Download Minireview: Pathophysiological Importance of Thyroid Hormone

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Hypothalamus wikipedia , lookup

Signs and symptoms of Graves' disease wikipedia , lookup

Hypothyroidism wikipedia , lookup

Growth hormone therapy wikipedia , lookup

Hyperthyroidism wikipedia , lookup

Transcript
M I N I R E V I E W
Minireview: Pathophysiological Importance of
Thyroid Hormone Transporters
Heike Heuer and Theo J. Visser
Neuroendocrinology Group, Leibniz Institute for Age Research/Fritz Lipmann Institute, D-07745 Jena, Germany;
and Department of Internal Medicine, Section of Endocrinology, Erasmus Medical Center, 3000 CA Rotterdam,
The Netherlands
Thyroid hormone metabolism and action are largely intracellular events that require transport of
iodothyronines across the plasma membrane. It has been assumed for a long time that this occurs
by passive diffusion, but it has become increasingly clear that cellular uptake and efflux of thyroid
hormone is mediated by transporter proteins. Recently, several active and specific thyroid hormone
transporters have been identified, including monocarboxylate transporter 8 (MCT8), MCT10, and
organic anion transporting polypeptide 1C1 (OATP1C1). The latter is expressed predominantly in
brain capillaries and transports preferentially T4, whereas MCT8 and MCT10 are expressed in
multiple tissues and are capable of transporting different iodothyronines. The pathophysiological
importance of thyroid hormone transporters has been established by the demonstration of
MCT8 mutations in patients with severe psychomotor retardation and elevated serum T3 levels.
MCT8 appears to play an important role in the transport of thyroid hormone in the brain, which
is essential for the crucial action of the hormone during brain development. It is expected that
more specific thyroid hormone transporters will be discovered in the near future, which will
lead to a better understanding of the tissue-specific regulation of thyroid hormone
bioavailability. (Endocrinology 150: 1078 –1083, 2009)
hyroid hormone is important for the growth and development of various tissues, in particular the brain, and for the
regulation of their basal metabolic rate throughout life. Many
actions of thyroid hormone are initiated by binding of the active
form of the hormone T3 to nuclear receptors in target cells (1).
These receptors are associated with the T3-responsive elements
in T3-sensitive genes, and binding of T3 to its receptors induces
changes in their interaction with corepressor and coactivator
proteins, resulting in the stimulation or the repression of gene
expression (1).
Although T3 is the major receptor-active form of thyroid hormone, T4 is the predominant iodothyronine secreted by the thyroid gland under normal conditions. As a consequence, T4 has
to be converted by so-called outer ring deiodination (ORD) to
T3. Alternatively, T4 is metabolized by inner ring deiodination
(IRD) to receptor-inactive rT3, and by the same reaction, T3
is inactivated to 3,3⬘-T2 (2). The latter is also produced by
ORD of rT3. Three iodothyronine deiodinases (D1–3) are involved in these reactions. D1 is expressed in liver, kidney, and
T
thyroid. Although it has both ORD and IRD activity, the
former prevails physiologically, contributing significantly to
the production of serum T3 and clearance of serum rT3 (2). D2
is expressed in brain, pituitary, brown adipose tissue, and
thyroid and, at least at the mRNA level, in skeletal muscle and
heart. D2 has only ORD activity and is important for local
production of T3 in brain, pituitary, and brown adipose tissue.
D2 in these and other tissues may also contribute to the production of circulating T3. D3 is expressed in adult brain and
skin and at high levels in multiple fetal tissues as well as in the
placenta and the uterus during pregnancy. It has only IRD
activity and is thus important for the negative control of local
and systemic T3 levels (2).
The biological activity of thyroid hormone is largely determined by the intracellular T3 concentration available for binding
to its nuclear receptors, which depends on a number of factors:
1) the circulating concentrations of T3 and its precursor T4, 2) the
activities of the deiodinases catalyzing the production or degradation of T3, and 3) the activities of transporters that facilitate
ISSN Print 0013-7227 ISSN Online 1945-7170
Printed in U.S.A.
Copyright © 2009 by The Endocrine Society
doi: 10.1210/en.2008-1518 Received October 30, 2008. Accepted December 15, 2008.
First Published Online January 29,2009
Abbreviations: AHDS, Allan-Herndon-Dudley syndrome; CRYM, ␮-crystallin; h, human;
IRD, inner ring deiodination; MCT, monocarboxylate transporter; OATP, organic anion
transporting polypeptide; ORD, outer ring deiodination; TAT1, T-type amino acid transporter; TLS, translation start site.
1078
endo.endojournals.org
Endocrinology, March 2009, 150(3):1078 –1083
Downloaded from endo.endojournals.org at Radcliffe Sci Library Acquisitions Department on February 8, 2010
Endocrinology, March 2009, 150(3):1078 –1083
cellular uptake and/or efflux of the different iodothyronines. It
has been thought for a long time that the lipophilic iodothyronines are capable of crossing the plasma membrane by simple diffusion, but it has become increasingly clear that this is impossible
without transporters (3).
