Вы находитесь на странице: 1из 9

J Inherit Metab Dis

DOI 10.1007/s10545-014-9712-9

ICIEM SYMPOSIUM 2013

Defects of thiamine transport and metabolism


Garry Brown

Received: 13 February 2014 / Revised: 25 March 2014 / Accepted: 31 March 2014


# SSIEM and Springer Science+Business Media Dordrecht 2014

Abstract Thiamine, in the form of thiamine pyrophosphate, supplementation in patients whose clinical presentation falls
is a cofactor for a number of enzymes which play important within the spectrum of documented cases.
roles in energy metabolism. Although dietary thiamine defi-
ciency states have long been recognised, it is only relatively
recently that inherited defects in thiamine uptake, activation
and the attachment of the active cofactor to target enzymes Introduction
have been described, and the underlying genetic defects iden-
tified. Thiamine is transported into cells by two carriers, Thiamine is an important vitamin in human nutrition and, in
THTR1 and THTR2, and deficiency of these results in the form of thiamine pyrophosphate (TPP), is the cofactor for
thiamine-responsive megaloblastic anaemia and biotin- a number of enzymes which play major roles in energy
responsive basal ganglia disease respectively. Defective syn- metabolism, including the -ketoacid dehydrogenase com-
thesis of thiamine pyrophosphate has been found in a small plexes. Defects in thiamine transport or the biosynthesis of
number of patients with episodic ataxia, delayed development thiamine pyrophosphate can lead to deficiency of these en-
and dystonia, while impaired transport of thiamine pyrophos- zymes, as can structural abnormalities in the enzymes them-
phate into the mitochondrion is associated with Amish lethal selves which impair cofactor binding. Patients have now been
microcephaly in most cases. In addition to defects in thiamine described with defects in all of these aspects of thiamine
uptake and metabolism, patients with pyruvate dehydrogenase metabolism and they present with a much wider range of
deficiency and maple syrup urine disease have been described clinical and biochemical manifestations than might be expect-
who have a significant clinical and/or biochemical response to ed from shared involvement in common biochemical path-
thiamine supplementation. In these patients, an intrinsic struc- ways. In this review, an outline of the biochemistry of thia-
tural defect in the target enzymes reduces binding of the mine will be presented, together with a summary of the
cofactor and this can be overcome at high concentrations. In manifestations of the different conditions in which this is
most cases, the clinical and biochemical abnormalities in these abnormal.
conditions are relatively non-specific, and the range of Thiamine is a dicyclic compound with thiazole and
recognised presentations is increasing rapidly at present as aminopyrimidine rings joined by a methylene bridge (Fig. 1).
new patients are identified, often by genome sequencing. There is an alcohol side chain on the thiazole ring and in the
These conditions highlight the value of a trial of thiamine activation of thiamine to its active cofactor derivative, this is
esterified with pyrophosphate. Thiamine is an essential vitamin
in humans and is found in most foods, albeit at relatively low
Communicated by: John Christodoulou levels. The most important source of thiamine in most diets is
unrefined cereal grains. Dietary thiamine is mainly in the form
Presented at the 12th International Congress of Inborn Errors of
Metabolism, Barcelona, Spain, September 36, 2013.
of phosphate derivatives and, before absorption, these are con-
verted to free thiamine by intestinal phosphatases. Although
G. Brown (*) normal requirements for thiamine can be met by a daily intake
Department of Biochemistry, University of Oxford, South Parks
Road, Oxford OX1 3QU, UK of 12 mg, there is no evidence of adverse effects of much
e-mail: garry.brown@bioch.ox.ac.uk higher, pharmacological doses, and this is an important
J Inherit Metab Dis

Fig. 1 Structure of thiamine. The


vitamin precursor, thiamine (top)
and the active cofactor derived
from it, thiamine pyrophosphate
+
(below) are shown

