10.01.2013 Views

Molecular pathogenesis of euthyroid and toxic multinodular goiter

Molecular pathogenesis of euthyroid and toxic multinodular goiter

Molecular pathogenesis of euthyroid and toxic multinodular goiter

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Endocrine Reviews. Published December 22, 2004 as doi:10.1210/er.2004-0005<br />

<strong>Molecular</strong> <strong>pathogenesis</strong> <strong>of</strong> <strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong><br />

<strong>multinodular</strong> <strong>goiter</strong><br />

KNUT KROHN 1,2 , DAGMAR FÜHRER 1 , YVONNE BAYER 1 , MARKUS ESZLINGER<br />

1 , VOLKER BRAUER 1 , SUSANNE NEUMANN 1 AND RALF PASCHKE 1<br />

1 III. Medical Department, University <strong>of</strong> Leipzig<br />

Ph.-Rosenthal-Str. 27, D-04103 Leipzig, Germany<br />

2 Interdisciplinary Center for Clinical Research Leipzig, University <strong>of</strong> Leipzig<br />

Inselstraße 22, D-04103 Leipzig, Germany<br />

Correspondence:<br />

R. Paschke, M.D.<br />

III. Medical Department, University <strong>of</strong> Leipzig<br />

Ph.-Rosenthal-Str. 27, D-04103 Leipzig, Germany<br />

Phone: 49-341-9713200<br />

Fax: 49-341-9713209<br />

email: pasr@medizin.uni-leipzig.de<br />

Copyright (C) 2004 by The Endocrine Society


Summary<br />

The purpose <strong>of</strong> this review is to summarize the current knowledge <strong>of</strong> the etiology <strong>of</strong><br />

<strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong> with respect to the epidemiology, clinical<br />

characteristics <strong>and</strong> molecular pathology.<br />

In reconstructing the line <strong>of</strong> events from early thyroid hyperplasia to <strong>multinodular</strong> <strong>goiter</strong> we<br />

will argue the predominant neoplastic character <strong>of</strong> nodular structures, the nature <strong>of</strong> known<br />

somatic mutations <strong>and</strong> the importance <strong>of</strong> mutagenesis. Furthermore, we outline direct <strong>and</strong><br />

indirect consequences <strong>of</strong> these somatic mutations for thyroid pathophysiology <strong>and</strong> summarize<br />

information concerning a possible genetic backround <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong>.<br />

Finally we discuss uncertainties/open questions in differential diagnosis <strong>and</strong> therapy <strong>of</strong><br />

<strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong>.<br />

I Definition <strong>and</strong> Epidemiology<br />

II Clinical aspects <strong>of</strong> <strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong><br />

A Differential diagnosis<br />

III Natural course <strong>of</strong> <strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong><br />

A Nodule Growth<br />

B Thyroid function<br />

IV Clonal origin <strong>of</strong> thyroid nodules<br />

V Hot thyroid nodules<br />

A Signal transduction <strong>of</strong> HTN with <strong>and</strong> without TSHR mutations<br />

B Secondary/indirect effects <strong>of</strong> activating TSHR mutations<br />

VI Cold thyroid nodules<br />

A Iodide transport <strong>and</strong> metabolism<br />

B Signaling proteins<br />

C Results <strong>of</strong> gene expression studies by arrays<br />

D Chromosomal aberrations<br />

VII Multinodular Goiter<br />

A Mutagenesis as the cause <strong>of</strong> nodular transformation<br />

B Etiology<br />

VIII Pathogenesis <strong>and</strong> genetic etiology <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong><br />

A Family <strong>and</strong> twin studies<br />

B C<strong>and</strong>idate loci<br />

C Linkage analysis<br />

IX Perspectives<br />

A Therapeutic implications<br />

B Diagnostic implications


I Definition <strong>and</strong> Epidemiology<br />

Benign nodular thyroid disease constitutes a heterogenous thyroid disorder, which is highly<br />

prevalent in iodine deficient areas. On a very general basis it can be divided into solitary<br />

nodular <strong>and</strong> <strong>multinodular</strong> thyroid disease. Histologically, benign thyroid nodules are<br />

distinguished as 1. encapsulated lesions (true adenomas) or adenomatous nodules, which lack<br />

a capsule, <strong>and</strong> 2. by morphological criteria according to the WHO classification (1). On<br />

functional grounds, nodules are classified as either “cold”, “normal” or “hot” depending on<br />

whether they show decreased, normal or increased uptake on scintiscan. Approximately 85%<br />

<strong>of</strong> all nodules are “cold”, 10% are normal <strong>and</strong> 5% are “hot” (2;3), although the prevalence<br />

may vary geographically with the ambient iodine supply. In contrast to solitary nodular<br />

thyroid disease, which has a more uniform clinical, pathological <strong>and</strong> molecular picture,<br />

<strong>euthyroid</strong> <strong>multinodular</strong> <strong>goiter</strong> (MNG) <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong> (TMNG) are a mixed<br />

group <strong>of</strong> nodular entities, i.e. one usually finds a combination <strong>of</strong> hyper-, hyp<strong>of</strong>unctional or<br />

normally functioning thyroid lesions within the same thyroid gl<strong>and</strong>. The overall balance <strong>of</strong><br />

functional properties <strong>of</strong> individual thyroid nodules within a <strong>multinodular</strong> <strong>goiter</strong> ultimately<br />

determine the functional status in the individual patient, which may be <strong>euthyroid</strong>ism (normal<br />

TSH <strong>and</strong> free thyroid hormone levels), subclinical hyperthyroidism (low or suppressed TSH<br />

<strong>and</strong> normal free thyroid hormone levels) or overt hyperthyroidism (suppressed TSH <strong>and</strong><br />

elevated free thyroid hormone levels). The term MNG is applied in the first scenario while<br />

TMNG refers to the latter situations. It is important to emphasize that this functional picture is<br />

not stationary but patients with TMNG usually have a history <strong>of</strong> long-st<strong>and</strong>ing MNG (4).<br />

Moreover, the status <strong>of</strong> TSH suppression in TMNG does not only imply clinical consequences<br />

for the patient but importantly also indicates that a critical level <strong>of</strong> thyroid autonomy i.e.<br />

independence <strong>of</strong> thyrotropin (TSH), the physiological regulator <strong>of</strong> thyroid function <strong>and</strong><br />

growth (5;6) has been reached. Constitutive activation <strong>of</strong> the cAMP signaling pathway is<br />

widely accepted as the biochemical driving force <strong>of</strong> thyroid autonomy as suggested by the<br />

3


presence <strong>of</strong> somatic activating TSH receptor (TSHR) mutations in scintigraphically non-<br />

suppressible foci in <strong>euthyroid</strong> <strong>goiter</strong>s in iodine deficient areas, the presence <strong>of</strong> somatic TSHR<br />

mutations <strong>and</strong> less frequently Gs alpha protein mutations in macroscopic <strong>toxic</strong> thyroid<br />

nodules both in solitary nodules <strong>and</strong> <strong>multinodular</strong> disease, the phenotype <strong>of</strong> patients with<br />

activating germline TSHR mutations <strong>and</strong> a number <strong>of</strong> animal models <strong>of</strong> thyroid autonomy<br />

(reviewed in (7-10)).<br />

Iodine deficiency is by far the best studied epidemiologic risk factor for nodular thyroid<br />

disease: the prevalence <strong>of</strong> nodular thyroid disease (as well as <strong>goiter</strong>) is inversely correlated<br />

with the population’s iodine intake (11;12). This has formerly been assessed clinically by<br />

palpation, nowadays considered highly inaccurate (13-15), but is also clearly documented by<br />

thyroid ultrasonography. Based on ultrasound investigation a frequency <strong>of</strong> thyroid nodular<br />

disease as high as 30-40% (women) <strong>and</strong> 20-30% (men) <strong>of</strong> the adult population has been<br />

reported in iodine deficient areas. Furthermore even minor differences in the ambient iodine<br />

supply may be reflected in the different prevalence <strong>of</strong> thyroid abnormalities: Knudsen et al.<br />

(16) found a difference in <strong>goiter</strong> prevalence (15% in mild <strong>and</strong> 22.6% in moderate deficiency)<br />

<strong>and</strong> nodule size (increased in the moderate iodine deficiency group). The prevalence <strong>of</strong><br />

thyroid nodules seems to increase with age (4;17;18). In a borderline iodine deficiency area<br />

MNG was present in 23% <strong>of</strong> the studied population <strong>of</strong> 2656 Danish people aged 41 to 71 yr.,<br />

<strong>and</strong> increased with age in women (20 to 46%) as well as men (7 to 23%) (3). In contrast, the<br />

relation between age <strong>and</strong> thyroid volume is less coherent, whereby in iodine deficient areas<br />

(except for severe deficiency), thyroid enlargement peaks around 40 years with no tendency<br />

for further increase (19). Interestingly, similar observations have been made in an iodine<br />

sufficient area. In the twenty year follow-up <strong>of</strong> the Whickham Survey the frequency <strong>of</strong> <strong>goiter</strong><br />

decreased with age (<strong>goiter</strong> prevalence initially: 23% women <strong>and</strong> 5% men; at 20 year follow-<br />

up <strong>of</strong> the same patients: 10% women <strong>and</strong> 2% men) (20).<br />

4


Thyroid nodules are found with higher frequency in enlarged thyroid gl<strong>and</strong>s though all<br />

clinicians will agree that they may also be present in an otherwise normal thyroid gl<strong>and</strong>.<br />

(4;18;21). The correlation between iodine supply <strong>and</strong> prevalence <strong>of</strong> nodular thyroid disease<br />

can similarly also applies to <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong>. The high frequency <strong>of</strong> thyroid<br />

autonomy, which accounts for up to 60% <strong>of</strong> cases <strong>of</strong> thyro<strong>toxic</strong>osis in iodine deficient areas is<br />

largely due to TMNG (~ 50%, solitary <strong>toxic</strong> nodules ~ 10%) (12;22). Prevalence <strong>of</strong> thyroid<br />

autonomy correlates with increased thyroid nodularity <strong>and</strong> increases with age (4;22). In<br />

contrast, thyroid autonomy is rare (3-10% <strong>of</strong> cases <strong>of</strong> thyro<strong>toxic</strong>osis) in regions with<br />

sufficient iodine supply (22;23;23). Correction <strong>of</strong> iodine deficiency in a population results in<br />

decrease <strong>of</strong> thyroid autonomy as demonstrated by the impressive 73% reduction in prevalence<br />

<strong>of</strong> TMNG only 15 yr. after the doubling <strong>of</strong> iodine content <strong>of</strong> salt in Switzerl<strong>and</strong> (12;24).<br />

While <strong>goiter</strong> <strong>and</strong> <strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> nodular thyroid disease share the common <strong>and</strong> important<br />

epidemiology <strong>of</strong> iodine deficiency (ID), it needs to be stressed that most epidemiological<br />

conclusions are derived from cross sectional studies.Thyroid nodules (<strong>and</strong> <strong>goiter</strong>) also occur<br />

in individuals without exposure to iodide deficiency <strong>and</strong> not all individuals in an iodine<br />

deficient region develop a <strong>goiter</strong> . Moreover there is a strong clustering <strong>of</strong> <strong>goiter</strong> in families<br />

(see chapter VIII).<br />

Screening has been performed for other “environmental factors” (19). Smoking has been<br />

proposed as a risk factor for <strong>goiter</strong> (25) <strong>and</strong> nodules were also found with higher prevalence<br />

in <strong>goiter</strong>s <strong>of</strong> smokers compared to non-smokers. The impact <strong>of</strong> smoking on thyroid disease<br />

could be due to i.e. increased thiocyanate levels in smokers exerting a competitive inhibitory<br />

effect on iodide uptake <strong>and</strong> organification (19;26). The association is more pronounced, again,<br />

in iodine deficiency (26). Radiation is another environmental risk factor not only for thyroid<br />

malignancy but also for benign nodular thyroid disease. An increased prevalence <strong>of</strong> nodular<br />

disease has been associated with exposure to radionuclear fallouts <strong>and</strong> therapeutic external<br />

radiation <strong>and</strong> is discussed by some authors also for occupational exposure to low-level<br />

5


adiation (27-31). Furthermore several studies suggest that thyroid volume is also<br />

significantly correlated with body weight <strong>and</strong> body mass index. In agreement with this a<br />

recent study has shown that in obese women weight loss <strong>of</strong> more than 10% may result in a<br />

significant decrease in thyroid volume (32).<br />

Nodular disease is more frequent (5-15 fold (33;34)) in women <strong>and</strong> yet the reasons for this are<br />

poorly understood. Thus at present one can only speculate as to a genetic susceptibility for<br />

thyroid disease (for details see chapter VIII) <strong>and</strong>/or a direct impact <strong>of</strong> steroid hormones.<br />

In fact, a growth promoting effect <strong>of</strong> estrogen has been described in vitro in rat FRTL-5 cells<br />

<strong>and</strong> thyroid cancer cell lines <strong>and</strong> has been proposed as a possible contributing, constitutional<br />

effect <strong>of</strong> gender (35;36). In addition, 17β- estradiol has been suggested to amplify growth<br />

factor induced signaling in normal thyroid <strong>and</strong> thyroid tumors (36). Interestingly, the use <strong>of</strong><br />

oral contraceptives which antagonise the physiological hormonal cycle has been reported to<br />

be associated with a decrease in <strong>goiter</strong> (but not nodules even though this may represent an age<br />

artefact <strong>of</strong> the studied population). On the other h<strong>and</strong> pregnancy related thyroid enlargement<br />

was clearly related to iodine deficiency (19) <strong>and</strong> in one German study increased MNG<br />

prevalence with parity was only observed in those women which had not taken iodine<br />

supplementation during earlier pregnancy (37).<br />

In summary the development <strong>of</strong> nodular disease is influenced by multiple environmental<br />

components in interaction with constitutional parameters <strong>of</strong> gender <strong>and</strong> age. However whether<br />

these factors actually result in <strong>goiter</strong> or nodular thyroid disease is a different matter ultimately<br />

decided on the genetic background <strong>of</strong> the individual patient, discussed below (see chapter<br />

VIII).<br />

II Clinical aspects <strong>of</strong> <strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong><br />

6


Clinical features in a patient with <strong>multinodular</strong> <strong>goiter</strong> can be attributed to thyroid enlargement<br />

<strong>and</strong> thyro<strong>toxic</strong>osis in case <strong>of</strong> TMNG. Thus, a patient may present with a lump or<br />

disfigurement <strong>of</strong> the neck, intolerance <strong>of</strong> tight necklaces or increase in collar size. Dysphagia<br />

or breathing difficulties due to local easophageal or tracheal compression may be apparent,<br />

especially with large <strong>goiter</strong> (33). Besides cosmetic aspects <strong>and</strong> compression signs, the daily<br />

challenge is to identify very rare thyroid malignancy in very frequent nodular thyroid disease<br />

<strong>and</strong> strategies to approach that goal have been reviewed in detail elsewhere (38-41).<br />

Alternatively, patients may present with symptoms suggestive <strong>of</strong> hyperthyroidism, the clinical<br />

presentation <strong>of</strong> which varies considerably with age. In a series <strong>of</strong> 84 French patients with<br />

overt hyperthyroidism, classical signs <strong>of</strong> thyro<strong>toxic</strong>osis e.g. nervousness, weight loss despite<br />

increased appetite, palpitations, tremor <strong>and</strong> heat intolerance were more frequently observed in<br />

younger patients (≤ 50 years) (42), while atrial fibrillation <strong>and</strong> anorexia dominated in the<br />

older age group (≥ 70 yr.). In addition, subclinical hyperthyroidism, defined by low or<br />

suppressed TSH with normal fT4 <strong>and</strong> fT3 levels is more commonly observed in older patients<br />

with TMNG (43). In fact the incidental finding <strong>of</strong> low or suppressed TSH levels on routine<br />

investigation in iodine deficient regions for other conditions is frequently a first indicator for<br />

presence <strong>of</strong> thyroid autonomy (4). Subclinical hyperthyroidism is more than “just” a low TSH<br />

status, since it is associated with increased prevalence <strong>of</strong> atrial fibrillation <strong>and</strong> bone density<br />

loss (43). In addition, an increased cardiovascular mortality rate in patients with low serum<br />

TSH levels has been described in a 10 yr. cohort-study in the UK (44). The management <strong>of</strong><br />

<strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> thyroid disease has recently been extensively reviewed by<br />

Hegedüs <strong>and</strong> coworkers (33).<br />

A Differential diagnosis<br />

7


Very rarely TMNG occurs as an autosomal dominantly inherited disease caused by activating<br />

germline mutations in the TSHR gene (45). A positive family history <strong>of</strong> recurrent<br />

hyperthyroidism <strong>and</strong> <strong>goiter</strong> with absence <strong>of</strong> typical diagnostic features <strong>of</strong> Graves’ disease,<br />

persistent neonatal thyro<strong>toxic</strong>osis <strong>and</strong> relapsing non-autoimmune thyro<strong>toxic</strong>osis in childhood<br />

are highly suggestive <strong>of</strong> the condition. So far more than 150 patients (10 families <strong>and</strong> 11<br />

children with sporadic occurrence <strong>of</strong> TSHR germline mutations) have been reported in the<br />

literature (http://www.uni-leipzig.de/innere/TSHR). Thyroid ablation is advocated as the first<br />

line treatment (surgery <strong>and</strong>/or radioiodine) to prevent relapses. <strong>Molecular</strong> analysis for<br />

germline TSHR mutations <strong>of</strong>fers the possibility for family screening, preclinical diagnosis<br />

