thyroid hormone-carbohydrate interaction at the hepatic nuclear
expression of UCP-1, their cooperative interaction results level. Fed. Proc. 41: 2674.)
in production of three to four times as much of this mito-
chondrial protein as the sum of their independent actions.
In addition, T3 increases the efficacy of norepinephrine to
release fatty acids from stored triglycerides and thus provides in hepatocytes, and glucose oxidation in liver, fat, and muscle
fuel for heat production. Brown adipose tissue also upregu- cells. No single or unique reaction in any pathway of carbohy-
lates type II deiodinase in response to sympathetic stimu- drate metabolism has been identified as the rate-determining
lation, and produces abundant T3 locally to meet its needs. target of T3 action. Rather, carbohydrate degradation
Adult humans have little brown fat, but may increase heat appears to be driven by other factors, such as increased
production through similar effects of UCP-2 and UCP-3 in demand for ATP, the content of carbohydrate in the diet,
white fat and muscle; however, supporting evidence for this or the nutritional state. Although T3 may induce synthesis
possibility is not available. of specific enzymes of carbohydrate and lipid metabolism
In rodents and other experimental animals, exposure (e.g., the malic enzyme, glucose 6-phosphate dehydrogenase,
to cold temperatures is an important stimulus for increased and 6-phosphogluconate dehydrogenase), it appears princi-
TSH secretion from the pituitary and the resultant increase pally to behave as an amplifier or gain control working in
in T4 and T3 secretion from the thyroid gland. Cold expo- conjunction with other signals (Figure 3.11). In the exam-
sure does not increase TSH section in humans except in the ple shown, induction of the malic enzyme in hepatocytes
newborn. In humans and experimental animals, however, was dependent both on the concentration of glucose in the
exposure to cold temperatures increases the abundance of culture medium and on the concentration of T3. T3 had lit-
the type II deiodinase in skeletal muscle as well as brown tle effect on enzyme induction when there was no glucose
fat. Consequently, the requirement for increased amounts of but amplified the effectiveness of glucose as an inducer of
T3 to satisfy demands imposed by the external environment genetic expression. This experiment provides a good example
is met by accelerated conversion of T4 to T3. Increased of how T3 can amplify readout of genetic information.
production of the type II deiodinase probably results from
increased sympathetic nervous activity, which leads to Lipid metabolism
increased cyclic AMP production in various tissues. Because glucose is the major precursor for fatty acid synthe-
sis in both liver and fat cells, it should not be surprising that
optimal amounts of thyroid hormone are necessary for lipo-
Carbohydrate metabolism genesis in these cells. Once again the primary determinant
T3 accelerates virtually all aspects of metabolism includ- of lipogenesis is not T3, but rather, the amount of avail-
ing carbohydrate utilization. It increases glucose absorption able carbohydrate or insulin (see Chapter 7), with thyroid
from the digestive tract, glycogenolysis and gluconeogenesis hormone acting as a gain control. Similarly, mobilization