consideration in providing supplementation for patients with The active cofactor of thiamine in animal cells, thiamine
defects in the bioavailability of the active cofactor. pyrophosphate (Fig. 1), is a cofactor for the -ketoacid dehy-
drogenase complexes (pyruvate dehydrogenase, -
ketoglutarate dehydrogenase and the branched chain -
ketoacid dehydrogenase), transketolase, an enzyme of the pen-
Thiamine transport and activation tose phosphate cycle, and, as recently described, 2-hydroxyacyl
CoA lyase, a peroxisomal enzyme involved in the -oxidation
Thiamine is absorbed into the body in the upper small intes- of branched fatty acids, such as phytanic acid (Casteels et al
tine by two transporters, THTR1 and THTR2, the products of 2007). The active TPP cofactor is generated within cells from
the SLC19A2 and SLC19A3 genes respectively (Ganapathy free thiamine by the enzyme thiamine pyrophosphokinase
et al 2004; Zhao and Goldman 2013). These transporters are (TPK) (Fig. 3). Biochemical studies indicate that TPP can be
closely related, with 48 % identity, and both facilitate a high converted to TMP by a thiamine pyrophosphatase and this can
affinity, pH dependent saturable transport across cell mem- be further hydrolysed back to free thiamine by a thiamine
branes. Both carriers are widely distributed in the body, but the monophosphatase, however genes for these enzymes have not
levels differ in different tissues and they have different kinetic yet been identified. The formation of TPP takes place in the
properties. They are also differentially distributed in polarised cytoplasm of the cell and the cofactor is immediately available
cells, such as those in the intestinal mucosa and renal tubule for association with transketolase. To provide active cofactor
(Boulware et al 2003; Rindi and Laforenza 2000). At very for the -ketoacid dehydrogenases, which are located in the
high concentrations, there is evidence that some thiamine may mitochondrial matrix, there is a TPP transporter in the
also be absorbed by diffusion. inner mitochondrial membrane (Kang and Samuels 2008;
While thiamine is mainly absorbed in the free form, there is
a mechanism for absorption of thiamine monophosphate RFC
(TMP), as this is a substrate for the reduced folate carrier TMP
(RFC1) (Zhao et al 2002). This is the product of the
SLC19A1 gene and is very closely related to the two thiamine TPP
THTR2 THTR1
carriers. The biological significance of this mechanism is T+ T+
unclear and it may actually provide a means for exporting
H+ H+
thiamine from cells. In addition, there is also some evidence
for direct uptake of TPP produced by gut bacteria into colonic
epithelial cells (Nabokina and Said 2012). The main mecha- LUMEN BLOOD
nisms for intestinal uptake of thiamine are shown in Fig. 2. At Fig. 2 Uptake of thiamine in the small intestine. Free thiamine is gener-
the luminal surface, THTR2 is the major transporter and this ated in the intestinal lumen from various phosphate derivatives present in
delivers thiamine into the cell in exchange for hydrogen ions. the diet. Thiamine is transported across the apical membrane by THTR2
At the basal surface, THTR1 is mainly responsible for release in exchange for hydrogen ions. At the basal membrane, thiamine is
released into the circulation by THTR1 in a similar exchange reaction.
of free thiamine into the circulation. Within the mucosal cells, In addition, some thiamine is converted by intestinal epithelial cells to
some thiamine can be converted to TPP and TMP and TMP and this can be released from the cell by the reduced folate carrier, a
exported into the circulation by RFC1. transporter which is closely related to the thiamine carriers
J Inherit Metab Dis

Fig. 3 Intra-cellular thiamine


metabolism. Thiamine is
transported into cells by THTR1 RFC1
and/or THTR2, depending on
their tissue distribution. Within
the cell, it is activated to TPP by
thiamine pyrophosphokinase. The
active cofactor can bind to TMP TPP
transketolase in the cytoplasm. It TMPase TPPase
is also attached to 2-hydroxyacyl
CoA lyase (HACL1) in the
cytoplasm before this enzyme is
transported into the peroxisome. T T TPP TPP transporter
TPP is transported into the THTR1 TPK
mitochondrion by a specific THTR2
transporter for attachment to the
-ketoacid dehydrogenase HACL1 TPP TPP
apoenzymes

Lindhurst et al 2006). TPP is attached to the hydroxyacyl not of major importance for intestinal absorption. The addition-
CoA lyase apoprotein before it is transported into the al clinical features show considerable overlap with mitochon-
peroxisome (Casteels et al 2007). drial diseases and this would be consistent with the important
roles of the -ketoacid dehydrogenase complexes in energy
metabolism. One patient has been described with raised blood
Defects in the thiamine transporters and cerebrospinal fluid lactate concentration. In this patient, a
muscle biopsy was morphologically normal, but there were
THTR1 deficiency reduced activities of complex I of the respiratory chain and
pyruvate dehydrogenase, which returned to normal with thia-
THTR1 deficiency results in thiamine-responsive megaloblas- mine supplementation (Scharfe et al 2000). Now that the
tic anaemia, also known as Rogers syndrome (Neufeld et al biochemical basis of this condition has been established, it is
2001; Ricketts et al 2006, OMIM 249270). This condition is anticipated that more systematic investigation of mitochondrial
characterised by a core triad of megaloblastic anaemia, diabe- function will be performed in future patients.
tes mellitus and sensorineural deafness. Patients usually pres- There has been some investigation of possible pathogenic
ent in infancy or early childhood and often the anaemia is the mechanisms responsible for the megaloblastic anaemia in
first recognised feature. The anaemia is often accompanied by THTR1 deficiency. The role of imbalance in DNA and RNA
thrombocytopaenia, and ringed sideroblasts are prominent in biosynthesis in megaloblastic erythropoiesis has led to a con-
bone marrow aspirates (Bazarbachi et al 1998). While the sideration of the activity of the pentose phosphate cycle in
diabetes usually develops during childhood, a number of general, and the enzyme transketolase in particular, in provi-
patients with neonatal diabetes have been reported (Shaw- sion of ribose-5-phosphate for these processes (Fig. 4).
Smith et al 2012). In addition to the core features, a number Ribose-5-phosphate can be formed from glucose either via
of other manifestations are associated with this condition, ribulose-5-phosphate, the product of the oxidative branch of
including cardiac abnormalities, optic atrophy, retinal abnor- the pathway, or from glycolytic intermediates through a
malities, stroke-like episodes and short stature. Cardiac in- series of carbohydrate interconversions. These interconver-
volvement appears to be relatively common, and patients have sions depend upon the activity of transketolase, with its
been described with various arrhythmias, congenital TPP cofactor. In vitro studies of ribose synthesis from
malformations and cardiomyopathy (Lorber et al 2003). glucose have been performed in cultured fibroblasts from
A variety of different mutations in the SLC19A2 gene have patients with THTR1 deficiency (Boros et al 2003). These
been identified in these patients confirming the deficiency of show that RNA ribose synthesis is significantly reduced in
the THTR1 transporter. However, as the focus of investigation thiamine-depleted medium and that this is due to enhanced
has been on the haematological aspects in most cases, the reliance on the oxidative branch of the pathway. The
biochemical consequences are rather poorly defined. The blood biochemical abnormalities are reversed when the cells are
thiamine concentration is normal, suggesting that THTR1 is grown in thiamine-supplemented medium.
J Inherit Metab Dis