<strong>and</strong> genetic counselling (7;46).<br />

In iodine deficient areas, the distinction between thyroid autonomy <strong>and</strong> Graves`disease (GD)<br />

can be complicated by absence <strong>of</strong> extrathyroidal signs <strong>of</strong> autoimmune thyroid disease <strong>and</strong><br />

“atypical” diagnostic findings. In this regard several possibilities may be encountered. Firstly,<br />

erroneous classification <strong>of</strong> Graves disease as TMNG due to presence <strong>of</strong> thyroid nodules<br />

observed in 10-15% <strong>of</strong> GD patients or a patchy scintiscan appearance compatible with TMNG<br />

(47;48). Secondly, failure to detect TSHR antibodies (TRAB) in Graves’ disease using less<br />

sensitive assays. This is illustrated by the detection <strong>of</strong> TRAB with highly sensitive 2 nd<br />

generation assays <strong>and</strong>/or bioassays in up to 56% <strong>of</strong> patients with scintiscan appearance <strong>of</strong><br />

TMNG <strong>and</strong> up to 22% <strong>of</strong> patients with diffuse uptake <strong>and</strong> absence <strong>of</strong> eye disease <strong>and</strong> negative<br />

TRAB results in older essays (hence erroneously classified as “diffuse thyroid autonomy”)<br />

(47-49). Thirdly, confusion <strong>of</strong> familial occurence <strong>of</strong> (autoimmune) hyperthyroidism with<br />

hereditary thyroid autonomy, which might clinically masquerade as Graves`disease. In this<br />

scenario, absence <strong>of</strong> TRAB is highly suggestive <strong>of</strong> familial non-autoimmune hyperthyroidism<br />

due to a constitutively activating TSHR germline mutation (46).<br />

8


III Natural course <strong>of</strong> <strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong><br />

A Nodule Growth<br />

From the epidemiological data discussed above one might expect an inherent progressive<br />

course <strong>of</strong> nodular thyroid disease. Studies aimed at accurate assessment <strong>of</strong> the nodules’ fate<br />

by ultrasonography differ in terms <strong>of</strong> follow-up period, definition <strong>of</strong> growth (increase in<br />

volume or nodule diameter), type <strong>of</strong> thyroid lesion (solid, cystic) <strong>and</strong> the background, in<br />

which they are conducted (e.g. environmental factors, specialised thyroid clinic). Moreover<br />

the inter-observer variability <strong>of</strong> long-term studies <strong>of</strong> nodule volumes is not known. With these<br />

caveats in mind the following observations have been reported: In iodine sufficient areas<br />

nodule “growth” has been reported in 35% <strong>of</strong> US patients over a follow-up period <strong>of</strong> 4.9 to<br />

5.6 yr (50). In another US study nodule growth (>15% increase in volume) was observed with<br />

similar frequency over a highly variable follow-up period (1 month to 5yr) (51). On long-term<br />

follow-up over 15 yr. in an area <strong>of</strong> iodine sufficiency only 1/3 <strong>of</strong> benign nodules showed<br />

growth as assessed by palpation <strong>and</strong> ultrasonography as opposed to the majority <strong>of</strong> nodules,<br />

which remained unchanged or even showed a decrease in size (52;53). In the German setting,<br />

for which the iodine deficit has been calculated at 30% <strong>of</strong> the recommended intake (54) a<br />

mean 3 yr. follow-up <strong>of</strong> 109 consecutive patients showed a steady <strong>and</strong> significant (> 30%<br />

volume) increase in nodular size in 50% <strong>of</strong> patients (55). In a Danish study only 4 (8%) <strong>of</strong> 45<br />

cold nodules in an area <strong>of</strong> borderline iodine deficiency showed a change in size (>5 mm in<br />

diameter) <strong>of</strong> which only 1 nodule actually increased whereas 3 nodules shrunk over a follow-<br />

up period <strong>of</strong> 2 yr. (table 1 (3)). The conclusion, which is suggested by these data is that both<br />

in an iodine deficient <strong>and</strong> sufficient setting a variable portion but most likely not all nodules<br />

will grow <strong>and</strong> the speed <strong>of</strong> growth is highly heterogeneous. Thus, identification <strong>of</strong> nodules<br />

with an increased growth potential is a challenge. This may also be relevant to the therapeutic<br />

management. In fact one could speculate that discrepant results reported in various treatment<br />

studies may actually reflect this heterogeneity <strong>of</strong> proliferation (<strong>and</strong>/or the potential to taper it<br />

9


down by treatment) rather than an evidence-based treatment effect <strong>of</strong> e.g. iodine vs. iodine<br />

plus l-thyroxine or l-thyroxine alone. Furthermore, results <strong>of</strong> currently available studies<br />

(3;52;53;55) do not allow conclusions as to whether nodule growth is associated with an<br />

increased risk <strong>of</strong> thyroid malignancy <strong>and</strong> thus the question arises as to the benefits <strong>of</strong> nodule<br />

volume reduction. In the authors’ opinion therapy, if efficient, is possibly better aimed at the<br />

primary prevention <strong>of</strong> the evolution <strong>of</strong> novel/further thyroid nodules in predisposed patients<br />

(56;57) with the long-term perspective to reduce ablative thyroid treatment for cosmetic<br />

reasons, compression symptoms <strong>and</strong> importantly thyroid malignancy. However, the pro<strong>of</strong> <strong>of</strong><br />

principle for any <strong>of</strong> these suggestions is (still) awaited.<br />

B Thyroid function<br />

Besides growth, transition from <strong>euthyroid</strong>ism to hyperthyroidism in a patient with<br />

<strong>multinodular</strong> thyroid disease is an even more relevant clinical issue. We know that<br />

hyperthyroidism in TMNG develops insidiously <strong>and</strong> that TMNG is usually preceeded by a<br />

long-st<strong>and</strong>ing <strong>euthyroid</strong> <strong>multinodular</strong> <strong>goiter</strong>. In fact autonomous areas have been described in<br />

up to 40% <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong>s in iodine deficient regions (58). The most accurate<br />

epidemiological data on evolution <strong>of</strong> hyperthyroidism have been published for solitary <strong>toxic</strong><br />

adenoma <strong>and</strong> most <strong>of</strong> these aspects can possibly also apply to MNG. The natural course is<br />

slow. An overall 4.1% annual incidence <strong>of</strong> thyro<strong>toxic</strong>osis was observed in a group <strong>of</strong> 375<br />

untreated <strong>euthyroid</strong> patients with <strong>toxic</strong> adenoma (TA) in Germany, who were followed for a<br />

mean period <strong>of</strong> 53 months (59). In two longitudinal studies an incidence <strong>of</strong> 9-10% <strong>of</strong> overt<br />

thyro<strong>toxic</strong>osis has been reported in patients with <strong>euthyroid</strong> MNG over a mean follow-up<br />

period <strong>of</strong> up to 12.2 years (60;61). There is a correlation between nodule size <strong>and</strong><br />

development <strong>of</strong> hyperthyroidism: In an American study 93.5% <strong>of</strong> patients with overt<br />

hyperthyroidism had TA > 3cm in size <strong>and</strong> patients with a <strong>euthyroid</strong> TA <strong>of</strong> > 3cm size carried<br />

10


a 20% risk <strong>of</strong> developing hyperthyroidism during a 6yr. follow-up period as opposed to a 2-<br />

5% risk <strong>of</strong> patients with nodules < 2.5cm in size (23). Similarly in areas with iodine<br />

deficiency an autonomous volume <strong>of</strong> 16 ml has been determined to be critical for clinical<br />

manifestation <strong>of</strong> hyperthyroidism (62). In TMNG the extent <strong>of</strong> thyroid nodularity (<strong>and</strong> hence<br />

the autonomous volume) is related to the prevalence <strong>of</strong> low or suppressed TSH levels <strong>and</strong><br />

both parameters are correlated with age (4). A sudden stimulation <strong>of</strong> thyroid function<br />

resulting in clinical manifestation as thyroid autonomy can be induced by the administration<br />

<strong>of</strong> excessive amounts <strong>of</strong> iodine e.g. in form <strong>of</strong> contrast media widely used for angiography<br />

<strong>and</strong> CT scans or by iodine containing drugs e.g. amiodarone (63). In the European Study<br />

Group <strong>of</strong> Hyperthyroidism a high proportion <strong>of</strong> iodine contamination was observed ranging<br />

from 18% in Graves disease to 54% in non-autoimmune hyperthyroidism (64). Severity <strong>of</strong><br />

iodine deficiency, autonomous thyroid cell mass, the quantity <strong>of</strong> administered iodine <strong>and</strong><br />

older age have been proposed as risk factors for the development <strong>of</strong> iodine induced<br />

hyperthyroidism (64).<br />

IV Clonal origin <strong>of</strong> thyroid nodules<br />

Studies that address the clonal expansion <strong>of</strong> a tumor have provided a valuable tool to decide<br />

the nature or etiology <strong>of</strong> a focal growth event being either neoplasia or hyperplasia. Whereas<br />

hyperplasia is a reversible outcome <strong>of</strong> an external trophic stimulus (e.g. iodine deficiency in<br />

the thyroid) neoplasia results from an intracellular defect (i.e. genetic alteration) <strong>and</strong> is<br />

irreversible (this definition <strong>of</strong> neoplasia in not equivalent with malignancy). For the thyroid<br />

gl<strong>and</strong> a critical review <strong>of</strong> the early work on clonal analysis was given by Thomas et al. (65).<br />

Later, heterozygous polymorphisms in X-chromosome-linked markers (66) have been<br />

extensively used to demonstrate a predominant clonal origin <strong>of</strong> tumor tissues including the<br />

thyroid gl<strong>and</strong> (67-71). Despite increasing technical concern (reviewed in (72)) clonal analysis<br />

is still a frequently used tool in tumor biology <strong>of</strong>ten seen as an intermediate step in pursuing<br />

11


the ultimate goal which is the detection <strong>of</strong> the molecular cause <strong>of</strong> neoplastic growth, namely<br />

mutations in the genomic DNA. Especially recent results <strong>of</strong> clonal analysis after PCR<br />

amplification <strong>of</strong> X-linked markers from micro-dissected tumors need to be interpreted with<br />

caution (73). The thyroid develops from a number <strong>of</strong> progenitor cells that migrate from the<br />

floor <strong>of</strong> the primitive pharynx called the median thyroid anlage (74). In females each<br />

progenitor shows a defined pattern <strong>of</strong> inactivation for most genes on one <strong>of</strong> the two X-<br />

chromosomes that is conferred to the progeny (75). Proliferation <strong>of</strong> these progenitors forms a<br />

cluster <strong>of</strong> cells (i.e. the thyroid patch) that share the same pattern <strong>of</strong> X-chromosome<br />

inactivation. If a sample for clonal analysis (e.g. from a micro-dissected tumor) lies entirely<br />

within such a patch/cluster an identical pattern <strong>of</strong> X-chromosome inactivation which implies<br />

monoclonality is not a reliable marker for neoplasia (72;76). Vice versa, without micro-<br />

dissection the distinction <strong>of</strong> monoclonal origin <strong>of</strong> samples from true neoplasia could be<br />

concealed by contamination with blood, connective, <strong>and</strong>, surrounding healthy tissue. In both<br />

cases a histochemical analysis <strong>of</strong> clonal origin that allows to examine the clonal architecture<br />

would be very helpful. However, available techniques can not be applied in general because<br />

the tissue under investigation has to meet several requirements (73;77). Nevertheless<br />

histology based clonal analysis (73;77) indicates a patch size in the thyroid gl<strong>and</strong> that is much<br />

smaller than patch sizes determined with PCR using paraffin-embedded tissue sections (78).<br />

Moreover, in line with the histology based data our own study using the PCR approach on<br />

microdissected thyroid follicles demonstrates a polyclonal origin in about 25% <strong>of</strong> single<br />

follicles (Krohn, unpublished). If a hyperplastic lesion is likely to arise from a single patch<br />

then clonal analysis with the current methodology would have a strong bias toward showing<br />

monoclonality for this lesion. It is therefore crucial to consider the conditions that would<br />

allow to assume that a hyperplastic nodule could arise from a single thyroid patch. This<br />

decision mainly depends on the extend <strong>of</strong> thyroid patch size <strong>and</strong> the growth potential <strong>of</strong> a<br />

hyperplastic lesion. If the thyroid patch size is small a higher growth potential would be<br />

12


necessary to allow a hyperplastic lesion to develop from a few follicles into a macroscopically<br />

detectable thyroid nodule. Because data that would allow to determine this growth potential in<br />

vivo are not directly available we instead like to consider data that show the extend <strong>of</strong> thyroid<br />

hyperplasia after goitrogenic stimulation in animal models. These date suggest a rather low<br />

growth potential because thyroid enlargement under extrinsic goitrogenic stimulation (e.g.<br />

iodine deficiency or extended TSH stimulation) is rarely higher than 3- to 5-fold (4;79-81). In<br />

contrast, intrinsic or intracellular growth stimulation caused by genetic manipulation in<br />

transgenic mice leads in some cases to increases <strong>of</strong> thyroid mass in the range <strong>of</strong> 100-fold<br />

(10;82;83). If this difference also applies to focal stimulation, it is very unlikely that a<br />

hyperplastic thyroid lesion (caused by extrinsic stimuli) that originates only from a single<br />

patch would reach the cell mass <strong>of</strong> a normal thyroid nodule. Therefore, it is more likely that a<br />

macroscopically detectable thyroid hyperplastic nodule originates from more than one patch.<br />

If so, this nodule should be detectable as polyclonal, if a large part <strong>of</strong> the respective tissue is<br />

studied for X-chromosome inactivation. Studies <strong>of</strong> clonal analysis in our group therefore used<br />

DNA extracted from the entire nodular tissue. This approach very likely reduces the a strong<br />

bias toward showing monoclonality for a hyperplastic lesion.<br />

Our investigations <strong>of</strong> the clonal origin <strong>of</strong> autonomously functioning thyroid nodules <strong>and</strong><br />

solitary cold thyroid nodules (both adenomas <strong>and</strong> adenomatous nodules) used a PCR<br />

approach to amplify the X-linked human <strong>and</strong>rogen receptor from genomic DNA <strong>of</strong> female<br />

patients (84-86). After thorough screening for somatic mutations in these thyroid nodules (for<br />

details see Chapter V) we could demonstrate that thyroid nodules with a somatic mutation are<br />

predominantly <strong>of</strong> clonal origin (84-86). This is not surprising because it is in full agreement<br />

with the widely accepted paradigm in tumor biology that neoplasia (for definition see above)<br />

originates from a single mutated cell (87). Moreover, we were specially interested in data that<br />

would elucidate the etiology <strong>of</strong> mutation negative nodules. Interestingly, more than 50% <strong>of</strong><br />

mutation negative cases from female patients show a monoclonal origin when tested for X-<br />

13


chromosome inactivation (84-86). This could indicate a neoplastic process with a mutation in<br />

a gene other than the TSHR, the Gsα protein or the ras family <strong>of</strong> oncogenes. Moreover, our<br />

finding <strong>of</strong> an overall frequency for the monoclonal origin <strong>of</strong> thyroid nodules at about 60-70%<br />

agrees with a number <strong>of</strong> other studies (67;68;70;71) <strong>and</strong> further underscores that thyroid<br />

nodules predominantly result from a neoplastic process with somatic mutations as the starting<br />

point (8).<br />

V Hot thyroid nodules<br />

A Signal transduction <strong>of</strong> HTN with <strong>and</strong> without TSHR mutations<br />

Both, growth <strong>and</strong> function <strong>of</strong> the thyroid are controlled by TSH (5). Although the activation<br />

<strong>of</strong> the TSHR preferentially leads to stimulation <strong>of</strong> the adenylyl cyclase via the Gsα-protein, at<br />

higher TSH concentrations an activation <strong>of</strong> the phospholipase C cascade by Gqα has also been<br />

shown (88;89). Moreover, there is evidence that the TSHR may be coupled to other members<br />

<strong>of</strong> the G protein family (88;90). However experimental data are frequently focused on the<br />

cAMP-branch <strong>of</strong> TSH signaling. Early work by Pisarev et al. demonstrated that cAMP<br />

elevation causes <strong>goiter</strong> (91). Moreover, in the thyroid gl<strong>and</strong> <strong>and</strong> cultured thyroid epithelial<br />

cells as well as other endocrine tissues it is widely accepted that cAMP stimulates<br />

proliferation (92-95). More recently, transgenic models were studied to further underst<strong>and</strong><br />

TSHR signaling in more detail: Chronic in vivo stimulation <strong>of</strong> the cAMP cascade stimulates<br />

epithelial cell proliferation in vivo (82;96;97). A dominant negative cAMP response element<br />

binding protein blocks signalling downstream <strong>of</strong> cAMP <strong>and</strong> causes severe growth retardation<br />

<strong>and</strong> primary hypothyroidism (98). TSH/TSHR signaling generally controls iodine metabolism<br />

but only affects growth in the adult thyroid gl<strong>and</strong> <strong>and</strong> not during embryonic development<br />

(99;100).<br />

Somatic point mutations that constitutively activate the TSHR were first identified by Parma<br />

