Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

1

REVIEW

Thyroid hormones and fetal neurological development


J Patel1,2, K Landers2, H Li2, R H Mortimer1,2,3 and K Richard1,2
1
School of Medicine, The University of Queensland, Herston, 4006 Brisbane, Queensland, Australia
2
Conjoint Endocrine Laboratory, Queensland Institute of Medical Research, Bancroft Centre, Royal Brisbane and Women’s Hospital, 300 Herston Road, Herston,
4029 Brisbane, Queensland, Australia
3
Disciplines of Medicine, Obstetrics and Gynaecology, The University of Queensland, Herston, 4006 Brisbane, Queensland, Australia
(Correspondence should be addressed to J Patel at Conjoint Endocrine Laboratory, Queensland Institute of Medical Research, Bancroft Centre, Royal Brisbane
and Women’s Hospital; Email: [email protected])

Abstract
The development of fetal thyroid function is dependent on such as the cerebral cortex at specific developmental stages.
the embryogenesis, differentiation, and maturation of the There are many known cell membrane thyroid hormone
thyroid gland. This is coupled with evolution of the transporters in fetal brain that play an essential role in
hypothalamic–pituitary–thyroid axis and thyroid hormone regulating thyroid hormone concentrations in key structures.
metabolism, resulting in the regulation of thyroid hormone They also provide the route for intracellular thyroid
action, production, and secretion. Throughout gestation hormone interaction with associated thyroid hormone
there is a steady supply of maternal thyroxine (T4) which has receptors, which activate their action. There is a growing
been observed in embryonic circulation as early as 4 weeks body of experimental evidence from rats and humans to
post-implantation. This is essential for normal early fetal suggest that even mild maternal hypothyroxinemia may lead
neurogenesis. Triiodothyronine concentrations remain very to abnormalities in fetal neurological development. Our
low during gestation due to metabolism via placental and review will focus on the ontogeny of thyroid hormone in
fetal deiodinase type 3. T4 concentrations are highly fetal development, with a focus on cell membrane
regulated to maintain low concentrations, essential for transporters and TR action in the brain.
protecting the fetus and reaching key neurological sites Journal of Endocrinology (2011) 209, 1–8

Introduction (HPT) axis and thyroid hormone metabolism by liver and


brain deiodinase enzymes (deiodinase type 2 (D2) and
While the impaired mental and physical development of deiodinase type 3 (D3)) to ensure basal levels are sustained
inhabitants of the iodine-deficient Alps was recognized as (Zoeller et al. 2007)).
cretinism hundreds of years ago, the link between iodine The fetal thyroid gland reaches maturity by week 11–12,
deficiency, hypothyroidism, and impaired neurologic close to the end of the first trimester and begins to secrete
development was made very much later (Cranefield 1962, thyroid hormones by about week 16 (Obregon et al. 2007).
Morreale de Escobar et al. 2004). Thyroid hormones During this period, an adequate supply of maternal thyroid
(thyroxine (T4) and triiodothyronine (T3)) are essential for hormones must be sustained to ensure normal neurological
the development and maintenance of normal physiological development. Hypothyroid fetuses suffer various postnatal
processes, especially those of the central nervous system, disorders including mental retardation, deafness, and spasti-
where thyroid hormones assist in brain maturation through- city. Severe iodine deficiency, which causes both maternal and
out gestation (Joffe & Sokolov 1994, Neale et al. 2007). fetal hypothyroidism, is, worldwide, the most common cause
Thyroid hormones primarily regulate genes involved in of mental retardation (Glinoer 2001, Morreale de Escobar
myelination and neuronal\glial cell differentiation (Bernal et al. 2004, Pearce 2009). If thyroid hormone replacement
2005). Delivery of thyroid hormones to the fetal brain is a for congenital hypothyroid babies is not initiated soon after
complex process requiring, at different times, expression of birth, further impairment of cognitive development occurs.
brain thyroid hormone receptors (TRs), materno-fetal More recent evidence suggests that even mild reductions in
thyroid hormone and iodide transport, an intricate system maternal thyroid hormone levels in early pregnancy are
of endocrine feedback (the hypothalamic–pituitary–thyroid associated with reduced IQ in offspring (LaFranchi & Austin

Journal of Endocrinology (2011) 209, 1–8 DOI: 10.1530/JOE-10-0444


0022–0795/11/0209–001 q 2011 Society for Endocrinology Printed in Great Britain Online version via http://www.endocrinology-journals.org
2 J PATEL and others . Thyroid hormone and neurological development

2007, Gyamfi et al. 2009). The molecular mechanisms by This emphasizes the need for maternal T4 levels to be
which thyroid hormones affect fetal neurological structures maintained to ensure normal fetal brain development. This
are still not well understood. This review will briefly discuss also explains why even minimally reduced maternal T4 levels
some current aspects of the role of thyroid hormones in fetal in early pregnancy can result in adverse outcomes to the
neurological development, focusing on TH transporters and offspring (Lavado-Autric et al. 2003, Auso et al. 2004).
receptor activation in the brain.