Fig. 4 Role of TPP in ribose GLUCOSE


metabolism. The pentose
phosphate shunt is an important 2NADP+
source of ribose-5-phosphate for
glucose-6-P ribulose-5-P
RNA and DNA biosynthesis.
This product can be generated 2NADPH
from ribulose-5-phosphate in the
oxidative part of this pathway, or
via a series of carbohydrate
interconversions involving the xylulose-5-P RIBOSE-5-P
enzymes transketolase (TK) and
transaldolase. The reactions
involving transketolase are fructose-6-P TK-TPP
dependent on the availability of
its cofactor, TPP
glyceraldehyde-3-P sedoheptulose-7-P
TK-TPP

glyceraldehyde-3-P erythrose-4-P fructose-6-P

Although this is a thiamine-responsive disorder, the differ- now been well defined (Alfadhel et al 2013; Tabarki et al 2013),
ent elements of the syndrome respond to a greater or lesser a number of additional patients with SLC19A3 mutations have
extent to thiamine supplementation. The anaemia and diabetes been described with different clinical presentations, so that the
usually respond very well initially to doses of thiamine be- recognised spectrum of this disorder has now expanded signif-
tween 2550 mg/day (although some patients have been treat- icantly and a new designation, thiamine metabolism dysfunc-
ed with considerably higher doses), and cardiac arrythmias tion syndrome 2 (OMIM 607483) has been proposed. Four
also generally respond well (Borgna-Pignatti et al 2009; patients from Japan have been described with atypical infantile
Ricketts et al 2006). On the other hand, the deafness and other spasms and delayed development, progressive cerebral atrophy
neurological manifestations respond poorly, although there and bilateral lesions in the thalami and basal ganglia (Yamada
may be a better outcome with early diagnosis and treatment et al 2010). One patient has been reported with severe neonatal
(Onal et al 2009). There is also evidence that thiamine- lactic acidosis and acute encephalopathy which responded im-
responsiveness decreases after puberty and adult patients mediately to thiamine and biotin (Perez-Duenas et al 2013).
may become transfusion- and insulin-dependent in spite of Although metabolic abnormalities in this patient were con-
previously adequate thiamine supplementation (Ricketts et al trolled with continuing thiamine supplementation, some neuro-
2006). logical sequelae remained. Several patients have presented later
in life in adolescence or as adults, including a 15 year old girl
THTR2 deficiency with a Leigh-like encephalopathy (Fassone et al 2013), two
adult brothers with a Wernicke-like encephalopathy (Kono et al
This was initially described in patients from Saudi Arabia as 2009), and two adult sisters with generalised dystonia and
biotin-responsive basal ganglia disease (Ozand et al 1998). seizures (Debs et al 2010).
Patients presented in childhood with a subacute encephalop- Recently, there have been two reports of SLC19A3 muta-
athy and symmetrical lesions in the basal ganglia, particularly tions in patients from eight unrelated families with an early
the caudate nucleus and putamen. Clinical manifestations onset, fatal encephalopathy and a characteristic pattern of brain
included confusion, difficulties with speech and swallowing, MRI abnormalities with cerebral atrophy and symmetrical le-
dystonia and rigidity. These responded very rapidly to biotin sions in the thalami, basal ganglia and brain stem (Gerards et al
supplementation at doses of 510 mg/day and the patients did 2013; Kevelam et al 2013). In some of these patients, various
not have neurological sequelae if recognised early and treated biochemical abnormalities have been documented, including
continuously. The genetic defect in SLC19A3, the gene for raised blood and cerebrospinal fluid lactate concentration and
THTR2 was identified in studies on this group of patients, in reduced activity of pyruvate and -ketoglutarate dehydroge-
which there is a high degree of parental consanguinity and a nase in muscle. In patients who received thiamine supplemen-
common missense mutation, p.thr422ala (Zeng et al 2005). tation, there was improved survival.
Although the clinical presentation and brain MRI changes In early-onset patients, the most effective treatment appears
in patients with biotin-responsive basal ganglia disease have to be a combination of biotin and thiamine. The range of doses
J Inherit Metab Dis