55

CH003-P373975.indd 55 6/18/2008 4:31:06 PM


Basic Medical Endocrinology

of fatty acids from storage depots in adipocytes is compro- Secretion of TSH by the pituitary gland is governed by
mised in the thyroid-deficient subject and increased above positive input from the hypothalamus by way of thyrotropin-
normal when thyroid hormones are present in excess. Once releasing hormone (TRH) and negative input from the thy-
again, T3 amplifies physiological signals for fat mobilization roid gland by way of T3 and T4. TRH increases expression
without itself acting as such a signal. of the genes for both the α- and β-subunits of TSH, and
Increased blood cholesterol (hypercholesterolemia), increases the post-translational incorporation of carbohy-
mainly in the form of low density lipoproteins (LDL), typi- drate that is required for normal potency of TSH. However,
cally is found in hypothyroidism. Thyroid hormones reduce dependence of the thyrotropes on TRH in not complete, as
cholesterol in the plasma of normal subjects and restore these processes can go on at a reduced level in the absence of
blood concentrations of cholesterol to normal in hypothy- TRH. Disruption of the TRH gene reduces the TSH con-
roid subjects. Hypercholesterolemia in hypothyroid sub- tent of mouse pituitaries to less than half that of wild-type
jects results from decreased ability to excrete cholesterol in litter mate controls. Blood levels of thyroid hormones and
bile rather than from overproduction of cholesterol. In fact, TSH were just below the normal range in a rare case of a
cholesterol synthesis is impaired in the hypothyroid indi- child with a genetic absence of TRH receptors. Growth was
vidual. T3 may facilitate hepatic excretion of cholesterol by somewhat retarded, but hypothyroidism was otherwise mild.
increasing the abundance of LDL receptors in hepatocyte Similarly, blood levels of thyroid hormones are below nor-
membranes, thereby enhancing uptake of cholesterol from mal in mice lacking a functional TRH gene, but they grow,
the blood. develop, and reproduce almost normally.
Maintaining constant levels of thyroid hormones in
Nitrogen metabolism blood depends upon negative feedback effects of T4 and T3,
which inhibit synthesis and secretion of TSH (Figure 3.12).
Body proteins are constantly being degraded and resynthe-
The contribution of free T4 in blood is quite significant in
sized. Both synthesis and degradation of protein are slowed
this regard. Because thyrotropes are rich in type II deiodi-
in the absence of thyroid hormones, and conversely, both are
nase, they can convert this more abundant form of thyroid
accelerated by thyroid hormones. In the presence of excess
hormone to T3 and thereby monitor the overall amount of
T4 or T3, the effects of degradation predominate, and often
free hormone in blood. High concentrations of thyroid hor-
there is severe catabolism of muscle. In hyperthyroid sub-
mones may shut off TSH secretion completely and, when
jects body protein mass decreases despite increased appetite
maintained over time, produce atrophy of the thyroid gland.
and ingestion of dietary proteins. With thyroid deficiency
Measurement of relative concentrations of TSH and thy-
there is a characteristic accumulation of a mucus-like mate-
roid hormones in the blood provide important information
rial consisting of protein complexed with hyaluronic acid
for diagnosing thyroid disease. For instance, low blood con-
and chondroitin sulfate in extracellular spaces, particu-
centrations of free T3 and T4 in the presence of elevated
larly in the skin. Because of its osmotic effect, this material
levels of TSH signal a primary defect in the thyroid gland,
causes water to accumulate in these spaces, giving rise to the
edema typically seen in hypothyroid individuals and to the
name myxedema for hypothyroidism. Hypothalamus

Pituitary
REGULATION OF THYROID HORMONE
SECRETION T3 ⴙ T4

As already indicated, secretion of thyroid hormones TSH


depends on stimulation of thyroid follicular cells by TSH,
which bears primary responsibility for integrating thyroid
function with bodily needs (see Chapter 2). In the absence
of TSH, thyroid cells are quiescent and atrophy, and as we Liver
have seen, administration of TSH increases both synthesis T4 ⴙT3
and secretion of T4 and T3. Patients who lack functional
TSH receptors present at birth with severe hypothyroidism Thyroid

accompanied by virtual absence of a functional thyroid FIGURE 3.12 Feedback regulation of thyroid hormone secretion. Green
gland, and high circulating levels of TSH. arrow ⫽ stimulation. Red arrow ⫽ inhibition.

56

CH003-P373975.indd 56 6/18/2008 4:31:07 PM


Thyroid Gland 3
PIP2
Daily T3 ⫹ T4
T4 (µg) (µg)
T4 PLC TRHR TRH
Nucleus 15 0 0
15 ⫹ 60
DAG IP3
T3 T3 TR
PKC
10