In the last decade, several transporters have been identified
that are capable of facilitating the cellular entry and/or efflux
of iodothyronine derivatives. These include the Na-taurocholate cotransporting polypeptide (NTCP), different members of the Na-independent organic anion transporting
polypeptide (OATP) family, and the L-type amino acid transporters LAT1 and LAT2 (4). We have also shown that fatty
acid translocase (FAT) expression in Xenopus oocytes stimulates iodothyronine uptake (5). However, FAT (also known as
CD36) is not a true transporter but may function as a cell surface
attractant for ligands of neighboring receptors and transporters
(6). Most of the above-mentioned organic anion transporters are
multispecific, accepting a wide variety of ligands. A notable exception is OATP1C1, which is almost exclusively expressed in
brain, in particular in microvessels and the choroid plexus, and
shows a high specificity for T4 (7–9). This transporter is thought
to be very important for the transport of T4 into the brain.
A series of studies by the group of Blondeau and Francon (10,
11) has strongly suggested the involvement of a T-type (aromatic) amino acid transporter in the cellular uptake of iodothyronines, in particular in erythrocytes. In 2001 and 2002, Kim
et al. (12, 13) reported on the cloning and characterization of
such a transporter in rats and humans, termed TAT1. They noted
that this transporter belongs to the family of monocarboxylate
transporters (MCTs); it is now better known as MCT10 (or
SLC16A10). Although Kim et al. (12, 13) clearly demonstrated
transport of the aromatic amino acids Phe, Tyr, and Trp, they
could not detect transport of T4 and T3 by MCT10. This led us
to hypothesize that another member of the MCT family most homologous to MCT10, namely MCT8, is the long-sought T-type
amino acid transporter that also transports thyroid hormone.
Indeed, MCT8 (SLC16A2) has been shown to be an active and
specific thyroid hormone transporter, but it does not transport aromatic amino acids (14, 15). Remarkably, we have
later demonstrated that, in contrast to the results of Kim et al.,
MCT10 is at least as good as MCT8 in transporting iodothyronines (see below) (16).
The MCT family consists of 14 homologous proteins and
earned its name because the first four members (MCT1– 4) have
been characterized as MCTs that facilitate the proton-dependent
cellular uptake and efflux of compounds such as lactate and
pyruvate (17, 18). MCT6 has been shown to transport the diuretic bumetadine, but its natural ligands have not been identified yet (19). The function of MCT5, -7, -9, and -11–14 also
remains to be elucidated. The proper expression and function of
MCT1– 4 in the plasma membrane depends on the oligomerization (e.g. heterotetramer formation) with the ancillary proteins
basigin (CD147) or embigin (gp70) (17, 20). This review focuses
on the biochemical and pathophysiologial aspects of thyroid hormone transport by MCT8 and MCT10.
endo.endojournals.org
1079
Biochemistry of MCT8
The MCT8 gene is located on human (h) chromosome Xq13.2
and consists of six exons and five introns, the first of which is
about 100,000 kb in size (17, 21). The mature mRNA is about
4.4 kb large and contains two possible translation start sites
(TLSs). Depending on which of these TLSs is used, proteins are
generated consisting of 613 or 539 amino acids. The most upstream TLS is absent in most species, including mouse and rat,
and the importance of the additional N-terminal sequence in the
longer hMCT8 protein is as yet unknown. The N-terminal domain of the shorter hMCT8 protein contains one PEST-domain
and the longer hMCT8 protein contains two PEST domains (21).
Based on this feature, MCT8 was initially named XPCT (Xlinked PEST-containing transporter). PEST domains are rich in
Pro, Glu, Ser and Thr residues and are often associated with rapid
protein degradation, but the function of the PEST domain(s) in
MCT8 is unknown.
As with all other MCTs and irrespective of the length of the
N terminus, hMCT8 contains 12 putative transmembrane domains, and both the N and C terminus are located intracellularly.
The hMCT8 amino acid sequence does not contain consensus
glycosylation sites, and immunoblots of cells transfected with
hMCT8 cDNA show a major protein with a molecular mass
corresponding to the nonglycosylated protein (15). In addition,
a weaker band is observed with a molecular mass suggestive of
the homodimer (15). Ancillary proteins such as basigin and embigin do not appear to be required for proper expression of
hMCT8 protein in the plasma membrane (Visser, W. E., unpublished results).
Initially, MCT8 was characterized as a thyroid hormone
transporter in studies where rat MCT8 was expressed in Xenopus oocytes (14). This resulted in an approximately 10-fold increase in the uptake of the iodothyronines T4, T3, rT3, and 3,3⬘T2, but it had no effect on the uptake of T4 sulfate, the aromatic
amino acids Phe, Tyr, and Trp, and lactate. Subsequent studies
in mammalian cells indicated marked stimulation of T4 and T3
uptake after transient transfection with hMCT8, but the magnitude of the response was much smaller than that observed
with rat MCT8 in oocytes (15). This was explained by the high
rate of iodothyronine efflux from hMCT8-transfected cells,
which could be prevented by cotransfection with the highaffinity cytoplasmic thyroid hormone-binding protein ␮-crystallin (CRYM). In the presence of CRYM, net uptake of T4
and T3 was greatly stimulated by hMCT8 expression in mammalian cells (15).
