Sei sulla pagina 1di 11

Journal of Neuroendocrinology

From Molecular to Translational Neurobiology


Journal of Neuroendocrinology 20, 784–794
ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd
REVIEW ARTICLE

Neurodevelopmental and Neurophysiological Actions of Thyroid Hormone


G. R. Williams
Molecular Endocrinology Group, Division of Medicine & MRC Clinical Sciences Centre, Imperial College, London, UK.

Journal of For over 100 years, thyroid hormones have been known to be essential for neonatal neurodevel-
opment but whether they are required by the foetal brain remains a matter of controversy. For
Neuroendocrinology decades, the prevailing view was that thyroid hormones are not necessary until after birth
because circulating levels in the foetus are very low and the placenta forms an efficient barrier
to their transfer from the mother. Clinical observations of good neurological outcome following
early treatment of congenital hypothyroidism were used to support the view that thyroid hor-
mones are not required early in neurodevelopment. Nevertheless, the issue remained contentious
because of findings that the severity of foetal neurological deficit due to maternal iodine defi-
ciency correlated with the degree of maternal thyroxine (T4) deficiency. Furthermore, neurologi-
cal damage in these cases could be prevented by correction of maternal T4 deficiency before
mid-gestation. This observation led to the opposing view, supported by epidemiological studies
of neurological cretinism, that maternal thyroid hormones are important and necessary for early
foetal neurodevelopment. It is now clear that thyroid hormones are essential for both foetal and
post-natal neurodevelopment and for the regulation of neuropsychological function in children
and adults. In recent years, this controversial subject has progressed very rapidly following
remarkable progress in understanding of the molecular mechanisms of thyroid hormone action.
Correspondence to: This article reviews the contributions of molecular biology and genetics to our new understand-
Graham R. Williams, Molecular
ing of the physiological effects of thyroid hormones on neurodevelopment and in the adult
Endocrinology Group, MRC Clinical
Sciences Centre, Hammersmith brain.
Hospital, Du Cane Road,
Key words: thyroid hormones, thyroid hormone receptor, brain, development, behaviour.
London W12 0NN, UK
(e-mail: graham.williams@imperial.
ac.uk). doi: 10.1111/j.1365-2826.2008.01733.x

eral signs of hypothyroidism (7). Although endemic cretinism can


Thyroid hormones and neurodevelopment
be prevented by public health measures, such as iodine supple-
Thyroid hormones have no influence on very early developmental mentation, that prevent or correct first trimester maternal hypo-
events, such as neural induction and establishment of polarity, thyroxinaemia, iodine deficiency remains the commonest
but regulate later processes, including neurogenesis, myelination, endocrine disorder worldwide and the most frequent cause of
dendrite proliferation and synapse formation (1–3). Numerous preventable mental retardation (4). By contrast, neurological fea-
thyroid hormone responsive genes have been identified (1) and tures in neonatal hypothyroidism are less severe, dependent on
the timing of the onset of thyroid hormone action in the devel- the severity of hypothyroidism and largely preventable by imme-
oping brain is crucial (3–6). For example, endemic neurological diate thyroid hormone replacement, although deficits in memory
cretinism is due to maternal iodine deficiency and the resulting and IQ may persist (3). Untreated neonates exhibit growth retar-
maternal hypothyroxinaemia, which is defined as thyroxine (T4) dation and general features of hypothyroidism with mental retar-
concentrations that are low for the stage of pregnancy. Low dation, tremor, spasticity and speech and language deficits (3, 7).
maternal T4 levels cause neurological hypothyroidism in the Differences between endemic cretinism and congenital hypothy-
foetus, which results in profound mental retardation, cerebral roidism illustrate that the timing of thyroid hormone action is
spastic diplegia, deaf-mutism and squint in the absence of gen- fundamental for neurodevelopment.
Thyroid hormone action in the brain 785

ptogenesis, together with the initiation of glial cell differentiation


Timing of thyroid hormone action in the brain
and migration and the onset of myelination (1, 7, 8). The third
Three stages of thyroid hormone dependent neurological develop- stage occurs in the neonatal and post-natal period when thyroid
ment can be recognised (Fig. 1). The first occurs before the hormone supplies to the brain are entirely derived from the child
onset of foetal thyroid hormone synthesis, which occurs at and critical for continuing maturation. During this period, migra-
16–20 weeks postconception in humans or by embryonic day tion of granule cells in the hippocampal dentate gyrus and cere-
E17.5–18 in the rat. During this period, thyroid hormone expo- bellum, pyramidal cells in the cortex and Purkinje cells in the
sure comes only from maternally synthesised hormone (4–6, 8) cerebellum are sensitive to thyroid hormones and thyroid hor-
and influences neuronal proliferation and migration of neurones mone-dependent gliogenesis and myelination continues (1, 7, 8).
in the cerebral cortex, hippocampus and medial ganglionic emi-
nence (9–12). The second stage occurs during the remainder of
Maternal and foetal thyroid physiology
pregnancy after the onset of foetal thyroid function when the
developing brain derives its supply of thyroid hormones from Before the onset of foetal thyroid function, low levels of T4 and
both the foetus and the mother (4–6, 8). During this period, 3,5,3¢-L-triiodothyronine (T3) of maternal origin are present in
thyroid hormone dependent processes include neurogenesis, neu- embryonic fluids and tissues, including the brain (13–16). Foetal
rone migration, axonal outgrowth, dendritic branching and syna- T4 and T3 levels correlate with maternal T4 rather than T3 (16),

1st Trimester 2nd Trimester 3rd Trimester Post-natal

14/40 28/40 Term 6/12


Neurone proliferation

Onset of neurone migration

Cortex
Hippocampus
Cochlea
Cerebellum

Myelination
Glial cell proliferation
Synapse formation
Axon & dendrite migration/branching
T4 levels
(pmol/l)
20
10
0
Maternal Maternal + Fetal Neonate
Expression
of TRs Apo-TRs Occupied TRs

Thyroid gland Thyroid hormone


formation production

D3 Fetal TSH D2

1st Trimester 2nd Trimester 3rd Trimester Post-natal

14/40 28/40 Term 6/12

Fig. 1. Relationship between thyroid hormone action and development of the brain. In the first trimester of pregnancy early neuronal proliferation and migra-
tion is dependent on maternal thyroxine (T4). In foetal tissues, inactivating type 3 deiodinase (D3) enzyme expression falls and development of the thyroid
gland commences. By the end of the first trimester, development of the hypothalamic-pituitary axis has occurred and a surge in thyroid-stimulating hormone
(TSH) secretion results in the onset of foetal thyroid hormone production, expression of the activating type 2 iodothyronine deiodinase enzyme (D2) and
increasing occupation of thyroid hormone receptors (TRs) by 3,5,3¢-L-triiodothyronine (T3). Continuing development of the brain in the second and third trimes-
ters relies increasingly on T4 produced by both the foetus and mother. Continued post-natal development is entirely dependent on neonatal thyroid hormone
production. Apo-TR, unliganded unoccupied thyroid hormone receptor.