<strong>and</strong> co-workers in hyperfunctioning thyroid adenomas (101). However, in different studies the<br />

14


prevalence <strong>of</strong> TSHR <strong>and</strong> Gsα mutations in autonomously functioning thyroid nodules has<br />

been reported to vary from 8 to 82% <strong>and</strong> 8 to 75%, respectively (101-112). These studies<br />

differ in the extent <strong>of</strong> mutation detection <strong>and</strong> the screening methods. A comparison with<br />

respect to the obvious differences between the studies has been done elsewhere (8;113;114).<br />

A comprehensive study <strong>of</strong> our group using the more sensitive denaturing gradient gel<br />

electrophoresis (115-117), revealed a frequency <strong>of</strong> 57% TSHR mutations <strong>and</strong> 3% Gsα<br />

mutations in 75 consecutive autonomously functioning thyroid nodules (86). These results<br />

raise the question <strong>of</strong> the molecular etiology <strong>of</strong> TSHR <strong>and</strong> Gsα mutation negative nodules. A<br />

possible answer is given by clonal analysis <strong>of</strong> these AFTNs which demonstrates a<br />

predominant clonal origin <strong>of</strong> thyroid nodules <strong>and</strong> implies a neoplastic process driven by<br />

genetic alteration (for details see IV). In a recent study (118) we found that AFTNs without a<br />

TSHR mutation show an increased expression <strong>of</strong> the tumor suppressor protein p53-binding<br />

protein 2, which interacts with p53 <strong>and</strong> specifically enhances p53-induced apoptosis but not<br />

cell cycle arrest (119). From this finding one could speculate that increased expression <strong>of</strong> this<br />

gene could increase apoptosis in AFTNs without a TSHR mutation <strong>and</strong> thus have a negative<br />

effect on the growth <strong>of</strong> the tumor. However, data on apoptosis in AFTNs do not allow a<br />

comparison with respect to the TSHR mutation status (120;121). Furthermore, the AFTNs<br />

without a TSHR mutation differ from the nodules harboring a TSHR mutation in their<br />

increased expression <strong>of</strong> two genes which are involved in the signal transduction <strong>of</strong> G protein<br />

coupled receptors: RGS 6 <strong>and</strong> GRK 2. Members <strong>of</strong> the RGS family have been shown to<br />

modulate the function <strong>of</strong> G proteins by activating the intrinsic GTPase activity <strong>of</strong> the alpha<br />

subunits (122), whereas G protein coupled receptor kinases play a role in the receptor<br />

desensitization (123). In general, a higher expression <strong>of</strong> these genes would rather restrict<br />

cAMP accumulation in AFTNs <strong>and</strong> could have an negative effect on functional autonomy.<br />

However, further experiments have to explore the importance <strong>of</strong> these genes in the etiology <strong>of</strong><br />

AFTNs without TSHR mutations.<br />

15


AFTNs with TSHR mutations lack a clear genotype/ phenotype correlation (113). A similar<br />

finding is evident for germline TSH receptor mutations (124). Variable phenotypes associated<br />

with the same TSHR mutation could be the result <strong>of</strong> influences on signaling downstream <strong>of</strong><br />

the TSHR. This modulation could have a number <strong>of</strong> targets like G protein coupling, receptor<br />

desensitization <strong>and</strong> internalization or cross-talk with other signaling cascades. Although our<br />

knowledge concerning these targets is far from complete recent findings are very promising.<br />

Firstly, the TSHR itself could be the subject <strong>of</strong> regulatory mechanisms that contribute to the<br />

etiology <strong>of</strong> AFTNs <strong>and</strong> the clinical phenotype. Voigt et al. (125) could show that ß-arrestins<br />

interact with the TSH receptor <strong>and</strong> are able to desensitize the receptor. Increased expression<br />

<strong>of</strong> beta-arrestin 2 in AFTNs could cause desensitization <strong>of</strong> the TSHR <strong>and</strong> thereby down<br />

regulate constitutive activation. A similar result could be caused by increased TSHR<br />

internalization due to increased expression <strong>of</strong> GRKs in AFTNs (118;126). Besides direct<br />

effects on the TSHR protein altered interaction with G proteins would be the next downstream<br />

level where modulation interferes with constitutive activation. For example the mutated<br />

TSHR could show a shift <strong>of</strong> the coupling specificity for G proteins (127). Such a shift could<br />

allow a more efficient activation <strong>of</strong> other downstream cascades (e.g. JAK/STAT pathway)<br />

through PKC in addition to cAMP <strong>and</strong> IP (128;129). Gene expression analysis in AFTNs<br />

with TSHR mutations in comparison to AFTNs without a TSHR mutation would support this<br />

hypothesis demonstrating an increased expression <strong>of</strong> JAK 1, protein kinase C beta 1 <strong>and</strong> zeta<br />

mRNA (118). Evidence for other cascades (e.g. RAS/RAF/MEK/ERK/MAP pathway) to play<br />

a role in constitutive TSHR signaling in AFTNs is currently missing.<br />

In addition to the intracellular signaling network that is connected to the TSHR, the<br />

extracellular action <strong>of</strong> different growth factors enhances the complexity <strong>of</strong> the signal flux into<br />

the thyroid cell. Growth factors like insulin-like growth factor I, epidermal growth factor,<br />

transforming growth factor β <strong>and</strong> fibroblast growth factor stimulate growth <strong>and</strong><br />

dedifferentiation <strong>of</strong> thyroid epithelial cells (130;131). Studies, which have been focused on<br />

16


insulin <strong>and</strong> insulin-like growth factor, show a permissive effect <strong>of</strong> insulin <strong>and</strong> IGF-I on TSH<br />

signaling (132-136) <strong>and</strong> a cooperative interaction <strong>of</strong> TSH <strong>and</strong> insulin/IGF-I (137). Signal<br />

modulation <strong>of</strong> the TSHR that would define the etiology <strong>of</strong> AFTNs <strong>and</strong> the clinical phenotype<br />

could therefore take part at a number <strong>of</strong> stages <strong>and</strong> very likely involves genetic/epigenetic,<br />

sex-related, <strong>and</strong> or environmental factors.<br />

B Secondary/indirect effects <strong>of</strong> activating TSHR mutations<br />

Because constitutively activating TSHR mutations disturb the coordinated signal transduction<br />

network <strong>of</strong> the thyroid in a drastic way, subsequent changes in the signal transduction network<br />

can be expected. These alterations based on the constitutive activation <strong>of</strong> the TSHR signaling<br />

can be described as indirect or secondary effects <strong>of</strong> the activating TSHR mutations. The use<br />

<strong>of</strong> the microarray technique <strong>of</strong>fers the advantage <strong>of</strong> a highly parallel analysis <strong>of</strong> gene<br />

expression to analyze changes between AFTNs <strong>and</strong> CTNs compared with their surrounding<br />

tissue. This approach also allows to evaluate which genes <strong>and</strong> groups <strong>of</strong> genes are most<br />

frequently affected in the molecular etiology <strong>of</strong> thyroid nodules <strong>and</strong> possibly deduce a<br />

molecular defect from the expression pattern. In a recent study using the Affymetrix<br />

GeneChip technology we could show a distinctly changed pattern <strong>of</strong> gene expression <strong>of</strong> the<br />

TGF-β signaling pathway between AFTNs with <strong>and</strong> without TSHR mutations <strong>and</strong> their<br />

normal surrounding tissue (figure 1 (118)). The type III TGF-β receptor, Smad 1, 3 <strong>and</strong> 4, as<br />

well as p300, a transcriptional co-activator, showed a decreased expression in AFTNs,<br />

whereas the inhibitory Smads 6 <strong>and</strong> 7 showed an increased expression in AFTNs. These<br />

findings suggest inactivation <strong>of</strong> TGF-ß signaling in AFTNs due constitutively activated<br />

TSHR (e.g. resulting from TSHR mutations). This assumption is supported by findings <strong>of</strong><br />

Gärtner et al. (138), who could show a decreased expression <strong>of</strong> TGF-β 1 mRNA after TSH<br />

stimulation <strong>of</strong> thyrocytes. Because TGF-β 1 has been shown to inhibit iodine uptake, iodine<br />

organification <strong>and</strong> thyroglobulin expression (139;140), as well as cell proliferation in different<br />

17


cell culture systems (141-144), these <strong>and</strong> our novel findings suggest that inactivation <strong>of</strong> TGF-<br />

β signaling is a major prerequisite for increased proliferation in AFTNs (120;145).<br />

Eggo et al. (146) have shown that enhanced production <strong>of</strong> insulin-like growth factor binding<br />

proteins (IGFBPs) is correlated with inhibition <strong>of</strong> thyroid function, whereas the TSH-cAMP<br />

signaling is capable to inhibit IGFBP production. Moreover, recent studies (118;147) reveal a<br />

significantly decreased expression <strong>of</strong> insulin-like growth factor (IGF)-II <strong>and</strong> IGFBP 5 <strong>and</strong> 6 in<br />

AFTNs in comparison to their normal surrounding tissue. Taken together, the decreased<br />

expression <strong>of</strong> the IGFBPs, <strong>and</strong> <strong>of</strong> IGF-II in AFTNs are most likely secondary effects <strong>of</strong> the<br />

increased TSHR-cAMP signaling in AFTNs.<br />

VI Cold thyroid nodules<br />

With a frequency <strong>of</strong> about 85% cold thyroid nodules (CTNs) constitute the most abundant<br />

thyroid nodular lesion (for detailed definition <strong>and</strong> epidemiology see chapter I). The term<br />

“cold” indicates that this thyroid lesion shows reduced uptake on scintiscan. Because<br />

histologic diagnosis is typically employed to exclude thyroid cancer many investigations <strong>of</strong><br />

thyroid nodules only refer to the histologic diagnosis <strong>of</strong> thyroid adenoma. This histologic<br />

entity should not be confounded with the scintigraphically characterized entity “cold nodule”,<br />

which like AFTNs or “warm nodules” (for the distinction see chapter I) can histologically<br />

appear as thyroid adenomas or adenomatous nodules according to the WHO classification (1).<br />

In this review we will focus on benign neoplastic lesions because substantial information<br />

concerning genetic events <strong>and</strong> molecular mechanism is available in the literature on human<br />

cancer in particular thyroid carcinoma that very likely also applies to benign neoplasia <strong>of</strong><br />

thyroid follicular cells. In contrast, focal hyperplasia is not very well explained on the<br />

molecular level <strong>and</strong> has been discussed in detail elsewhere as the cause <strong>of</strong> thyroid tumors<br />

(148;149). As detailed in chapter IV, a monoclonal origin has been detected for the majority<br />

<strong>of</strong> thyroid nodules which implies nodular development from a single mutated thyroid cell.<br />

18


Hypotheses in studies that aim to underst<strong>and</strong> the molecular or genetic causes <strong>of</strong> human<br />

cancer in general (150;151) or AFTN (8) <strong>and</strong> thyroid carcinomas (152) in detail have <strong>of</strong>ten<br />

also been applied to studies <strong>of</strong> cold thyroid nodules (e.g. ras mutations (153;154), for review<br />

see Wynford-Thomas (155)). In contrast to thyroid carcinomas where a number <strong>of</strong> genes have<br />

been implicated in the <strong>pathogenesis</strong> <strong>of</strong> these lesions (152;156) <strong>and</strong> in contrast to AFTN where<br />

constitutively activating TSHR mutations are very prevalent genetic events (7;8) knowledge<br />

concerning the molecular etiology <strong>of</strong> CTNs is limited.<br />

A Iodide transport <strong>and</strong> metabolism<br />

With reference to their functional status (i.e. reduced iodine uptake) failure in the iodide<br />

transport system or failure <strong>of</strong> the organic binding <strong>of</strong> iodide have been detected as functional<br />

aberrations <strong>of</strong> cold thyroid nodules long before the molecular components <strong>of</strong> the iodine<br />

metabolism were known (for review see Paschke <strong>and</strong> Neumann (157)). Later, a decreased<br />

expression <strong>of</strong> the Na + /I - symporter (NIS) in thyroid carcinoma <strong>and</strong> benign cold thyroid<br />

nodules suggested the molecular mechanism for the failure <strong>of</strong> the iodide transport (reviewed<br />

in (158-160)). Although the extent <strong>of</strong> decrease in NIS mRNA expression <strong>of</strong> cold thyroid<br />

nodules varies in different studies (160-163) reduction is in many cases very likely the result<br />

<strong>of</strong> hypermethylation in the NIS promoter (160). Moreover, in vitro studies suggest that<br />

reduced NIS mRNA expression could be caused by constitutive activation <strong>of</strong> RET or RAS<br />

genes (164-166). However, reduced NIS mRNA expression does not necessarily lead to<br />

reduced NIS protein expression (figure 2) (160). Furthermore <strong>and</strong> in contrast to other thyroid<br />

disorders with congenital iodide transport defects (for review, see (167;168)) no NIS gene<br />

mutation that would render this protein non-functional was detectable in CTNs (160).<br />

Therefore, the recently identified defective cell membrane targeting <strong>of</strong> the NIS protein is a<br />

more likely molecular mechanism that could account for the failure <strong>of</strong> the iodine uptake in<br />

CTNs (158;160;169). However the ultimate cause <strong>of</strong> this defect is currently unknown.<br />

19


Compared to iodine transport the organic binding <strong>of</strong> iodine is a multistep process with a<br />

number <strong>of</strong> protein components that still awaits final characterization (170). mRNA expression<br />

<strong>of</strong> enzymatic components (e.g thyroid peroxidase (TPO) or flavoproteins) <strong>and</strong> the substrate <strong>of</strong><br />

iodination (i.e. thyroglobulin (TG)) have been quantified in CTNs without significant<br />

differences to normal follicular tissue (163;171). TPO, TG <strong>and</strong> thyroid specific oxidases<br />

(THOX) have been successfully screened for molecular defects especially in congenital<br />

hypothyroidism (172). Although, cold thyroid nodules could be considered as a form <strong>of</strong> focal<br />

hypothyroidism, somatic mutations in enzymes that catalyze organic binding <strong>of</strong> iodine would<br />

need to exert a growth advantage on the affected cell to cause the development <strong>of</strong> a thyroid<br />

nodule. At least in the case <strong>of</strong> inactivating mutations in the TPO or THOX genes growth<br />

advantage could result from a lack <strong>of</strong> enzyme activity which would not only reduce thyroid<br />

hormone synthesis but also follicular iodide trapping in organic iodocompounds. Because<br />

these compounds have been shown to inhibit thyroid epithelial cell proliferation<br />

(133;173;174) reduced synthesis could have a proliferative effect. Therefore, somatic TPO or<br />

THOX mutations could be a molecular cause <strong>of</strong> CTN. However mutations in the TPO gene<br />

could not be detected (175) <strong>and</strong> an ongoing screening for mutations in the THOX genes is<br />

also negative so far (Krohn, Paschke, Ris-Stalpers, unpublished).<br />

B Signaling proteins<br />

In addition to a failure in metabolic proteins that might explain the development <strong>of</strong> CTNs on<br />

the molecular level pathologic changes <strong>of</strong> signaling molecules might reprogram the growth<br />

stimulus <strong>and</strong> lead to clonal expansion <strong>of</strong> thyroid epithelial cells. Although not a particular<br />

subject <strong>of</strong> this review, much can be learned from findings in thyroid carcinoma. In this regard<br />

genetic changes (i.e. point mutations) that cause constitutive activation <strong>of</strong> the<br />

RAS/RAF/MEK/ERK/MAP pathway have been suggested as a key mechanism during tumor<br />

initiation or progression in thyroid follicular cells (for review, see (155)). So far, the only<br />

known molecular event that evidently causes such an activation in thyroid carcinomas <strong>and</strong><br />

20


cold thyroid nodules is a mutation in one <strong>of</strong> the small RAS oncogenes (153). Recently BRAF<br />

mutations first detected in melanomas <strong>and</strong> with lower frequency in other cancers (176) have<br />

been detected in thyroid papillary carcinomas (177). They can also activate this pathway <strong>and</strong><br />

might therefore also cause benign follicular lesions. Strikingly, both in colorectal <strong>and</strong> thyroid<br />

cancers BRAF mutations occur only in tumors that do not carry mutations in a RAS gene. In a<br />

recent study <strong>of</strong> 40 cold thyroid adenoma <strong>and</strong> adenomatous nodules we detected ras mutations<br />

in only a single case (85). Moreover, in the same set <strong>of</strong> CTNs we did not detect point<br />

mutations in the mutational hot spots <strong>of</strong> the BRAF gene (178). This is in line with the lack <strong>of</strong><br />

BRAF mutations in benign follicular adenoma in other studies (177;179;180). So far only one<br />

study detected a single BRAF mutation in a set <strong>of</strong> 51 follicular adenoma (181). Instead <strong>of</strong><br />

RAS <strong>and</strong> BRAF mutations there could be other molecular events that could constitutively<br />

activate the RAS/RAF/MEK/ERK/MAP pathway. Such c<strong>and</strong>idate molecules include other<br />

members <strong>of</strong> the RAF gene family like RAF-1 or downstream genes like ERK or MAP<br />

kinases. However mutations in these genes have not been reported in benign thyroid lesion so<br />

far. Furthermore, molecular events that lead to activation <strong>of</strong> other cascades that exert synergy<br />

with MAP kinase signaling (e.g. cAMP signaling) or inactivation <strong>of</strong> independent cascades<br />

that restrict proliferation (e.g. TGF-ß signaling) could explain cold thyroid nodules. In<br />

addition to the TSHR, several G protein is<strong>of</strong>orms like Gi2alpha (182) or Gq, <strong>and</strong> G11 (183) as<br />

well as some c<strong>and</strong>idate genes mediating downstream cAMP signaling like Epac <strong>and</strong> Rap1<br />