Metabolism of iodothyronines in intact deiodinase-transfected cells is strongly enhanced by cotransfection with hMCT8
(15). This has been shown for T4 metabolism by D2 or D3, for
T3 metabolism by D3, for rT3 metabolism by D1 or D2, and for
3,3⬘-T2 metabolism by D3. Although a different topography has
been suggested for D3, the active centers of the different deiodinases are localized in the cytoplasm (2). These findings, therefore, demonstrate that hMCT8 greatly increases the intracellular availability of the different iodothyronines. Presumably,
MCT8 also facilitates the supply of T3 to its nuclear receptors,
but this remains to be demonstrated.
Downloaded from endo.endojournals.org at Radcliffe Sci Library Acquisitions Department on February 8, 2010
1080
Heuer and Visser
Minireview
Endocrinology, March 2009, 150(3):1078 –1083
As demonstrated by Northern blotting and RT-PCR, MCT8
mRNA is widely expressed in human and rat tissues, including
brain, heart, liver, kidney, adrenal gland, and thyroid (22, 23).
Rat MCT8 protein has been localized by immunohistochemistry
in the basolateral membrane of hepatocytes, renal tubule cells,
and thyrocytes (Philp, N., unpublished observations). Because
little is known about the mechanism of hormone secretion from
the thyroid gland, it is unclear whether and how MCT8 plays a
role in this process.
Analysis of the MCT8 mRNA expression pattern in the
mouse brain by in situ hybridization revealed a distinct localization of this transporter in specific neuronal populations
known to be highly dependent on proper thyroid hormone supply (24). For instance, pronounced transcript levels were detected in hippocampal pyramidal neurons and granule cells, in
layers 2–3 and 5 of the cerebral cortex, in basal ganglia, throughout the amygdala, and in cerebellar Purkinje cells, indicating that
MCT8 may play a critical role in the uptake of T3 into neuronal
cells (Fig. 1). Moreover, high transcript levels for MCT8 were
observed in choroid plexus structures and in capillary endothelial cells, suggesting that MCT8 also contributes to the passage
of thyroid hormones via the blood-brain barrier and/or via the
blood-cerebrospinal fluid barrier (24, 25) (Fig. 1), a hypothesis
that was confirmed by the analysis of MCT8-deficient mice (see
below).
Clinical Relevance of MCT8
Since 2004, several male patients have been reported in the literature with a particular combination of severe neurological deficits and abnormal serum thyroid hormone levels (26 –36). The
neurological phenotype includes in most patients central hypotonia, with poor head control; initially also peripheral hypoto-
Oatp1c1
MCT8
T2
D3
T4
?
T4
D2
T3
BBB
T3
T3
?
?
astrocyte
neuron
FIG. 1. A model of the two routes of T3 supply to central neurons. One consists
of 1) T4 transport across the blood-brain barrier (BBB) involving OATP1C1, 2) T4
uptake in astrocytes via an unknown transporter, 3) T4 conversion to T3 by D2, 4)
T3 release from astrocytes via an unknown transporter, and 5) T3 uptake by
neurons via MCT8 or another transporter. The second route consists of 1) T3
transport across the BBB by MCT8 and 2) uptake into neurons by MCT8 or
another transporter. Neuronal T3 uptake through other transporters than MCT8
may be more important in mice than in humans. Adapted from Heuer et al. (24).
nia, which evolves into spastic quadriplegia; inability to sit,
stand, or walk independently; severe mental retardation; and
absence of speech. For a detailed description of the clinical phenotype, the reader is referred to previous articles (37, 38). This
severe form of X-linked psychomotor retardation has already
been described in 1944 (39) and since then also named after the
original authors as the Allan-Herndon-Dudley syndrome
(AHDS). What was not known at that time was the association
with abnormal thyroid parameters. Patients with AHDS invariably have markedly elevated serum T3 and low T4, free T4, and
rT3 levels. Serum TSH varies between normal and somewhat
elevated.
The severe neurological impairment in these patients and the
elevated serum T3 levels, in view of the well-known importance
of thyroid hormone for brain development, suggested that this
syndrome represented some form of thyroid hormone resistance.
Because the X-linked inheritance of the disorder was not obvious
when we encountered this syndrome in 2002 in the first two
patients, the THRA and THRB genes, which encode the T3 receptors, were examined for possible pathogenic mutations with
negative results (27). Also the DIO1, DIO2, and DIO3 genes,
which code for D1, D2, and D3, respectively, were screened for
possible mutations that were not found either (27). This
prompted us to hypothesize that the AHDS syndrome represents
a novel form of thyroid hormone resistance due to a lack of
thyroid hormone entry in target cells. Because we had just discovered MCT8 as an active and specific thyroid hormone transporter, which gene is located on the X chromosome, we tested
this hypothesis by sequencing the MCT8 gene in the first two
patients and, indeed, identified different mutations in this gene.