used has been quite wide, with biotin doses between 2 and simple screening test for this condition. While the primary
10 mg/day and thiamine doses of 100400 mg/day being the defect should theoretically affect all enzymes which have TPP
most common (Alfadhel et al 2013; Tabarki et al 2013). In the as cofactor, the most significant effects appear to be on pyru-
Japanese patients with infantile spasms and delayed develop- vate dehydrogenase and -ketoglutarate dehydrogenase.
ment, biotin was ineffective and they were treated with thia- When assayed in the presence of TPP, pyruvate dehydroge-
mine alone. Similarly, the patients presenting with a nase activity in muscle was normal.
Wernicke-like encephalopathy also responded to thiamine
alone. A significant number of different mutations in the
SLC19A3 gene have been identified and there is no clear Mitochondrial TPP transporter deficiency
genotype-phenotype correlation to inform treatment.
The most striking aspect of this condition is the response The mitochondrial TPP transporter is the product of the
to biotin supplementation, given that THTR2 does not trans- SLC25A19 gene and was originally described as the mito-
port biotin (Subramanian et al 2006). An explanation for the chondrial deoxynucleotide carrier (Dolce et al 2001; Kang and
responsiveness may come from studies of the role of biotin Samuels 2008). Deficiency of this carrier was first described
in gene expression. In addition to being a cofactor for in the condition of Amish lethal microcephaly (OMIM
various biotin-containing enzymes, biotin is also involved 607196), a condition with a characteristic facial appearance,
in the expression of a number of genes including those for brain MRI findings and increased urinary excretion of -
enzymes involved in glucose metabolism, biotin transport, ketoglutarate. All patients from the Amish community have
carboxylase apoenzymes, cytokines and oncogenes a common missense mutation, p.Gly177Ala (Rosenberg et al
(Dakshinamurti 2005; Rodriguez-Melendez and Zempleni 2002). While this condition is usually associated with early
2003). The mechanism for the regulation of gene expression lethality, there is one report of a patient of Amish ancestry who
involves activation of cGMP signalling pathways by has survived into late childhood (Siu et al 2010).
biotinyl-AMP and biotinylation of histones H3 and H4 by Subsequent identification of mutations in the SLC25A19
holocarboxylase synthase (Rodriguez-Melendez and gene in patients outside of this inbred community suggests
Zempleni 2003). In a study of human subjects made mar- that the spectrum of this condition is likely to be much
ginally biotin deficient, Vlasova et al demonstrated a signif- broader. Four siblings have been described with a similar
icant reduction in expression of the SLC19A3 gene (Vlasova presentation of acute episodes of flaccid paralysis and enceph-
et al 2005). In patients with THTR2 deficiency due to alopathy precipitated by intercurrent illness (Spiegel et al
mutations in this gene, limited availability of biotin may 2009). During acute episodes, they developed a motor and
be sufficient to aggravate the basic defect and this could be sensory neuropathy, with a raised cerebrospinal fluid lactate
overcome by dietary supplementation. concentration. Between episodes, there was some residual
weakness, and a progressive polyneuropathy. There was no
cognitive impairment. Brain MRI revealed basal ganglia
Thiamine pyrophosphokinase deficiency changes affecting the caudate nucleus and putamen, with
sparing of the globus pallidus. Excretion of -ketoglutarate
This condition has only recently been reported in five patients was not increased, and in one patient, activity of respiratory
from three families (Mayr et al 2011, OMIM 606370). Onset chain complexes and pyruvate dehydrogenase in muscle were
was during early childhood, in some cases after a period of normal. This condition has been designated thiamine metab-
normal development. Common features were episodic ataxia, olism syndrome 4 (bilateral striatal degeneration and progres-
psychomotor retardation and dystonia, with additional spas- sive polyneuropathy type, OMIM 613710).
ticity and seizures in one of the patients. Brain MRI changes
were quite variable with global atrophy, and abnormal signal
in the basal ganglia, cerebellum and brain stem. While two Thiamine-responsive -ketoacid dehydrogenase
patients died in childhood, three are currently being treated deficiencies
with thiamine 100200 mg/day and a high fat diet. One of
these attends normal school and has normal development. All A clinical and/or biochemical response to pharmacological
patients have mutations in the TPK1 gene. doses of thiamine has been reported in a small number of
These patients have all had extensive biochemical investi- pyruvate dehydrogenase deficiency (OMIM 312170) and ma-
gation which revealed consistent elevation of blood and cere- ple syrup urine disease (OMIM 248600) patients. This respon-
brospinal fluid lactate concentration during episodes of ataxia, siveness suggests a structural abnormality in the complexes
and enhanced excretion of -ketoglutarate in urine. All pa- which impairs TPP binding, but which can be overcome in the
tients had reduced TPP concentration in both blood and mus- presence of a high concentration of the cofactor. From the
cle and measurement of the concentration in blood provides a published reports, it is difficult to assess the significance of the
J Inherit Metab Dis