(184) have been screened in cold thyroid nodules for mutations. However, only a single<br />

somatic mutation in the Gi2alpha gene (182) was found in follicular adenomas.<br />

C Results <strong>of</strong> gene expression studies by arrays<br />

Currently, expression pr<strong>of</strong>iling <strong>of</strong> signaling proteins using microarray methodology is a<br />

promising approach which may contribute to further underst<strong>and</strong>ing the molecular events that<br />

lead to the development <strong>of</strong> AFTNs (147;185) or thyroid carcinoma (186-188). Functional<br />

21


characteristics <strong>of</strong> cold thyroid nodules suggest that mechanisms initiating growth but not<br />

leading to hyperfunction need to be defined. As far as future screenings for genetic defects are<br />

concerned expression pr<strong>of</strong>iling could describe the molecular mechanism <strong>and</strong> rule out a<br />

number <strong>of</strong> possible targets (e.g. because they are not expressed) or unmask alternative<br />

c<strong>and</strong>idates. Moreover, as demonstrated for AFTNs, results <strong>of</strong> expression pr<strong>of</strong>iling might shift<br />

attention to other signaling cascades (for details on AFTNs see chapter V). For differentially<br />

expressed genes within these cascades a sequencing approach might then be warranted. In<br />

addition, knowledge <strong>of</strong> the molecular signature <strong>of</strong> CTNs <strong>and</strong> benign thyroid tumors in general<br />

could be very helpful to define differences between benign <strong>and</strong> malignant thyroid disease with<br />

diagnostic or therapeutic relevance.<br />

Recently, our group investigated 588 genes by cDNA expression arrays in three AFTNs <strong>and</strong><br />

three CTNs as well as corresponding normal surrounding tissue. In general, changes in the<br />

expression <strong>of</strong> several signal transducing components were detected. Although this seems to<br />

reflect a disturbed signaling system, the results <strong>of</strong> that limited study did not allow to identify<br />

specific signal transduction cascades which might be involved in nodular development (147).<br />

To gain a higher resolution we compared gene expression for approximately 10,000 full-<br />

length genes between CTNs <strong>and</strong> their corresponding normal surrounding tissue (Eszlinger,<br />

Krohn <strong>and</strong> Paschke, submitted). Here regulation <strong>of</strong> gene expression in CTNs was most<br />

consistent for a group <strong>of</strong> several histone mRNAs. Increased expression <strong>of</strong> these histone<br />

mRNAs <strong>and</strong> <strong>of</strong> cell cycle associated genes like cyclin D1, cyclin H/cyclin dependent kinase<br />

(CDK) 7 <strong>and</strong> cyclin B most likely reflect a molecular setup for an increased proliferation in<br />

CTNs (189). In line with the low prevalence <strong>of</strong> ras mutations in CTNs (85), we find a reduced<br />

expression <strong>of</strong> ras-MAPK cascade associated genes which might suggests a minor importance<br />

<strong>of</strong> this signaling cascade.<br />

22


D Chromosomal aberrations<br />

Loss <strong>of</strong> heterozygocity (LOH), microsatellite instability <strong>and</strong> more recently gene<br />

rearrangements <strong>and</strong> chromosomal translocations as different forms <strong>of</strong> chromosomal aberration<br />

are considered important steps in carcinogenesis <strong>and</strong> have been investigated as potential<br />

markers to discern benign from malignant nodular disease. Findings <strong>of</strong> chromosomal<br />

aberrations <strong>and</strong> microsatellite instability in benign thyroid tumors although sometimes<br />

sporadic suggest that there is a difference in the extent <strong>of</strong> these DNA changes (190;191).<br />

Alternatively these results could stem from errors in histologic characterisation (192).<br />

Especially gene rearrangements unique to thyroid adenomas have recently been the focus<br />

(reviewed in (193)). These studies led to the identification <strong>of</strong> the thyroid adenoma associated<br />

gene (THADA) that encodes a death receptor interacting protein (194).<br />

Although also reported for thyroid follicular carcinoma (195) our finding <strong>of</strong> (LOH) at the<br />

TPO locus is characteristic for some CTNs (about 15%) but rather points to defects in a gene<br />

near TPO on the short arm <strong>of</strong> chromosome 2 (175). Moreover, after identification in a<br />

significant portion <strong>of</strong> follicular carcinomas (196) Pax-8/PPARγ gene rearrangement have also<br />

been reported for cold thyroid nodules (197) but seem to be a rare finding (198-200) or due to<br />

histologic misclassification <strong>of</strong> the thyroid nodules (192). Although the frequency <strong>of</strong> each <strong>of</strong><br />

these DNA aberrations is rather low together these chromosomal changes need to be<br />

considered in the further elucidation <strong>of</strong> the molecular etiology <strong>of</strong> CTNs.<br />

VII Multinodular Goiter<br />

Multinodular <strong>goiter</strong> refers to an enlargement <strong>of</strong> the thyroid with deformation <strong>of</strong> the normal<br />

parenchymal structure by the presence <strong>of</strong> nodules. These nodules vary considerably in size,<br />

morphology <strong>and</strong> function (for detailed definition <strong>and</strong> epidemiology see chapter I). In areas<br />

without endemic <strong>goiter</strong> MNG is <strong>of</strong>ten referred to as sporadic non-<strong>toxic</strong> <strong>goiter</strong>. MNG usually<br />

develops in an already enlarged thyroid independent <strong>of</strong> the cause <strong>of</strong> hyperplasia (for review<br />

23


see (149). Over time (sometimes decades) many <strong>euthyroid</strong> <strong>multinodular</strong> <strong>goiter</strong>s enlarge<br />

further, some develop subclinical hyperthyroidism <strong>and</strong> subsequently present as TMNG<br />

(4;201). The main epidemiologic determinants outlined in detail in chapter I for the<br />

development <strong>of</strong> MNG <strong>and</strong> TMNG are iodine deficiency (22), age, sex <strong>and</strong> duration <strong>of</strong> <strong>goiter</strong><br />

in iodine deficient (4;17;18) <strong>and</strong> also in iodine sufficient areas (for review, see (202)). It is<br />

widely accepted that the basis for the development <strong>of</strong> nodular structures is an early stimulus<br />

that causes enlargement <strong>of</strong> the thyroid. However, clinical manifestations <strong>of</strong> MNG might only<br />

appear after a long period <strong>of</strong> time (sometimes up to 30 <strong>and</strong> more years). In general<br />

development <strong>of</strong> MNG proceeds in two phases: global activation <strong>of</strong> thyroid epithelial cell<br />

proliferation (e.g., as the result <strong>of</strong> iodine deficiency or other goitrogenic stimuli) leading to<br />

<strong>goiter</strong> <strong>and</strong> a focal increase <strong>of</strong> thyroid epithelial cell proliferation causing thyroid nodules. So<br />

far, the most common stimulus for local proliferation are somatic mutations (see chapter V<br />

<strong>and</strong> VI).<br />

A Mutagenesis as the cause <strong>of</strong> nodular transformation<br />

From animal models <strong>of</strong> hyperplasia caused by iodine depletion (79;203;204) we learn that<br />

besides an increase in functional activity a tremendous increase in thyroid cell number occurs.<br />

These two events very likely orchestrate a burst <strong>of</strong> mutation events. Although the enzymatic<br />

setup awaits further characterization (171) it is known that thyroid hormone synthesis goes<br />

along with increased H2O2 production <strong>and</strong> free radical formation (205), which may damage<br />

genomic DNA <strong>and</strong> cause mutations (206). As a consequence, the spontaneous mutation rate in<br />

the thyroid is almost 10-times higher than in other organs (e.g. compared to liver, Krohn<br />

unpublished). Together with a higher spontaneous mutation rate a higher replication rate will<br />

more <strong>of</strong>ten prevent mutation repair <strong>and</strong> increase the mutagenic load <strong>of</strong> the thyroid, thereby<br />

also r<strong>and</strong>omly affecting genes crucial for thyrocyte physiology. Mutations, that confer a<br />

growth advantage (e.g. TSHR or Gsα protein mutations) very likely initiate focal growth.<br />

Hence autonomously functioning thyroid nodules are likely to develop from small cell clones,<br />

24


that contain advantageous mutations as shown for the TSHR in ‘hot’ microscopic regions <strong>of</strong><br />

<strong>euthyroid</strong> <strong>goiter</strong>s (9).<br />

B Etiology<br />

Epidemiologic studies, animal models <strong>and</strong> molecular/genetic data outline a general theory <strong>of</strong><br />

nodular transformation. Based on the identification <strong>of</strong> somatic mutations <strong>and</strong> the predominant<br />

clonal origin <strong>of</strong> AFTNs we propose the following sequence <strong>of</strong> events that could lead to<br />

thyroid nodular transformation in three steps (figure 3). In the first step, iodine deficiency,<br />

nutritional goitrogens or autoimmunity cause diffuse thyroid hyperplasia. Then, at this stage<br />

<strong>of</strong> thyroid hyperplasia increased proliferation together with a possible DNA damage due to<br />

H2O2 action causes a higher mutational load with a higher number <strong>of</strong> cells bearing a mutation.<br />

Some <strong>of</strong> these spontaneous mutations confer constitutive activation <strong>of</strong> the cAMP cascade (e.g.<br />

TSH-R <strong>and</strong> Gsα mutations) that stimulate growth <strong>and</strong> function. Finally, in a proliferating<br />

thyroid growth factor expression (e.g. IGF-I, TGF-ß1 or EGF) is increased. As a result <strong>of</strong><br />

growth factor co-stimulation all cells divide <strong>and</strong> form small clones. After increased growth<br />

factor expression ceases small clones with activating mutations will further proliferate if they<br />

can achieve self-stimulation. They could thus form small foci, which will develop into thyroid<br />

nodules. This mechanism could explain AFTNs by advantageous mutations that both initiate<br />

growth <strong>and</strong> function <strong>of</strong> the affected thyroid cells as well as CTNs by mutations that stimulate<br />

proliferation only (e.g. ras mutations or other mutations in the RAS/RAF/MEK/ERK/MAP<br />

cascade). Moreover, nodular transformation <strong>of</strong> thyroid tissue due to TSH secreting pituitary<br />

adenomas (207), nodular transformation <strong>of</strong> thyroid tissue in Graves’ disease (208) <strong>and</strong> in<br />

<strong>goiter</strong>s <strong>of</strong> patients with acromegaly (209) could follow a similar mechanism because thyroid<br />

pathology in these patients is characterized by early thyroid hyperplasia.<br />

As an alternative to the increase <strong>of</strong> cell mass <strong>and</strong> as illustrated by those individuals who do<br />

not develop a <strong>goiter</strong> when exposed to iodine deficiency the thyroid might also adapt to iodine<br />

deficiency without extended hyperplasia (210). Although the mechanism that allows this<br />

25


adaptation is poorly understood preliminary data from a mouse model suggest an increase <strong>of</strong><br />

mRNA expression <strong>of</strong> TSH-R, NIS <strong>and</strong> TPO in response to iodine deficiency which might be a<br />

sign for increased iodine turnover in the thyroid cell in iodine deficiency (Krohn<br />

unpublished).<br />

VIII Pathogenesis <strong>and</strong> genetic etiology <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong><br />

Considering recent advances in the methodology <strong>of</strong> genetic analysis the genetic etiology <strong>of</strong><br />

<strong>goiter</strong> is under-investigated. This applies especially to studies that target the genetic basis for<br />

<strong>euthyroid</strong> <strong>and</strong> <strong>toxic</strong> MNG. As outlined in the preceding chapter the development <strong>of</strong> nodular<br />

<strong>goiter</strong> is very likely a continuous process that starts with thyroid hyperplasia <strong>and</strong> simple<br />

<strong>goiter</strong>. Therefore, defects in genes that play an important role in thyroid physiology <strong>and</strong><br />

hormone synthesis (see chapter ‘C<strong>and</strong>idate loci’) could be genetic factors that predispose for<br />

the mechanisms that lead to <strong>multinodular</strong> <strong>goiter</strong>. Such defects likely lead to<br />

dyshormonogenesis as an immediate response <strong>and</strong> might not directly explain nodular<br />

transformation <strong>of</strong> the thyroid. In this chapter we therefore also consider genetic studies that<br />

concern other forms <strong>of</strong> <strong>goiter</strong>.<br />

A Family <strong>and</strong> twin studies<br />

Although lack <strong>of</strong> iodine is the most prevalent factor for simple <strong>goiter</strong> as well as endemic<br />

<strong>goiter</strong>, other causes are likely. Familial clustering <strong>of</strong> <strong>goiter</strong>s <strong>and</strong> the female predominance <strong>of</strong><br />

<strong>goiter</strong>s are the two major arguments suggesting a genetic background for <strong>euthyroid</strong> <strong>goiter</strong>s.<br />

Family <strong>and</strong> twin pair studies in endemic <strong>and</strong> non endemic areas clearly demonstrated a<br />

genetic predisposition for <strong>goiter</strong> development. Within a Greek region endemic <strong>goiter</strong> affects<br />

some families more than others (211). This could provide evidence for a genetic etiology,<br />

though environmental factors that differ between families must also be considered (212). The<br />

familial aggregation <strong>of</strong> <strong>goiter</strong>s in Greece was confirmed in a subsequent study (213). The<br />

26


progeny <strong>of</strong> affected persons were more <strong>of</strong>ten affected by <strong>goiter</strong> than the descendants <strong>of</strong><br />

unaffected subjects. Likewise, other <strong>euthyroid</strong> <strong>goiter</strong> family studies in Greece, Slovakia <strong>and</strong><br />

Africa also lead to the conclusion that a genetic predisposition is present in the affected<br />

individuals (211;214;215). In addition, more rapidly growing <strong>goiter</strong>s in a subgroup <strong>of</strong> school<br />

children (15-20%) in spite <strong>of</strong> iodine supplementation (214) <strong>and</strong> differences in thyroid volume<br />

<strong>of</strong> adolescent siblings with sufficient iodine intake in Slovakia (216) also supports a genetic<br />

influence on thyroid growth. Moreover, studies in Greek populations have shown the<br />

persistence <strong>of</strong> endemic <strong>goiter</strong>s in certain regions despite iodine supplementation (217).<br />

Familial occurrence <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong> in an iodine replete region in Sweden was reported for<br />

41% <strong>of</strong> the patients with <strong>goiter</strong> with an even higher frequency <strong>of</strong> familial occurrence in those<br />

individuals with prepubertal development <strong>of</strong> the <strong>goiter</strong> (218). Even though, family studies are<br />

a reliable method to determine <strong>goiter</strong>s in many family members over several generations, it is<br />

impossible to definitively conclude whether the members shared the same susceptible genetic<br />

make up or the same environment. Therefore, twin studies are more informative in<br />

demonstrating a genetic component in the etiology <strong>of</strong> <strong>goiter</strong>. Hence, several investigations<br />

have provided evidence that there is a predisposing genetic background for <strong>goiter</strong> in twins. In<br />

endemic as well as nonendemic areas female monozygotic twins (80% (213;219) <strong>and</strong> 42%<br />

(220), respectively) have a higher concordance rate for <strong>goiter</strong> than female dizygotic twins (40-<br />

50%, (213;219) <strong>and</strong> 13%, (220). Twins <strong>of</strong> the same sex are supposed to share the same family<br />

environment. Therefore, the increased concordance was attributed to greater genetic similarity<br />

characterizing the monozygotic twins. Contribution <strong>of</strong> genetic susceptibility to the<br />

development <strong>of</strong> <strong>goiter</strong> was calculated to be 39% in endemic regions (219). Moreover, a study<br />

<strong>of</strong> 5479 monozygotic <strong>and</strong> dizygotic twins (220) performed by path analyses (structural<br />

equation modelling) suggests that the genetic predisposition to develop <strong>goiter</strong> is 82 % with 18<br />

% according to individual environmental factors in a nonendemic area. However, the<br />

27


previously reported twin studies show the importance <strong>of</strong> both hereditary <strong>and</strong> environmental<br />

factors (Hegedus, Bonnema, & Bennedbaek 2003).<br />

B C<strong>and</strong>idate loci<br />

Because <strong>of</strong> their important role in thyroid physiology <strong>and</strong> hormone synthesis, thyroglobulin<br />

(TG) <strong>and</strong> thyroid peroxidase (TPO), the sodium–iodide–symporter (NIS), pendrin gene (PDS)<br />

<strong>and</strong> the TSHR are major c<strong>and</strong>idate genes for familial <strong>euthyroid</strong> <strong>goiter</strong>s.<br />

Studies <strong>of</strong> hypothyroid <strong>goiter</strong>s have identified several genetic defects in TG <strong>and</strong> TPO (172).<br />

Congenital <strong>goiter</strong> <strong>and</strong> hypothyroidism caused by qualitative <strong>and</strong> quantitative defects <strong>of</strong> the<br />