To date, we have demonstrated different mutations in 15
unrelated AHDS families (40), and worldwide over 40 different
mutations have been identified. Especially the groups of
Schwartz and Refetoff (41, 42) have also contributed considerably to the clinical and genetic investigation of AHDS patients.
These mutations include large deletions resulting in the loss of
one or more exons, smaller frame-shift deletions, 3-nucleotide
(1-amino acid) deletions or insertions, nonsense mutations resulting in truncation of the MCT8 protein, and missense mutations resulting in 1-amino acid substitutions. Many of these mutations are obviously detrimental for hMCT8 function, but for
the 1-amino acid deletions, insertions, and substitutions, this is
not immediately clear. We have, therefore, introduced a variety
of such mutations found in patients into the hMCT8 cDNA and
compared the function of mutated and wild-type MCT8 in transfected cells (30, 34, 43). Cells transfected with the MCT8 variants alone were tested for T3 uptake, and cells cotransfected with
D3 were tested for T3 metabolism.
In both tests, most mutations resulted in a complete inactivation of MCT8 function. However, significant residual activity
was observed with a number of MCT8 mutations, and some of
these were associated with a milder clinical phenotype (30, 34,
43). There appear to be different mechanisms by which mutations affect MCT8 function. With some mutations, the generation of hMCT8 protein is strongly diminished, perhaps due to
incorrect protein processing and rapid protein degradation.
With other mutations, hMCT8 protein generation is not dimin-
Downloaded from endo.endojournals.org at Radcliffe Sci Library Acquisitions Department on February 8, 2010
Endocrinology, March 2009, 150(3):1078 –1083
ished, but it does not undergo proper trafficking to the plasma
membrane. Yet other hMCT8 mutants are expressed at the
plasma membrane but appear to have lost their iodothyroninetransporting activity (43).
The effects of AHDS-causing mutations on MCT8 transporter function can also be studied in fibroblasts cultured from
patients’ skin biopsies (34). Apparently, MCT8 is the major thyroid hormone transporter expressed in these cells, because fibroblasts from AHDS patients show markedly decreased T4 and
T3 uptake compared with control fibroblasts. We have studied
fibroblasts from four AHDS patients, three of whom had devastating MCT8 mutations, and one had a Phe501del mutation.
Fibroblasts from the latter patient showed T3 uptake and efflux
rates in between those of the other three patients and the controls.
This patient also presented the mildest clinical phenotype and least
abnormal serum T4, T3, and rT3 levels in our patient cohort. Results
obtained with cells transfected with the MCT8-Phe501del mutant
were in close agreement with the fibroblast findings. These and
other results suggest a genotype-phenotype correlation for AHDScausing mutations in MCT8 (34, 43).
Delayed myelinization is often mentioned in magnetic resonance imaging studies of patients with MCT8 mutations, but this
appears not to be a general characteristic. Interestingly, however,
a study was reported recently in patients with the PelizaeusMerzbacher syndrome, who have X-linked mental retardation
because of a myelinization defect. In most patients, this is caused
by a mutation in the PLP1 gene, which encodes proteolipid protein 1, an important component of myelin. Vaurs-Barrière et al.
(44) screened serum thyroid parameters and the MCT8 gene
sequence in 53 patients with Pelizaeus-Merzbacher-like syndrome without mutations in PLP1 and identified six patients
with high serum T3 levels and different MCT8 mutations. This
study underscores how difficult it is to categorize patients with
X-linked mental retardation on the basis of clinical characteristics. It also supports the relevance of screening such patients for high serum T3 levels to identify those with possible
mutations in MCT8.
endo.endojournals.org
1081
absence of MCT8 in mice results in a strongly diminished uptake
of T3 into the brain. As a consequence, the brain was found to be
in a rather hypothyroid state as reflected by decreased T4 and T3
concentrations and increased D2 and decreased D3 activities.
Because the transport of T4 into the brain of MCT8-null mice
is not compromised, an increase in local T3 production is sufficient to provide certain neuronal cells such as cerebellar Purkinje
cells with adequate amounts of thyroid hormone, thereby preventing serious neurological damage. Other neurons such as
RC3-expressing cells in the striatum exhibit signs of hypothyroidism indicating that either local T3 production or neuronal
uptake of T3 into these cells is affected in the absence of MCT8.
Furthermore, strongly elevated transcript levels for TRH in the
hypothalamic paraventricular nucleus points to a strong hypothyroid state of these cells in the absence of MCT8. This increase
in TRH expression can be successfully suppressed with a high
dose of T4, demonstrating that paraventricular nucleus neurons
still respond to locally produced T3.