TSH (µU/ml)
TSHα-mRNA Ca2⫹
TRHRmRNA TSHβ-mRNA ER

PreTSH 5
Downregulation
Processing
TRH
TRHR 0
Golgi apparatus
Ca2⫹ 0 30 60 90 120 180
TRHR Minutes
Secretory vesicles
FIGURE 3.14 Effect of treatment of normal young men with
thyroid hormones for 3 to 4 weeks on the response of the pituitary to
thyrotropin-releasing hormone (TRH) as measured by changes in plasma
TSH concentrations of thyroid-stimulating hormone (TSH). The six subjects
received 25 mg of TRH at time 0 as indicated by the green arrow. Values
FIGURE 3.13 Effects of TRH, T3, and T4 on the thyrotrope. T3 are expressed as means⫾SEM. (From Snyder, P.J. and Utiger, R.D. (1972)
downregulates expression of genes for TRH receptors (TRHR) and Inhibition of thyrotropin response to thyrotropin-releasing hormone by
both subunits of TSH (red arrows). TRH effects (shown in green) include small quantities of thyroid hormones. J. Clin. Invest. 52: 2077.)
upregulation of TSH gene expression, enhanced TSH glycosylation, and
accelerated secretion. TR ⫽ thyroid hormone receptor.

deiodinase. T3 enters the nucleus, binds to its receptors, and


while high concentrations of free T3 and T4 accompanied downregulates transcription of the genes for both the α-
by high concentrations of TSH reflect a defect in the pitui- and β-subunits of TSH and for TRH receptors. In addition,
tary or hypothalamus. As already noted, the high concentra- T3 inhibits release of stored hormone and accelerates TRH
tions of T4 and T3 seen in Graves’ disease are accompanied receptor degradation. The net consequence of these actions
by very low concentrations of TSH in blood as a result of of T3 is a reduction in the sensitivity of the thyrotropes to
negative feedback inhibition of TSH secretion. TRH (Figure 3.14).
Negative feedback inhibition of TSH secretion results
from actions of thyroid hormones exerted both on TRH
neurons in the paraventricular nuclei of the hypothalamus
and on thyrotropes in the pituitary. Results of animal studies MECHANISM OF THYROID HORMONE ACTION
indicate that T3 and T4 inhibit TRH synthesis and secre-
tion. Events thought to occur within the thyrotropes are As must already be obvious, virtually all cells appear to
illustrated in Figure 3.13. TRH binds its G-protein coupled require optimal amounts of thyroid hormone for normal
receptors (see Chapter 1) on the surface of thyrotropes. The operation, even though different aspects of function may be
resulting activation of phospholipase C generates the sec- affected in different cells. T3 formed by deiodination of T4
ond messengers inositol trisphosphate (IP3) and diacylglyc- within target cells mixes freely with T3 taken up from the
erol (DAG). IP3 promotes calcium mobilization, and DAG plasma and enters the nucleus where it binds to its receptors
activates protein kinase C, both of which rapidly stimulate (see Chapter 1). Thyroid hormone receptors are members of
release of stored hormone. This effect is augmented by the the nuclear receptor superfamily of transcription factors and
influx of extracellular calcium following activation of mem- bind T3 about ten times more avidly than T4. They contain
brane calcium channels. In addition, transcription of genes about 400 amino acids arranged into regions or domains
for both subunits of TSH is increased. TRH also promotes involved in ligand binding, dimerization with other recep-
processing of the carbohydrate components of TSH nec- tors, DNA binding, and protein–protein interactions with
essary for maximum biological activity. Meanwhile, both coactivator or corepressor complexes (see Chapter 1).
T4 and T3 are transported into thyrotropes at rates deter- The biology of thyroid hormone receptors is complex
mined by their free concentrations in blood plasma. T4 is and incompletely understood, and much of what is known
deiodinated to T3 in the cytoplasm by the abundant type II has been learned from genetically manipulated mice. Thyroid