TG gene were described by Medeiros-Neto (221). Other studies have also shown a link<br />

between the TG gene <strong>and</strong> congenital <strong>goiter</strong> <strong>and</strong> hypothyroidism (222-229). Furthermore, an<br />

inherited abnormality in TG synthesis leading to a lower content <strong>of</strong> TG in the thyroid gl<strong>and</strong><br />

was reported by Yoshida et al. (230) <strong>and</strong> a single amino acid substitution in the TG protein<br />

(Leu2366Pro) causes endoplasmic reticulum storage disease as determined in the cog/cog<br />

mouse (231). Although TG was postulated to be a major c<strong>and</strong>idate gene for <strong>euthyroid</strong> simple<br />

<strong>goiter</strong> only one genetic variation associated with <strong>euthyroid</strong> <strong>goiter</strong> has been identified in the<br />

TG gene up to date. Corral et al. (232) found a G - to T substitution at position 2610 <strong>of</strong> the<br />

TG cDNA. This resulted in replacement <strong>of</strong> histidine for glutamine at codon 870. This<br />

sequence alteration was located within exon 10 <strong>of</strong> the TG gene <strong>and</strong> was present in 25 <strong>of</strong> 26<br />

members <strong>of</strong> 3 families affected by <strong>euthyroid</strong> <strong>goiter</strong>. However, Perez–Centeno et al. (233)<br />

found the same point mutation in thyroglobulin exon 10 gene only in one <strong>of</strong> 36 patients with<br />

endemic <strong>euthyroid</strong> <strong>goiter</strong>. Hishinuma et al. (226;229) found two novel cystein substitutions in<br />

thyroglobulin, which caused defects in the intracellular transport <strong>of</strong> thyroglobulin in patients<br />

with a variant type <strong>of</strong> adenomatous <strong>euthyroid</strong> <strong>goiter</strong>. Gonzalez–Sarmiento et al. (234)<br />

identified a large heterozygous deletion within the TG gene in a study <strong>of</strong> 50 cases affected<br />

with nonendemic <strong>goiter</strong>. The deletion involved the promotor region <strong>and</strong> the exons 1 to 11 <strong>of</strong><br />

the TG gene <strong>and</strong> was associated with <strong>euthyroid</strong> <strong>goiter</strong>.<br />

28


Mutations responsible for dyshormogenesis have also been described in the TPO gene. TPO<br />

catalyzes the oxidation <strong>of</strong> iodide to an iodination species that forms iodothyrosines <strong>and</strong><br />

iodothyronines. Defects <strong>of</strong> TPO synthesis caused by a heterogenous spectrum <strong>of</strong> TPO<br />

mutations (235-240) have been reported to result in reduced TPO activity in combination with<br />

total iodide organification defect (TIOD). Hagen et al. (241) described an intelligent,<br />

<strong>euthyroid</strong> child with <strong>goiter</strong>. Together with her affected sister she showed no iodide<br />

peroxidation or thyrosine iodination activity. Likewise, a <strong>euthyroid</strong> woman with a recurrent<br />

<strong>goiter</strong> <strong>and</strong> partial iodide discharge was described by Pommier et al. (242). She had normal<br />

iodide peroxidation but deficient thyroglobulin iodination. However, these are the only two<br />

examples for TPO mutation resulting in <strong>euthyroid</strong> <strong>goiter</strong>s. Most <strong>of</strong> the previously reported<br />

homozygous or compound heterozygous mutations in the TPO gene lead to <strong>goiter</strong> <strong>and</strong><br />

hypothyroidism (236;243;244).<br />

Since the cloning <strong>and</strong> molecular characterization <strong>of</strong> the human NIS gene (245) several defects<br />

in this gene have been detected in patients with different phenotypes <strong>of</strong> thyroid diseases (246).<br />

A heterozygous T354P mutation results in congenital hypothyroidism <strong>and</strong> <strong>goiter</strong> (247-251).<br />

However, two studies reported that the homozygous T354P mutation is associated with<br />

<strong>euthyroid</strong>ism <strong>and</strong> <strong>goiter</strong> (252;253). Moreover, other allelic variants producing deletion,<br />

missense or truncation <strong>of</strong> the NIS protein have been described (254;255).<br />

Mutations in the PDS gene cause Pendred syndrome characterized by congenital sensorineural<br />

hearing loss combined with <strong>goiter</strong>. In Pendred syndrome the thyroid enlargement typically<br />

begins in childhood <strong>and</strong> can vary between <strong>and</strong> within families (256;257). Reported <strong>goiter</strong><br />

sizes vary from small nodules to large <strong>multinodular</strong> <strong>goiter</strong>s (258). Almost all affected<br />

individuals are clinically <strong>and</strong> biochemically <strong>euthyroid</strong>. Positive perchlorate tests suggest that<br />

the PDS defects impair the organification <strong>of</strong> iodide. Different PDS mutations, each<br />

segregating with the disease in the families in which they occurred, have been identified (259-<br />

263)(85). Most <strong>of</strong> them are loss-<strong>of</strong>-function mutations which directly cause thyroid disease in<br />

29


Pendred syndrome. Therefore, the PDS gene is a c<strong>and</strong>idate gene for development <strong>of</strong> <strong>euthyroid</strong><br />

<strong>goiter</strong>.<br />

Furthermore, the TSHR on chromsome 14q31 could be a c<strong>and</strong>idate gene for <strong>euthyroid</strong> <strong>goiter</strong><br />

according to its central role for thyroid function <strong>and</strong> growth. TSHR germline mutations have<br />

been found in rare cases <strong>of</strong> <strong>euthyroid</strong> familial <strong>goiter</strong> (7) (see also chapter IA). Moreover, a<br />

germline genetic variation in codon 727 <strong>of</strong> the TSHR gene (Asp ->Glu (264)) has been<br />

associated with <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong> in iodine deficient areas (265). However, the wild<br />

type TSHR response to TSH was very low in this study <strong>and</strong> this in vitro finding was not<br />

confirmed in a further study (266;267). Moreover, the putative predisposition for <strong>toxic</strong><br />

<strong>multinodular</strong> <strong>goiter</strong> was based on functional analysis <strong>of</strong> the TSHR codon 727 variation which<br />

revealed a higher cAMP response compared to the wild type receptor. However, this in vitro<br />

finding was not confirmed in a recent study (266). Recently Peeters et al. (268) reported that<br />

the homozygous C variant (coding for aspartic acid) <strong>of</strong> the D727E polymorphism was<br />

associated with a lower serum TSH level in 156 healthy blood donors. Although this finding<br />

supports a possible functional relevance <strong>of</strong> this polymorphism the role <strong>of</strong> a lower TSH level in<br />

the development <strong>of</strong> MNG is unknown.<br />

C Linkage analysis<br />

Over the last years linkage analyses became a reliable method to identify novel susceptibility<br />

loci for both mendelian <strong>and</strong> complex diseases in large families or affected sib pairs by using<br />

genetic markers for microsatellite DNA or repetitive sequences in the entire human genome.<br />

Several different susceptible areas have been discovered in large families with non-mendelian<br />

transmission <strong>of</strong> <strong>euthyroid</strong> familial <strong>goiter</strong>. The <strong>multinodular</strong>-<strong>goiter</strong>-1 locus MNG-1 on<br />

chromosome 14q31 was first reported by Bignell (269) as the result <strong>of</strong> a genome wide linkage<br />

analysis <strong>of</strong> a large Canadian family with 18 patients affected with <strong>multinodular</strong> <strong>goiter</strong>. A<br />

maximum two point LOD score <strong>of</strong> 3.8 at D14S1030 <strong>and</strong> a multipoint LOD score <strong>of</strong> 4.88,<br />

defined by D14S1062 <strong>and</strong> D14S267 was calculated. In a further study a family with recurrent<br />

30


<strong>euthyroid</strong> <strong>goiter</strong>s was investigated for linkage to the same c<strong>and</strong>idate region (270). According<br />

to a dominant pattern <strong>of</strong> inheritance with full penetrance, indication for linkage was obtained<br />

by a maximum two point LOD score <strong>of</strong> 1.5 at marker D14S1030 at the MNG-1 locus.<br />

Moreover, a maximum multipoint LOD score <strong>of</strong> 1.49 was obtained for the region between the<br />

TSHR <strong>and</strong> the MNG-1 c<strong>and</strong>idate loci. The haplotype cosegregation <strong>of</strong> microsatellite markers<br />

confirmed the entire chromosomal segment between both loci on chromosome 14q31 as a<br />

positional c<strong>and</strong>idate region for non<strong>toxic</strong> <strong>goiter</strong>. Although the TSHR was a first line c<strong>and</strong>idate<br />

gene sequence analysis <strong>of</strong> the TSHR only revealed several previously reported<br />

polymorphisms. In the study by Bignell et al. the TSHR was previously clearly excluded as a<br />

c<strong>and</strong>idate gene (269). Another study reported the analysis <strong>of</strong> an Italian - three generation<br />

pedigree, including 10 affected females <strong>and</strong> 2 affected males (271). An X-linked dominant<br />

pattern <strong>of</strong> inheritance was observed. The investigation <strong>of</strong> 18 markers spaced at 10 cM<br />

intervals on the X-chromosome revealed evidence for linkage at marker DXS1226 with a<br />

significant LOD score <strong>of</strong> 4.73. These findings led to the conclusion that defects in the Xp22<br />

region caused thyroid disease in this family. Moreover, the haplotype inspection reduced the<br />

critical interval to 9.6 cM between the markers DXS1052 <strong>and</strong> DXS8039.<br />

In view <strong>of</strong> the different susceptibility loci for <strong>euthyroid</strong> <strong>goiter</strong> a heterogenous mode <strong>of</strong><br />

inheritance for <strong>euthyroid</strong> <strong>goiter</strong> is very likely. Linkage analysis for the thyroid c<strong>and</strong>idate<br />

genes has been performed in four German <strong>and</strong> one Slovakian family (272). The c<strong>and</strong>idate<br />

genes were also analyzed assuming different recombination fractions for the microsatellite<br />

markers in two point <strong>and</strong> multipoint analysis. Linkage analysis results <strong>of</strong> this study were not<br />

significant enough to definitely exclude or confirm linkage to the investigated c<strong>and</strong>idate genes<br />

TG, TPO <strong>and</strong> NIS. To date, there is no evidence for or against susceptibility <strong>of</strong> the<br />

investigated c<strong>and</strong>idate genes for <strong>euthyroid</strong> familial <strong>goiter</strong>. Since linkage to MNG-1 (14q31)<br />

was previously reported in two families (269;270) <strong>and</strong> Xp22 in a single family (271), the four<br />

German families <strong>and</strong> one Slovakian family were also investigated to test a more general<br />

31


validity <strong>of</strong> these c<strong>and</strong>idate regions (272). However, the absence <strong>of</strong> a correlation <strong>of</strong> inheritance<br />

patterns for the investigated markers in the families <strong>and</strong> the nonsignificant LOD scores<br />

determined according to the L<strong>and</strong>er-Kruglyak guide suggested a lower probability for MNG-1<br />

<strong>and</strong> Xp22 as major monogenic causes for the etiology <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong>. Moreover, in two<br />

families a very weak indication for linkage to PDS <strong>and</strong> Xp22, respectively, was identified<br />

(272). Furthermore, the nonsignificant LOD scores calculated in this study suggest, that the<br />

strongest genetic locus detectable by linkage is unknown to date <strong>and</strong> that it is probable that<br />

different c<strong>and</strong>idate genes or loci cause <strong>euthyroid</strong> <strong>goiter</strong> in different families. In conclusion,<br />

these studies gave further indications for genetic heterogeneity <strong>of</strong> <strong>euthyroid</strong> familial <strong>goiter</strong>s.<br />

To discover novel <strong>and</strong> more general c<strong>and</strong>idate regions or genes we performed a genome wide<br />

scan to detect susceptibility loci that predispose for <strong>euthyroid</strong> <strong>goiter</strong> using 450 microsatellite<br />

markers in 18 Danish, German <strong>and</strong> one Slovakian family, comprising 79 affected <strong>and</strong> 68<br />

unaffected family members (273). Assuming genetic heterogeneity <strong>and</strong> a dominant pattern <strong>of</strong><br />

inheritance four novel c<strong>and</strong>idate loci on chromosomes 2q, 3p, 7q <strong>and</strong> 8p were identified. Four<br />

families showed linkage to the 3p locus whereas the loci 2q, 7q <strong>and</strong> 8p each showed linkage<br />

in one family. The haplotype inspection delimited a critical interval <strong>of</strong> 16 cM on 3p (figure 4).<br />

Within this interval the thyroid hormone receptor beta (THRB) is mapped. Our mutation<br />

screen also included the two thyroid hormone receptor interactor genes 6 <strong>and</strong> 12 on 7q <strong>and</strong> 2q<br />

in addition to the THRB gene (273). However, sequencing <strong>of</strong> all these c<strong>and</strong>idate genes<br />

revealed no germline mutations, that would co-segregate with the <strong>goiter</strong> in the affected<br />

families. In conclusion, these genetic studies confirm that genetic heterogeneity is likely to<br />

explain the identification <strong>of</strong> different c<strong>and</strong>idate loci like MNG-1 (269;270) <strong>and</strong> Xp22 (271)<br />

in several families.<br />

Most cases <strong>of</strong> familial <strong>goiter</strong> present an autosomal dominant pattern <strong>of</strong> inheritance. However<br />

for the majority <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong> cases a multifactorial genesis with complex interactions <strong>of</strong><br />

environmental factors like iodine deficiency, cigarette smoking, age, sex <strong>and</strong> certain drugs or<br />

32


emotional stress on a genetic background is more likely. Loci from linkage analysis or<br />

association studies could provide important genetic risk factors in a more complex genetic<br />

background.<br />

A Therapeutic implications<br />

IX Perspectives<br />

It is still poorly understood, how genes interact with different environmental factors (33).<br />

Therefore we have actually no ideal treatment for <strong>euthyroid</strong> benign (multi-)nodular <strong>goiter</strong>.<br />

Most clinical trials (56;57;274-281) investigated the efficacy <strong>of</strong> levothyroxine to suppress<br />

TSH <strong>and</strong> to arrest further growth or reduce the size <strong>of</strong> thyroid nodules. Thyroxine suppressive<br />

therapy is given with the hope that nodules might decrease in size. However, these studies<br />

reported contradictory results concerning the investigated endpoint (i.e. reducing the size <strong>of</strong><br />

thyroid nodules). Moreover, the benefit <strong>of</strong> arresting growth or reducing the size <strong>of</strong> a thyroid<br />

nodule has not been conclusively answered because there are controversial reports on a<br />

possible correlation between thyroid nodule size <strong>and</strong> the development <strong>of</strong> thyroid epithelial<br />

cell carcinomas (52;282-284).<br />

Moreover, a decrease in serum TSH is related to increasing nodularity <strong>and</strong> size <strong>of</strong> the thyroid<br />

(4). Cross-section studies provide no evidence that the stimulation <strong>of</strong> thyroid growth or<br />

thyroid function through serum TSH is responsible for thyroid nodule growth (285;286)<br />

because patients with benign, cold thyroid nodules did not exhibit elevated TSH levels in<br />

comparison to controls (56;57;274;277-279).<br />

Furthermore, TSH suppression may lead to hyperthyroidism, reduced bone density <strong>and</strong> atrial<br />

fibrillation (287;288) <strong>and</strong> levothyroxine therapy can lower the intrathyroidal iodine content<br />

(289-291). The pathophysiological rationale for levothyroxine therapy <strong>of</strong> thyroid nodules with<br />

the aim <strong>of</strong> reducing their volume is therefore questionable.<br />

33


Moreover there is an uncertainty about predictors <strong>of</strong> response like clonality (8;84), growth<br />

(52), size at time <strong>of</strong> diagnosis (282-284), or cell-rich nodules (292)).<br />

Since thyroid nodules, thyroid autonomy <strong>and</strong> thyroid cancer were more <strong>of</strong>ten detected in<br />

iodine deficiency areas than in iodine sufficient areas (2;18;33;293), area wide iodine<br />

supplementation became the first choice in thyroid nodule prevention (24). Although iodine<br />

supplementation is an adequate therapy for nodular <strong>goiter</strong> (291;294) this option is <strong>of</strong>ten<br />

ignored. Possible benefits <strong>of</strong> treating or preventing thyroid nodule (growth) could be the<br />

avoidance <strong>of</strong> thyroid nodules/<strong>goiter</strong> associated symptoms (hoarseness, pain, hyperthyroidism,<br />

hypothyroidism), more rarely prevention <strong>of</strong> thyroid malignancy, prevention <strong>of</strong> surgical<br />

intervention <strong>and</strong> it’s related risks (295). In addition, this could lead to reduction <strong>of</strong> costs for<br />

common surgical interventions <strong>and</strong> postoperative pharmacotherapy (296). Therefore, the<br />

benefit <strong>of</strong> treating or preventing thyroid nodules is more likely prevention <strong>of</strong> clinical disease<br />

rather than reduction <strong>of</strong> nonclinical disease (295). In the authors’ view future studies should<br />

include patient-relevant outcomes like thyroid cancer incidence, health-related quality <strong>of</strong> life<br />

<strong>and</strong> costs. Multicentre studies are needed to investigate whether thyroid nodule growth is<br />

associated with an increased frequency <strong>of</strong> thyroid malignancies.<br />

B Diagnostic implications<br />

Evaluation <strong>of</strong> patients with nodular thyroid disease is directed at two aspects: exclusion <strong>of</strong><br />

thyroid malignancy <strong>and</strong> definition <strong>of</strong> the functional <strong>and</strong> if possible pathomorphologic<br />

character <strong>of</strong> the nodule to stratify the best treatment approach.<br />