In comparison with the hypothyroid state of the hypothalamus, the pituitary appears to be surprisingly normal with no
alterations in the expression levels of thyroid hormone-responsive genes. Furthermore, TSH levels are only slightly elevated in
MCT8-null mice. However, when MCT8-null mice are rendered
hypothyroid, only high concentrations of T3 are able to suppress
TSH expression, suggesting that not only the hypothalamus but
also the pituitary is rather insensitive to the circulating T3 levels.
This central resistance to thyroid hormone has also been found
in patients with MCT8 mutations (47). However, the underlying
mechanism is not understood yet and needs further investigation.
The fact that uptake of T3 into the brain is strongly diminished in MCT8-null animals raises the question of why the transport of T4 is not also impaired. We speculate that OATP1C1,
which exhibits a striking substrate specificity toward T4 and rT3,
is critically involved in the transport of T4 via the blood-brain
and blood-cerebrospinal fluid barrier because this transporter is
highly enriched in capillary endothelial cells as well as in the
choroid plexus (9, 25). This hypothesis, however, needs to be
ultimately validated by the analysis of MCT8/OATP1C1-deficient animals.
Analysis of MCT8-Null Mice
The generation and analysis of mouse mutants deficient in
MCT8 provided further information as to the role of this thyroid
hormone transporter in specific tissues (45, 46). The most remarkable phenotype of MCT8-null animals is the lack of any
overt neurological abnormalities, a rather unexpected finding in
light of the severe human phenotype. However, MCT8-null mice
exhibit a marked increase in serum T3 and a decrease in serum T4
and rT3. Thus, these mice fully replicate the endocrinological
aberrances diagnosed in patients with inactivating MCT8
mutations.
How do the abnormal serum thyroid hormone concentrations affect metabolism and function of organs that are dependent on proper thyroid hormone supply? Analysis of the thyroid
state of the liver revealed increased D1 activities as well as an
increased T3 content in MCT8-null mice, indicating that hepatocytes fully sense the rise in circulating T3 levels. In contrast,
MCT10
Of all MCT family members, the MCT10 amino acid sequence
is most homologous to that of MCT8, suggesting that they are
derived from a common ancestor (16, 17). This is supported by
the presence of single genes in different insects that code for
proteins highly homologous to MCT8/10 (16). In Drosophila,
the gene is known as kar and codes for a putative transporter for
Trp or another precursor for an ommochrome pigment; kar is
mutated in flies with the karmoisin eye color phenotype (16, 48).
The hMCT10 gene is located on human chromosome 6q21-q22
and has the same structure as the hMCT8 gene. The mature
mRNA is about 2.6 kb in size, has a single TLS, and codes for a
protein of 515 amino acids, containing 12 putative transmembrane domains. Like hMCT8, there is no consensus glycosylation site in the hMCT10 amino acid sequence. Transfection of
Downloaded from endo.endojournals.org at Radcliffe Sci Library Acquisitions Department on February 8, 2010
1082
Heuer and Visser
Minireview
cells with hMCT10 cDNA results in the generation of a major
approximately 55-kDa protein, the expected molecular mass for
the nonglycosylated protein. Immunoblots also show a higher
band, suggestive of the formation of the homodimer (16).
Initially, rat and subsequently hMCT10 have been characterized in Xenopus oocytes as a T-type amino acid transporter
(TAT1), which facilitates cellular uptake and efflux of aromatic
amino acids such as Trp, Phe, Tyr, and 3,4-dihydroxyphenylalanine (DOPA) (12, 13). MCT10 is expressed in different human and rodent tissues, in particular intestine, kidney, liver, skeletal muscle, heart, and placenta (12, 13, 23, 49). MCT10 protein
has been localized by immunohistochemistry in the basolateral membranes of perivenous hepatocytes, renal proximal
tubule cells, and intestinal villi cells (49). Because MCT10
facilitates the efflux of aromatic amino acids much better than
their uptake, the transporter appears to serve an important
function in the intestinal and renal resorption of these amino
acids by mediating their efflux into the bloodstream (49).
Transfection of mammalian cells with hMCT10 cDNA results in a marked induction of iodothyronine uptake, which is
strongly augmented if the cells are cotransfected with CRYM
(16). The latter is explained by the inhibition of iodothyronine
efflux in the presence of the cytosolic thyroid hormone-binding
protein. These findings indicate that hMCT10 facilitates both
cellular uptake and efflux of the different iodothyronines. As is
the case with MCT8, expression of hMCT10 also largely stimulates the deiodination of iodothyronines in cells cotransfected
with the different deiodinases through an increase in the intracellular availability of these substrates (16).
The physiological relevance of MCT10 for thyroid hormone
action and metabolism in different tissues remains to be elucidated. Because MCT10 is at least as active in transporting thyroid hormone as MCT8, it is to be expected that inactivating
mutations in MCT10 will also result in significant changes in
intracellular thyroid hormone concentrations in different tissues.
The magnitude and direction of these changes will not only depend on the expression of MCT10 but also on that of other
transporters in these tissues.
