Download 16) Clifton-Bligh RJ, de Zegher F, Wagner RL, Collingwood TN

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

Hormone replacement therapy (menopause) wikipedia , lookup

Hormone replacement therapy (male-to-female) wikipedia , lookup

Iodine-131 wikipedia , lookup

Hypothyroidism wikipedia , lookup

Growth hormone therapy wikipedia , lookup

Hyperthyroidism wikipedia , lookup

Androgen insensitivity syndrome wikipedia , lookup

Transcript
1
Resistance to Thyroid Hormone - emerging definition of a disorder of thyroid
2
hormone action
3
4
Carla Moran and Krishna Chatterjee
5
6
University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC, Institute of
7
Metabolic Science, Addenbrooke’s Hospital, Cambridge CB2 0QQ, United Kingdom
8
9
Abbreviated title: RTH.
10
11
Key terms: Resistance to Thyroid Hormone, dominant negative inhibition; THRA mutation
12
13
Disclosure statement: The authors have nothing to disclose
14
15
Word Count: text (1651); figures 1
16
17
Address for correspondence and reprint requests:
18
V Krishna K Chatterjee, University of Cambridge, Metabolic Research Laboratories, Wellcome Trust-
19
MRC Institute of Metabolic Science, Level 4, Box 289, Addenbrooke’s Hospital, Cambridge, CB2
20
0QQ. Tel: (44)-1223-336842; Fax: (44)-1223-330598; Email: [email protected]
21
22
Funding: Our research is supported by funding from the Wellcome Trust (095564/Z/11/Z to KC) and
23
National Institute for Health Research Cambridge Biomedical Research Centre (CM, KC).
24
25
26
27
1
28
Thyroid hormones (TH) act via nuclear receptors, encoded by separate genes (THRA, THRB), with
29
differing tissue distributions (TR1: central nervous system, myocardium, skeletal muscle,
30
gastrointestinal tract; TR1: liver, kidney; TR2: hypothalamus, pituitary, cochlea, retina).
31
Alternative splicing of THRA generates a variant, non TH-binding, protein (TR2), with a divergent
32
carboxyterminal domain, whose function is not understood (1).
33
Resistance to thyroid hormone  (RTH), usually due to heterozygous mutations in THRB, is
34
characterised by raised T4 and T3 levels, non-suppressed TSH and a variable phenotype
35
encompassing both hyperthyroid (e.g. failure to thrive, raised metabolic rate, tachycardia, low bone
36
density, anxiety) and hypothyroid (e.g. dyslipidaemia) features (2). Consonant with its dominant
37
mode of inheritance, dominant negative inhibition of wild type receptor function by mutant TR is
38
central to the pathogenesis of this disorder (3). Although TR and  proteins exhibit marked
39
aminoacid sequence similarity, such that introduction of a  receptor mutation causing RTH into the
40
murine TR gene is associated with an abnormal phenotype (e.g. growth retardation, locomotor
41
abnormalities) (4), the homologous human disorder (Resistance to Thyroid Hormone , RTH),
42
eluded discovery for over a decade probably because it was not associated with a readily-recognisable
43
diagnostic signature.
44
RTH, due to a highly deleterious mutation (E403X), truncating the carboxyterminal region of TR1,
45
was first identified in a six yr old girl with hypothyroid features (e.g. skeletal dysplasia with growth
46
retardation, neurodevelopmental delay, constipation) in selected target tissues, but associated with
47
near-normal (borderline low or normal T4, borderline high or normal T3, normal TSH) thyroid
48
function tests (5). Subsequently, further childhood plus adult male and female cases, harbouring
49
frameshift/premature stop mutations also involving the carboxyterminus of TR1 were found, with
50
recognition of additional, potentially pathognomonic, features (macrocephaly, anaemia, dysmorphic,
51
facies) (6,7). Indeed, recognition of a distinct, shared, phenotype encompassing macrocephaly and
52
dysmorphic features in a single clinic, led to the identification of a further five cases with similar,
53
carboxyterminal TR1 mutations (8). Where functionally characterised, these TR1 mutants
54
exhibited negligible hormone binding and functional activity, with failure to dissociate from
2
55
transcriptional corepressor and recruit coactivator proteins – properties that are highly analogous to