Deiodination of thyroid hormones


Ontogenesis of thyroid hormone action in fetal
development As mentioned above, locally generated T3 in the brain from
maternally transported T4 has been reported to be essential for
There is growing evidence that thyroid hormones act on normal early brain development (Zoeller 2010). Almost
embryological and fetal tissues early in development. Thyroid 80% of brain T3 is produced locally by D2 (Crantz et al. 1982).
hormone and associated receptors are already found in human D2 is found almost exclusively in astrocytes, whereas TRs
fetal tissues prior to the production and secretion of fetal are highly expressed in oligodendrocytes and neurons
thyroid hormones at 16–18 weeks of gestation, as evidenced (Guadano-Ferraz et al. 1997). In the fetal rat, D2 expression
by detection of T4 and T3 in the human cerebral cortex by is first seen at E16.5 and increases steadily until P15. In human
week 12 gestation (Calvo et al. 2002, Kester et al. 2004). This brain, D2 expression is first detectable in the cerebral cortex
is confirmation that active transport of maternal thyroid in the first trimester of pregnancy at the same time that T3 can
hormone across the placenta is occurring during this crucial be measured there (Chan et al. 2002). D2 is largely responsible
period of gestation and highlights the need for maternal for maintaining appropriate concentrations of T3 during fetal
thyroid hormones to be at optimal levels at that time (Fig. 1; brain development (Guadano-Ferraz et al. 1999). D2 knock-
Bernal 2007). Following onset of active T4 secretion by the out (KO) mice also demonstrate impaired cochlea and visual
fetus, levels of T4 in fetal tissues parallel those in fetal plasma. development, poor thermal regulation, reduced anxiety, and
T4 levels are, however, low, reflecting active type 3 crucially pituitary resistance to T4, further highlighting the
deiodination in the fetus (Ruiz de Ona et al. 1988, Obregon importance of D2 in developmental processes (Obregon et al.
et al. 2007). D3 converts T4 to the biologically inactive reverse 1991). In cases of hypothyroidism, D2 expression and activity
T3. In contrast to tissue T4 levels, T3 concentrations vary in are up-regulated, enhancing T3 supply, whereas in hyperthy-
different tissues, for example levels are low in fetal liver and roidism, the opposite is true (Burmeister et al. 1997).
plasma and high in brain and brown adipose tissue (Obregon Interestingly, however, a more recent study in D2KO mice
et al. 2007). These differences have been attributed to demonstrated that locomotor activity and learning and
variations in the activity of the D2, which converts T4 to memory skills were normal despite low local T3 generation
biologically active T3. This suggests an important role for T3 (Galton et al. 2007).
in brain developmental and maturation processes. D3 is also Raised brain levels of thyroid hormone in the fetus can also
active in placenta, ensuring the fetus is not exposed to cause neurological damage. D3 plays an important role in fetal
excessive amounts of maternal T4 (Galton 2005). neurological development by ensuring that safe levels of
Much of the information known about thyroid hormones thyroid hormones are maintained (Gereben et al. 2008). D3 is
and brain development has been derived from rodent highly expressed in neurons and in contrast to D2 is positively
experiments. As in humans, normal neurological develop- regulated by thyroid hormone. D3KO mice demonstrate
ment depends on thyroid hormone. TRs and deiodinase significant increases in perinatal mortality and an abnormal
enzymes are expressed in the early brain before the thyroid HPT axis (Hernandez et al. 2006, 2007). Furthermore, D3KO
gland develops (Obregon et al. 2007). The critical time period mice are born hypothyroid with impaired growth and fertility
for thyroid hormone action in rat brain is estimated to extend (Galton 2005). They exhibit excessive T3-responsive gene
from around embryonic day 18 (E18) to postnatal day 21–25 activation during development and reduced activation later in
(P21–25; Porterfield & Hendrich 1993). Abnormalities in life (Horn & Heuer 2010).
brain development in hypothyroid rats are mostly seen in the
postnatal period and are demonstrated by reduced maturation
of key structures such as the cerebellum, where delayed Thyroid hormone transporters in the brain
granular cell migration and Purkinje cell maturation are
prevented (Koibuchi et al. 2003). Although this may suggest Over the last decade, the notion of thyroid hormone uptake
that rat brain is more affected by thyroid hormone action after into cells by passive diffusion has been dismissed following the
birth, it should be recognized that TRs are seen very early in discovery of a number of thyroid hormone cell membrane
rat development (discussed below). Rat studies also indicate transporters. Thyroid hormones are amino acid derivatives,
that the developing brain is dependent on a supply of T4, and several classes of transmembrane transporter proteins can
which is locally deiodinated to T3, and that replacement with transport thyroid hormones, including organic anion
T3 does not adequately replenish brain T3 levels (Calvo et al. 1990). transporters 2 and 3 (Oatp2 and Oatp3), L-type amino acid

Journal of Endocrinology (2011) 209, 1–8 www.endocrinology-journals.org


Thyroid hormone and neurological development . J PATEL and others 3

A Maternal thyroid hormone

Increasing D2 and T3 in cerebral cortex

T3 binding to brain nuclear receptors

Nuclear receptors in brain, liver, lung, kidney, etc.