Diagnosis <strong>of</strong> thyroid malignancy is ultimately based on the histological examination but can<br />

be strongly suggested clinically e.g. by the presence <strong>of</strong> a rapid growing nodule, cervical<br />

lymph nodes, sudden onset <strong>of</strong> hoarseness <strong>and</strong> almost established on the basis <strong>of</strong> a malignant<br />

FNAC <strong>of</strong> the thyroid nodule (38;39;41). However, despite this clear-cut approach, the<br />

34


ultimate challenge in past <strong>and</strong> present thyroidology remains the identification <strong>of</strong> generally<br />

very rare thyroid cancer amongst the highly prevalent condition <strong>of</strong> nodular thyroid disease.<br />

This is not only reason for concern <strong>of</strong> the affected patients, <strong>and</strong> a daily task for all doctors<br />

dealing with thyroid disease, but increasingly poses an economic problem in times <strong>of</strong> limited<br />

health care budgets.<br />

Hence many studies <strong>and</strong> reviews have been dedictated to the resolution <strong>of</strong> this problem: There<br />

is agreement that both ultrasonography <strong>and</strong> thyroid scintiscan add little to nothing to the<br />

clarification <strong>of</strong> the benign or malignant nature <strong>of</strong> a nodule, with the exception that “hot” i.e.<br />

autonomously functioning thyroid nodules very rarely represent malignancy (38;39;41).<br />

Furthermore, it is widely acknowledged that fine needle aspiration cytology represents the<br />

most sensitive <strong>and</strong> specific means for pre-operative diagnosis <strong>of</strong> thyroid malignancy (40;297).<br />

The drawback remains that FNAC is only reliable if performed <strong>and</strong> analysed by a thyroid<br />

expert team (40;41). Besides it will be impossible to perform FNAC in all patients with<br />

nodular thyroid disease, which in countries with iodine deficiency may affect up to 30% <strong>and</strong><br />

more <strong>of</strong> the adult population (18). Guidelines have therefore defined a nodule size <strong>of</strong> at least<br />

10-15 mm <strong>and</strong>/or hyp<strong>of</strong>unctionality as indications for FNAC (38;39;41). However, it is<br />

unclear how to proceed in case <strong>of</strong> the much more frequent <strong>multinodular</strong> <strong>goiter</strong>s, which<br />

probably harbor the same malignancy risk as solitary lesions (2), <strong>and</strong> a pragmatic approach<br />

here may be to perform FNAC <strong>of</strong> the prominent “cold” nodule (33) or to recommend thyroid<br />

surgery in cases <strong>of</strong> diagnostic uncertainty. The same pragmatic but not evidence guided<br />

approach is to perform surgery in patients with risk constellations e.g. past history <strong>of</strong><br />

radiation, family history <strong>of</strong> thyroid cancer etc.<br />

Furthermore, even in an ideal setting e.g. in a specialist thyroid clinic, FNAC may be “non-<br />

diagnostic” or “suspicious” in more than 20% <strong>of</strong> cases. What are the options then? If FNAC<br />

has been non-diagnostic it needs to be repeated (298-300). “A number <strong>of</strong> at least six clusters<br />

35


<strong>of</strong> thyrocytes on each <strong>of</strong> at least two slides prepared from separate aspirates" has been<br />

proposed by Hamburger as a quality criterion for a diagnostic thyroid FNAC (301). However,<br />

recently Oertel has critically discussed that adequacy <strong>of</strong> the specimen cannot be based solely<br />

on the cell count (292). In case <strong>of</strong> ”suspicious” FNACs, which represent 10-20% <strong>of</strong> all<br />

FNACs (41) markers could be helpful, in particular to discriminate follicular adenoma from<br />

follicular carcinoma from follicular variant <strong>of</strong> papillary carcinoma <strong>and</strong> to distinguish Huerthle<br />

cell adenoma <strong>and</strong> cancer etc. (302). However, since much but still too little is known about<br />

the molecular etiology <strong>of</strong> the “common” thyroid nodule, it is even more difficult to define a<br />

marker <strong>of</strong> benignity versus malignancy although many c<strong>and</strong>idates have been screened <strong>and</strong><br />

proposed, some <strong>of</strong> which indeed seem promising e.g. Galectin-3, thyroperoxidase (MoAb47),<br />

PAX-8/PPARγ rearrangements, BRAF mutation (152;303-305), but none <strong>of</strong> which have<br />

made their way into routine diagnostics yet. In contrast novel technologies in particular<br />

microarray <strong>and</strong> proteomics methodolgies will almost certainly contribute to change this rather<br />

pessimistic state-<strong>of</strong>-the-art situation. Through these methods, which are increasingly applied<br />

in research labs everywhere, we have the possibility for the first time, to perform genome <strong>and</strong><br />

proteome wide screening studies <strong>and</strong> despite all drawbacks they may in fact be ideally suited<br />

to identify useful markers similary to the in vivo situation, they are also performed as “cross<br />

sectional” studies. Contribution will also come from the likewise many genome wide linkage<br />

studies, which contributed to identify a number <strong>of</strong> disease “genes” in the past (306). One<br />

example is familial <strong>euthyroid</strong> <strong>goiter</strong>, discussed in chapter VIII.<br />

Which diagnostic markers would then be needed besides the important cytological<br />

differentiation <strong>of</strong> the above discussed entities <strong>of</strong> thyroid pathology?<br />

It would be ideal to have markers which indicate which nodules, on the long term, may turn<br />

into thyroid malignancy: More realistically, it could be feasible to define markers which<br />

correlate with augmented nodule growth or which correlate with ongoing thyroid de-<br />

differentiation. Clarification <strong>of</strong> the molecular characteristics <strong>and</strong> application <strong>of</strong> markers which<br />

36


define increased nodule growth <strong>and</strong> de-differention will allow formation <strong>of</strong> nodule subgroups.<br />

This would be the basis for a therapeutic strategy study to determine whether e.g. iodide<br />

supplementation, combination <strong>of</strong> L-T4 <strong>and</strong> iodine, no therapy, ethanol injection, radioiodine<br />

or surgery are indicated for which nodule subgroup.<br />

37


Figure legends<br />

Figure 1<br />

Differential mRNA expression in components <strong>of</strong> TGF β signaling in AFTNs (118)<br />

Gene expression analysis revealed a significantly decreased expression (green colored boxes)<br />

<strong>of</strong> the type III TGF-β receptor, Smad 1, 3 <strong>and</strong> 4 <strong>and</strong> the E1A-associated protein p300 in<br />

AFTNs in comparison to their normal surrounding tissue, whereas the inhibitory Smad 6 <strong>and</strong><br />

7 are characterized by a significantly increased expression (red colored boxes). These findings<br />

argue for an inactivation <strong>of</strong> the TGF-β signaling cascade in AFTNs, which is normally<br />

responsible for inhibition <strong>of</strong> iodine uptake, iodine organification, thyroglobulin expression<br />

<strong>and</strong> cell proliferation in thyroid cells.<br />

Figure 2<br />

Immunohistochemical analysis <strong>of</strong> NIS expression in thyroid nodules <strong>and</strong> surrounding<br />

tissues (307)<br />

A: CTN 65 - nodule tissue; predominant intracytoplasmic NIS staining, only partly cell<br />

membrane immunoreactivity, B: CTN 65 - surrounding tissue; hNIS protein immunoreactivity<br />

is very low <strong>and</strong> heterogeneous, C: CTN 13 - nodule tissue, mostly<br />

intracytoplasmic immunoreactivity, D: <strong>toxic</strong> thyroid nodule; NIS cell membrane<br />

immunoreactivity is predominant<br />

Figure 3<br />

Hypothesis for thyroid nodular transformation. The starting point for the development <strong>of</strong><br />

MNG is hyperplasia induced by goitrogenic stimuli (e.g. iodine deficiency). Iodine deficiency<br />

increases mutagenesis directly (production <strong>of</strong> H2O2/free radicals) or indirectly (proliferation<br />

<strong>and</strong> increased number <strong>of</strong> cell divisions). Subsequently, hyperplasia forms cell clones. Some <strong>of</strong><br />

38


them contain somatic mutations <strong>of</strong> the TSH-R leading to autonomously functioning thyroid<br />

nodules (red spots) or contain mutations that lead to dedifferentiation <strong>and</strong> therefore cold<br />

thyroid nodules or cold adenoma (blue dots).<br />

Figure 4<br />

The figure shows the mapping strategy for susceptible chromosomal regions responsible for<br />

<strong>euthyroid</strong> <strong>goiter</strong> at chromosome 3p. a) highly polymorphic microsatellite markers on 3p, used<br />

to decide linkage <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong> to this locus. b) shows the location <strong>of</strong> the peak NPL<br />

score which indicates linkage <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong>.<br />

39


Table 1: Natural course <strong>of</strong> benign thyroid nodules in studies in iodine deficient <strong>and</strong> iodine sufficient areas with more than 2yr. follow-up.<br />

study country number <strong>of</strong><br />

patients<br />

Papini et al.<br />

(56)<br />

Br<strong>and</strong>er et<br />

al. (50)<br />

Alex<strong>and</strong>er et<br />

al. (51)<br />

Kuma et<br />

al.(53))<br />

Quadbeck et<br />

al. (55)<br />

Knudsen et<br />

al. (3)<br />

Italy 41 multicentre<br />

study<br />

location assessment follow-up definition <strong>of</strong><br />

growth<br />

10MHz<br />

ultrasound<br />

inl<strong>and</strong> 34 Cohort study 7.5MHz<br />

ultrasound<br />

USA 268 thyroid<br />

nodule clinic<br />

5-15MHz<br />

ultrasound<br />

Japan 134 Hospital Palpation<br />

<strong>and</strong> 7.5MHz<br />

ultrasound<br />

Germany 109 Endocrine<br />

Clinic<br />

7.5MHz<br />

ultrasound<br />

Denmark 45 Cohort study 7.5MHz<br />

ultrasound<br />

5 yr. > 12% increase in<br />

volume<br />

growth comment<br />

56% placebo group in L-T4<br />

study<br />

4.9-5.6 yr. not defined 35% investigation also <strong>of</strong><br />

lesions < 10mm<br />

1 month to 5<br />

yr.<br />

> 15% increase in<br />

volume<br />

39% growth predominantly in<br />

solid nodules<br />

9-11 yr. not defined 21% > 40% (80% <strong>of</strong> cystic<br />

nodules) <strong>of</strong> nodules<br />

decreased or disappeared<br />

3-12 yr. > 30% increase in<br />

volume<br />

2 yr. > 5mm change in<br />

diameter<br />

50% no correlation with nodule<br />

function, age <strong>and</strong> gender<br />

2% 10% decreased only<br />

„cold“ nodules studied


Reference List<br />

1. Hedinger C, Williams ED, Sobin LH 1989 The WHO histological classification<br />

<strong>of</strong> thyroid tumors: a commentary on the second edition. Cancer 63:908-911<br />

2. Belfiore A, La Rosa GL, La Porta GA, Giuffrida D, Milazzo G, Lupo L,<br />

Regalbuto C, Vigneri R 1992 Cancer risk in patients with cold thyroid nodules:<br />

relevance <strong>of</strong> iodine intake, sex, age, <strong>and</strong> <strong>multinodular</strong>ity. Am J Med 93:363-369<br />

3. Knudsen N, Perrild H, Christiansen E, Rasmussen S, Dige-Petersen H,<br />

Jorgensen T 2000 Thyroid structure <strong>and</strong> size <strong>and</strong> two-year follow-up <strong>of</strong> solitary<br />

cold thyroid nodules in an unselected population with borderline iodine<br />

deficiency. Eur J Endocrinol 142:224-230<br />

4. Berghout A, Wiersinga WM, Smits NJ, Touber JL 1990 Interrelationships<br />

between age, thyroid volume, thyroid nodularity, <strong>and</strong> thyroid function in<br />

patients with sporadic non<strong>toxic</strong> <strong>goiter</strong>. Am J Med 89:602-608<br />

5. Vassart G, Dumont JE 1992 The thyrotropin receptor <strong>and</strong> the regulation <strong>of</strong><br />

thyrocyte function <strong>and</strong> growth. Endocr Rev 13:596-611<br />

6. Dumont JE, Lamy F, Roger P, Maenhaut C 1992 Physiological <strong>and</strong><br />

pathological regulation <strong>of</strong> thyroid cell proliferation <strong>and</strong> differentiation by<br />

thyrotropin <strong>and</strong> other factors. Physiol Rev 72:667-697<br />

7. Paschke R, Ludgate M 1997 The thyrotropin receptor in thyroid diseases. N<br />

Engl J Med 337:1675-1681<br />

8. Krohn K, Paschke R 2001 Progress in underst<strong>and</strong>ing the etiology <strong>of</strong> thyroid<br />

autonomy. J Clin Endocrinol Metab 86:3336-3345<br />

9. Krohn K, Wohlgemuth S, Gerber H, Paschke R 2000 Hot microscopic areas <strong>of</strong><br />

iodine deficient <strong>euthyroid</strong> <strong>goiter</strong>s contain constitutively activating TSH<br />

receptor mutations. J Pathology 192:37-42<br />

10. Ledent C, Coppee F, Dumont JE, Vassart G, Parmentier M 1996 Transgenic<br />

models for proliferative <strong>and</strong> hyperfunctional thyroid diseases. Exp Clin<br />

Endocrinol Diabetes 104 Suppl 3:43-46<br />

11. Delange F 1994 The disorders induced by iodine deficiency. Thyroid 4:107-128<br />

12. Delange F, de Benoist B, Pretell E, Dunn JT 2001 Iodine deficiency in the<br />

world: where do we st<strong>and</strong> at the turn <strong>of</strong> the century? Thyroid 11:437-447<br />

13. Jarlov AE, Nygaard B, Hegedus L, Hartling SG, Hansen JM 1998 Observer<br />

variation in the clinical <strong>and</strong> laboratory evaluation <strong>of</strong> patients with thyroid<br />

dysfunction <strong>and</strong> <strong>goiter</strong>. Thyroid 8:393-398<br />

14. Tan GH, Gharib H 1997 Thyroid incidentalomas: management approaches to<br />

nonpalpable nodules discovered incidentally on thyroid imaging. Ann Intern<br />

Med 126:226-231


15. Hegedus L 2001 Thyroid ultrasound. Endocrinol Metab Clin North Am 30:339-<br />

344<br />

16. Knudsen N, Bulow I, Jorgensen T, Laurberg P, Ovesen L, Perrild H 2000<br />

Goitre prevalence <strong>and</strong> thyroid abnormalities at ultrasonography: a<br />

comparative epidemiological study in two regions with slightly different iodine<br />

status. Clin Endocrinol (Oxf) 53:479-485<br />

17. Aghini-Lombardi F, Antonangeli L, Martino E, Vitti P, Maccherini D, Leoli F,<br />

Rago T, Grasso L, Valeriano R, Balestrieri A, Pinchera A 1999 The spectrum <strong>of</strong><br />

thyroid disorders in an iodine-deficient community: the Pescopagano survey. J<br />

Clin Endocrinol Metab 84:561-566<br />

18. Hampel R, Kulberg T, Klein K, Jerichow JU, Pichmann EG, Clausen V,<br />

Schmidt I 1995 [Goiter incidence in Germany is greater than previously<br />

suspected]. Med Klin 90:324-329<br />

19. Knudsen N, Laurberg P, Perrild H, Bulow I, Ovesen L, Jorgensen T 2002 Risk<br />

factors for <strong>goiter</strong> <strong>and</strong> thyroid nodules. Thyroid 12:879-888<br />

20. V<strong>and</strong>erpump MP, Tunbridge WM, French JM, Appleton D, Bates D, Clark F,<br />

Grimley EJ, Hasan DM, Rodgers H, Tunbridge F, . 1995 The incidence <strong>of</strong><br />

thyroid disorders in the community: a twenty-year follow-up <strong>of</strong> the Whickham<br />

Survey. Clin Endocrinol (Oxf) 43:55-68<br />

21. Gärtner R, Bechtner G, Rafferzeder M, Greil W 1997 Comparison <strong>of</strong> urinary<br />

iodine excretion <strong>and</strong> thyroid volume in students with or without constant<br />

iodized salt intake. Exp Clin Endocrinol Diabetes 105 Suppl 4:43-45<br />

22. Laurberg P, Pedersen KM, Vestergaard H, Sigurdsson G 1991 High incidence<br />

<strong>of</strong> <strong>multinodular</strong> <strong>toxic</strong> goitre in the elderly population in a low iodine intake area<br />

vs. high incidence <strong>of</strong> Graves' disease in the young in a high iodine intake area:<br />

comparative surveys <strong>of</strong> thyro<strong>toxic</strong>osis epidemiology in East-Jutl<strong>and</strong> Denmark<br />