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
786 G. R. Williams

and T3 derived from maternal or foetal serum barely contributes


Delivery of thyroid hormones to target tissues
to brain T3 levels (13). Importantly, during the first trimester,
there is a surge in maternal T4 accompanied by inhibition of Thyroid hormones are lipophilic and poorly soluble in water. They bind
thyroid-stimulating hormone (TSH) that results from high concen- reversibly to plasma transport proteins of varying affinity, which
trations of placental chorionic gonadotrophin at approximately include thyroxine-binding globulin (TBG), transthyretin (previously
10–12 weeks of gestation (17). This has been interpreted to called thyroxine-binding prealbumin), albumin and various lipopro-
reflect an increase in maternal thyroid hormone production teins. The free-fractions of circulating thyroid hormones are depen-
imposed by the conceptus, which thus ensures an adequate sup- dent on the concentrations and saturations of binding proteins and
ply of maternal T4 to the foetus during the critical period of in normal serum free T4 represents 0.02% of the total T4 concentra-
early thyroid hormone-dependent neurodevelopment (4, 5). This tion, whereas free T3 is 0.3% of total T3 because of its lower affinity
need for increased thyroid hormone production and heightened for TBG. As a result, total circulating concentrations of T4 are 50–
vulnerability to maternal thyroid status also arises because of 60-fold higher than total T3, whereas free T4 levels are only approxi-
the increased volume of distribution of thyroid hormones in mately four-fold higher than free T3. Circulating T4 is derived solely
plasma, their increased metabolism and turnover in pregnancy from synthesis and secretion by the thyroid gland, whereas 80% of
and the effects of oestrogens on thyroid hormone binding pro- circulating T3 is produced in peripheral tissues by enzymatic removal
teins. Accordingly, maternal hypothyroxinaemia rather than of an outer ring 5¢-iodine atom from T4. Both T4 and T3 are trans-
T3-deficiency prior to the onset of foetal thyroid function results ported into target tissues with equal efficiency and do not compete
in abnormal neuronal migration and neocortical cyto-architecture for uptake. Until recently, the mechanism of cellular entry of free thy-
(12, 18). The necessity for increased thyroid hormone require- roid hormones was not clear but was presumed to occur by passive
ments in pregnancy has also been highlighted by clinical studies diffusion because of their lipophilic nature (26). In fact, the hormones
demonstrating the need for increased thyroid hormone replace- enter target cells and cross the placenta via an energy-dependent,
ment in pregnant women with hypothyroidism (19, 20). ATP-requiring, stereospecific and saturable transport mechanism that
is mediated by the monocarboxylate transporter-8 (MCT8) (27) and
other transporter proteins such as OATP1c1, a member of the Na+-
Utero–placental transfer of thyroid hormones
independent organic anion transporter protein (OATP) family (28, 29).
Surprisingly, levels of total T4 in foetal fluids are 100-fold lower
than levels in maternal serum, and T3 concentrations are lower
Thyroid hormone action
still (15, 16). This paradox is resolved by the finding that free
T4 levels in the foetus are comparable to maternal concentrations Thyroid hormone receptors (TRs) bind T3 with high affinity and
(15). The free T4 fraction in the foetus is higher than in the function as ligand-inducible transcription factors that regulate
mother because of differences in T4 binding proteins. Thus, the expression of T3-responsive target genes. TRa and TRb are members
availability of free T4 to foetal tissues is dependent on maternal of the steroid ⁄ thyroid hormone receptor superfamily and were orig-
T4 levels and decreases significantly in situations of maternal hyp- inally cloned in 1986 (30, 31). The TRa gene (THRA, NR1A1) encodes
othyroxinaemia. By analogy, if the total thyroid hormone levels in three C-terminal variants in mammals: TRa1 binds T3 and DNA and
the mother and foetus were similar, foetal tissues would be is a functional receptor, whereas TRa2 and TRa3 do not bind
exposed to elevated levels of free T4 and free T3 (4, 5) that are T3 and are weak dominant negative antagonists in vitro, although
also detrimental to the critical temporal sequence of thyroid hor- their roles in vivo are unclear (32). A promoter that generates two
mone responses during foetal development (21). These consider- truncated variants, TRDa1 and TRDa2, has also been identified in
ations underlie the requirement for an efficient but incomplete intron 7 of murine Thra. TRDa1 and TRDa2 are potent repressors
utero–placental barrier to maternal thyroid hormone transfer that of TR function in vitro but their physiological significance
limits supplies and ensures that ‘euthyroid’ free hormone concen- is also unknown (33). The TRb gene (THRB, NR1A2) encodes two
trations are maintained in foetal fluids and tissues. Following the N-terminal variants in all vertebrates TRb1 and TRb2, both of which
onset of foetal thyroid hormone production, levels of total and are functional receptors. Additional variants, TRb3 and TRDb3, are
free T3 remain very low in the foetus compared to the mother, expressed in rats but their physiological role is not known (34, 35).
whereas total and free T4 concentrations reach adult levels by the Unlike most nuclear receptors, unliganded apo-TRs compete with
beginning of the third trimester (22). Despite the increasing con- liganded TRs for DNA response elements and act as potent repres-
centrations of T4 in the foetus as gestation progresses, the foetal sors that exert important physiological roles during the develop-
thyroid reserve remains low and the gland does not mature fully ment of specific tissues including the brain (36–39). Apo-TRs
until birth (23). Thus, maternal thyroid hormones continue to con- interact with co-repressor proteins, which recruit histone deacety-
tribute to foetal T4 levels until birth, as demonstrated in neonates lases and maintain a nonpermissive chromatin structure to inhibit
who cannot synthesise their own thyroid hormones because of gene transcription. By contrast, liganded TRs bind to coactivators in
complete organification defects (24). Likewise, hypothyroxinaemia a T3-dependent manner. Coactivators possess intrinsic histone ace-
in premature babies results from a complete absence of maternal tyl transferase activity, which facilitates formation of permissive
T4 and may account in part for their increased risk of cerebral nucleosomes and activation of gene expression. Thus, the opposing
palsy and neurological deficit (3, 4, 25). effects of unoccupied and occupied TRs result in a greatly increased

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
Thyroid hormone action in the brain 787

amplitude of transcriptional response to T3 (32, 37). TRa1 and TRb1 ties acts as a critical homeostatic regulator of T3 availability to the
are expressed in most tissues but their relative abundance varies cell nucleus even at extremes of thyroid function. In the brain, D2
and this may represent a mechanism for control of T3 action in a activity is markedly up-regulated in the presence of low thyroid
temporo-spatial specific manner (40). Although circulating levels of hormone levels (54), whereas D3 activity is strongly decreased (55).
free T4 are four-fold higher than free T3, the TR has at least a This adaptation is thought to protect the susceptible brain from
15-fold greater affinity for T3 (41), indicating that T4 is a prohor- normal fluctuations in circulating thyroid hormone levels as well as
mone that must be converted to T3 prior to the onset of thyroid to some extent protecting from the detrimental effects of hypothy-
hormone action (42). roidism or hyperthyroidism, particularly during development (13, 29,
56). Thus, local regulation of thyroid status in specific regions of
the brain is achieved by the coordinated regional expression of the
Thyroid hormone metabolism
D2 and D3 enzymes (57) and T3 homeostasis results from compen-
The iodothyronine deiodinases are selenoenzymes that activate or satory reciprocal changes in activities of the D2 and D3 enzymes
inactivate thyroid hormones. The type 2 deiodinase (D2) catalyses (44, 58, 59).
removal of an outer ring iodine atom from the prohormone T4 with
an apparent Michaelis constant (Km) of 10)9M to generate the
Control of T3 availability and action in the brain
active product T3. By contrast, the D3 isozyme inactivates T3, or
prevents T4 being activated, by catalysing removal of an inner ring The main cellular site of T3 action in the brain is the neurone, but
iodine with a Km of 10)9M to generate 3,3¢-diiodothyronine (T2) or T3 must gain access to neurones by a circuitous route that is sub-
3,3¢,5¢-triiodothyronine (reverse T3, rT3) respectively. The D1 enzyme ject to local regulation (29, 60, 61) (Fig. 2). The highly specific iodo-
is inefficient with a Km of 10)6–10)7M and catalyses removal of thyronine transporters OATP1c1 and MCT8 are both expressed in
inner or outer ring iodine atoms in equimolar proportions to gener- the central nervous system (CNS) and, in the last 5 years, our
ate T3, rT3 or T2 depending on the substrate. Most circulating T3 is
derived from T4 by the actions of D1 in liver and kidney, although
D2 in skeletal muscle also contributes (42–44). Nevertheless, the T4
OATP1c1
primary action of D2 is to determine the intra-cellular concentra-
tion of T3 and level of saturation of the nuclear TR. Its lower Km Blood Endothelium
T4 T3
enables efficient local generation of T3 at times of T4 deprivation
and D2 is thought to protect vital structures from periods of hypo- OATP1c1 ?
thyroidism. Accordingly, T4 treatment of cells coexpressing the T4
T4 Astrocyte
MCT8 transporter and D2 activating enzyme results in increased T3 T3 CP D2 T3
MCT8 ?
production and hormone responsiveness (27), demonstrating a
functional link between thyroid hormone uptake and metabolism in T3
the regulation of T3-action. By contrast, the inactivating D3 iso- TR
zyme prevents thyroid hormone access to specific tissues at critical Neurone
T4 T4 T3 T3 T3
times and reduces TR-saturation (42, 44). MCT8 D2
MCT8 D3
Astrocyte T2