Thyroid gland Thyroid hormone secretion

B
Neurological cretinism Myxedermatous cretinism

Maternal hypothyroxinemia Prematurity Congenital hypothyrodism

C
Striatum

Neocortex

Cerebellum, dentate gyrus granular cells

Cochlea

Myelination

Glial cell proliferation

Synapse formation

Axon and dendrite sprouting


Neuronal migration

Neuronal proliferation

0 5 10 15 20 25 30 35 40 1 2 3 4
Birth

Weeks Years
Figure 1 (A) The ontogeny of fetal thyroid function and expression of thyroid hormone receptors and deiodinase enzymes during gestation
and early years postnatal are demonstrated. (B) Critical time points during gestation that require thyroid hormone action for fetal
neurological development. Neurological abnormalities can be seen if maternal or fetal hypothyroidism is present during gestation. (C) Rodent
studies, where time-specific actions of thyroid hormones on precise neurological and auditory structures were observed. Adapted image from
Bernal (2007).

transporters (Lat1 and Lat2), and monocarboxylate transpor- Horn & Heuer 2010). The T4-binding protein, transthyretin,
ters 8 and 10 (MCT8 and MCT10) (Abe et al. 1998, Friesema produced in significant amounts by the choroid plexus, has
et al. 2001, 2003, 2008). For thyroid hormone to gain access been implicated in this transport, and transthyretin may be
to the brain during maturation, it must pass through many involved in delivery of CSF thyroid hormone to the brain
different cell types which express the various cell membrane (Patel et al. 2010). T4 transported to the brain via blood
transporters. For rodents, the preferred route of TH entry is circulation passes through endothelial cells and is taken up by
through the cerebral circulation and its blood–brain barrier, astrocytes, probably through the Oatp1c1 cell membrane
with some thyroid hormone entering the cerebrospinal transporter (Fig. 2; Hernandez et al. 2007). Within the
fluid (CSF) via the choroid plexus (Dratman et al. 1991, astrocyte, T4 is deionated via D2 to produce T3, which then

www.endocrinology-journals.org Journal of Endocrinology (2011) 209, 1–8


4 J PATEL and others . Thyroid hormone and neurological development

T4

Oatp1c1

Nucleus

T4

D2 T2
T3
D3
Astrocyte T3
T 3 TR
A
MCT8 mRN

in
Neuron Prote

Figure 2 T4 is transported to the brain thyroid hormone-binding proteins such as transthyretin (TTR), where T4 then
passes out through endothelial cells lining the blood vessels. T4 is rapidly transported through the cell membrane
transporter, Oatp1c1, located on the surface of astrocytes. T4 is then metabolized intracellularly by D2 to T3, where it
can then be transported out from astrocytes by an as yet unidentified cell membrane transporter. Within the brain
parenchyma, T3 is then promptly uptaked by neurons and oligodendrocytes via the MCT8 cell membrane transporter.
Within the cell, T3 can either translocate and bind to thyroid hormone receptors (TRs), resulting in thyroid hormone
action, or be metabolized via D3 to biologically inactive T2.

exits the cell possibly via MCT8 (yet to be elucidated) and is MCT8 expression, cell surface translocation, and specific
taken up via MCT8 by oligodendrocytes and neurons (Fig. 2; substrate transport deficits, consequently producing a
Heuer et al. 2005). complete loss-of-function phenotype in patients (Kinne
In humans, the lack of MCT8 leads to greater neurological et al. 2009). MCT8 may transport other molecules essential
damage than in rodents, suggesting that other but as yet for brain maturation and which could explain the difference
undefined transporters are present in rodents. Lat2 has been between human and rodent brain maturation in the presence
proposed as a candidate, which although expressed in of MCT8 transporter mutations.
developing rat brain is not present in human developing
neurons (Wirth et al. 2009). In humans, the absence of the
MCT8 transporter (the Allan–Herndon–Dudley syndrome) TR genes and activity
results in X-linked moderate to severe mental retardation and
muscle hypotonia and hypoplasia (Dumitrescu et al. 2004, There are two TR genes (THRA and THRB), which encode
Friesema et al. 2004, Schwartz et al. 2005). Raised serum T3 for the four isotypes of TR (TRa1, TRb1, TRb2, and
concentrations and low T4 levels are present, but TSH levels TRb3) (Bernal 2007). The four isotypes all bind to T3 and
are normal suggesting a role for MCT8 in pituitary thyroid DNA and drive intracellular thyroid hormone action. Several
hormone uptake (Friesema et al. 2006). Patients also display isoforms of the TR from alternate RNA splicing are also seen.
altered and delayed maturation of myelination (Namba et al. Two of these are the TRa2 (c-erbAa2) and truncated TRb3
2008). More than two dozen MCT8 gene mutations have (DTRb3), which are known as nonreceptor proteins (Bassett
been reported linked to the X-chromosome (Friesema et al. et al. 2003). TRa2 has a conserved DNA-binding region but
2010). Almost all are missense mutations resulting in reduced is unable to bind T3. Conversely, DTRb3 is able to bind T3