thiamine-responsiveness, as thiamine supplementation concentration. Initial reports suggested that assaying the com-
alone has usually been insufficient to control symptoms and plex from cultured cells with a range of TPP concentrations
has had to be combined with other therapeutic measures. An was sufficient to differentiate responsive and non-responsive
extremely wide range of doses, 501200 mg/day, has been patients (Naito et al 2002b). In our experience, this is often not
used, and only in about half of the cases has a clear and the case, and patient fibroblasts may have normal activity even
sustained response been demonstrated, either clinically or when cultured and assayed in very low thiamine concentra-
biochemically. There are very few reports of long term follow tions (van Dongen et al 2014). In these cases, the diagnosis
up, but several patients have survived to late childhood and are may be missed unless more extensive investigations to expose
attending normal school. impaired cofactor binding are performed.
Thiamine-responsive pyruvate dehydrogenase deficiency As thiamine responsiveness is not easily demonstrated by
has been reported in over 20 patients (Di Rocco et al 2000; biochemical analysis, attempts have also been made to iden-
Lee et al 2006; Naito et al 2002a, 2002b; Pastoris et al 1996; tify specific mutations which are associated with the respon-
Sedel et al 2008). These patients are usually at the less severe sive phenotype. Although the TPP binding site is in the
end of the spectrum of this condition, presenting typically in interface between the and subunits of the E1 component
late infancy with hypotonia and delayed development, or of the complex, so far all mutations in thiamine-responsive
episodes of ataxia associated with intercurrent illness. Blood patients have been located in the PDHA1 gene which encodes
and cerebrospinal fluid lactate concentrations are usually the E1 subunit. In general, these mutations do not involve
raised during acute episodes, but may be normal or moderate- amino acid residues which form the actual TPP binding site,
ly elevated at other times. In almost all cases, brain MRI instead, most are located in one of two adjacent regions. These
imaging reveals symmetrical lesions in basal ganglia and brain mutations presumably affect cofactor binding indirectly,
stem, typical of Leigh syndrome. Several patients have been through interactions with the amino acids which form the site
described with prolonged survival, well preserved cognitive itself (Fig. 5). While most cases of thiamine-responsive pyru-
function, and a clinical presentation dominated by peripheral vate dehydrogenase deficiency have mutations in these re-
neuropathy, rather than the more common central neurological gions, other mutations in the PDHA1 gene which are well
deficits (Bachmann-Gagescu et al 2009; Lee et al 2006; away from the TPP binding site have also been implicated in
Marsac et al 1997). some patients. These may result in altered stability of the
Because of variability in the clinical and biochemical re- complex which may be overcome by high concentrations of
sponse to thiamine, underlying biochemical defects in these the cofactor.
patients have been investigated in vitro in an attempt to Thiamine-responsive maple syrup urine disease has been
confirm that there is impaired cofactor binding to the complex reported in over ten patients (Simon et al 2006). These again
and that this can be overcome by increasing the thiamine usually present in late infancy with delayed development and

I87M, R88S, R88C, G89S

TPP
Y118, R119

F205L, M210V, L216F

D196, G197, A198


N225, Y227

Fig. 5 Amino acid substitutions associated with thiamine-responsive subunit and the pyrophosphate attached to the E1 subunit by the amino
pyruvate dehydrogenase deficiency. The region of the E1 subunit sur- acid residues highlighted in black. The majority of amino acid substitu-
rounding the thiamine pyrophosphate binding site is shown. The cofactor tions which are associated with thiamine responsiveness involve residues
(shown in ball and stick form) is bound in the interface between the E1 adjacent to the binding site on either side (shown in red)
and E1 subunits, with the thiamine attached primarily to the E1
J Inherit Metab Dis