56
those of dominant negative TRmutants in RTH (3). From these reports, a biochemical signature for
57
RTH also seemed to emerge; although TH levels could be in the normal range, a computed T4/T3
58
ratio or reverse T3 levels (where ascertained) were subnormal (5,8,9). Following this, a missense
59
THRA mutation (A263V), located in a receptor region common to both TR1 and TR2 proteins, was
60
identified in three members of a family with similar clinical (growth and developmental retardation,
61
constipation, macrocephaly) features. The A263V TR1 mutant showed partial loss-of-function,
62
being transcriptionally impaired at low TH concentrations in vitro, with reversal of this at higher T3
63
levels. It was tempting to speculate that such partial preservation of mutant TR1 function accounted
64
for a milder phenotype in this family. However, the possibility that thyroxine treatment of these cases
65
since early childhood had also contributed to amelioration of the phenotype, could not be discounted.
66
Concordant with no measurable gain- or loss-of-function of A263V mutant TR2 compared with its
67
normal counterpart, no added phenotypes attributable to mutated TR2, could be discerned in these
68
cases (10). In contrast, some unusual added clinical features (micrognathia, clavicular agenesis,
69
metacarpal fusion and syndactyly of digits, hyperparathyroidism, chronic diarrhoea) were reported in
70
an adult female harbouring a different mutation (N359Y), common to both TR1 and TR2 proteins
71
(11).
72
In this context, the report by Demir and colleagues in the current issue of JCEM (12), describing three
73
different THRA mutations (C380fs387X, R384H, A263S) in 10 RTH patients from 3 different
74
families, together with associated clinical features, clarifies several aspects of this disorder. The
75
authors were able to discern a gradation in phenotype associated with different genotypes: a child,
76
harbouring a highly deleterious mutation (C380fs387X) involving the carboxyterminus of TR1, is
77
growth retarded with cognitive and motor deficits that result in severe handicap; moderate cognitive
78
deficit, motor abnormalities and growth retardation in infancy but less evident in adulthood, were
79
recorded in a child and mother with a partial loss-of-function mutation (R384H) in TR1; in a large
80
family, harbouring A263S mutant TR1 with slightly impaired function, mild growth retardation and
3
81
slight developmental delay in childhood culminated in average/low-average adult IQ, with
82
constipation being a relatively conserved clinical feature.
83
The A263S mutation in THRA reported by Demir et al, as well as another RTH-associated mutation
84
(D211G) reported recently by the same group, also involve the TR2 protein (12,13). No added
85
phenotypic characteristics were observed in nine affected individuals harbouring one or other of these
86
mutated residues. Accordingly, it is conceivable that the unusual skeletal and other abnormalities
87
unique to the patient with the N359Y THRA mutation, are attributable to a defect in another pathway;
88
although whole exome sequencing was undertaken in this case, a pathogenic abnormality (for
89
example in a non-coding gene region) has not been excluded (11). Alternatively, as depicted when
90
RTH mutations reported hitherto are mapped on the crystal structure of TR1, the Asparagine to
91
Tyrosine change at codon 359 (N359Y), involves a residue that is in an unusual location in the
92
receptor protein compared to the position of other RTH-associated mutations (Figure 2); therefore,
93
the possibility of the N359Y substitution inducing a specific functional alteration in the receptor that
94
is linked to unique, additional phenotypes, cannot be discounted.
95
With the identification of a more functionally diverse repertoire of TR1 mutations, a specific
96
biochemical signature for RTH seems less clearcut. Specifically, in some cases, particularly
97
associated with mild TR mutations (e.g. A263S, D211G), normal reverse T3 levels have been
98
documented (12,13). The T4/T3 or T3/rT3 ratio remains abnormal in most cases, overlapping