Journal of Endocrinology (2011) 209, 1–8 www.endocrinology-journals.org


Thyroid hormone and neurological development . J PATEL and others 5

but not to DNA. The physiological role of these nonreceptor rhombencephalon (Bradley et al. 1992). It is believed that the
proteins is still unknown (Forrest & Vennstrom 2000, O’Shea TRs found in these regions mediate the biological effects of T3
& Williams 2002, Bernal 2007). that has been locally generated from transported maternal T4,
TRs mediate their actions following homodimerization early in gestation. Conversely, TRb isoforms are expressed
or heterodimerization with retinoic acid receptors (RXR), more postnatally within specific neuronal populations such as
which then bind to specific sequences known as thyroid hippocampal pyramidal and granule cells, paraventricular
response elements (TREs) in the regulatory regions of target hypothalamic neurons, and cerebellar Purkinje cells (Bradley
genes (Wagner et al. 1995, Feng et al. 1998). A multitude of et al. 1989, Horn & Heuer 2010). Studies in TRb KO mice
transcription factors are involved. Without the presence of have shown that these isoforms are predominant in mediating
T3, the unliganded receptor (aporeceptor) recruits corepres- thyroid hormone effects on the development of the vision and
sors, such as nuclear receptor corepressors (NCoR) or auditory systems (Jones et al. 2003).
silencing mediator for retinoic and thyroid receptor There is a growing body of evidence suggesting that
(SMRT) and histone deacetylases, which retain the chromatin deficiencies in TR may not lead to abnormal brain
in a compact repressed position (Bernal 2007). However, in development as seen in hypothyroidism, which suggests that
the presence of T3, the binding of the hormone to TR the lack of the ligand (T3) is more detrimental than being TR
initiates transcription by the release of corepressors deficient (Bernal 2007). This has been demonstrated by Bernal
and recruiting coactivators (steroid receptor coactivators 1, et al. in TRa1 KO mice, which have normal cerebellar
SRC-1), histone acetylases (CREB-binding protein, CBP; development, even though the mice were hypothyroid
p300; and mammalian homolog of the yeast transcriptional (Morte et al. 2002). Another interesting study has demon-
activator GCN5, pCAF), and other mediators, all of which strated that TRa1 deletion did not affect cerebellar granule cell
assist in the access of transcription apparatus to the promoter migration during development, which is now known to take
regions (Fig. 3; Bernal 2007, Cheng et al. 2010). place in the absence of both receptor and T3 (Yacubova &
Both TR genes, THRA and THRB, are found within the Komuro 2002). It has been postulated by Bernal et al. that this
brain. In adult rat brain, more than 70–80% of all TRs result could well demonstrate that in vivo thyroid hormones are
present are in the TRa1 isoform. The receptor proteins are permissive when the TR are present, releasing any blocking
predominantly found in the cerebrum and cerebellum function that TR may be regulating (Bernal 2007). In contrast,
(Ercan-Fang et al. 1996). It has been postulated that the the absence of ligand could lead to aberrant receptor signaling,
TRa1 isoform also plays an important role during fetal brain which has been demonstrated with hypothyroid wild-type
development, both in humans and in rodents. This is animals with cell migration defects. This phenomenon has also
evidenced by detection in fetal rat brain of mRNA E11.5 been demonstrated using pheochromocytoma PC12 cells.
in the neural tube and E12.5 in the diencephalon and ventral When exposed to nerve growth factors, these cells differentiate
into neurons. Specifically, T3 has no effect on this process;
A Repression however, exogenous unliganded TRa1 will block this process
Corepressors T3 unless T3 is added to the culture medium (Munoz et al. 1993).
(CoR)
This has led to the hypothesis that the aporeceptor has
Transcriptional some transcriptional activity that may play an as yet unreco-
RXR TR machinery
gnized role in developmental processes. This also poses the
possibility of the aporeceptor possibly repressing specific
TRE
gene expression until T3 becomes available to the cells in a
B Activation time-specific manner.
Further analysis into the role of TR and brain development
Coactivators
(CoA) has been studied using mouse mutants expressing dominant
T3 Transcriptional
negative TR, where DNA binding is conserved with reduced
RXR TR machinery or absent T3-binding capacity. In two different models of
TRb mutant mice, impaired cerebellar Purkinje cell
TRE development and maturation and motor deficits were
observed, with cerebellar morphology similar to that seen
Figure 3 Schematic representation of thyroid hormone receptor in hypothyroidism (Forrest & Vennstrom 2000). This also
(TR) action. (A) Without the presence of the T3 ligand, corepressors
confirms the dominant role of TRb isoforms within the
(CoR) (nuclear receptor corepressors (NCoR) or silencing mediator
for retinoic and thyroid receptor (SMRT)) and histone deacetylases Purkinje cells of the brain. In TRa mutant models where
are recruited by TR to prevent transcription and translation of target T3-binding affinity was reduced by almost tenfold, mice
genes is demonstrated. (B) A bound TR with T3 results in the showed significant growth retardation and cardiac abnorm-
recruitment of coactivators (CoA) (steroid receptor coactivators 1, alities, with the heterozygous strain also demonstrating similar
SRC-1) and histone acetylases (CREB-binding protein, CBP; p300;
and mammalian homolog of the yeast transcriptional activator cerebellar abnormalities seen in hypothyroidism (Tinnikov
GCN5, pCAF) as well as the release of CoR, resulting in changes in et al. 2002). These mice also displayed striking anxiety-related
expression of target genes is demonstrated. behavior as evidenced by time spent motionless and reduced