episodes of ketoacidosis or ataxia. Again a wide range of administered and the requirement for additional dietary ma-
thiamine doses has been administered, up to 1000 mg/day, nipulation, all make it difficult to determine the extent to
and patients have also received dietary branched chain amino which true thiamine responsiveness exists in these conditions.
acid restriction. Several patients remain healthy as adults, It is anticipated that the spectrum of the thiamine metabo-
without episodes of ketoacidosis and with normal cognitive lism dysfunction syndromes will continue to expand as new
function on this regimen. Although the TPP binding site of the patients are identified, either by biochemical or genetic anal-
branched chain -ketoacid dehydrogenase is in the interface ysis and that a clearer idea of the true incidence will be
between the E1 and E1 subunits, as in pyruvate dehydro- obtained. Although, at present, it appears that the individual
genase, almost all thiamine-responsive maple syrup urine disorders of thiamine metabolism are quite rare, they all
disease patients have been found to have mutations in the emphasise the importance of a trial of thiamine supplementa-
DBT gene for the E2 component of the complex (Chuang tion in appropriate patients, both for potential therapy and to
et al 2006). While the E2 protein is not directly involved in indicate possible diagnoses for specific investigation.
TPP binding, in vitro studies suggest that the affinity of the
E1 subunit for TPP is influenced by binding to this compo-
Compliance with Ethics Guidelines
nent, which forms the core of the complex (Chuang et al
2004). No responsive patients with mutations in the Conflict of interest None.
BCKDHA gene for the E1 subunit have been described,
but there is a recent report of a responsive patient with a Animal Rights This article does not contain any studies with human or
mutation in the BCKDHB gene which codes for the E1 animal subjects performed by the author.
subunit (Yang et al 2012).

Conclusion References

The two thiamine transporters, THTR1 and THTR2, are both Alfadhel M, Almuntashri M, Jadah RH et al (2013) Biotin-responsive
widely distributed, however, differences in their expression basal ganglia disease should be renamed biotin-thiamine-responsive
basal ganglia disease: a retrospective review of the clinical, radio-
may account for the different manifestations of deficiencies. logical and molecular findings of 18 new cases. Orphanet J Rare Dis
Thiamine is converted to the active thiamine pyrophosphate 8:83
cofactor in the cytoplasm of the cell, and a specific transporter Bachmann-Gagescu R, Merritt JL, 2nd, Hahn SH (2009) A cognitively
then delivers this to the mitochondrion. Apart from THTR1 normal PDH-deficient 18-year-old man carrying the R263G muta-
tion in the PDHA1 gene. J Inherit Metab Dis 32 Suppl 1 doi: 10.
deficiency, which results in a well-defined condition of 1007/s10545-009-1101-4
thiamine-responsive megaloblastic anaemia, consequences Bazarbachi A, Muakkit S, Ayas M, Taher A, Salem Z, Solh H, Haidar JH
of defects in thiamine transport and metabolism are quite (1998) Thiamine-responsive myelodysplasia. Br J Haematol 102:
variable and often non-specific, with considerable overlap 10981100
Borgna-Pignatti C, Azzalli M, Pedretti S (2009) Thiamine-responsive
with various mitochondrial disorders. Biochemical abnormal- megaloblastic anemia syndrome: long term follow-up. J Pediatr
ities are inconstant, although raised blood and cerebrospinal 155:295297
fluid lactate concentrations and increased urinary excretion of Boros LG, Steinkamp MP, Fleming JC, Lee WN, Cascante M, Neufeld EJ
-ketoglutarate are among the most frequent findings. In the (2003) Defective RNA ribose synthesis in fibroblasts from patients
with thiamine-responsive megaloblastic anemia (TRMA). Blood
absence of specific diagnostic features, patients with this 102:35563561
group of conditions are increasingly being recognised by Boulware MJ, Subramanian VS, Said HM, Marchant JS (2003) Polarized
genome sequencing. expression of members of the solute carrier SLC19A gene family of
In addition to defects in thiamine transport and activation, water-soluble multivitamin transporters: implications for physiolog-
ical function. Biochem J 376:4348
there are also defects in the binding of TPP to some of the Casteels M, Sniekers M, Fraccascia P, Mannaerts GP, Van Veldhoven PP
enzymes for which this is a cofactor. Patients with thiamine- (2007) The role of 2-hydroxyacyl-CoA lyase, a thiamin
responsive pyruvate dehydrogenase deficiency and maple pyrophosphate-dependent enzyme, in the peroxisomal metabolism
syrup urine disease have been described, but it is often diffi- of 3-methyl-branched fatty acids and 2-hydroxy straight-chain fatty
acids. Biochem Soc Trans 35:876880
cult to correlate a clinical response with the results of either Chuang JL, Wynn RM, Moss CC, Song JL, Li J, Awad N, Mandel H,
biochemical or genetic analysis. In general, responsive pa- Chuang DT (2004) Structural and biochemical basis for novel
tients have less severe illness and a more prolonged course, mutations in homozygous Israeli maple syrup urine disease patients:
however follow up studies are very limited and it is not known a proposed mechanism for the thiamin-responsive phenotype. J Biol
Chem 279:1779217800
if long term problems may still develop. Few of these patients Chuang DT, Chuang JL, Wynn RM (2006) Lessons from genetic disor-
have been fully normal, or have reached adulthood. Genetic ders of branched-chain amino acid metabolism. J Nutr 136:243S
heterogeneity, great variation in the dose of thiamine 249S
J Inherit Metab Dis