<strong>and</strong> Icel<strong>and</strong>. J Intern Med 229:415-420<br />

23. Hamburger JI 1980 Evolution <strong>of</strong> <strong>toxic</strong>ity in solitary non<strong>toxic</strong> autonomously<br />

functioning thyroid nodules. J Clin Endocrinol Metab 50:1089-1093<br />

24. Baltisberger BL, Minder CE, Bürgi H 1995 Decrease <strong>of</strong> incidence <strong>of</strong> <strong>toxic</strong><br />

nodular goitre in a region <strong>of</strong> Switzerl<strong>and</strong> after full correction <strong>of</strong> mild iodine<br />

deficiency . Eur J Endocrinol 132:546-549<br />

25. Christensen SB, Ericsson UB, Janzon L, Tibblin S, Mel<strong>and</strong>er A 1984 Influence<br />

<strong>of</strong> cigarette smoking on <strong>goiter</strong> formation, thyroglobulin, <strong>and</strong> thyroid hormone<br />

levels in women. J Clin Endocrinol Metab 58:615-618<br />

26. Knudsen N, Bulow I, Laurberg P, Ovesen L, Perrild H, Jorgensen T 2002<br />

Association <strong>of</strong> tobacco smoking with <strong>goiter</strong> in a low-iodine-intake area. Arch<br />

Intern Med 162:439-443<br />

27. Nagataki S, Nystrom E 2002 Epidemiology <strong>and</strong> primary prevention <strong>of</strong> thyroid<br />

cancer. Thyroid 12:889-896<br />

42


28. Kazakov VS, Demidchik EP, Astakhova LN 1992 Thyroid cancer after<br />

Chernobyl. Nature 359:21<br />

29. Shibata Y, Yamashita S, Masyakin VB, Panasyuk GD, Nagataki S 2001 15<br />

years after Chernobyl: new evidence <strong>of</strong> thyroid cancer. Lancet 358:1965-1966<br />

30. Ron E, Lubin JH, Shore RE, Mabuchi K, Modan B, Pottern LM, Schneider<br />

AB, Tucker MA, Boice JD, Jr. 1995 Thyroid cancer after exposure to external<br />

radiation: a pooled analysis <strong>of</strong> seven studies. Radiat Res 141:259-277<br />

31. Hahn K, Schnell-Inderst P, Grosche B, Holm LE 2001 Thyroid cancer after<br />

diagnostic administration <strong>of</strong> iodine-131 in childhood. Radiat Res 156:61-70<br />

32. Sari R, Balci MK, Altunbas H, Karayalcin U 2003 The effect <strong>of</strong> body weight<br />

<strong>and</strong> weight loss on thyroid volume <strong>and</strong> function in obese women. Clin<br />

Endocrinol (Oxf) 59:258-262<br />

33. Hegedus L, Bonnema SJ, Bennedbaek FN 2003 Management <strong>of</strong> simple nodular<br />

<strong>goiter</strong>: current status <strong>and</strong> future perspectives. Endocr Rev 24:102-132<br />

34. Reinwein D, Benker G, Konig MP, Pinchera A, Schatz H, Schleusener A 1988<br />

The different types <strong>of</strong> hyperthyroidism in Europe. Results <strong>of</strong> a prospective<br />

survey <strong>of</strong> 924 patients. J Endocrinol Invest 11:193-200<br />

35. Furlanetto TW, Nguyen LQ, Jameson JL 1999 Estradiol increases proliferation<br />

<strong>and</strong> down-regulates the sodium/iodide symporter gene in FRTL-5 cells.<br />

Endocrinology 140:5705-5711<br />

36. Manole D, Schildknecht B, Gosnell B, Adams E, Derwahl M 2001 Estrogen<br />

promotes growth <strong>of</strong> human thyroid tumor cells by different molecular<br />

mechanisms. J Clin Endocrinol Metab 86:1072-1077<br />

37. Struve C, Ohlen S 1990 [The effect <strong>of</strong> earlier pregnancies on the prevalence <strong>of</strong><br />

<strong>goiter</strong> <strong>and</strong> nodules in thyroid-healthy women]. Dtsch Med Wochenschr<br />

115:1050-1053<br />

38. Singer PA, Cooper DS, Daniels GH, Ladenson PW, Greenspan FS, Levy EG,<br />

Braverman LE, Clark OH, McDougall IR, Ain KV, Dorfman SG 1996<br />

Treatment guidelines for patients with thyroid nodules <strong>and</strong> well-differentiated<br />

thyroid cancer. American Thyroid Association. Arch Intern Med 156:2165-<br />

2172<br />

39. Mazzaferri EL 1993 Management <strong>of</strong> a solitary thyroid nodule. N Engl J Med<br />

328:553-559<br />

40. Blum M, Hussain MA 2003 Evidence <strong>and</strong> thoughts about thyroid nodules that<br />

grow after they have been identified as benign by aspiration cytology. Thyroid<br />

13:637-641<br />

41. Giuffrida D, Gharib H 1995 Controversies in the management <strong>of</strong> cold, hot, <strong>and</strong><br />

occult thyroid nodules. Am J Med 99:642-650<br />

43


42. Trivalle C, Doucet J, Chassagne P, L<strong>and</strong>rin I, Kadri N, Menard JF, Berc<strong>of</strong>f E<br />

1996 Differences in the signs <strong>and</strong> symptoms <strong>of</strong> hyperthyroidism in older <strong>and</strong><br />

younger patients. J Am Geriatr Soc 44:50-53<br />

43. T<strong>of</strong>t AD 2001 Clinical practice. Subclinical hyperthyroidism. N Engl J Med<br />

345:512-516<br />

44. Parle JV, Maisonneuve P, Sheppard MC, Boyle P, Franklyn JA 2001 Prediction<br />

<strong>of</strong> all-cause <strong>and</strong> cardiovascular mortality in elderly people from one low serum<br />

thyrotropin result: a 10-year cohort study. Lancet 358:861-865<br />

45. Duprez L, Parma J, Van S<strong>and</strong>e J, Allgeier A, Leclere J, Schvartz C, Delisle MJ,<br />

Decoulx M, Orgiazzi J, Dumont J 1994 Germline mutations in the thyrotropin<br />

receptor gene cause non-autoimmune autosomal dominant hyperthyroidism.<br />

Nat Genet 7:396-401<br />

46. Führer D, Warner J, Sequeira M, Paschke R, Gregory J, Ludgate M 2000<br />

Novel TSHR germline mutation (Met463Val) masquerading as Graves' disease<br />

in a large Welsh kindred with hyperthyroidism. Thyroid 10:1035-1041<br />

47. Kraiem Z, Glaser B, Yigla M, Pauker J, Sadeh O, Sheinfeld M 1987 Toxic<br />

<strong>multinodular</strong> <strong>goiter</strong>: a variant <strong>of</strong> autoimmune hyperthyroidism. J Clin<br />

Endocrinol Metab 65:659-664<br />

48. Wallasch<strong>of</strong>ski H, Orda C, Georgi P, Miehle K, Paschke R 2001 Distinction<br />

between autoimmune <strong>and</strong> non-autoimmune hyperthyroidism by determination<br />

<strong>of</strong> TSH-receptor antibodies in patients with the initial diagnosis <strong>of</strong> <strong>toxic</strong><br />

<strong>multinodular</strong> <strong>goiter</strong>. Horm Metab Res 33:504-507<br />

49. Meller J, Jauho A, Hufner M, Gratz S, Becker W 2000 Disseminated thyroid<br />

autonomy or Graves' disease: reevaluation by a second generation TSH<br />

receptor antibody assay. Thyroid 10:1073-1079<br />

50. Br<strong>and</strong>er AE, Viikinkoski VP, Nickels JI, Kivisaari LM 2000 Importance <strong>of</strong><br />

thyroid abnormalities detected at US screening: a 5-year follow-up. Radiology<br />

215:801-806<br />

51. Alex<strong>and</strong>er EK, Hurwitz S, Heering JP, Benson CB, Frates MC, Doubilet PM,<br />

Cibas ES, Larsen PR, Marqusee E 2003 Natural history <strong>of</strong> benign solid <strong>and</strong><br />

cystic thyroid nodules. Ann Intern Med 138:315-318<br />

52. Kuma K, Matsuzuka F, Kobayashi A, Hirai K, Morita S, Miyauchi A,<br />

Katayama S, Sugawara M 1992 Outcome <strong>of</strong> long st<strong>and</strong>ing solitary thyroid<br />

nodules. World J Surg 16:583-587<br />

53. Kuma K, Matsuzuka F, Yokozawa T, Miyauchi A, Sugawara M 1994 Fate <strong>of</strong><br />

untreated benign thyroid nodules: results <strong>of</strong> long-term follow-up. World J Surg<br />

18:495-498<br />

54. Manz F, Bohmer T, Gartner R, Grossklaus R, Klett M, Schneider R 2002<br />

Quantification <strong>of</strong> iodine supply: representative data on intake <strong>and</strong> urinary<br />

excretion <strong>of</strong> iodine from the German population in 1996. Ann Nutr Metab<br />

46:128-138<br />

44


55. Quadbeck B, Pruellage J, Roggenbuck U, Hirche H, Janssen OE, Mann K,<br />

Hoermann R 2002 Long-term follow-up <strong>of</strong> thyroid nodule growth. Exp Clin<br />

Endocrinol Diabetes 110:348-354<br />

56. Papini E, Petrucci L, Guglielmi R, Panunzi C, Rinaldi R, Bacci V, Crescenzi A,<br />

Nardi F, Fabbrini R, Pacella CM 1998 Long-term changes in nodular <strong>goiter</strong>: a<br />

5-year prospective r<strong>and</strong>omized trial <strong>of</strong> levothyroxine suppressive therapy for<br />

benign cold thyroid nodules. J Clin Endocrinol Metab 83:780-783<br />

57. Wemeau JL, Caron P, Schvartz C, Schlienger JL, Orgiazzi J, Cousty C,<br />

Vlaeminck-Guillem V 2002 Effects <strong>of</strong> thyroid-stimulating hormone suppression<br />

with levothyroxine in reducing the volume <strong>of</strong> solitary thyroid nodules <strong>and</strong><br />

improving extranodular nonpalpable changes: a r<strong>and</strong>omized, double-blind,<br />

placebo-controlled trial by the French Thyroid Research Group. J Clin<br />

Endocrinol Metab 87:4928-4934<br />

58. Bähre M, Hilgers R, Lindemann C, Emrich D 1988 Thyroid autonomy:<br />

sensitive detection in vivo <strong>and</strong> estimation <strong>of</strong> its functional relevance using<br />

quantified high-resolution scintigraphy. Acta Endocrinol (Copenh ) 117:145-<br />

153<br />

59. S<strong>and</strong>rock D, Olbricht T, Emrich D, Benker G, Reinwein D 1993 Long-term<br />

follow-up in patients with autonomous thyroid adenoma. Acta Endocrinol<br />

(Copenh) 128:51-55<br />

60. Elte JW, Bussemaker JK, Haak A 1990 The natural history <strong>of</strong> <strong>euthyroid</strong><br />

<strong>multinodular</strong> goitre. Postgrad Med J 66:186-190<br />

61. Wiener JD 1987 Long-term follow-up in untreated Plummer's disease<br />

(autonomous <strong>goiter</strong>). Clin Nucl Med 12:198-203<br />

62. Emrich D, Erlenmaier U, Pohl M, Luig H 1993 Determination <strong>of</strong> the<br />

autonomously functioning volume <strong>of</strong> the thyroid. Eur J Nucl Med 20:410-414<br />

63. Stanbury JB, Ermans AE, Bourdoux P, Todd C, Oken E, Tonglet R, Vidor G,<br />

Braverman LE, Medeiros-Neto G 1998 Iodine-induced hyperthyroidism:<br />

occurrence <strong>and</strong> epidemiology. Thyroid 8:83-100<br />

64. Reinwein D, Benker G, Konig MP, Pinchera A, Schatz H, Schleusener H 1986<br />

Hyperthyroidism in Europe: clinical <strong>and</strong> laboratory data <strong>of</strong> a prospective<br />

multicentric survey. J Endocrinol Invest 9 Suppl 2:1-36<br />

65. Thomas GA, Williams D, Williams ED 1989 Clonal origin <strong>of</strong> thyroid tumours.<br />

In: Wynford-Thomas D, Williams ED (eds) Thyroid tumors. <strong>Molecular</strong> basis <strong>of</strong><br />

<strong>pathogenesis</strong>.Churchill Livingstone, London38-56<br />

66. Vogelstein B, Fearon ER, Hamilton SR, Preisinger AC, Willard HF, Michelson<br />

AM, Riggs AD, Orkin SH 1987 Clonal analysis using recombinant DNA probes<br />

from the X-chromosome. Cancer Res 47:4806-4813<br />

67. Namba H, Matsuo K, Fagin JA 1990 Clonal composition <strong>of</strong> benign <strong>and</strong><br />

malignant human thyroid tumors. J Clin Invest 86:120-125<br />

45


68. Aeschimann S, Kopp PA, Kimura ET, Zbaeren J, Tobler A, Fey MF, Studer H<br />

1993 Morphological <strong>and</strong> functional polymorphism within clonal thyroid<br />

nodules. J Clin Endocrinol Metab 77:846-851<br />

69. Fey MF, Peter HJ, Hinds HL, Zimmermann A, Liechti-Gallati S, Gerber H,<br />

Studer H, Tobler A 1992 Clonal analysis <strong>of</strong> human tumors with M27 beta, a<br />

highly informative polymorphic X chromosomal probe. J Clin Invest 89:1438-<br />

1444<br />

70. Kopp P, Kimura ET, Aeschimann S, Oestreicher M, Tobler A, Fey MF, Studer<br />

H 1994 Polyclonal <strong>and</strong> monoclonal thyroid nodules coexist within human<br />

<strong>multinodular</strong> <strong>goiter</strong>s. J Clin Endocrinol Metab 79:134-139<br />

71. Hicks DG, LiVolsi VA, Neidich JA, Puck JM, Kant JA 1990 Clonal analysis <strong>of</strong><br />

solitary follicular nodules in the thyroid. Am J Pathol 137:553-562<br />

72. Levy A 2001 Monoclonality <strong>of</strong> endocrine tumours: What does it mean? Trends<br />

Endocrinol Metab 12:301-307<br />

73. Novelli M, Cossu A, Oukrif D, Quaglia A, Lakhani S, Poulsom R, Sasieni P,<br />

Carta P, Contini M, Pasca A, Palmieri G, Bodmer W, T<strong>and</strong>a F, Wright N 2003<br />

X-inactivation patch size in human female tissue confounds the assessment <strong>of</strong><br />

tumor clonality. Proc Natl Acad Sci U S A 100:3311-3314<br />

74. Romert P, Gauguin J 1973 The early development <strong>of</strong> the median thyroid gl<strong>and</strong><br />

<strong>of</strong> the mouse. A light-, electron-microscopic <strong>and</strong> histochemical study. Z Anat<br />

Entwicklungsgesch 139:319-336<br />

75. Lyon MF 1972 X-chromosome inactivation <strong>and</strong> developmental patterns in<br />

mammals. Biol Rev Camb Philos Soc 47:1-35<br />

76. Levy A 2000 Is monoclonality in pituitary adenomas synonymous with<br />

neoplasia? Clin Endocrinol (Oxf) 52:393-397<br />

77. Thomas GA, Williams D, Williams ED 1988 The demonstration <strong>of</strong> tissue<br />

clonality by X-linked enzyme histochemistry. J Pathol 155:101-108<br />

78. Jovanovic L, Delahunt B, McIver B, Eberhardt NL, Grebe SK 2003 Thyroid<br />

gl<strong>and</strong> clonality revisited: the embryonal patch size <strong>of</strong> the normal human<br />

thyroid gl<strong>and</strong> is very large, suggesting X-chromosome inactivation tumor<br />

clonality studies <strong>of</strong> thyroid tumors have to be interpreted with caution. J Clin<br />

Endocrinol Metab 88:3284-3291<br />

79. Many MC, Denef JF, Hamudi S, Cornette C, Haumont S, Beckers C 1986<br />

Effects <strong>of</strong> iodide <strong>and</strong> thyroxine on iodine-deficient mouse thyroid: a<br />

morphological <strong>and</strong> functional study. J Endocrinol 110:203-210<br />

80. Stübner D, Gärtner R, Greil W, Gropper K, Brabant G, Permanetter W, Horn<br />

K, Pickardt CR 1987 Hypertrophy <strong>and</strong> hyperplasia during goitre growth <strong>and</strong><br />

involution in rats - separate bioeffects <strong>of</strong> TSH <strong>and</strong> iodine. Acta Endocrinol<br />

(Copenh ) 116:537-548<br />

46


81. Wynford-Thomas D, Stringer BM, Williams ED 1982 Dissociation <strong>of</strong> growth<br />

<strong>and</strong> function in the rat thyroid during prolonged goitrogen administration.<br />

Acta Endocrinol (Copenh ) 101:210-216<br />

82. Ledent C, Dumont JE, Vassart G, Parmentier M 1992 Thyroid expression <strong>of</strong> an<br />

A2 adenosine receptor transgene induces thyroid hyperplasia <strong>and</strong><br />

hyperthyroidism. EMBO J 11:537-542<br />

83. Ledent C, Franc B, Parmentier M 1998 [Transgenic mouse models. Their<br />

interest in thyroid tumors]. Arch Anat Cytol Pathol 46:31-37<br />

84. Krohn K, Fuhrer D, Holzapfel HP, Paschke R 1998 Clonal origin <strong>of</strong> <strong>toxic</strong><br />

thyroid nodules with constitutively activating thyrotropin receptor mutations. J<br />

Clin Endocrinol Metab 83:130-134<br />

85. Krohn K, Reske A, Ackermann F, Paschke R 2001 Ras mutations are rare in<br />

solitary cold <strong>and</strong> <strong>toxic</strong> thyroid nodules. Clinical Endocrinology 55:241-248<br />