www.endocrinology-journals.org Journal of Endocrinology (2011) 209, 1–8


6 J PATEL and others . Thyroid hormone and neurological development

exploratory behavior compared to the wild-type controls Bassett JH, Harvey CB & Williams GR 2003 Mechanisms of thyroid
(Tinnikov et al. 2002). This evidence further strengthens the hormone receptor-specific nuclear and extra nuclear actions. Molecular and
Cellular Endocrinology 213 1–11. (doi:10.1016/j.mce.2003.10.033)
hypothesis that TR play roles in fetal brain development and Bernal J 2005 Thyroid hormones and brain development. Vitamins and
maturation, especially within the hippocampus, and in Hormones 71 95–122. (doi:10.1016/S0083-6729(05)71004-9)
maintaining adult brain function (Bernal 2007). Further Bernal J 2007 Thyroid hormone receptors in brain development and function.
insights into phenotypic differences between these mutant Nature Clinical Practice. Endocrinology & Metabolism 3 249–259. (doi:10.1038/
ncpendmet0424)
mice strains are of particular interest, with additional work now
Bradley DJ, Young WS III & Weinberger C 1989 Differential expression of
being conducted in generating cell-specific TR mutant alpha and beta thyroid hormone receptor genes in rat brain and pituitary.
models (Quignodon et al. 2007, Flamant & Quignodon 2010). PNAS 86 7250–7254. (doi:10.1073/pnas.86.18.7250)
TRs have also been observed within the cytosol, where Bradley DJ, Towle HC & Young WS III 1992 Spatial and temporal expression
they interact with p85, a regulatory subunit of phosphatidyl- of alpha- and beta-thyroid hormone receptor mRNAs, including the beta
2-subtype, in the developing mammalian nervous system. Journal of
inositol 3-kinase (Moeller et al. 2006). This affects the AKT Neuroscience 12 2288–2302.
pathway, resulting in induction of nitric oxide synthesis in Burmeister LA, Pachucki J & St Germain DL 1997 Thyroid hormones inhibit
endothelial cells in response to middle cerebral artery type 2 iodothyronine deiodinase in the rat cerebral cortex by both pre- and
occlusion (Hiroi et al. 2006). This highlights the need for posttranslational mechanisms. Endocrinology 138 5231–5237. (doi:10.1210/
en.138.12.5231)
further investigation into the complex roles that TR play in
Calvo R, Obregon MJ, Ruiz de Ona C, Escobar del Rey F & Morreale de
physiological and pathological processes. Escobar G 1990 Congenital hypothyroidism, as studied in rats. Crucial role
of maternal thyroxine but not of 3,5,3 0 -triiodothyronine in the protection
of the fetal brain. Journal of Clinical Investigation 86 889–899. (doi:10.1172/
JCI114790)
Calvo RM, Jauniaux E, Gulbis B, Asuncion M, Gervy C, Contempre B &
Conclusion Morreale de Escobar G 2002 Fetal tissues are exposed to biologically
relevant free thyroxine concentrations during early phases of development.
Our understanding of the complex processes involved in Journal of Clinical Endocrinology and Metabolism 87 1768–1777. (doi:10.1210/
ensuring normal fetal development is increasing. Knowledge jc.87.4.1768)
Chan S, Kachilele S, McCabe CJ, Tannahill LA, Boelaert K, Gittoes NJ,
of the key relationships between thyroid hormone and brain Visser TJ, Franklyn JA & Kilby MD 2002 Early expression of thyroid
development has progressed significantly over the past decade. hormone deiodinases and receptors in human fetal cerebral cortex. Brain
However, many issues regarding TR and thyroid hormone Research. Developmental Brain Research 138 109–116. (doi:10.1016/
cell membrane transporters have yet to be clarified. S0165-3806(02)00459-5)
Cheng SY, Leonard JL & Davis PJ 2010 Molecular aspects of thyroid hormone
Differences between rodent and human development are
actions. Endocrine Reviews 31 139–170. (doi:10.1210/er.2009-0007)
significant and need to be further explored to ensure that the Cranefield PF 1962 The discovery of cretinism. Bulletin of the History of
relationships between thyroid hormone and human brain Medicine 36 489–511.
development are better understood. Crantz FR, Silva JE & Larsen PR 1982 An analysis of the sources and quantity
of 3,5,3 0 -triiodothyronine specifically bound to nuclear receptors in rat
cerebral cortex and cerebellum. Endocrinology 110 367–375. (doi:10.1210/
Declaration of interest endo-110-2-367)
Dratman MB, Crutchfield FL & Schoenhoff MB 1991 Transport of
iodothyronines from bloodstream to brain: contributions by blood:brain
The authors declare that there is no conflict of interest that could be perceived
and choroid plexus:cerebrospinal fluid barriers. Brain Research 554 229–236.
as prejudicing the impartiality of the research reported.
(doi:10.1016/0006-8993(91)90194-Z)
Dumitrescu AM, Liao XH, Best TB, Brockmann K & Refetoff S 2004 A
novel syndrome combining thyroid and neurological abnormalities is
Funding associated with mutations in a monocarboxylate transporter gene. American
Journal of Human Genetics 74 168–175. (doi:10.1086/380999)
This research did not receive any specific grant from any funding agency in the Ercan-Fang S, Schwartz HL & Oppenheimer JH 1996 Isoform-specific
public, commercial or not-for-profit sector. 3,5,3 0 -triiodothyronine receptor binding capacity and messenger ribonu-
cleic acid content in rat adenohypophysis: effect of thyroidal state and
comparison with extrapituitary tissues. Endocrinology 137 3228–3233.
(doi:10.1210/en.137.8.3228)
Feng W, Ribeiro RC, Wagner RL, Nguyen H, Apriletti JW, Fletterick RJ,
References Baxter JD, Kushner PJ & West BL 1998 Hormone-dependent coactivator
binding to a hydrophobic cleft on nuclear receptors. Science 280 1747–1749.
Abe T, Kakyo M, Sakagami H, Tokui T, Nishio T, Tanemoto M, Nomura H, (doi:10.1126/science.280.5370.1747)
Hebert SC, Matsuno S, Kondo H et al. 1998 Molecular characterization Flamant F & Quignodon L 2010 Use of a new model of transgenic mice to
and tissue distribution of a new organic anion transporter subtype (oatp3) clarify the respective functions of thyroid hormone receptors in vivo. Heart
that transports thyroid hormones and taurocholate and comparison with Failure Reviews 15 117–120. (doi:10.1007/s10741-008-9121-y)
oatp2. Journal of Biological Chemistry 273 22395–22401. (doi:10.1074/jbc. Forrest D & Vennstrom B 2000 Functions of thyroid hormone receptors in
273.35.22395) mice. Thyroid 10 41–52. (doi:10.1089/thy.2000.10.41)
Auso E, Lavado-Autric R, Cuevas E, Del Rey FE, Morreale De Escobar G & Friesema EC, Docter R, Moerings EP, Verrey F, Krenning EP, Hennemann G
Berbel P 2004 A moderate and transient deficiency of maternal thyroid & Visser TJ 2001 Thyroid hormone transport by the heterodimeric human
function at the beginning of fetal neocorticogenesis alters neuronal system L amino acid transporter. Endocrinology 142 4339–4348. (doi:10.
migration. Endocrinology 145 4037–4047. (doi:10.1210/en.2004-0274) 1210/en.142.10.4339)