Tissue thyroid status CSF


T3
D3 is expressed in foetal tissues and the placenta where it initially
prevents maternal thyroid hormone access to the developing foetus
(45). At this time, unoccupied TRs are critical factors that generally Tanycyte
maintain cell proliferation and prevent differentiation (37, 46). The
sharp rise in T3 availability at birth in mammals is analogous to the Fig. 2. Delivery of thyroid hormones to neurones. Circulating thyroid hor-
mones enter the cerebrospinal fluid via the choroid plexus, which expresses
T3-dependent metamorphosis climax in amphibians (47) and a simi-
both monocarboxylate transporter-8 (MCT8) and Na+-independent organic
lar period at hatching in birds (48), and depends on tightly regu- anion transporter protein 1c1 (OATP1c1) thyroid hormone-specific trans-
lated temporo-spatial expression of D2 and D3 (49). Increased porter proteins. The prohormone, T4 is transported across the blood–brain
pituitary D2 expression correlates with maturation of the hypotha- barrier via OATP1c1 in endothelial cells or MCT8 in tanycytes lining the third
lamic-pituitary-thyroid (HPT) axis whereas its expression in ventricle. Thyroxine (T4) enters glial cells including astrocytes via an
T3-target tissues, concomitant with reduced expression of D3, unknown mechanism and is activated to 3,5,3¢-L-triiodothyronine (T3) via
results in conversion of unoccupied TRs into occupied TRs and the the activating type 2 iodothyronine deiodinase (D2) enzyme. T3 is exported
from glial cells by an unknown transporter to facilitate MCT8-dependent
initiation of cell differentiation (46, 47, 50–53) (Fig. 1). Thus, the TR
entry into neurones. T3 may also enter neurones directly from blood or
acts as a deiodinase-dependent developmental switch that regu- cerebrospinal fluid (CSF) by a poorly defined route. T3 acts via thyroid hor-
lates maturation of T3-dependent tissues. Expression of D2 is mone receptors (TRs) expressed in neurones or is metabolised by the inacti-
increased in hypothyroidism whereas D3 expression is increased in vating type 3 deiodinase (D3) enzyme to inactive 3,3¢-diiodothyronine (T2).
thyrotoxicosis, ensuring that the balance between D2 and D3 activi- CP, choroid plexus;?, unknown transporter protein.

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
788 G. R. Williams

understanding of thyroid hormone uptake into the brain has pro- or modestly elevated TSH. The characteristic abnormalities are
gressed remarkably. Most likely, control of local T3 levels in brain accompanied by a severe, X-linked, psychomotor retardation syn-
tissue takes place in functional units of astrocytes and neurones drome that presents in early childhood and is thought to result
(62). T4 (and T3) must first cross the blood–brain barrier, and the from defective neuronal T3 entry causing abnormal T3 action and
T4-specific transporter OATP1c1 is ideally placed to achieve this as metabolism (62). There is global developmental delay including poor
it is expressed at high levels in capillary endothelium throughout communication skills, no speech development, poor head control,
the CNS (63, 64). Some T3 transport across the blood–brain barrier mental retardation and varying degrees of truncal hypotonia, athe-
may also be mediated by MCT8 (65). Thyroid hormones also enter toid movements and motor deficiency, which may include spastic
via the choroid plexus–cerobrospinal fluid (CSF) barrier. OATP-1 and quadriplegia in severe cases. The neurological phenotype of X-linked
MCT-8 are expressed in choroid plexus and likely to mediate trans- psychomotor retardation in patients with SLC16A2 mutations clo-
port of T4 (65), whilst MCT8 and D2 are coexpressed in tanycytes sely resembles Allan–Herndon–Dudley syndrome (OMIM 300523),
lining the third ventricle and facilitate access of thyroid hormones originally described in 1944 (62, 76–79).
to hypothalamic nuclei and thyrotrophin-releasing hormone (TRH)
neurones (56, 65–67). Having entered the CNS, T4 is taken up by
MCT8-deficient mice
astrocytes via an unidentified transporter, where it is converted to
T3 via local activity of D2 (66, 67). T3 generated in astrocytes is To investigate the role of MCT8, two laboratories generated MCT8-
then transported out of the cell via an unknown transporter before deficient mice (80, 81). Similar to patients with SLC16A2 mutations,
active uptake into neurones is mediated via MCT8 (65, 67). T3 then MCT8-deficient mice had increased circulating T3, reduced T4 and
exerts its major actions directly in neurones by regulating expres- reverse T3 with modestly elevated or normal levels of TSH. Hepatic
sion of T3-target genes (2, 68, 69). T3 is finally metabolised and D1 activity was increased and an important contributor to increased
degraded by D3 in neurones (59, 67). This model, which proposes circulating T3 (80, 81). MCT8-deficient mice had reduced uptake of
that T3 supply to neurones is dependent on D2 activity in astro- radiolabelled T3 into brain tissue, whereas T4 entry was not
cytes, has been challenged recently by observations in mutant mice impaired (81). Nevertheless, tissue concentrations of T4 and T3 in
lacking D2. Compared with wild-type littermates D2-knockout mice brain were reduced. Despite increased D2 activity and reduced D3
have a very mild neurological phenotype, indicating that T3 supply clearance (80, 81), the presence of tissue hypothyroidism demon-
to the brain can also be obtained directly from serum and CSF to strates that increased local T3 production and decreased T3 catabo-
compensate for the lack of D2 activity in astrocytes (70). lism only compensates partially for MCT8 deficiency (81). Despite
these metabolic changes, no neurological abnormalities were
observed in MCT8-deficient mice, suggesting there are only minor
Thyroid hormone transport in the brain
consequences to impaired neuronal T3 entry in development of the
In situ hybridisation studies have shown MCT8 mRNA is expressed murine CNS. In particular, the cerebellum, which is especially sensi-
at high levels in choroid plexus, olfactory bulb, cerebral cortex, hip- tive to thyroid hormones during development, was unaffected with
pocampus and amygdala, at moderate levels in striatum and cere- normal proliferation, migration and dendrite formation in Pukinje
bellum and at low levels in some neuroendocrine nuclei. and granule cells (81). Nevertheless, markedly elevated hypotha-
Colocalisation studies revealed that MCT8 is predominantly lamic TRH expression that did not respond to injected T3 was iden-
expressed in neurones. Together with a spatiotemporal pattern of tified in MCT8 mutants, confirming the presence of impaired
MCT8 expression during the perinatal period, these data indicate neuronal T3 entry. By contrast, TRH was inhibited by T4 injection,
that MCT8 plays an important role in CNS development by trans- indicating that TRH neurones still respond to T4, which acts
porting thyroid hormones into neurones (65). MCT8 is also presumably following D2-mediated local conversion of T4 to T3.
expressed in pituitary folliculo-stellate cells (71), the same cells that Similarly, pituitary TSH responsiveness to T3 was impaired in MCT8-
express TSH receptor and may be involved in ultra-short feedback deficient mice (80, 81). These data suggest the presence of
control of TSH secretion (72, 73). In support of this view, MCT8 additional neuronal thyroid hormone transporters in developing
protein is expressed in human hypothalamic paraventricular, supra- mouse brain (29, 60).
optic and infundibular nuclei and in the lining of ependymal cells
of the third ventricle, which are all locations involved in negative
Thyroid hormone metabolism in the brain
feedback of TRH (67).

Type 2 deiodinase
MCT8 mutations in humans
The activating D2 enzyme is expressed in glial cells, third ventricle
A key physiological role for thyroid hormone transport was con- tanycytes, astrocytes and some sensory neurones including nuclei
firmed in patients with mutations in SLC16A2 (previously MCT8) within the trigeminal, auditory and visual pathways (56, 58, 66).
located on chromosome Xq13.2 (74, 75). Affected boys have an Understanding of key neurodevelopmental roles for D2 has come
unusual thyroid status characterised by elevated T3 concentrations from a series of elegant studies in mice. In the cochlea, D2 is
with reduced T4 and reverse T3 levels and inappropriately normal expressed in periosteal connective tissue surrounding the internal

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
Thyroid hormone action in the brain 789