Dakshinamurti K (2005) Biotina regulator of gene expression. J Nutr Onal H, Baris S, Ozdil M et al (2009) Thiamine-responsive megaloblastic
Biochem 16:419423 anemia: early diagnosis may be effective in preventing deafness.
Debs R, Depienne C, Rastetter A et al (2010) Biotin-responsive basal Turkish J Pediatr 51:301304
ganglia disease in ethnic Europeans with novel SLC19A3 muta- Ozand PT, Gascon GG, Al Essa M et al (1998) Biotin-responsive basal
tions. Arch Neurol 67:126130 ganglia disease: a novel entity. Brain 121:12671279
Di Rocco M, Lamba LD, Minniti G, Caruso U, Naito E (2000) Outcome Pastoris O, Savasta S, Foppa P, Catapano M, Dossena M (1996) Pyruvate
of thiamine treatment in a child with Leigh disease due to thiamine- dehydrogenase deficiency in a child responsive to thiamine treat-
responsive pyruvate dehydrogenase deficiency. Eur J Paediatr ment. Acta Paediatr 85:625628
Neurol 4:115117 Perez-Duenas B, Serrano M, Rebollo M, Muchart J, Gargallo E, Dupuits
Dolce V, Fiermonte G, Runswick MJ, Palmieri F, Walker JE (2001) The C, Artuch R (2013) Reversible lactic acidosis in a newborn with
human mitochondrial deoxynucleotide carrier and its role in the thiamine transporter-2 deficiency. Pediatrics 131:e1670e1675
toxicity of nucleoside antivirals. Proc Natl Acad Sci U S A 98: Ricketts CJ, Minton JA, Samuel J, Ariyawansa I, Wales JK, Lo IF, Barrett
22842288 TG (2006) Thiamine-responsive megaloblastic anaemia syndrome:
Fassone E, Wedatilake Y, DeVile CJ, Chong WK, Carr LJ, Rahman S long-term follow-up and mutation analysis of seven families. Acta
(2013) Treatable Leigh-like encephalopathy presenting in adoles- Paediatr 95:99104
cence. BMJ Case Rep 2013:200838 Rindi G, Laforenza U (2000) Thiamine intestinal transport and related
Ganapathy V, Smith SB, Prasad PD (2004) SLC19: the folate/thiamine issues: recent aspects. Proc Soc Exp Biol Med 224:246255
transporter family. Pflugers Archiv: Eur J Physiol 447:641646 Rodriguez-Melendez R, Zempleni J (2003) Regulation of gene expres-
Gerards M, Kamps R, van Oevelen J et al (2013) Exome sequencing sion by biotin (review). J Nutr Biochem 14:680690
reveals a novel Moroccan founder mutation in SLC19A3 as a new Rosenberg MJ, Agarwala R, Bouffard G et al (2002) Mutant
cause of early-childhood fatal Leigh syndrome. Brain 136:882890 deoxynucleotide carrier is associated with congenital microcephaly.
Kang J, Samuels DC (2008) The evidence that the DNC (SLC25A19) is Nat Genet 32:175179
not the mitochondrial deoxyribonucleotide carrier. Mitochondrion 8: Scharfe C, Hauschild M, Klopstock T, Janssen AJ, Heidemann
103108 PH, Meitinger T, Jaksch M (2000) A novel mutation in the
Kevelam SH, Bugiani M, Salomons GS et al (2013) Exome sequencing thiamine responsive megaloblastic anaemia gene SLC19A2 in
reveals mutated SLC19A3 in patients with an early-infantile, lethal a patient with deficiency of respiratory chain complex I. J
encephalopathy. Brain 136:15341543 Med Genet 37:669673
Kono S, Miyajima H, Yoshida K, Togawa A, Shirakawa K, Suzuki H Sedel F, Challe G, Mayer JM, Boutron A, Fontaine B, Saudubray JM,
(2009) Mutations in a thiamine-transporter gene and Wernickes-like Brivet M (2008) Thiamine responsive pyruvate dehydrogenase de-
encephalopathy. New Engl J Med 360:17921794 ficiency in an adult with peripheral neuropathy and optic neuropa-
Lee EH, Ahn MS, Hwang JS, Ryu KH, Kim SJ, Kim SH (2006) A thy. J Neurol Neurosurg Psychiatr 79:846847
Korean female patient with thiamine-responsive pyruvate dehydro- Shaw-Smith C, Flanagan SE, Patch AM et al (2012) Recessive
genase complex deficiency due to a novel point mutation (Y161C)in SLC19A2 mutations are a cause of neonatal diabetes mellitus
the PDHA1 gene. J Korean Med Sci 21:800804 in thiamine-responsive megaloblastic anaemia. Pediatr