99
minimally with values in unaffected family members (12), but this abnormal thyroid function test
100
pattern is a recognised feature of other disorders (e.g. MCT8 deficiency) (14); likewise, although mild
101
anaemia is almost universal in RTH, this parameter is unhelpfully nonspecific. Studies in a
102
transgenic model of RTH raised the possibility of circulating trace element (selenium) levels having
103
diagnostic utility (15), but did not translate readily into the human context (7). In RTH, even subtle
104
TR mutants exhibit impaired negative feedback regulation of hypothalamo-pituitary target genes
105
(TRH, TSH) (16), reflected in pathognomonic, raised, TH levels. It has been suggested that a
106
combination of raised type 1 deiodinase and reduced type 3 deiodinase enzyme activities accounts for
107
thyroid biochemical abnormalities in RTH17); whether another TH metabolite that reflects such
4
108
altered deiodinase activity more consistently or a biomarker associated with another universally
109
dysregulated pathway in this disorder, can be identified, remains to be seen.
110
With the estimated prevalence of RTH being ~1 in 40,000, with over 160 different TR mutations
111
recorded hitherto, it is likely that RTH is more common, but underdiagnosed. Both maternal and
112
paternal transmission of TR mutations has now been recorded in several families (6, 8, 10, 12, 13),
113
suggesting that, unlike in a murine transgenic model (18), fertility in either gender is not unduly
114
compromised. However, screening for THRA defects based on current, documented, phenotypes may
115
artificially enrich for identification of particular TR mutations (e.g. involving the carboxyterminal
116
receptor region). Supporting this notion, whole exome sequencing (WES), identified a TR variant
117
(R384C), known to be pathogenic in transgenic mice (4), associated with autism spectrum disorder –
118
an unexpected phenotype (19). Indeed, interrogation of exome databases (e.g. ExAC;
119
http://exac.broadinstitute.org/gene/ENSG00000126351) reveals many rare, nonsyonymous THRA
120
variants including several in TR1 predicted to be damaging, based either on homology to known
121
RTH mutations or structural modelling. However, in the absence of definitive diagnostic criteria
122
(clinical, biochemical) for RTH, it may be necessary to develop functional assays to assess the
123
putative pathogenicity of receptor variants, as has been done for PPARG variants identified in WES of
124
type 2 diabetes cohorts (20). TR mutations causing RTH cluster within three regions of its hormone
125
binding domain and, as illustrated in Figure 1, many RTH-associated TR mutations have TR
126
counterparts, involving the same aminoacids, which localise to these clusters. As the repertoire of
127
RTH-associated TR mutations grows, it will be interesting to see whether they also localise to
128
particular receptor regions. One functional constraint on RTH-associated TR mutations is
129
preservation of the ability to exert dominant negative activity, such that receptor regions mediating
130
functions (DNA binding, interaction with corepressor and RXR) that are required for such activity are
131
devoid of naturally-occuring mutations (3); if dominant negative inhibition is also central to the
132
pathogenesis of RTH, this property may also constrain localisation of RTHmutations within TR.
133
Seven out of fourteen TR mutations recorded hitherto have occurred de novo, with mutations
134
involving some residues (e.g. Alanine 263, Arginine 384, Glutamic Acid 403) occurring more than
5
135
once in unrelated families; as has been shown in RTH (21), it is possible that nucleotide substitutions
136
involving mutation-prone, CpG dinucleotides will occur more frequently, resulting in some aminoacid
137
changes being overrepresented.
138
Although not identified via neonatal screening (in a program with both T4 and TSH measurement),
139
thyroxine therapy from age 18 months has been beneficial for growth, motor development and
140
constipation in a childhood case with a partial loss-of-function mutation (D211G) (13). Despite the