Journal of Endocrinology (2011) 209, 1–8 www.endocrinology-journals.org


Thyroid hormone and neurological development . J PATEL and others 7

Friesema EC, Ganguly S, Abdalla A, Manning Fox JE, Halestrap AP & Kester MH, Martinez de Mena R, Obregon MJ, Marinkovic D, Howatson A,
Visser TJ 2003 Identification of monocarboxylate transporter 8 as a specific Visser TJ, Hume R & Morreale de Escobar G 2004 Iodothyronine levels in
thyroid hormone transporter. Journal of Biological Chemistry 278 the human developing brain: major regulatory roles of iodothyronine
40128–40135. (doi:10.1074/jbc.M300909200) deiodinases in different areas. Journal of Clinical Endocrinology and Metabolism
Friesema EC, Grueters A, Biebermann H, Krude H, von Moers A, Reeser M, 89 3117–3128. (doi:10.1210/jc.2003-031832)
Barrett TG, Mancilla EE, Svensson J, Kester MH et al. 2004 Association Kinne A, Roth S, Biebermann H, Kohrle J, Gruters A & Schweizer U 2009
between mutations in a thyroid hormone transporter and severe X-linked Surface translocation and tri-iodothyronine uptake of mutant MCT8
psychomotor retardation. Lancet 364 1435–1437. (doi:10.1016/S0140- proteins are cell type-dependent. Journal of Molecular Endocrinology 43
6736(04)17226-7) 263–271. (doi:10.1677/JME-09-0043)
Friesema EC, Jansen J, Heuer H, Trajkovic M, Bauer K & Visser TJ 2006 Koibuchi N, Jingu H, Iwasaki T & Chin WW 2003 Current perspectives on
Mechanisms of disease: psychomotor retardation and high T3 levels caused the role of thyroid hormone in growth and development of cerebellum.
by mutations in monocarboxylate transporter 8. Nature Clinical Practice. Cerebellum 2 279–289. (doi:10.1080/14734220310011920)
Endocrinology and Metabolism 2 512–523. (doi:10.1038/ncpendmet0262) LaFranchi SH & Austin J 2007 How should we be treating children with
Friesema EC, Jansen J, Jachtenberg JW, Visser WE, Kester MH & Visser TJ congenital hypothyroidism? Journal of Pediatric Endocrinology and Metabolism
2008 Effective cellular uptake and efflux of thyroid hormone by human 20 559–578.
monocarboxylate transporter 10. Molecular Endocrinology 22 1357–1369. Lavado-Autric R, Auso E, Garcia-Velasco JV, Arufe Mdel C, Escobar del Rey
(doi:10.1210/me.2007-0112) F, Berbel P & Morreale de Escobar G 2003 Early maternal hypothyrox-
Friesema EC, Visser WE & Visser TJ 2010 Genetics and phenomics of inemia alters histogenesis and cerebral cortex cytoarchitecture of the
thyroid hormone transport by MCT8. Molecular and Cellular Endocrinology progeny. Journal of Clinical Investigation 111 1073–1082. (doi:10.1172/
322 107–113. (doi:10.1016/j.mce.2010.01.016) JCI200316262)
Galton VA 2005 The roles of the iodothyronine deiodinases in mammalian Moeller LC, Cao X, Dumitrescu AM, Seo H & Refetoff S 2006 Thyroid
development. Thyroid 15 823–834. (doi:10.1089/thy.2005.15.823) hormone mediated changes in gene expression can be initiated by
Galton VA, Wood ET, St Germain EA, Withrow CA, Aldrich G, St Germain cytosolic action of the thyroid hormone receptor beta through
GM, Clark AS & St Germain DL 2007 Thyroid hormone homeostasis and the phosphatidylinositol 3-kinase pathway. Nuclear Receptor Signaling
action in the type 2 deiodinase-deficient rodent brain during development. 4 e020. (doi:10.1621/nrs.04020)
Endocrinology 148 3080–3088. (doi:10.1210/en.2006-1727) Morreale de Escobar G, Obregon MJ & Escobar del Rey F 2004 Role of
Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, Simonides WS, thyroid hormone during early brain development. European Journal of
Zeold A & Bianco AC 2008 Cellular and molecular basis of Endocrinology 151 (Supplement 3) U25–37. (doi:10.1530/eje.0.151U025)
deiodinase-regulated thyroid hormone signaling. Endocrine Reviews 29 Morte B, Manzano J, Scanlan T, Vennstrom B & Bernal J 2002 Deletion of
898–938. (doi:10.1210/er.2008-0019) the thyroid hormone receptor alpha 1 prevents the structural alterations
Glinoer D 2001 Pregnancy and iodine. Thyroid 11 471–481. (doi:10.1089/ of the cerebellum induced by hypothyroidism. PNAS 99 3985–3989.
105072501300176426) (doi:10.1073/pnas.062413299)
Guadano-Ferraz A, Escamez MJ, Morte B, Vargiu P & Bernal J 1997 Munoz A, Wrighton C, Seliger B, Bernal J & Beug H 1993 Thyroid hormone
Transcriptional induction of RC3/neurogranin by thyroid hormone: receptor/c-erbA: control of commitment and differentiation in the