57

CH003-P373975.indd 57 6/18/2008 4:31:07 PM


Basic Medical Endocrinology

hormone receptors are encoded in two genes, designated A ⫺T3


TRα and TRβ. The TRα gene resides on chromosome 17
in humans and gives rise to two isoforms, TRα1 and TRα2. General
Other corepressors
Alternate RNA splicing deletes the T3 binding site from the transcription
TRα2 isoform so that it cannot act as a hormone receptor, HDAC
(⫺)
complex
HDM HMT
but nevertheless plays a vital physiological role (see later).
SMRT
Two additional truncated isoforms of the TRα gene, Δα1 RXR
and Δα2, arise by transcription from a downstream promoter TR DNA
TRE TATA
and lack both DNA and ligand binding domains. Their
physiological role is uncertain, but they may act as inhibitors
B ⫹T3
of TRα1. The human TRβ gene maps to chromosome 3 and
also gives rise to two isoforms, TRβ1 and TRβ2, by alternate
Other
splicing, and both contain the hormone binding site. TRα1 coactivators
and TRβ1 are distributed widely throughout the body and HA General
(⫹) transcription
are present in different ratios in the nuclei of all target tis- SRC-1 complex
sues examined, but TRβ2 appears to be expressed primarily T3 RXR
in the anterior pituitary gland and the brain. TR DNA
Unlike most other nuclear receptors, thyroid hor- TRE TATA
mone receptors bind to specific nucleotide sequences (thy-
FIGURE 3.15 Models of the effects of thyroid hormone receptor (TR)
roid response elements or TREs) in the genes they regulate on gene transcription. A. In the absence of T3 the TR in heterodimeric
whether or not the hormone is present. They may bind as union with the retinoic acid receptor (RXR) binds to the thyroid hormone
monomers, but more typically as homodimers composed of response element (TRE) in DNA. The unliganded TR also binds to a
corepressor, in this case the silencing mediator retinoic acid and TR (SMRT),
two thyroid hormone receptors or as heterodimers formed which recruits factors that remodel chromatin such as histone deacetylase
with the receptor for an isomer of retinoic acid (RXR) or (HDAC), histone demethylase (HDM), histone methyl transferase (HMT)
other nuclear receptor family members. In the absence of T3, and other factors that interfere with attachment of the RNA polymerase
in the general transcription complex to the TATA box in the promoter of
the unoccupied receptor, in conjunction with a corepressor a thyroid hormone regulated gene. B. Upon binding T3, the TR changes
protein complex, inhibits T3-dependent gene expression by its conformation, sheds the corepressor complex and replaces it with a
maintaining the DNA in a tightly coiled configuration that coactivator, in this case steroid receptor coactivator-1 (SRC-1), which recruits
such chromatin remodeling factors as histone acetylase (HA) and other
bars access of transcription activators or RNA polymerase.
coactivators that make the gene accessible to the polymerase complex.
Upon binding T3, the configuration of the receptor is modi-
fied in a way that causes it to release the corepressor and bind
instead to a coactivator (Figure 3.15). Although T3 acts in
an analogous way to suppress expression of some genes, the the consequences of T4 hypersecretion. This disease typi-
underlying mechanism for negative control of gene expres- cally results from mutations in the TRβ gene.
sion is not understood. Manipulation of the gene for TRα so that only the α2
The question of which T3 responses are mediated by isoform can be expressed produced no deleterious effects on
each form of the T3 receptor has been addressed by exam- lifespan or fertility even though this isoform cannot bind T3.
ining the consequences of disrupting all or parts of the However, lack of the TRα1 isoform in these mice resulted
THRα or β genes in mouse embryos. Mice that lack both in low heart rate and low body temperature. When the TRα
TRβ isoforms are fertile, exhibit no obvious behavioral gene was knocked out so that neither the α1 nor the α2 iso-
abnormalities, and have minimal developmental deficien- form could be expressed, the animals stopped growing after
cies except for retinal and cochlear development. However, about two weeks and died shortly after weaning with appar-
these animals have abnormally high concentrations of TSH, ent failure of intestinal development. Although relatively few
T4, and T3 in their blood presumably because TRβ2 medi- symptoms of hypothyroidism result from knockout of any of
ates the negative feedback action of T3. These symptoms the three TR receptors that are capable of binding T3, loss of
are remarkably similar to those seen in a rare genetic dis- the α2 isoform produced devastating effects, suggesting that
ease that is characterized by resistance to thyroid hormone. products of the TRα gene play critical, though perhaps T3-
Like the knockout mice, patients with this disease exhibit independent, roles in gene transcription and development.
few abnormalities but have increased circulating levels of It is possible that the α and β forms of the receptors
TSH, T4, and T3. Their thyroid glands are enlarged (goiter) regulate different complements of genes as suggested in the
in response to the high TSH levels, but they suffer none of mouse studies, but the observed effects may simply reflect