Conclusion and Perspectives
Through the characterization of MCT8, MCT10, and OATP1C1
as active thyroid hormone transporters, and the recognition of
the severe clinical phenotype caused by mutations in MCT8, the
concept that plasma membrane transporters are essential for
action and metabolism of thyroid hormone has been firmly established. It is to be expected that more thyroid hormone transporters will be discovered in the near future. For instance, there
is strong evidence for the existence of high-affinity, Na-dependent iodothyronine transporters in the liver, but the identity of
these transporters is still elusive. Also, different transporters may
be involved in thyroid hormone transport in different cell types
and different regions in the brain. Endothelium is a first barrier
that has to be crossed in the transfer of thyroid hormone from the
blood into the tissues, but very little is still known about the
nature of the transporters involved. A lot remains to be investi-
Endocrinology, March 2009, 150(3):1078 –1083
gated before we begin to understand all the ins and outs of thyroid hormone transporters.
Acknowledgments
These studies were supported by grants from the American Thyroid
Association and The Netherlands Organization for Scientific Research.
Address all correspondence and requests for reprints to: Theo J. Visser, Department of Internal Medicine, Section of Endocrinology, Erasmus Medical College, Room Ee 502, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands. E-mail: [email protected].
Disclosure Statement: The authors have nothing to disclose.
References
1. Yen PM 2001 Physiological and molecular basis of thyroid hormone action.
Physiol Rev 81:1097–1142
2. Gereben B, Zavacki AM, Ribich S, Kim BW, Huang SA, Simonides WS, Zeold
A, Bianco AC 2008 Cellular and molecular basis of deiodinase-regulated thyroid hormone signaling. Endocr Rev 29:898 –938
3. Hennemann G, Docter R, Friesema EC, de Jong M, Krenning EP, Visser TJ
2001 Plasma membrane transport of thyroid hormones and its role in thyroid
hormone metabolism and bioavailability. Endocr Rev 22:451– 476
4. Friesema EC, Jansen J, Milici C, Visser TJ 2005 Thyroid hormone transporters.
Vitam Horm 70:137–167
5. van der Putten HH, Friesema EC, Abumrad NA, Everts ME, Visser TJ 2003
Thyroid hormone transport by the rat fatty acid translocase. Endocrinology
144:1315–1323
6. Benton R, Vannice KS, Vosshall LB 2007 An essential role for a CD36-related
receptor in pheromone detection in Drosophila. Nature 450:289 –293
7. Tohyama K, Kusuhara H, Sugiyama Y 2004 Involvement of multispecific
organic anion transporter, Oatp14 (Slc21a14), in the transport of thyroxine
across the blood-brain barrier. Endocrinology 145:4384 – 4391
8. Pizzagalli F, Hagenbuch B, Stieger B, Klenk U, Folkers G, Meier PJ 2002
Identification of a novel human organic anion transporting polypeptide as a
high affinity thyroxine transporter. Mol Endocrinol 16:2283–2296
9. Sugiyama D, Kusuhara H, Taniguchi H, Ishikawa S, Nozaki Y, Aburatani H,
Sugiyama Y 2003 Functional characterization of rat brain-specific organic
anion transporter (Oatp14) at the blood-brain barrier: high affinity transporter
for thyroxine. J Biol Chem 278:43489 – 43495
10. Zhou Y, Samson M, Francon J, Blondeau JP 1992 Thyroid hormone concentrative uptake in rat erythrocytes. Involvement of the tryptophan transport
system T in countertransport of tri-iodothyronine and aromatic amino acids.
Biochem J 281(Pt 1):81– 86
11. Zhou Y, Samson M, Osty J, Francon J, Blondeau JP 1990 Evidence for a close
link between the thyroid hormone transport system and the aromatic amino
acid transport system T in erythrocytes. J Biol Chem 265:17000 –17004
12. Kim DK, Kanai Y, Chairoungdua A, Matsuo H, Cha SH, Endou H 2001
Expression cloning of a Na⫹-independent aromatic amino acid transporter
with structural similarity to H⫹/monocarboxylate transporters. J Biol Chem
276:17221–17228
13. Kim DK, Kanai Y, Matsuo H, Kim JY, Chairoungdua A, Kobayashi Y, Enomoto
A, Cha SH, Goya T, Endou H 2002 The human T-type amino acid transporter-1:
characterization, gene organization, and chromosomal location. Genomics 79:
95–103
14. Friesema EC, Ganguly S, Abdalla A, Manning Fox JE, Halestrap AP, Visser TJ
2003 Identification of monocarboxylate transporter 8 as a specific thyroid
hormone transporter. J Biol Chem 278:40128 – 40135
15. Friesema EC, Kuiper GG, Jansen J, Visser TJ, Kester MH 2006 Thyroid hormone transport by the human monocarboxylate transporter 8 and its ratelimiting role in intracellular metabolism. Mol Endocrinol 20:2761–2772
16. Friesema EC, Jansen J, Jachtenberg JW, Visser WE, Kester MH, Visser TJ 2008
Effective cellular uptake and efflux of thyroid hormone by human monocarboxylate transporter 10. Mol Endocrinol 22:1357–1369