sensory tissues, with enzyme activity peaking before the onset of to TRH and TSH, respectively (85, 86). Accordingly, neonatal
hearing. TR expression, however, is localised to the cochlea sensory D3-deficient mice, which have elevated circulating T3 but low T4
epithelium, suggesting that periosteal D2 provides a spatiotempo- levels, show evidence of tissue thyrotoxicosis in the brain that is
rally regulated supply of T3 to the sensory epithelium that is neces- accompanied by increased D2 activity (presumably due to low
sary for correct timing of the development and maturation of the circulating T4 levels). This increase in D2 activity in the face of
cochlea (50). This hypothesis was supported by the finding that elevated circulating T3 concentrations results in increased tissue T3
D2-deficient mice exhibit delayed cochlea development and defec- levels and increased expression of T3-target genes (85), confirming
tive auditory function despite circulating levels of thyroid hormones the key role for D3 in regulation of tissue thyroid status during
that normally are permissive for development of hearing. Thus, brain development and its intimate reciprocal relationship with the
D2-dependent local generation of T3 to the cochlea is essential for activating enzyme D2.
auditory function (52). In this case, the activating D2-enzyme func-
tions as a local amplifier of T3 action to regulate sensory develop-
Thyroid hormone receptors in the brain
ment. By contrast, the inactivating D3 enzyme can influence
spatiotemporal development of sensory pathways by inhibiting
TR expression
T3 action locally. For example, D3 regulates localised asymmetrical
growth of the dorsal retina during development of the eye in Xeno- In the brain, TRs are expressed prior to the onset of foetal thyroid
pus by reducing local T3 concentrations to inhibit T3-dependent hormone production (87). TRa1 is the major isoform expressed during
proliferation of lateral projecting ganglion cells (82). foetal life (68, 87) but, prior to birth, there is increased expression of
D2-deficient mice also have elevated circulating T4 and TSH lev- TRb1 (87, 88), which is distributed widely as development proceeds
els but normal T3 concentrations and display an impaired negative (68, 69, 89). Nevertheless, TRa has been estimated to account for
feedback TSH response to T4 but not T3, demonstrating that D2 is 70–80% of total TR expression in the brain (90). TRa1 and TRb1 also
required for local T3 generation in the pituitary and essential for exhibit differential spatiotemporal expression in neurones throughout
normal control of the HPT axis (83). Additionally, neonatal D2 the post-natal and adult brain (68), suggesting discrete roles for the
knockout mice have 25–50% reduced tissue T3 concentrations two isoforms during development and in the mature CNS. For exam-
throughout the brain, although levels of expression of T3-target ple, in cerebellum, TRa1 is expressed in granular cells whereas both
genes (RC3, TrkB, Hairless, Srg1) were either unaffected or much TRa1 and TRb1 are present in Purkinje cells. Accordingly, T3 acts via
less affected when compared to alterations in hypothyroid brain TRa1 to regulate granular cell migration and via both TRa1 and TRb1
(70). Consequently, and although D2 expression is increased mark- to control Purkinje cell differentiation (91). Similarly, differences in
edly in neonatal rat brain during the critical period of neuronal levels of TRa1 and TRb1 expression in GABAergic interneurones in
development (49), D2-deficient mice exhibit a very mild general cerebral cortex and hippocampus correlate with behavioural pheno-
neurological phenotype compared with abnormalities seen in hypo- types characterised in TR mutant mice (92). Furthermore TRa1, but
thyroid mice. Thus, although neuronal T3 is thought to be derived not TRb, has been shown to regulate the onset of oligodendrocyte
primarily from D2-dependent metabolism of T4 in glial cells, these precursor cell differentiation and control the timing of oligodendro-
findings suggest compensatory mechanisms can ameliorate the cyte maturation and migration in the optic nerve (93, 94). The actions
neurological consequences of D2 deficiency and that other sources of TRa1 and TRb1 in the brain, however, are not necessarily discrete;
of T3 are available to the brain during its development (70) (Fig. 2). recent studies have revealed cooperative interactions between the
Expression of MCT8 in cerebral cortex and cerebellum was similar in two isoforms during astrocyte maturation (95). Expression of the
wild-type, D2 knockout and hypothyroid mice, indicating that com- TRb2 isoform, by contrast, is localised and restricted to the hypothal-
pensation for D2 deficiency is unlikely to involve increased MCT8- amus (69, 96–98), anterior pituitary (99, 100), developing cochlea
mediated T3 uptake (70). (101) and neural retina (102).

Type 3 deiodinase Regulation of the HPT axis by TRs


The inactivating D3 enzyme is highly expressed in the developing In the hypothalamus and pituitary, TRb controls the HPT axis. Mice
rat brain (49) and is also present in neurones throughout the adult lacking all TRb isoforms or harbouring a dominant negative knockin
rat brain especially in pyramidal cells of the hippocampus, granule mutation of Thrb display the biochemical features of resistance to
cells in the dentate and in cerebral cortex (59). In early studies, hetero- thyroid hormone (RTH) seen in patients with mutations in THRB
geneous levels of D3 enzyme activity were detected throughout the (103–108). TRb knockout mice or mice expressing dominant-nega-
brain (84). Studies in D3-deficient mice have also revealed a critical tive TRb proteins display defective HPT axis regulation in both
role for this enzyme in maturation and activity of the HPT axis at hypothalamus and pituitary resulting in elevated T3 and T4 concen-
the levels of the hypothalamus, pituitary and thyroid gland. As a trations and inappropriately normal or elevated TSH (109, 110).
result, neonatal D3-deficient mice are thyrotoxic because of delayed Gene transfer experiments also demonstrate an important role for
T3 clearance, whereas, after post-natal day 15, mutants exhibit cen- TRb in the regulation of TRH (111, 112). More specifically, TRb2-
tral hypothyroidism with impaired pituitary and thyroid responses deficient mice exhibit a similar degree of central RTH to mice with

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
790 G. R. Williams

complete deletion of all TRb isoforms, with defects also demon- a novel role for TRa1 in the development and maintenance of
strated at the level of both pituitary and hypothalamus (96, 104). behavioural responses and motor coordination and may provide new
Together, these data indicate a major role for TRb2 in regulation insight into the molecular pathogenesis of neuronal damage result-
and determination of the set-point of the HPT axis in vivo. ing from congenital hypothyroidism or endemic cretinism.

TRs and sensory development The future


In the auditory system, although the organ of Corti within the New details regarding the molecular basis for thyroid hormone
cochlea expresses both TR isoforms, TRb has the major functional transport, metabolism and action has resulted in a transformation
role. TRb knockout but not TRa1-deficient mice display abnormal of our understanding of neurological development and behaviour.
cochlear development and defective auditory function (113, 114), There are many new opportunities for the future, however, as
although additional deletion of TRa1 in compound mutants wors- understanding remains in its infancy. For example, most of the
ens the cochlear abnormalities to reveal a cooperative role for TRa1 T3-target genes identified in the brain are regulated during early
(113, 115). Both TRb1 and TRb2 are expressed in similar patterns in development (1) and a remaining challenge will be to identify
the cochlea but mice with selective deletion of TRb2 display normal T3-regulated genes in the post-natal period and those that are
auditory function (104, 116), indicating a primary role for TRb1 or associated with behavioural responses in adults. Understanding of
the ability of TRb1 to compensate for lack of TRb2 in development the role of MCT8 in the brain is advanced; the precise role of
of hearing. TRb-deficient mice also display increased susceptibility OATP1c1 is less clear but abundant evidence suggests the presence
to audiogenic seizures (117). The THRB gene is also important for of additional transporters mediating thyroid hormone influx and
hearing in humans as severe deafness was described in a family efflux, especially in glial cells. The neurobiology of thyroid hormone
with recessive RTH caused by a deletion of THRB, whereas milder transporters will be an expanding field in the near future. New
sensori-neural deafness is a well-described feature in patients with insights into TR-isoform specific actions in different regions of the
autosomal dominant RTH due to dominant negative mutations in brain have developed alongside an understanding of TR-isoform
THRB (108). In the retina, TRb2 is expressed in developing photore- specific actions in other tissues. These breakthroughs have led to
ceptors and deletion results in a selective loss of M-cones but pres- the development of TR-isoform selective analogues (122). Thus, the
ervation of normal total cone number. All cones in TRb2 knockout TRb-selective agonists GC-1 and KB2115 have beneficial effects on
mice are S-opsin expressing, indicating that TRb2 is essential for cholesterol and lipid metabolism but lack the detrimental effects of
commitment of a sub-population of developing cones to the T3 to the heart and skeleton (123, 124). As a result, safety and effi-
M-opsin expressing phenotype (116, 118). cacy studies have begun with KB2115 in humans (124). TRa1
antagonists have the potential for the selective treatment of car-
diac arrhythmias or the prevention of bone loss (40, 122, 125).
TRs and behaviour
Despite this progress, behavioural and motor defects identified in
Behavioural analysis of TR mutant mice has provided new insight TRa1 mutant mice and behavioural abnormalities in TRb mutants
into the regulation of cognition, memory and emotion by thyroid emphasise the need for further characterisation of the role of TR
hormones. Transgenic mice expressing a dominant-negative TRb dis- isoforms in the adult brain. The effects of TR analogues on behav-
play stereotypic behavioural features reminiscent of attention-defi- iour will be an important avenue for the future. Along these lines,
cit-hyperactivity disorder (119), a condition that also occurs in it has always been intriguing that patients with RTH have muta-
human RTH (108). Female TR knockout mice display abnormal mat- tions restricted to the THRB gene encoding TRb (108). No individu-
ing; TRa1-deficient mice have suppression of female receptiveness, als with mutations in THRA have been defined. Close analysis of
whereas this behaviour is enhanced in TRb mutants (120). In the unexpected neurological and behavioural phenotypes of TRa1
behavioural tests, including the open field test, Morris water maze, mutant mice may lead to the identification of individuals with
Y maze and contextual fear conditioning, TRb knockout mice THRA mutations and provide a place for TRs in the arena of molec-
perform normally (107). By contrast, TRa1 deficient mice display ular psychiatry.
freezing responses to open-field testing and a poor response to con- This review has focused on the traditional genomic mechanisms
textual fear conditioning. These findings suggest TRa1 deficient mice of thyroid hormone action mediated via nuclear receptors. Space
display high emotionality and negative memory persistence (92). has not allowed discussion of nongenomic and rapid membrane
Studies in mice with a point mutation in TRa1 (121) revealed evi- responses to thyroid hormones (126, 127). These have hardly been
dence of anxiety with reduced exploratory behaviour and memory studied in the brain, but it is likely they will have important contri-
deficiencies, which are defects that could be improved following butions for the future. There is excitement ahead for thyroid
T3 administration at very specific time periods (38). Further analysis research in neuroendocrinology!
demonstrated abnormalities of motor skills and locomotion in adults
that were dependent on adequate supplies of thyroid hormone dur- Received: 13 March 2008,
ing early and late foetal development as well as continuing supplies revised 27 March 2008,
in the post-natal period and adulthood (39). These recent data reveal accepted 28 March 2008