141
first recorded patient harbouring a more deleterious E403X TR mutation (5), we have observed
142
similar benefit following five years of thyroxine therapy (Moran, Chatterjee and Dattani unpublished
143
observations). Accordingly, despite the relative paucity of data, a trial of thyroxine therapy in RTH
144
cases harbouring both mild and severe loss-of-function TR mutations may be indicated. However,
145
careful surveillance to not only assess therapeutic benefit but also monitor for unwanted tissue toxicity
146
(e.g in liver, which expresses mainly TR), is warranted. In this context, as in RTH, reliable
147
biomarkers of tissue responses to TH are needed. In the future, it also conceivable that genetic
148
stratification may guide therapy of RTH patients, with thyroxine or selective thyromimetics being
149
used in patients harbouring TR mutations with residual functional activity, with alternative
150
approaches (e.g. blocking formation or activity of receptor-transcriptional corepressor complexes) in
151
cases with severe, hormone-refractory, TR defects.
152
153
ACKNOWLEDGEMENTS
154
155
We thank Dr Louise Fairall, Department of Molecular and Cell Biology, University of Leicester, UK
156
for the crystallographic modelling shown in Figure 2.
157
6
158
159
REFERENCES
160
161
1) Lazar MA. Thyroid hormone receptors: multiple forms, multiple possibilities. Endocr Rev 1993;
162
14:184-193.
163
164
2) Refetoff S, Weiss RE, Usala SJ. The syndromes of resistance to thyroid hormone. Endocr Rev
165
1993; 14:348-399.
166
167
3) Gurnell M, Visser T, Beck-Peccoz P and Chatterjee K. Resistance to thyroid hormone. In: Jameson
168
JL & De Groot LJ, eds. Endocrinology. 7th ed. Philadelphia, PA: Saunders Elsevier; 2015:1648-1665.
169
170
4) Tinnikov A, Nordström K, Thorén P, Kindblom JM, Malin S, Rozell B, Adams M, Rajanayagam O,
171
Pettersson S, Ohlsson C, Chatterjee K, Vennström B. Retardation of post-natal development caused
172
by a negatively acting thyroid hormone receptor 1. EMBO J 2002; 21:5079-5087.
7
173
174
5) Bochukova E, Schoenmakers N, Agostini M, Schoenmakers E, Rajanayagam O, Keogh JM,
175
Henning E, Reinemund J, Gevers E, Sarri M, Downes K, Offiah A, Albanese A, Halsall D, Schwabe J,
176
Bain M, Lindley K, Muntoni F, Vargha-Khadem F, Dattani M, Farooqi S, Gurnell M, Chatterjee K. A
177
dominant negative mutation in the thyroid hormone receptor α gene. N Engl J Med 2012; 366:243-249
178
179
6) van Mullem A, Van Heerebeek R, Chrysis D, Visser E, Medici M, Andrikoula M, Tsatsoulis A,
180
Peeters R, Visser TJ. Clinical phenotype and mutant TRα1. N Engl J Med 2012; 366:1451-1453.
181
182
7) Moran C, Schoenmakers N, Agostini M, Schoenmakers E, Offiah A, Kydd A, Kahaly G, Mohr-
183
Kahaly S, Rajanayagam O, Lyons G, Wareham N, Halsall D, Dattani M, Hughes S, Gurnell M, Park
184
SM, Chatterjee K. An Adult Female With Resistance to Thyroid Hormone Mediated by Defective
185
Thyroid Hormone Receptor α. J Clin Endocrinol Metab 2013; 98: 4254-4261.
186
187
8) Tylki-Szymańska A, Acuna-Hidalgo R, Krajewska-Walasek M, Lecka-Ambroziak A, Steehouwer
188
M, Gilissen C, Brunner HG, Jurecka A, Różdżyńska-Świątkowska A, Hoischen A, Chrzanowska KH.
189
Thyroid hormone resistance syndrome due to mutations in the thyroid hormone receptor α gene
190
(THRA). J Med Gen 2015; 52: 312-316.
191
192
9) van Mullem AA, Chrysis D, Eythimiadou A, Chroni E, Tsatsoulis A, de Rijke YB, Visser WE,
193
Visser TJ, Peeters RP. Clinical phenotype of a new type of thyroid hormone resistance caused by a
194
mutation of the TRα1 receptor; consequences of LT4 treatment. J Clin Endocrinol Metab 2013;
195
98:3029-3038.
196
197
10) Moran C, Agostini M, Visser E, Schoenmakers E, Schoenmakers N, Offiah AC, Poole K,
198
Rajanayagam O, Lyons G, Halsall D, Gurnell M, Chrysis D, Efthymiadou A, Buchanan C, Aylwin S,
199
Chatterjee KK. Resistance to Thyroid Hormone caused by a mutation in thyroid hormone receptor
200
(TR) alpha1 and alpha2: clinical, biochemical and genetic analyses of three related patients. Lancet
8
201
Diabetes Endocrinol 2014; 2:619-626.
202
203
11) Espiard S, Savagner F, Flamant F, Vlaeminck-Guillem V, Guyot R, Munier M, d'Herbomez M,