58

CH003-P373975.indd 58 6/18/2008 4:31:09 PM


Thyroid Gland 3
differences in distribution of the α and β thyroid hormone Suggested reading
receptors in the various affected cell types. The combined
absence of TRα1, TRβ1, and TRβ2 produces more pro- Braverman, L.E. and Utiger, R.D., eds. (2005) Werner and Ingbar’s The
Thyroid, 9th ed. Lippincott Williams and Wilkins, Philadelphia. (This
nounced symptoms of hypothyroidism than lack of either
book provides excellent coverage of a broad range of basic and clinical
TRα1 or the TRβ isoforms, suggesting that these recep- topics.)
tors have redundant or overlapping functions. However, the Bianco, A.C. and Kim, B.W. (2006) Deiodinases: Implications of the local
hypothyroid symptoms are mild compared to those seen control of thyroid hormone action. J. Clin. Invest. 116: 2571–2579.
de Escobar, G.M., Obregón, M.J., and Escobar del Rey, F. (2004) Role of
when the complement of TRs is normal but thyroid hor- thyroid hormone during early brain development. Eur. J. Endocrinol.
mones are absent. These and related observations in geneti- 151: U25–U37.
cally manipulated cultured cells gave rise to the idea that De La Vieja, A., Dohan, O., Levy, O., and Carrasco, N. (2000) Molecular
analysis of the sodium/iodide symporter: Impact on thyroid and
unoccupied thyroid hormone receptors may repress gene extrathyroid pathophysiology. Physiol. Rev. 80: 1083–1105.
expression and therefore produce harmful effects. Thus at Flamant, F., Gauthire, K., and Samarut, J. (2007) Thyroid hormones sign-
least one of the physiological roles of T3 may be to coun- aling is getting more complex: STORMS are coming. Mole. Endocrinol.
21: 321–333.
teract the consequences of the silencing of some genes by
Gershengorn, M.C. and Osman, R. (1996) Molecular and cellular biology
unoccupied thyroid hormone receptors. of thyrotropin-releasing hormone receptors. Physiol. Rev. 76: 175–191.
Although extensive evidence indicates that T3 and Kahaly, G.J. and Dilmann, W.H. (2005) Thyroid hormone action in the
T4 produce the majority of their actions through nuclear heart. Endocr. Rev. 26: 704–728.
Koibuchi, N. and Chin, W.W. (2000) Thyroid hormone action and brain
receptors, it is likely that these receptors do not mediate all development. Trends Endocrinol. Metab. 11: 123–128.
the actions of thyroid hormones. Extranuclear specific bind- Oetting, A. and Yen, P.M. (2007) New insights into thyroid hormone
ing proteins for thyroid hormones have been found in the action. Best Practice & Research Clinical Endocrinology and Metabolism
21: 193–208.
cytosol and mitochondria, although the functions, if any, Rapoport, B., Chazenbalk, D., Jaume, J.C., and McLachlan, S.M. (1998)
of these proteins are not known. In addition, some rapid The thyrotropin (TSH) receptor: Interactions with TSH and autoanti-
effects of T3 and T4 that may not involve the genome also bodies. Endocr. Rev. 19: 673–716.
Shao, Y.Y., Wang, L., and Ballock, R.T. (2006) Thyroid hormone and the
have been described. It is highly likely that T3 and T4 have
growth plate. Rev. Endocr. Metab. Disord. 7: 265–271.
physiologically important actions that are not dependent on Silva, J.E. (2006) Thermogenic mechanisms and their hormonal regulation.
nuclear events, but detailed understanding will require fur- Physiol. Rev. 86: 435–464.
ther research. Thyroid hormones are critical factors in both Weiss, R.E. and Refetoff, S. (2000) Resistance to thyroid hormone. Rev.
Endocr. Metab. Disord. 1: 97–108.
prenatal and anti-natal development and participate in a Yen, P.M. (2001) Physiological and molecular basis of thyroid hormone
wide range of homeostatic processes, perhaps playing differ- action. Physiol. Rev. 81: 1097–1142.
ent roles at different stages of development. It should not be
surprising therefore that multiple mechanisms are employed
in expressing all the actions of these hormones or in the
conclusion that much remains to be learned in this regard.

59

CH003-P373975.indd 59 6/18/2008 4:31:10 PM

You might also like