17. Halestrap AP, Meredith D 2004 The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond.
Pflugers Arch 447:619 – 628
Downloaded from endo.endojournals.org at Radcliffe Sci Library Acquisitions Department on February 8, 2010
Endocrinology, March 2009, 150(3):1078 –1083
18. Meredith D, Christian HC 2008 The SLC16 monocarboxylate transporter
family. Xenobiotica 38:1072–1106
19. Murakami Y, Kohyama N, Kobayashi Y, Ohbayashi M, Ohtani H, Sawada Y,
Yamamoto T 2005 Functional characterization of human monocarboxylate
transporter 6 (SLC16A5). Drug Metab Dispos 33:1845–1851
20. Wilson MC, Meredith D, Fox JE, Manoharan C, Davies AJ, Halestrap AP
2005 Basigin (CD147) is the target for organomercurial inhibition of monocarboxylate transporter isoforms 1 and 4: the ancillary protein for the insensitive MCT2 is EMBIGIN (gp70). J Biol Chem 280:27213–27221
21. Lafreniere RG, Carrel L, Willard HF 1994 A novel transmembrane transporter
encoded by the XPCT gene in Xq13.2. Hum Mol Genet 3:1133–1139
22. Price NT, Jackson VN, Halestrap AP 1998 Cloning and sequencing of four new
mammalian monocarboxylate transporter (MCT) homologues confirms the
existence of a transporter family with an ancient past. Biochem J 329(Pt 2):
321–328
23. Nishimura M, Naito S 2008 Tissue-specific mRNA expression profiles of
human solute carrier transporter superfamilies. Drug Metab Pharmacokinet
23:22– 44
24. Heuer H, Maier MK, Iden S, Mittag J, Friesema EC, Visser TJ, Bauer K 2005
The monocarboxylate transporter 8 linked to human psychomotor retardation
is highly expressed in thyroid hormone-sensitive neuron populations. Endocrinology 146:1701–1706
25. Roberts LM, Woodford K, Zhou M, Black DS, Haggerty JE, Tate EH, Grindstaff
KK, Mengesha W, Raman C, Zerangue N 2008 Expression of the thyroid
hormone transporters monocarboxylate transporter-8 (SLC16A2) and organic ion transporter-14 (SLCO1C1) at the blood-brain barrier. Endocrinology 149:6251– 6261
26. Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S 2004 A novel
syndrome combining thyroid and neurological abnormalities is associated
with mutations in a monocarboxylate transporter gene. Am J Hum Genet
74:168 –175
27. Friesema EC, Grueters A, Biebermann H, Krude H, von Moers A, Reeser M,
Barrett TG, Mancilla EE, Svensson J, Kester MH, Kuiper GG, Balkassmi S,
Uitterlinden AG, Koehrle J, Rodien P, Halestrap AP, Visser TJ 2004 Association between mutations in a thyroid hormone transporter and severe X-linked
psychomotor retardation. Lancet 364:1435–1437
28. Frints SG, Lenzner S, Bauters M, Jensen LR, Van Esch H, des Portes V, Moog
U, Macville MV, van Roozendaal K, Schrander-Stumpel CT, Tzschach A,
Marynen P, Fryns JP, Hamel B, van Bokhoven H, Chelly J, Beldjord C, Turner
G, Gecz J, Moraine C, Raynaud M, Ropers HH, Froyen G, Kuss AW 2008
MCT8 mutation analysis and identification of the first female with AllanHerndon-Dudley syndrome due to loss of MCT8 expression. Eur J Hum Genet
16:1029 –1037
29. Herzovich V, Vaiani E, Marino R, Dratler G, Lazzati JM, Tilitzky S, Ramirez
P, Iorcansky S, Rivarola MA, Belgorosky A 2007 Unexpected peripheral markers of thyroid function in a patient with a novel mutation of the MCT8 thyroid
hormone transporter gene. Horm Res 67:1– 6
30. Jansen J, Friesema EC, Kester MH, Milici C, Reeser M, Gruters A, Barrett TG,
Mancilla EE, Svensson J, Wemeau JL, Busi da Silva Canalli MH, Lundgren J,
McEntagart ME, Hopper N, Arts WF, Visser TJ 2007 Functional analysis of
monocarboxylate transporter 8 mutations identified in patients with X-linked
psychomotor retardation and elevated serum triiodothyronine. J Clin Endocrinol Metab 92:2378 –2381
31. Maranduba CM, Friesema EC, Kok F, Kester MH, Jansen J, Sertie AL, PassosBueno MR, Visser TJ 2006 Decreased cellular uptake and metabolism in
Allan-Herndon-Dudley syndrome (AHDS) due to a novel mutation in the
MCT8 thyroid hormone transporter. J Med Genet 43:457– 460
32. Namba N, Etani Y, Kitaoka T, Nakamoto Y, Nakacho M, Bessho K, Miyoshi
endo.endojournals.org
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
1083
Y, Mushiake S, Mohri I, Arai H, Taniike M, Ozono K 2008 Clinical phenotype
and endocrinological investigations in a patient with a mutation in the MCT8
thyroid hormone transporter. Eur J Pediatr 167:785–791
Schwartz CE, May MM, Carpenter NJ, Rogers RC, Martin J, Bialer MG,
Ward J, Sanabria J, Marsa S, Lewis JA, Echeverri R, Lubs HA, Voeller K,
Simensen RJ, Stevenson RE 2005 Allan-Herndon-Dudley syndrome and the
monocarboxylate transporter 8 (MCT8) gene. Am J Hum Genet 77:41–53
Visser WE, Jansen J, Friesema EC, Kester MH, Mancilla E, Lundgren J, van der
Knaap MS, Lunsing RJ, Brouwer OF, Visser TJ 17 July 2008 Novel pathogenic
mechanism suggested by ex vivo analysis of MCT8 (SLC16A2) mutations.