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
Thyroid hormone action in the brain 791

during pregnancy in women with hypothyroidism. N Engl J Med 2004;


References 351: 241–249.
1 Bernal J, Guadano-Ferraz A, Morte B. Perspectives in the study of thy- 20 Mandel SJ. Hypothyroidism and chronic autoimmune thyroiditis in the
roid hormone action on brain development and function. Thyroid 2003; pregnant state: maternal aspects. Best Pract Res Clin Endocrinol Metab
13: 1005–1012. 2004; 18: 213–224.
2 Bernal J. Thyroid hormone receptors in brain development and function. 21 Anselmo J, Cao D, Karrison T, Weiss RE, Refetoff S. Fetal loss associated
Nat Clin Pract Endocrinol Metab 2007; 3: 249–259. with excess thyroid hormone exposure. JAMA 2004; 292: 691–695.
3 Zoeller RT, Rovet J. Timing of thyroid hormone action in the developing 22 Thorpe-Beeston JG, Nicolaides KH, Felton CV, Butler J, McGregor AM.
brain: clinical observations and experimental findings. J Neuroendocri- Maturation of the secretion of thyroid hormone and thyroid-stimulat-
nol 2004; 16: 809–818. ing hormone in the fetus. N Engl J Med 1991; 324: 532–536.
4 de Escobar GM, Obregon MJ, del Rey FE. Maternal thyroid hormones 23 van den Hove MF, Beckers C, Devlieger H, de Zegher F, De Nayer P. Hor-
early in pregnancy and fetal brain development. Best Pract Res Clin mone synthesis and storage in the thyroid of human preterm and term
Endocrinol Metab 2004; 18: 225–248. newborns: effect of thyroxine treatment. Biochimie 1999; 81: 563–570.
5 Morreale de Escobar G, Obregon MJ, Escobar del Rey F. Role of thyroid 24 Vulsma T, Gons MH, de Vijlder JJ. Maternal-fetal transfer of thyroxine
hormone during early brain development. Eur J Endocrinol. 2004; 151 in congenital hypothyroidism due to a total organification defect or
Suppl 3u25–37. thyroid agenesis. N Engl J Med 1989; 321: 13–16.
6 Obregon MJ, Calvo RM, Del Rey FE, de Escobar GM. Ontogenesis of thy- 25 LaFranchi S. Thyroid function in the preterm infant. Thyroid 1999; 9:
roid function and interactions with maternal function. Endocr Dev. 71–78.
2007; 10: 86–98. 26 Friesema EC, Jansen J, Milici C, Visser TJ. Thyroid hormone transporters.
7 Porterfield SP, Hendrich CE. The role of thyroid hormones in prenatal Vitam Horm 2005; 70: 137–167.
and neonatal neurological development – current perspectives. Endocr 27 Friesema EC, Ganguly S, Abdalla A, Manning Fox JE, Halestrap AP, Visser
Rev 1993; 14: 94–106. TJ. Identification of monocarboxylate transporter 8 as a specific thyroid
8 Morreale de Escobar G, Obregon MJ, Escobar del Rey F. Is neuropsycho- hormone transporter. J Biol Chem 2003; 278: 40128–40135.
logical development related to maternal hypothyroidism or to maternal 28 Jansen J, Friesema EC, Milici C, Visser TJ. Thyroid hormone transporters
hypothyroxinemia? J Clin Endocrinol Metab 2000; 85: 3975–3987. in health and disease. Thyroid 2005; 15: 757–768.
9 Narayanan CH, Narayanan Y. Cell formation in the motor nucleus and 29 Heuer H. The importance of thyroid hormone transporters for brain
mesencephalic nucleus of the trigeminal nerve of rats made hypothy- development and function. Best Pract Res Clin Endocrinol Metab 2007;
roid by propylthiouracil. Exp Brain Res 1985; 59: 257–266. 21: 265–276.
10 Lucio RA, Garcia JV, Ramon Cerezo J, Pacheco P, Innocenti GM, Berbel 30 Sap J, Munoz A, Damm K, Goldberg Y, Ghysdael J, Leutz A, Beug
P. The development of auditory callosal connections in normal and H, Vennstrom B. The c-erb-A protein is a high-affinity receptor for thy-
hypothyroid rats. Cereb Cortex 1997; 7: 303–316. roid hormone. Nature 1986; 324: 635–640.
11 Cuevas E, Auso E, Telefont M, Morreale de Escobar G, Sotelo C, Berbel 31 Weinberger C, Thompson CC, Ong ES, Lebo R, Gruol DJ, Evans RM. The
P. Transient maternal hypothyroxinemia at onset of corticogenesis c-erb-A gene encodes a thyroid hormone receptor. Nature 1986; 324:
alters tangential migration of medial ganglionic eminence-derived neu- 641–646.
rons. Eur J Neurosci 2005; 22: 541–551. 32 Harvey CB, Williams GR. Mechanism of thyroid hormone action. Thyroid
12 Auso E, Lavado-Autric R, Cuevas E, Del Rey FE, Morreale De Escobar G, 2002; 12: 441–446.
Berbel P. A moderate and transient deficiency of maternal thyroid func- 33 Chassande O, Fraichard A, Gauthier K, Flamant F, Legrand C, Savatier P,
tion at the beginning of fetal neocorticogenesis alters neuronal migra- Laudet V, Samarut J. Identification of transcripts initiated from an
tion. Endocrinology 2004; 145: 4037–4047. internal promoter in the c-erbA alpha locus that encode inhibitors of
13 Calvo R, Obregon MJ, Ruiz de Ona C, Escobar del Rey F, Morreale de retinoic acid receptor-alpha and triiodothyronine receptor activities.
Escobar G. Congenital hypothyroidism, as studied in rats. Crucial role of Mol Endocrinol 1997; 11: 1278–1290.
maternal thyroxine but not of 3,5,3¢-triiodothyronine in the protection 34 Williams GR. Cloning and characterization of two novel thyroid hor-
of the fetal brain. J Clin Invest 1990; 86: 889–899. mone receptor beta isoforms. Mol Cell Biol 2000; 20: 8329–8342.
14 Woods RJ, Sinha AK, Ekins RP. Uptake and metabolism of thyroid hor- 35 Harvey CB, Bassett JH, Maruvada P, Yen PM, Williams GR. The rat thy-
mones by the rat foetus in early pregnancy. Clin Sci (Lond) 1984; 67: roid hormone receptor (TR) Deltabeta3 displays cell-, TR isoform-, and
359–363. thyroid hormone response element-specific actions. Endocrinology
15 Calvo RM, Jauniaux E, Gulbis B, Asuncion M, Gervy C, Contempre B, 2007; 148: 1764–1773.
Morreale de Escobar G. Fetal tissues are exposed to biologically relevant 36 Hashimoto K, Curty FH, Borges PP, Lee CE, Abel ED, Elmquist JK, Cohen
free thyroxine concentrations during early phases of development. J RN, Wondisford FE. An unliganded thyroid hormone receptor causes
Clin Endocrinol Metab 2002; 87: 1768–1777. severe neurological dysfunction. Proc Natl Acad Sci USA 2001; 98:
16 Contempre B, Jauniaux E, Calvo R, Jurkovic D, Campbell S, de Escobar GM. 3998–4003.
Detection of thyroid hormones in human embryonic cavities during the 37 Chassande O. Do unliganded thyroid hormone receptors have physio-
first trimester of pregnancy. J Clin Endocrinol Metab 1993; 77: 1719–1722. logical functions? J Mol Endocrinol 2003; 31: 9–20.
17 Glinoer D. The regulation of thyroid function in pregnancy: pathways 38 Venero C, Guadano-Ferraz A, Herrero AI, Nordstrom K, Manzano J,
of endocrine adaptation from physiology to pathology. Endocr Rev de Escobar GM, Bernal J, Vennstrom B. Anxiety, memory impairment, and
1997; 18: 404–433. locomotor dysfunction caused by a mutant thyroid hormone receptor alpha1
18 Lavado-Autric R, Auso E, Garcia-Velasco JV, Arufe Mdel C, Escobar del can be ameliorated by T3 treatment. Genes Dev 2005; 19: 2152–2163.
Rey F, Berbel P, Morreale de Escobar G. Early maternal hypothyroxin- 39 Wallis K, Sjogren M, van Hogerlinden M, Silberberg G, Fisahn A, Nord-
emia alters histogenesis and cerebral cortex cytoarchitecture of the strom K, Larsson L, Westerblad H, Morreale de Escobar G, Shupliakov O,
progeny. J Clin Invest 2003; 111: 1073–1082. Vennstrom B. Locomotor deficiencies and aberrant development of sub-
19 Alexander EK, Marqusee E, Lawrence J, Jarolim P, Fischer GA, Larsen PR. type-specific GABAergic interneurons caused by an unliganded thyroid
Timing and magnitude of increases in levothyroxine requirements hormone receptor alpha1. J Neurosci 2008; 28: 1904–1915.