differential neuronal sensitivity is not correlated with thyroid hormone neuronal/chromaffin progenitor line PC12. Journal of Cell Biology 121
receptor distribution in the brain. Brain Research. Molecular Brain Research 49 423–438. (doi:10.1083/jcb.121.2.423)
37–44. (doi:10.1016/S0169-328X(97)00119-8) Namba N, Etani Y, Kitaoka T, Nakamoto Y, Nakacho M, Bessho K, Miyoshi
Guadano-Ferraz A, Escamez MJ, Rausell E & Bernal J 1999 Expression of Y, Mushiake S, Mohri I, Arai H et al. 2008 Clinical phenotype and
type 2 iodothyronine deiodinase in hypothyroid rat brain indicates an endocrinological investigations in a patient with a mutation in the MCT8
important role of thyroid hormone in the development of specific primary thyroid hormone transporter. European Journal of Pediatrics 167 785–791.
sensory systems. Journal of Neuroscience 19 3430–3439. (doi:10.1007/s00431-007-0589-6)
Gyamfi C, Wapner RJ & D’Alton ME 2009 Thyroid dysfunction in Neale DM, Cootauco AC & Burrow G 2007 Thyroid disease in pregnancy.
pregnancy: the basic science and clinical evidence surrounding the Clinics in Perinatology 34 543–557, v-vi. (doi:10.1016/j.clp.2007.10.003)
controversy in management. Obstetrics and Gynecology 113 702–707. Obregon MJ, Ruiz de Ona C, Calvo R, Escobar del Rey F
(doi:10.1097/AOG.0b013e3181996fe5) & Morreale de Escobar G 1991 Outer ring iodothyronine deiodinases and
Hernandez A, Martinez ME, Fiering S, Galton VA & St Germain D 2006 thyroid hormone economy: responses to iodine deficiency in the rat
Type 3 deiodinase is critical for the maturation and function of the thyroid fetus and neonate. Endocrinology 129 2663–2673. (doi:10.1210/endo-129-
axis. Journal of Clinical Investigation 116 476–484. (doi:10.1172/JCI26240) 5-2663)
Hernandez A, Martinez ME, Liao XH, Van Sande J, Refetoff S, Galton VA & Obregon MJ, Calvo RM, Del Rey FE & de Escobar GM 2007 Ontogenesis of
St Germain DL 2007 Type 3 deiodinase deficiency results in functional thyroid function and interactions with maternal function. Endocrine
abnormalities at multiple levels of the thyroid axis. Endocrinology 148 Development 10 86–98. (doi:10.1159/000106821)
5680–5687. (doi:10.1210/en.2007-0652) O’Shea PJ & Williams GR 2002 Insight into the physiological actions of
Heuer H, Maier MK, Iden S, Mittag J, Friesema EC, Visser TJ & Bauer K thyroid hormone receptors from genetically modified mice. Journal of
2005 The monocarboxylate transporter 8 linked to human psychomotor Endocrinology 175 553–570. (doi:10.1677/joe.0.1750553)
retardation is highly expressed in thyroid hormone-sensitive neuron Patel J, Landers K, Mortimer RH & Richard K 2010 Regulation of hypoxia
populations. Endocrinology 146 1701–1706. (doi:10.1210/en.2004-1179) inducible factors (HIF) in hypoxia and normoxia during placental
Hiroi Y, Kim HH, Ying H, Furuya F, Huang Z, Simoncini T, Noma K, development. Placenta 31 951–957. (doi:10.1016/j.placenta.2010.08.008)
Ueki K, Nguyen NH, Scanlan TS et al. 2006 Rapid nongenomic actions of Pearce EN 2009 What do we know about iodine supplementation in
thyroid hormone. PNAS 103 14104–14109. (doi:10.1073/pnas. pregnancy? Journal of Clinical Endocrinology and Metabolism 94 3188–3190.
0601600103) (doi:10.1210/jc.2009-1512)
Horn S & Heuer H 2010 Thyroid hormone action during brain development: Porterfield SP & Hendrich CE 1993 The role of thyroid hormones in prenatal
more questions than answers. Molecular and Cellular Endocrinology 315 and neonatal neurological development – current perspectives. Endocrine
19–26. (doi:10.1016/j.mce.2009.09.008) Reviews 14 94–106. (doi:10.1210/edrv-14-1-94)
Joffe RT & Sokolov ST 1994 Thyroid hormones, the brain, and affective Quignodon L, Vincent S, Winter H, Samarut J & Flamant F 2007 A point
disorders. Critical Reviews in Neurobiology 8 45–63. mutation in the activation function 2 domain of thyroid hormone receptor
Jones I, Srinivas M, Ng L & Forrest D 2003 The thyroid hormone receptor alpha1 expressed after CRE-mediated recombination partially recapitulates
beta gene: structure and functions in the brain and sensory systems. Thyroid hypothyroidism. Molecular Endocrinology 21 2350–2360. (doi:10.1210/me.
13 1057–1068. (doi:10.1089/105072503770867228) 2007-0176)