Hum Mutat 10.1002/humu.20808
Papadimitriou A, Dumitrescu AM, Papavasiliou A, Fretzayas A, Nicolaidou P,
Refetoff S 2008 A novel monocarboxylate transporter 8 gene mutation as a
cause of severe neonatal hypotonia and developmental delay. Pediatrics 121:
e199 – e202
Kakinuma H, Itoh M, Takahashi H 2005 A novel mutation in the monocarboxylate transporter 8 gene in a boy with putamen lesions and low free T4 levels
in cerebrospinal fluid. J Pediatr 147:552–554
Brockmann K, Dumitrescu AM, Best TT, Hanefeld F, Refetoff S 2005 Xlinked paroxysmal dyskinesia and severe global retardation caused by defective MCT8 gene. J Neurol 252:663– 666
Holden KR, Zuniga OF, May MM, Su H, Molinero MR, Rogers RC, Schwartz
CE 2005 X-linked MCT8 gene mutations: characterization of the pediatric
neurologic phenotype. J Child Neurol 20:852– 857
Allan W, Herndon CN, Dudley FC 1944 Some examples of the inheritance of
mental deficiency: apparently sex-linked idiocy and microcephaly. Am J Mental Defic 48:325–334
Visser WE, Friesema EC, Jansen J, Visser TJ 2008 Thyroid hormone transport
in and out of cells. Trends Endocrinol Metab 19:50 –56
Schwartz CE, Stevenson RE 2007 The MCT8 thyroid hormone transporter
and Allan-Herndon-Dudley syndrome. Best Pract Res Clin Endocrinol Metab
21:307–321
Refetoff S, Dumitrescu AM 2007 Syndromes of reduced sensitivity to thyroid
hormone: genetic defects in hormone receptors, cell transporters and deiodination. Best Pract Res Clin Endocrinol Metab 21:277–305
Jansen J, Friesema EC, Kester MH, Schwartz CE, Visser TJ 2008 Genotypephenotype relationship in patients with mutations in thyroid hormone transporter MCT8. Endocrinology 149:2184 –2190
Vaurs-Barrière C, Deville M, Sarret C, Giraud G, Des Portes V, Prats-Vinas
JM, De Michele G, Dan B, Brady AF, Boespflug-Tanguy O, Touraine R,
Pelizaeus-Merzbacher-like disease presentation of MCT8 mutated males. Ann
Neurol, in press
Dumitrescu AM, Liao XH, Weiss RE, Millen K, Refetoff S 2006 Tissue-specific
thyroid hormone deprivation and excess in monocarboxylate transporter
(mct) 8-deficient mice. Endocrinology 147:4036 – 4043
Trajkovic M, Visser TJ, Mittag J, Horn S, Lukas J, Darras VM, Raivich G,
Bauer K, Heuer H 2007 Abnormal thyroid hormone metabolism in mice lacking the monocarboxylate transporter 8. J Clin Invest 117:627– 635
Biebermann H, Ambrugger P, Tarnow P, von Moers A, Schweizer U, Grueters
A 2005 Extended clinical phenotype, endocrine investigations and functional
studies of a loss-of-function mutation A150V in the thyroid hormone specific
transporter MCT8. Eur J Endocrinol 153:359 –366
Reed RD, Nagy LM 2005 Evolutionary redeployment of a biosynthetic module: expression of eye pigment genes vermilion, cinnabar, and white in butterfly
wing development. Evol Dev 7:301–311
Ramadan T, Camargo SM, Summa V, Hunziker P, Chesnov S, Pos KM, Verrey
F 2006 Basolateral aromatic amino acid transporter TAT1 (Slc16a10) functions as an efflux pathway. J Cell Physiol 206:771–779
Downloaded from endo.endojournals.org at Radcliffe Sci Library Acquisitions Department on February 8, 2010