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
792 G. R. Williams

40 O’Shea PJ, Bassett JH, Cheng SY, Williams GR. Characterization of skele- deiodinases in different areas. J Clin Endocrinol Metab 2004; 89: 3117–
tal phenotypes of TRalpha1 and TRbeta mutant mice: implications for 3128.
tissue thyroid status and T3 target gene expression. Nucl Recept Signal 58 Tu HM, Kim SW, Salvatore D, Bartha T, Legradi G, Larsen PR, Lechan
2006; 4: E011. RM. Regional distribution of type 2 thyroxine deiodinase messenger
41 Lin KH, Fukuda T, Cheng SY. Hormone and DNA binding activity of a ribonucleic acid in rat hypothalamus and pituitary and its regulation by
purified human thyroid hormone nuclear receptor expressed in Escheri- thyroid hormone. Endocrinology 1997; 138: 3359–3368.
chia coli. J Biol Chem 1990; 265: 5161–5165. 59 Tu HM, Legradi G, Bartha T, Salvatore D, Lechan RM, Larsen PR. Regio-
42 Bianco AC, Kim BW. Deiodinases: implications of the local control of nal expression of the type 3 iodothyronine deiodinase messenger ribo-
thyroid hormone action. J Clin Invest 2006; 116: 2571–2579. nucleic acid in the rat central nervous system and its regulation by
43 Maia AL, Kim BW, Huang SA, Harney JW, Larsen PR. Type 2 iodothyro- thyroid hormone. Endocrinology 1999; 140: 784–790.
nine deiodinase is the major source of plasma T3 in euthyroid humans. 60 Visser WE, Friesema EC, Jansen J, Visser TJ. Thyroid hormone transport
J Clin Invest 2005; 115: 2524–2533. by monocarboxylate transporters. Best Pract Res Clin Endocrinol Metab
44 Bianco AC, Salvatore D, Gereben B, Berry MJ, Larsen PR. Biochemistry, 2007; 21: 223–236.
cellular and molecular biology, and physiological roles of the iodothyro- 61 Fliers E, Alkemade A, Wiersinga WM, Swaab DF. Hypothalamic thyroid
nine selenodeiodinases. Endocr Rev 2002; 23: 38–89. hormone feedback in health and disease. Prog Brain Res 2006; 153:
45 Wasco EC, Martinez E, Grant KS, St Germain EA, St Germain DL, Galton 189–207.
VA. Determinants of iodothyronine deiodinase activities in rodent 62 Friesema EC, Jansen J, Heuer H, Trajkovic M, Bauer K, Visser TJ. Mecha-
uterus. Endocrinology 2003; 144: 4253–4261. nisms of disease: psychomotor retardation and high T3 levels caused
46 Flamant F, Poguet AL, Plateroti M, Chassande O, Gauthier K, Streichen- by mutations in monocarboxylate transporter 8. Nat Clin Pract Endocri-
berger N, Mansouri A, Samarut J. Congenital hypothyroid Pax8() ⁄ )) nol Metab 2006; 2: 512–523.
mutant mice can be rescued by inactivating the TRalpha gene. Mol 63 Sugiyama D, Kusuhara H, Taniguchi H, Ishikawa S, Nozaki Y, Aburatani
Endocrinol 2002; 16: 24–32. H, Sugiyama Y. Functional characterization of rat brain-specific organic
47 Huang H, Cai L, Remo BF, Brown DD. Timing of metamorphosis and the anion transporter (Oatp14) at the blood–brain barrier: high affinity
onset of the negative feedback loop between the thyroid gland and the transporter for thyroxine. J Biol Chem 2003; 278: 43489–43495.
pituitary is controlled by type II iodothyronine deiodinase in Xenopus 64 Tohyama K, Kusuhara H, Sugiyama Y. Involvement of multispecific
laevis. Proc Natl Acad Sci USA 2001; 98: 7348–7353. organic anion transporter, Oatp14 (Slc21a14), in the transport of thyrox-
48 Van der Geyten S, Van den Eynde I, Segers IB, Kuhn ER, Darras VM. Dif- ine across the blood-brain barrier. Endocrinology 2004; 145: 4384–4391.
ferential expression of iodothyronine deiodinases in chicken tissues 65 Heuer H, Maier MK, Iden S, Mittag J, Friesema EC, Visser TJ, Bauer K.
during the last week of embryonic development. Gen Comp Endocrinol The monocarboxylate transporter 8 linked to human psychomotor retar-
2002; 128: 65–73. dation is highly expressed in thyroid hormone-sensitive neuron popula-
49 Bates JM, St Germain DL, Galton VA. Expression profiles of the three tions. Endocrinology 2005; 146: 1701–1706.
iodothyronine deiodinases, D1, D2, and D3, in the developing rat. Endo- 66 Guadano-Ferraz A, Obregon MJ, St Germain DL, Bernal J. The type 2
crinology 1999; 140: 844–851. iodothyronine deiodinase is expressed primarily in glial cells in the neo-
50 Campos-Barros A, Amma LL, Faris JS, Shailam R, Kelley MW, Forrest D. natal rat brain. Proc Natl Acad Sci USA 1997; 94: 10391–10396.
Type 2 iodothyronine deiodinase expression in the cochlea before the 67 Alkemade A, Friesema EC, Unmehopa UA, Fabriek BO, Kuiper GG, Leon-
onset of hearing. Proc Natl Acad Sci USA 2000; 97: 1287–1292. ard JL, Wiersinga WM, Swaab DF, Visser TJ, Fliers E. Neuroanatomical
51 Mai W, Janier MF, Allioli N, Quignodon L, Chuzel T, Flamant F, Samarut pathways for thyroid hormone feedback in the human hypothalamus. J
J. Thyroid hormone receptor alpha is a molecular switch of cardiac Clin Endocrinol Metab 2005; 90: 4322–4334.
function between fetal and postnatal life. Proc Natl Acad Sci USA 68 Mellstrom B, Naranjo JR, Santos A, Gonzalez AM, Bernal J. Independent
2004; 101: 10332–10337. expression of the alpha and beta c-erbA genes in developing rat brain.
52 Ng L, Goodyear RJ, Woods CA, Schneider MJ, Diamond E, Richardson Mol Endocrinol 1991; 5: 1339–1350.
GP, Kelley MW, Germain DL, Galton VA, Forrest D. Hearing loss and 69 Bradley DJ, Towle HC, Young WS III. Spatial and temporal expression of
retarded cochlear development in mice lacking type 2 iodothyronine alpha- and beta-thyroid hormone receptor mRNAs, including the beta
deiodinase. Proc Natl Acad Sci USA 2004; 101: 3474–3479. 2-subtype, in the developing mammalian nervous system. J Neurosci
53 Sachs LM, Damjanovski S, Jones PL, Li Q, Amano T, Ueda S, Shi YB, 1992; 12: 2288–2302.
Ishizuya-Oka A. Dual functions of thyroid hormone receptors during 70 Galton VA, Wood ET, St Germain EA, Withrow CA, Aldrich G, St Germain
Xenopus development. Comp Biochem Physiol B Biochem Mol Biol GM, Clark AS, St Germain DL. Thyroid hormone homeostasis and action
2000; 126: 199–211. in the type 2 deiodinase-deficient rodent brain during development.
54 Burmeister LA, Pachucki J, St Germain DL. Thyroid hormones inhibit type Endocrinology 2007; 148: 3080–3088.
2 iodothyronine deiodinase in the rat cerebral cortex by both pre- and 71 Alkemade A, Friesema EC, Kuiper GG, Wiersinga WM, Swaab DF, Visser
posttranslational mechanisms. Endocrinology 1997; 138: 5231–5237. TJ, Fliers E. Novel neuroanatomical pathways for thyroid hormone
55 Friedrichsen S, Christ S, Heuer H, Schafer MK, Mansouri A, Bauer K, action in the human anterior pituitary. Eur J Endocrinol 2006; 154:
Visser TJ. Regulation of iodothyronine deiodinases in the Pax8) ⁄ ) 491–500.
mouse model of congenital hypothyroidism. Endocrinology 2003; 144: 72 Prummel MF, Brokken LJ, Wiersinga WM. Ultra short-loop feedback
777–784. control of thyrotropin secretion. Thyroid 2004; 14: 825–829.
56 Guadano-Ferraz A, Escamez MJ, Rausell E, Bernal J. Expression of type 73 Brokken LJ, Bakker O, Wiersinga WM, Prummel MF. Functional thyrotro-
2 iodothyronine deiodinase in hypothyroid rat brain indicates an impor- pin receptor expression in the pituitary folliculo-stellate cell line TtT ⁄ GF.
tant role of thyroid hormone in the development of specific primary Exp Clin Endocrinol Diabetes 2005; 113: 13–20.
sensory systems. J Neurosci 1999; 19: 3430–3439. 74 Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S. A novel
57 Kester MH, Martinez de Mena R, Obregon MJ, Marinkovic D, Howatson syndrome combining thyroid and neurological abnormalities is associ-
A, Visser TJ, Hume R, Morreale de Escobar G. Iodothyronine levels in ated with mutations in a monocarboxylate transporter gene. Am J Hum
the human developing brain: major regulatory roles of iodothyronine Genet 2004; 74: 168–175.