www.endocrinology-journals.org Journal of Endocrinology (2011) 209, 1–8


8 J PATEL and others . Thyroid hormone and neurological development

Ruiz de Ona C, Obregon MJ, Escobar del Rey F & Morreale de Escobar G Mct8, the neuronal T3 transporter mutated in Allan–Herndon–Dudley
1988 Developmental changes in rat brain 5 0 -deiodinase and thyroid syndrome. Journal of Neuroscience 29 9439–9449. (doi:10.1523/JNEUR-
hormones during the fetal period: the effects of fetal hypothyroidism and OSCI.6055-08.2009)
maternal thyroid hormones. Pediatric Research 24 588–594. Yacubova E & Komuro H 2002 Intrinsic program for migration of cerebellar
Schwartz CE, May MM, Carpenter NJ, Rogers RC, Martin J, Bialer MG, granule cells in vitro. Journal of Neuroscience 22 5966–5981.
Ward J, Sanabria J, Marsa S, Lewis JA et al. 2005 Allan–Herndon–Dudley Zoeller RT 2010 New insights into thyroid hormone action in the developing
syndrome and the monocarboxylate transporter 8 (MCT8) gene. American brain: the importance of T3 degradation. Endocrinology 151 5089–5091.
Journal of Human Genetics 77 41–53. (doi:10.1086/431313) (doi:10.1210/en.2010-0926)
Tinnikov A, Nordstrom K, Thoren P, Kindblom JM, Malin S, Rozell B, Zoeller RT, Tan SW & Tyl RW 2007 General background on the
Adams M, Rajanayagam O, Pettersson S, Ohlsson C et al. 2002 Retardation hypothalamic–pituitary–thyroid (HPT) axis. Critical Reviews in Toxicology 37
of post-natal development caused by a negatively acting thyroid hormone 11–53. (doi:10.1080/10408440601123446)
receptor alpha1. EMBO Journal 21 5079–5087. (doi:10.1093/emboj/
cdf523)
Wagner RL, Apriletti JW, McGrath ME, West BL, Baxter JD & Fletterick RJ
1995 A structural role for hormone in the thyroid hormone receptor. Received in final form 21 December 2010
Nature 378 690–697. (doi:10.1038/378690a0) Accepted 6 January 2011
Wirth EK, Roth S, Blechschmidt C, Holter SM, Becker L, Racz I, Zimmer A,
Klopstock T, Gailus-Durner V, Fuchs H et al. 2009 Neuronal 3 0 ,3,5- Made available online as an Accepted Preprint
triiodothyronine (T3) uptake and behavioral phenotype of mice deficient in 6 January 2011

Journal of Endocrinology (2011) 209, 1–8 www.endocrinology-journals.org

You might also like