Lindhurst MJ, Fiermonte G, Song S et al (2006) Knockout of Slc25a19 Diabetes 13:314321
causes mitochondrial thiamine pyrophosphate depletion, embryonic Simon E, Flaschker N, Schadewaldt P, Langenbeck U, Wendel U (2006)
lethality, CNS malformations, and anemia. Proc Natl Acad Sci U S Variant maple syrup urine disease (MSUD)the entire spectrum. J
A 103:1592715932 Inherit Metab Dis 29:716724
Lorber A, Gazit AZ, Khoury A, Schwartz Y, Mandel H (2003) Cardiac Siu VM, Ratko S, Prasad AN, Prasad C, Rupar CA (2010) Amish
manifestations in thiamine-responsive megaloblastic anemia syn- microcephaly: Long-term survival and biochemical characteriza-
drome. Pediatr Cardiol 24:476481 tion. Am J Med Genet A 152A:17471751
Marsac C, Benelli C, Desguerre I et al (1997) Biochemical and genetic Spiegel R, Shaag A, Edvardson S et al (2009) SLC25A19 mutation as a
studies of four patients with pyruvate dehydrogenase E1 alpha cause of neuropathy and bilateral striatal necrosis. Ann Neurol 66:
deficiency. Hum Genet 99:785792 419424
Mayr JA, Freisinger P, Schlachter K et al (2011) Thiamine Subramanian VS, Marchant JS, Said HM (2006) Biotin-responsive basal
pyrophosphokinase deficiency in encephalopathic children with defects ganglia disease-linked mutations inhibit thiamine transport via
in the pyruvate oxidation pathway. Am J Hum Genet 89:806812 hTHTR2: biotin is not a substrate for hTHTR2. Am J Physiol Cell
Nabokina SM, Said HM (2012) A high-affinity and specific carrier- Physiol 291:C851C859
mediated mechanism for uptake of thiamine pyrophosphate by Tabarki B, Al-Shafi S, Al-Shahwan S et al (2013) Biotin-responsive basal
human colonic epithelial cells. Am J Physiol Gastrointest Liver ganglia disease revisited: clinical, radiologic, and genetic findings.
Physiol 303:G389G395 Neurology 80:261267
Naito E, Ito M, Yokota I et al (2002a) Thiamine-responsive pyruvate Van Dongen S, Brown RM, Brown GK, Thorburn DR, Boneh A (2014)
dehydrogenase deficiency in two patients caused by a point muta- Diagnosis of thiamine-responsive and thiamine-non-responsive pa-
tion (F205L and L216F) within the thiamine pyrophosphate binding tients with pyruvate deydrogenase complex deficiency. JIMD Rep.
region. Biochim Biophys Acta 1588:7984 doi:10.1007/8904_2014_293
Naito E, Ito M, Yokota I, Saijo T, Ogawa Y, Kuroda Y (2002b) Diagnosis Vlasova TI, Stratton SL, Wells AM, Mock NI, Mock DM (2005)
and molecular analysis of three male patients with thiamine- Biotin deficiency reduces expression of SLC19A3, a potential
responsive pyruvate dehydrogenase complex deficiency. J Neurol biotin transporter, in leukocytes from human blood. J Nutr
Sci 201:3337 135:4247
Neufeld EJ, Fleming JC, Tartaglini E, Steinkamp MP (2001) Thiamine- Yamada K, Miura K, Hara K et al (2010) A wide spectrum of clinical and
responsive megaloblastic anemia syndrome: a disorder of high- brain MRI findings in patients with SLC19A3 mutations. BMC Med
affinity thiamine transport. Blood Cell Mol Dis 27:135138 Genet 11:171
J Inherit Metab Dis

Yang N, Han L, Gu X et al (2012) Analysis of gene mutations in Chinese Zhao R, Goldman ID (2013) Folate and thiamine transporters mediated
patients with maple syrup urine disease. Mol Genet Metab 106:412 by facilitative carriers (SLC19A1-3 and SLC46A1) and folate re-
418 ceptors. Mol Aspects Med 34:373385
Zeng WQ, Al-Yamani E, Acierno JS Jr et al (2005) Biotin-responsive Zhao R, Gao F, Goldman ID (2002) Reduced folate carrier transports
basal ganglia disease maps to 2q36.3 and is due to mutations in thiamine monophosphate: an alternative route for thiamine delivery
SLC19A3. Am J Hum Genet 77:1626 into mammalian cells. Am J Physiol Cell Physiol 282:C1512C1517

Вам также может понравиться