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
Thyroid hormone action in the brain 793

75 Friesema EC, Grueters A, Biebermann H, Krude H, von Moers A, Reeser 92 Guadano-Ferraz A, Benavides-Piccione R, Venero C, Lancha C, Venn-
M, Barrett TG, Mancilla EE, Svensson J, Kester MH, Kuiper GG, Balkassmi strom B, Sandi C, DeFelipe J, Bernal J. Lack of thyroid hormone receptor
S, Uitterlinden AG, Koehrle J, Rodien P, Halestrap AP, Visser TJ. Associa- alpha1 is associated with selective alterations in behavior and hippo-
tion between mutations in a thyroid hormone transporter and severe campal circuits. Mol Psychiatry 2003; 8: 30–38.
X-linked psychomotor retardation. Lancet 2004; 364: 1435–1437. 93 Billon N, Jolicoeur C, Tokumoto Y, Vennstrom B, Raff M. Normal timing
76 Holden KR, Zuniga OF, May MM, Su H, Molinero MR, Rogers RC, Sch- of oligodendrocyte development depends on thyroid hormone receptor
wartz CE. X-linked MCT8 gene mutations: characterization of the pedi- alpha 1 (TRalpha1). EMBO J 2002; 21: 6452–6460.
atric neurologic phenotype. J Child Neurol 2005; 20: 852–857. 94 Billon N, Tokumoto Y, Forrest D, Raff M. Role of thyroid hormone
77 Schwartz CE, May MM, Carpenter NJ, Rogers RC, Martin J, Bialer MG, receptors in timing oligodendrocyte differentiation. Dev Biol 2001; 235:
Ward J, Sanabria J, Marsa S, Lewis JA, Echeverri R, Lubs HA, Voeller K, 110–120.
Simensen RJ, Stevenson RE. Allan–Herndon–Dudley syndrome and the 95 Morte B, Manzano J, Scanlan TS, Vennstrom B, Bernal J. Aberrant matu-
monocarboxylate transporter 8 (MCT8) gene. Am J Hum Genet 2005; ration of astrocytes in thyroid hormone receptor alpha 1 knockout mice
77: 41–53. reveals an interplay between thyroid hormone receptor isoforms. Endo-
78 Schwartz CE, Stevenson RE. The MCT8 thyroid hormone transporter and crinology 2004; 145: 1386–1391.
Allan–Herndon–Dudley syndrome. Best Pract Res Clin Endocrinol Metab 96 Abel ED, Ahima RS, Boers ME, Elmquist JK, Wondisford FE. Critical role
2007; 21: 307–321. for thyroid hormone receptor beta2 in the regulation of paraventricular
79 Refetoff S, Dumitrescu AM. Syndromes of reduced sensitivity to thyroid thyrotropin-releasing hormone neurons. J Clin Invest 2001; 107: 1017–
hormone: genetic defects in hormone receptors, cell transporters and 1023.
deiodination. Best Pract Res Clin Endocrinol Metab 2007; 21: 277–305. 97 Lechan RM, Qi Y, Jackson IM, Mahdavi V. Identification of thyroid hor-
80 Dumitrescu AM, Liao XH, Weiss RE, Millen K, Refetoff S. Tissue-specific mone receptor isoforms in thyrotropin-releasing hormone neurons of
thyroid hormone deprivation and excess in monocarboxylate transporter the hypothalamic paraventricular nucleus. Endocrinology 1994; 135:
(mct) 8-deficient mice. Endocrinology 2006; 147: 4036–4043. 92–100.
81 Trajkovic M, Visser TJ, Mittag J, Horn S, Lukas J, Darras VM, Raivich G, 98 Cook CB, Kakucska I, Lechan RM, Koenig RJ. Expression of thyroid hor-
Bauer K, Heuer H. Abnormal thyroid hormone metabolism in mice lacking mone receptor beta 2 in rat hypothalamus. Endocrinology 1992; 130:
the monocarboxylate transporter 8. J Clin Invest 2007; 117: 627–635. 1077–1079.
82 Marsh-Armstrong N, Huang H, Remo BF, Liu TT, Brown DD. Asymmetric 99 Wood WM, Ocran KW, Gordon DF, Ridgway EC. Isolation and character-
growth and development of the Xenopus laevis retina during metamor- ization of mouse complementary DNAs encoding alpha and beta thy-
phosis is controlled by type III deiodinase. Neuron 1999; 24: 871–878. roid hormone receptors from thyrotrope cells: the mouse pituitary-
83 Schneider MJ, Fiering SN, Pallud SE, Parlow AF, St Germain DL, Galton specific beta 2 isoform differs at the amino terminus from the corre-
VA. Targeted disruption of the type 2 selenodeiodinase gene (DIO2) sponding species from rat pituitary tumor cells. Mol Endocrinol 1991;
results in a phenotype of pituitary resistance to T4. Mol Endocrinol 5: 1049–1061.
2001; 15: 2137–2148. 100 Hodin RA, Lazar MA, Wintman BI, Darling DS, Koenig RJ, Larsen PR,
84 Kaplan MM, McCann UD, Yaskoski KA, Larsen PR, Leonard JL. Anatomi- Moore DD, Chin WW. Identification of a thyroid hormone receptor that
cal distribution of phenolic and tyrosyl ring iodothyronine deiodinases is pituitary-specific. Science 1989; 244: 76–79.
in the nervous system of normal and hypothyroid rats. Endocrinology 101 Bradley DJ, Towle HC, Young WS III. Alpha and beta thyroid hormone
1981; 109: 397–402. receptor (TR) gene expression during auditory neurogenesis: evidence
85 Hernandez A, Martinez ME, Fiering S, Galton VA, St Germain D. Type 3 for TR isoform-specific transcriptional regulation in vivo. Proc Natl
deiodinase is critical for the maturation and function of the thyroid Acad Sci USA 1994; 91: 439–443.
axis. J Clin Invest 2006; 116: 476–484. 102 Sjoberg M, Vennstrom B, Forrest D. Thyroid hormone receptors in chick
86 Hernandez A, Martinez ME, Liao XH, Van Sande J, Refetoff S, Galton retinal development: differential expression of mRNAs for alpha and
VA, St Germain DL. Type 3 deiodinase deficiency results in functional N-terminal variant beta receptors. Development 1992; 114: 39–47.
abnormalities at multiple levels of the thyroid axis. Endocrinology 2007; 103 Kaneshige M, Kaneshige K, Zhu X, Dace A, Garrett L, Carter TA,
148: 5680–5687. Kazlauskaite R, Pankratz DG, Wynshaw-Boris A, Refetoff S, Weintraub
87 Forrest D, Hallbook F, Persson H, Vennstrom B. Distinct functions for B, Willingham MC, Barlow C, Cheng S. Mice with a targeted mutation
thyroid hormone receptors alpha and beta in brain development indi- in the thyroid hormone beta receptor gene exhibit impaired growth and
cated by differential expression of receptor genes. EMBO J 1991; 10: resistance to thyroid hormone. Proc Natl Acad Sci USA 2000; 97:
269–275. 13209–13214.
88 Forrest D, Sjoberg M, Vennstrom B. Contrasting developmental and tis- 104 Abel ED, Boers ME, Pazos-Moura C, Moura E, Kaulbach H, Zakaria M,
sue-specific expression of alpha and beta thyroid hormone receptor Lowell B, Radovick S, Liberman MC, Wondisford F. Divergent roles for
genes. EMBO J 1990; 9: 1519–1528. thyroid hormone receptor beta isoforms in the endocrine axis and audi-
89 Strait KA, Schwartz HL, Perez-Castillo A, Oppenheimer JH. Relationship tory system. J Clin Invest 1999; 104: 291–300.
of c-erbA mRNA content to tissue triiodothyronine nuclear binding 105 Gothe S, Wang Z, Ng L, Kindblom JM, Barros AC, Ohlsson C, Vennstrom
capacity and function in developing and adult rats. J Biol Chem 1990; B, Forrest D. Mice devoid of all known thyroid hormone receptors are
265: 10514–10521. viable but exhibit disorders of the pituitary-thyroid axis, growth, and
90 Schwartz HL, Strait KA, Ling NC, Oppenheimer JH. Quantitation of rat bone maturation. Genes Dev 1999; 13: 1329–1341.
tissue thyroid hormone binding receptor isoforms by immunoprecipita- 106 Gauthier K, Chassande O, Plateroti M, Roux JP, Legrand C, Pain B,
tion of nuclear triiodothyronine binding capacity. J Biol Chem 1992; Rousset B, Weiss R, Trouillas J, Samarut J. Different functions for the
267: 11794–11799. thyroid hormone receptors TRalpha and TRbeta in the control of thyroid
91 Morte B, Manzano J, Scanlan T, Vennstrom B, Bernal J. Deletion of the hormone production and post-natal development. EMBO J 1999; 18:
thyroid hormone receptor alpha 1 prevents the structural alterations of 623–631.
the cerebellum induced by hypothyroidism. Proc Natl Acad Sci USA 107 Forrest D, Hanebuth E, Smeyne RJ, Everds N, Stewart CL, Wehner JM,
2002; 99: 3985–3989. Curran T. Recessive resistance to thyroid hormone in mice lacking