204
Bourguet W, Pinto G, Rose C, Rodien P, Wémeau JL. A novel mutation in THRA gene associated with
205
an atypical phenotype of resistance to thyroid hormone. J Clin Endocrinol Metab 2015; 100:2841-2848.
206
207
12) Demir K, van Gucht A, Buyukinan M, Catli G, Ahan Y, Bas V, Dundar B, Ozkan B, Meima M,
208
Visser WE, Peeters R, Visser TJ. Diverse genotypes and phenotypes of three novel thyroid hormone
209
receptor alpha mutations J Clin Endocrinol Metab 2016; doi:10.1210/jc.2016-1404.
210
211
13) Van Gucht ALM, Meima ME, Zwaveling-Soonawala N, Visser WE, Fliers E, Wennink JMB,
212
Henny C, Visser TJ, Peeters RP, van Trotsenburg ASP. Resistance to Thyroid Hormone Alpha in an
213
18-month-old Girl: Clinical, Therapeutic and Molecular Characteristics. Thyroid 2016; 26:338-346.
214
215
216
14) Moran C, Chatterjee K. Resistance to Thyroid Hormone due to defective thyroid receptor alpha.
217
Best Pract Res Clin Endocrinol Metab 2015; 29:647-657.
218
219
15) Mittag J, Behrends T, Hoefig CS, Vennström B, Schomburg L. Thyroid hormones regulate
220
selenoprotein expression and selenium status in mice. PLoS ONE 2010; 5:e12931.
221
222
16) Clifton-Bligh RJ, de Zegher F, Wagner RL, Collingwood TN, Francois I, Van Helvoirt M,
223
Fletterick RJ, Chatterjee VK. A novel TR beta mutation (R383H) in resistance to thyroid hormone
224
syndrome predominantly impairs corepressor release and negative transcriptional regulation. Mol
225
Endocrinol 1998; 12:609-621.
226
227
17) Zavacki AM, Larsen PR. RTH, a newly recognised phenotype of the Resistance to Thyroid
9
228
Hormone (RTH) syndrome in patients with THRA mutations J Clin Endocrinol Metab 2013; 98:2684-
229
2686.
230
231
18) Kaneshige M, Suzuki H, Kaneshige K, Cheng J, Wimbrow H, Barlow C, Willingham MC, Cheng
232
S. A targeted dominant negative mutation of the thyroid hormone alpha 1 receptor causes increased
233
mortality, infertility, and dwarfism in mice. Proc Natl Acad Sci U S A 2001; 98:15095-100.
234
235
19) Yuen RK, Thiruvahindrapuram B, Merico D, Walker S, Tammimies K, Hoang N, Chrysler C,
236
Nalpathamkalam T, Pellecchia G, Liu Y, Gazzellone MJ, D'Abate L, Deneault E, Howe JL, Liu RS,
237
Thompson A, Zarrei M, Uddin M, Marshall CR, Ring RH, Zwaigenbaum L, Ray PN, Weksberg R,
238
Carter MT, Fernandez BA, Roberts W, Szatmari P, Scherer SW. Whole-genome sequencing of quartet
239
families with autism spectrum disorder. Nat Medicine 2015; 21:185-191.
240
241
20) Majithia AR, Flannick J, Shahinian P, Guo M, Bray MA, Fontanillas P, Gabriel SB; GoT2D
242
Consortium; NHGRI JHS/FHS Allelic Spectrum Project; SIGMA T2D Consortium; T2D-GENES
243
Consortium, Rosen ED, Altshuler D. Rare variants in PPARG with decreased activity in adipocyte
244
differentiation are associated with increased risk of type 2 diabetes. Proc Natl Acad Sci U S A 2014;
245
111:13127-32.
246
247
21) Weiss RE, Weinberg M, Refetoff S. Identical mutations in unrelated families with generalized
248
resistance to thyroid hormone occur in cytosine-guanine-rich areas of the thyroid hormone receptor β
249
gene. J Clin Invest 1993; 91:2408–2415.
250
10
251
FIGURE LEGENDS
252
Figure 1.
253
Schematic alignment of TR1, TR2 and TR showing functional regions (N-terminal, DNA
254
binding domain DBD, hormone binding domains); the divergent carboxyterminus of TRis cross-
255
hatched. The location of recorded RTH mutations, involving either TR1 alone or both TR1 and
256
2 proteins, is superimposed. Three regions of TR1 (aminoacids 234 to 282, 309 to 353 and 426 to
257
460), within which RTH mutations cluster are depicted. Within these clusters, published or our
258
unpublished (D265A) RTH mutations, involving aminoacids homologous to residues mutated in
259
RTH, are shown.
260
261
Figure 2: The crystal structure of the hormone binding domain of human TR1 bound to T3 (PDB
262
2H77, magenta) is shown. The location of aminoacids which are mutated (missense or premature stop
263
or frameshift/premature stop) in RTH is superimposed (red balls). The carboxyterminal region
264
(distal helix 11 to helix 12) within which RTH mutations occur, is highlighted in yellow.
265
11