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794
794 G. R. Williams

thyroid hormone receptor beta: evidence for tissue-specific modulation 118 Applebury ML, Farhangfar F, Glosmann M, Hashimoto K, Kage K, Rob-
of receptor function. EMBO J 1996; 15: 3006–3015. bins JT, Shibusawa N, Wondisford FE, Zhang H. Transient expression of
108 Weiss RE, Refetoff S. Resistance to thyroid hormone. Rev Endocr Metab thyroid hormone nuclear receptor TRbeta2 sets S opsin patterning dur-
Disord 2000; 1: 97–108. ing cone photoreceptor genesis. Dev Dyn 2007; 236: 1203–1212.
109 Abel ED, Kaulbach HC, Campos-Barros A, Ahima RS, Boers ME, Hashimoto 119 Wong R, Vasilyev VV, Ting YT, Kutler DI, Willingham MC, Weintraub BD,
K, Forrest D, Wondisford FE. Novel insight from transgenic mice into thy- Cheng S. Transgenic mice bearing a human mutant thyroid hormone
roid hormone resistance and the regulation of thyrotropin. J Clin Invest beta 1 receptor manifest thyroid function anomalies, weight reduction,
1999; 103: 271–279. and hyperactivity. Mol Med 1997; 3: 303–314.
110 Abel ED, Moura EG, Ahima RS, Campos-Barros A, Pazos-Moura CC, Bo- 120 Dellovade TL, Chan J, Vennstrom B, Forrest D, Pfaff DW. The two thy-
ers ME, Kaulbach HC, Forrest D, Wondisford FE. Dominant inhibition of roid hormone receptor genes have opposite effects on estrogen-stimu-
thyroid hormone action selectively in the pituitary of thyroid hormone lated sex behaviors. Nat Neurosci 2000; 3: 472–475.
receptor-beta null mice abolishes the regulation of thyrotropin by thy- 121 Tinnikov A, Nordstrom K, Thoren P, Kindblom JM, Malin S, Rozell B,
roid hormone. Mol Endocrinol 2003; 17: 1767–1776. Adams M, Rajanayagam O, Pettersson S, Ohlsson C, Chatterjee K,
111 Dupre SM, Guissouma H, Flamant F, Seugnet I, Scanlan TS, Baxter JD, Vennstrom B. Retardation of post-natal development caused by a
Samarut J, Demeneix BA, Becker N. Both thyroid hormone receptor negatively acting thyroid hormone receptor alpha1. EMBO J 2002; 21:
(TR)beta 1 and TR beta 2 isoforms contribute to the regulation of hypo- 5079–5087.
thalamic thyrotropin-releasing hormone. Endocrinology 2004; 145: 122 Flamant F, Gauthier K, Samarut J. Thyroid hormones signaling is getting
2337–2345. more complex: STORMs are coming. Mol Endocrinol 2007; 21: 321–333.
112 Guissouma H, Becker N, Seugnet I, Demeneix BA. Transcriptional repres- 123 Johansson L, Rudling M, Scanlan TS, Lundasen T, Webb P, Baxter J,
sion of TRH promoter function by T3: analysis by in vivo gene transfer. Angelin B, Parini P. Selective thyroid receptor modulation by GC-1
Biochem Cell Biol 2000; 78: 155–163. reduces serum lipids and stimulates steps of reverse cholesterol trans-
113 Rusch A, Ng L, Goodyear R, Oliver D, Lisoukov I, Vennstrom B, Richard- port in euthyroid mice. Proc Natl Acad Sci USA 2005; 102: 10297–
son G, Kelley MW, Forrest D. Retardation of cochlear maturation and 10302.
impaired hair cell function caused by deletion of all known thyroid hor- 124 Berkenstam A, Kristensen J, Mellstrom K, Carlsson B, Malm J, Rehnmark
mone receptors. J Neurosci 2001; 21: 9792–9800. S, Garg N, Andersson CM, Rudling M, Sjoberg F, Angelin B, Baxter JD.
114 Forrest D, Erway LC, Ng L, Altschuler R, Curran T. Thyroid hormone The thyroid hormone mimetic compound KB2115 lowers plasma LDL
receptor beta is essential for development of auditory function. Nat cholesterol and stimulates bile acid synthesis without cardiac effects in
Genet 1996; 13: 354–357. humans. Proc Natl Acad Sci USA 2008; 105: 663–667.
115 Gauthier K, Plateroti M, Harvey CB, Williams GR, Weiss RE, Refetoff S, 125 Bassett JH, O’Shea PJ, Sriskantharajah S, Rabier B, Boyde A, Howell PG,
Willott JF, Sundin V, Roux JP, Malaval L, Hara M, Samarut J, Chassande O. Weiss RE, Roux JP, Malaval L, Clement-Lacroix P, Samarut J, Chassande
Genetic analysis reveals different functions for the products of the O, Williams GR. Thyroid hormone excess rather than thyrotropin defi-
thyroid hormone receptor alpha locus. Mol Cell Biol 2001; 21: 4748–4760. ciency induces osteoporosis in hyperthyroidism. Mol Endocrinol 2007;
116 Ng L, Hurley JB, Dierks B, Srinivas M, Salto C, Vennstrom B, Reh TA, 21: 1095–1107.
Forrest D. A thyroid hormone receptor that is required for the 126 Bassett JH, Harvey CB, Williams GR. Mechanisms of thyroid hormone
development of green cone photoreceptors. Nat Genet 2001; 27: 94–98. receptor-specific nuclear and extra nuclear actions. Mol Cell Endocrinol
117 Ng L, Pedraza PE, Faris JS, Vennstrom B, Curran T, Morreale de Escobar 2003; 213: 1–11.
G, Forrest D. Audiogenic seizure susceptibility in thyroid hormone 127 Davis PJ, Leonard JL, Davis FB. Mechanisms of nongenomic actions of
receptor beta-deficient mice. Neuroreport 2001; 12: 2359–2362. thyroid hormone. Front Neuroendocrinol. 2008; 29: 211–218.

ª 2008 The Author. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20, 784–794

Potrebbero piacerti anche