86. Trülzsch B, Krohn K, Wonerow P, Chey S, Holzapfel HP, Ackermann F,<br />

Führer D, Paschke R 2001 Detection <strong>of</strong> thyroid-stimulating hormone receptor<br />

<strong>and</strong> Gsalpha mutations: in 75 <strong>toxic</strong> thyroid nodules by denaturing gradient gel<br />

electrophoresis. J Mol Med 78:684-691<br />

87. Knudson AG 1973 Mutation <strong>and</strong> human cancer. Advances in Cancer Research<br />

17:317-352<br />

88. Laugwitz KL, Allgeier A, Offermanns S, Spicher K, Van S<strong>and</strong>e J, Dumont JE,<br />

Schultz G 1996 The human thyrotropin receptor: a heptahelical receptor<br />

capable <strong>of</strong> stimulating members <strong>of</strong> all four G protein families. Proc Natl Acad<br />

Sci U S A 93:116-120<br />

89. Corvilain B, Laurent E, Lecomte M, Vans<strong>and</strong>e J, Dumont JE 1994 Role <strong>of</strong> the<br />

cyclic adenosine 3',5'-monophosphate <strong>and</strong> the phosphatidylinositol-Ca2+<br />

cascades in mediating the effects <strong>of</strong> thyrotropin <strong>and</strong> iodide on hormone<br />

synthesis <strong>and</strong> secretion in human thyroid slices. J Clin Endocrinol Metab<br />

79:152-159<br />

90. Allgeier A, Laugwitz KL, Van S<strong>and</strong>e J, Schultz G, Dumont JE 1997 Multiple<br />

G-protein coupling <strong>of</strong> the dog thyrotropin receptor. Mol Cell Endocrinol<br />

127:81-90<br />

91. Pisarev MA, DeGroot LJ, Wilber JF 1970 Cyclic-AMP production <strong>of</strong> <strong>goiter</strong>.<br />

Endocrinology 87:339-342<br />

92. Roger PP, Hotimsky A, Moreau C, Dumont JE 1982 Stimulation by<br />

thyrotropin, cholera toxin <strong>and</strong> dibutyryl cyclic AMP <strong>of</strong> the multiplication <strong>of</strong><br />

differentiated thyroid cells in vitro. Mol Cell Endocrinol 26:165-176<br />

93. Wynford-Thomas D, Stringer BM, Harach HR, Williams ED 1983 Control <strong>of</strong><br />

growth in the rat thyroid--an example <strong>of</strong> specific desensitization to trophic<br />

hormone stimulation. Experientia 39:421-423<br />

47


94. Dumont JE, Roger P, Van Heuverswyn B, Erneux C, Vassart G 1984 Control <strong>of</strong><br />

growth <strong>and</strong> differentiation by known intracellular signal molecules in<br />

endocrine tissues: the example <strong>of</strong> the thyroid gl<strong>and</strong>. Adv Cyclic Nucleotide<br />

Protein Phosphorylation Res 17:337-42:337-342<br />

95. Roger P, Taton M, Van S<strong>and</strong>e J, Dumont JE 1988 Mitogenic effects <strong>of</strong><br />

thyrotropin <strong>and</strong> adenosine 3',5'-monophosphate in differentiated normal<br />

human thyroid cells in vitro. J Clin Endocrinol Metab 66:1158-1165<br />

96. Michiels FM, Caillou B, Talbot M, Dessarps-Freichey F, Maunoury MT,<br />

Schlumberger M, Mercken L, Monier R, Feunteun J 1994 Oncogenic potential<br />

<strong>of</strong> guanine nucleotide stimulatory factor alpha subunit in thyroid gl<strong>and</strong>s <strong>of</strong><br />

transgenic mice. Proc Natl Acad Sci U S A 91:10488-10492<br />

97. Zeiger MA, Saji M, Gusev Y, Westra WH, Takiyama Y, Dooley WC, Kohn LD,<br />

Levine MA 1997 Thyroid-specific expression <strong>of</strong> cholera toxin A1 subunit causes<br />

thyroid hyperplasia <strong>and</strong> hyperthyroidism in transgenic mice. Endocrinology<br />

138:3133-3140<br />

98. Nguyen LQ, Kopp P, Martinson F, Stanfield K, Roth SI, Jameson JL 2000 A<br />

dominant negative CREB (cAMP response element-binding protein) is<strong>of</strong>orm<br />

inhibits thyrocyte growth, thyroid-specific gene expression, differentiation, <strong>and</strong><br />

function. Mol Endocrinol 14:1448-1461<br />

99. Postiglione MP, Parlato R, Rodriguez-Mallon A, Rosica A, Mithbaokar P,<br />

Maresca M, Marians RC, Davies TF, Zannini MS, De Felice M, Di Lauro R<br />

2002 Role <strong>of</strong> the thyroid-stimulating hormone receptor signaling in<br />

development <strong>and</strong> differentiation <strong>of</strong> the thyroid gl<strong>and</strong>. Proc Natl Acad Sci U S A<br />

99:15462-15467<br />

100. Marians RC, Ng L, Blair HC, Unger P, Graves PN, Davies TF 2002 Defining<br />

thyrotropin-dependent <strong>and</strong> -independent steps <strong>of</strong> thyroid hormone synthesis by<br />

using thyrotropin receptor-null mice. Proc Natl Acad Sci U S A 99:15776-15781<br />

101. Parma J, Duprez L, Van S<strong>and</strong>e J, Cochaux P, Gervy C, Mockel J, Dumont J,<br />

Vassart G 1993 Somatic mutations in the thyrotropin receptor gene cause<br />

hyperfunctioning thyroid adenomas. Nature 365:649-651<br />

102. Führer D, Holzapfel HP, Wonerow P, Scherbaum WA, Paschke R 1997<br />

Somatic mutations in the thyrotropin receptor gene <strong>and</strong> not in the Gs alpha<br />

protein gene in 31 <strong>toxic</strong> thyroid nodules. J Clin Endocrinol Metab 82:3885-3891<br />

103. Führer D, Kubisch C, Scheibler U, Lamesch P, Krohn K, Paschke R 1998 The<br />

extracellular thyrotropin receptor domain is not a major c<strong>and</strong>idate for<br />

mutations in <strong>toxic</strong> thyroid nodules. Thyroid 8:997-1001<br />

104. Lyons J, L<strong>and</strong>is CA, Harsh G, Vallar L, Grunewald K, Feichtinger H, Duh QY,<br />

Clark OH, Kawasaki E, Bourne HR 1990 Two G protein oncogenes in human<br />

endocrine tumors. Science 249:655-659<br />

105. O'Sullivan C, Barton CM, Staddon SL, Brown CL, Lemoine NR 1991<br />

Activating point mutations <strong>of</strong> the gsp oncogene in human thyroid adenomas.<br />

Mol Carcinog 4:345-349<br />

48


106. Parma J, Duprez L, Van S<strong>and</strong>e J, Hermans J, Rocmans P, Van Vliet G,<br />

Costagliola S, Rodien P, Dumont JE, Vassart G 1997 Diversity <strong>and</strong> prevalence<br />

<strong>of</strong> somatic mutations in the thyrotropin receptor <strong>and</strong> Gs alpha genes as a cause<br />

<strong>of</strong> <strong>toxic</strong> thyroid adenomas. J Clin Endocrinol Metab 82:2695-2701<br />

107. Paschke R, Tonacchera M, Van S<strong>and</strong>e J, Parma J, Vassart G 1994<br />

Identification <strong>and</strong> functional characterization <strong>of</strong> two new somatic mutations<br />

causing constitutive activation <strong>of</strong> the thyrotropin receptor in hyperfunctioning<br />

autonomous adenomas <strong>of</strong> the thyroid. J Clin Endocrinol Metab 79:1785-1789<br />

108. Porcellini A, Ciullo I, Laviola L, Amabile G, Fenzi G, Avvedimento VE 1994<br />

Novel mutations <strong>of</strong> thyrotropin receptor gene in thyroid hyperfunctioning<br />

adenomas. Rapid identification by fine needle aspiration biopsy. J Clin<br />

Endocrinol Metab 79:657-661<br />

109. Russo D, Arturi F, Wicker R, Chazenbalk GD, Schlumberger M, DuVillard JA,<br />

Caillou B, Monier R, Rapoport B, Filetti S 1995 Genetic alterations in thyroid<br />

hyperfunctioning adenomas. J Clin Endocrinol Metab 80:1347-1351<br />

110. Russo D, Arturi F, Suarez HG, Schlumberger M, Du VJ, Crocetti U, Filetti S<br />

1996 Thyrotropin receptor gene alterations in thyroid hyperfunctioning<br />

adenomas. J Clin Endocrinol Metab 81:1548-1551<br />

111. Vanvooren V, Uchino S, Duprez L, Costa MJ, V<strong>and</strong>ekerckhove J, Parma J,<br />

Vassart G, Dumont JE, Van S<strong>and</strong>e J, Noguchi S 2002 Oncogenic mutations in<br />

the thyrotropin receptor <strong>of</strong> autonomously functioning thyroid nodules in the<br />

Japanese population. Eur J Endocrinol 147:287-291<br />

112. Georgopoulos NA, Sykiotis GP, Sgourou A, Papachatzopoulou A, Markou KB,<br />

Kyriazopoulou V, Papavassiliou AG, Vagenakis AG 2003 Autonomously<br />

functioning thyroid nodules in a former iodine-deficient area commonly harbor<br />

gain-<strong>of</strong>-function mutations in the thyrotropin signaling pathway. Eur J<br />

Endocrinol 149:287-292<br />

113. Arturi F, Scarpelli D, Coco A, Sacco R, Bruno R, Filetti S, Russo D 2003<br />

Thyrotropin receptor mutations <strong>and</strong> thyroid hyperfunctioning adenomas ten<br />

years after their first discovery: unresolved questions. Thyroid 13:341-343<br />

114. Vassart G 2004 Activating mutations <strong>of</strong> the TSH receptor. Thyroid 14:86-87<br />

115. Garcia-Delgado M, Gonzalez-Navarro CJ, Napal MC, Baldonado C, Vizmanos<br />

JL, Gullon A 1998 Higher sensitivity <strong>of</strong> denaturing gradient gel electrophoresis<br />

than sequencing in the detection <strong>of</strong> mutations in DNA from tumor samples.<br />

Biotechniques 24:72, 74, 76<br />

116. Fischer SG, Lerman LS 1983 DNA fragments differing by single base-pair<br />

substitutions are separated in denaturing gradient gels: correspondence with<br />

melting theory. Proc Natl Acad Sci U S A 80:1579-1583<br />

117. Trülzsch B, Krohn K, Wonerow P, Paschke R 1999 DGGE is more sensitive for<br />

the detection <strong>of</strong> somatic point mutations than direct sequencing. Biotechniques<br />

27:266-268<br />

49


118. Eszlinger M, Krohn K, Frenzel R, Kropf S, Tonjes A, Paschke R 2004 Gene<br />

expression analysis reveals evidence for inactivation <strong>of</strong> the TGF-beta signaling<br />

cascade in autonomously functioning thyroid nodules. Oncogene 23:795-804<br />

119. Samuels-Lev Y, O'Connor DJ, Bergamaschi D, Trigiante G, Hsieh JK, Zhong<br />

S, Campargue I, Naumovski L, Crook T, Lu X 2001 ASPP proteins specifically<br />

stimulate the apoptotic function <strong>of</strong> p53. Mol Cell 8:781-794<br />

120. Deleu S, Allory Y, Radulescu A, Pirson I, Carrasco N, Corvilain B, Salmon I,<br />

Franc B, Dumont JE, Van S<strong>and</strong>e J, Maenhaut C 2000 Characterization <strong>of</strong><br />

autonomous thyroid adenoma: metabolism, gene expression, <strong>and</strong> pathology.<br />

Thyroid 10:131-140<br />

121. Mezosi E, Yamazaki H, Bretz JD, Wang SH, Arscott PL, Utsugi S, Gauger PG,<br />

Thompson NW, Baker JR, Jr. 2002 Aberrant apoptosis in thyroid epithelial<br />

cells from <strong>goiter</strong> nodules. J Clin Endocrinol Metab 87:4264-4272<br />

122. Watson N, Linder ME, Druey KM, Kehrl JH, Blumer KJ 1996 RGS family<br />

members: GTPase-activating proteins for heterotrimeric G-protein alphasubunits.<br />

Nature 383:172-175<br />

123. Freedman NJ, Lefkowitz RJ 1996 Desensitization <strong>of</strong> G protein-coupled<br />

receptors. Recent Prog Horm Res 51:319-353<br />

124. Führer D, Mix M, Wonerow P, Richter I, Willgerodt H, Paschke R 1999<br />

Variable phenotype associated with Ser505Asn-activating thyrotropin-receptor<br />

germline mutation. Thyroid 9:757-761<br />

125. Voigt C, Holzapfel H, Paschke R 2000 Expression <strong>of</strong> ß-arrestins in <strong>toxic</strong> <strong>and</strong><br />

cold thyroid nodules. FEBS Lett 486:208-212<br />

126. Voigt C, Holzapfel HP, Meyer S, Paschke R 2004 Increased expression <strong>of</strong> Gprotein<br />

coupled receptor kinases 3 <strong>and</strong> 4 in hyperfunctioning thyroid nodules. J<br />

Endocrinol 182:173-182<br />

127. Biebermann H, Schoneberg T, Schulz A, Krause G, Gruters A, Schultz G,<br />

Gudermann T 1998 A conserved tyrosine residue (Y601) in transmembrane<br />

domain 5 <strong>of</strong> the human thyrotropin receptor serves as a molecular switch to<br />

determine G-protein coupling. FASEB J 12:1461-1471<br />

128. Park Y, Park E, Kim M, Kim T, Lee H, Lee S, Jang I, Shong M, Park D, Cho B<br />

2002 Involvement <strong>of</strong> the protein kinase C pathway in thyrotropin-induced<br />

STAT3 activation in FRTL-5 thyroid cells. Mol Cell Endocrinol 194:77<br />

129. Park ES, Kim H, Suh JM, Park SJ, You SH, Chung HK, Lee KW, Kwon OY,<br />

Cho BY, Kim YK, Ro HK, Chung J, Shong M 2000 Involvement <strong>of</strong> JAK/STAT<br />

(Janus kinase/signal transducer <strong>and</strong> activator <strong>of</strong> transcription) in the<br />

thyrotropin signaling pathway. Mol Endocrinol 14:662-670<br />

130. Dumont JE, Maenhaut C, Pirson I, Baptist M, Roger PP 1991 Growth factors<br />

controlling the thyroid gl<strong>and</strong>. Baillieres Clin Endocrinol Metab 5:727-754<br />

50


131. Van S<strong>and</strong>e J, Parma J, Tonacchera M, Swillens S, Dumont J, Vassart G 1995<br />

Somatic <strong>and</strong> germline mutations <strong>of</strong> the TSH receptor gene in thyroid diseases. J<br />

Clin Endocrinol Metab 80:2577-2585<br />

132. Brenner-Gati L, Berg KA, Gershengorn MC 1989 Insulin-like growth factor-I<br />

potentiates thyrotropin stimulation <strong>of</strong> adenylyl cyclase in FRTL-5 cells.<br />

Endocrinology 125:1315-1320<br />

133. Dugrillon A, Bechtner G, Uedelhoven WM, Weber PC, Gärtner R 1990<br />

Evidence that an iodolactone mediates the inhibitory effect <strong>of</strong> iodide on thyroid<br />

cell proliferation but not on adenosine 3',5'-monophosphate formation.<br />

Endocrinology 127:337-343<br />

134. Gärtner R 1992 Thyroid growth in vitro. Exp Clin Endocrinol 100:32-35<br />

135. Roger PP, Servais P, Dumont JE 1983 Stimulation by thyrotropin <strong>and</strong> cyclic<br />

AMP <strong>of</strong> the proliferation <strong>of</strong> quiescent canine thyroid cells cultured in a defined<br />

medium containing insulin. FEBS Lett 157:323-329<br />

136. Smith P, Wynford-Thomas D, Stringer BM, Williams ED 1986 Growth factor<br />

control <strong>of</strong> rat thyroid follicular cell proliferation. Endocrinology 119:1439-1445<br />

137. Eggo MC, Bachrach LK, Burrow GN 1990 Interaction <strong>of</strong> TSH, insulin <strong>and</strong><br />

insulin-like growth factors in regulating thyroid growth <strong>and</strong> function. Growth<br />

Factors 2:99-109<br />

138. Gärtner R, Schopohl D, Schaefer S, Dugrillon A, Erdmann A, Toda S, Bechtner<br />

G 1997 Regulation <strong>of</strong> transforming growth factor beta 1 messenger ribonucleic<br />

acid expression in porcine thyroid follicles in vitro by growth factors, iodine, or<br />

delta-iodolactone. Thyroid 7:633-640<br />

139. Pang XP, Park M, Hershman JM 1992 Transforming growth factor-beta blocks<br />

protein kinase-A-mediated iodide transport <strong>and</strong> protein kinase-C-mediated<br />

DNA synthesis in FRTL-5 rat thyroid cells. Endocrinology 131:45-50<br />

140. Taton M, Lamy F, Roger PP, Dumont JE 1993 General inhibition by<br />

transforming growth factor beta 1 <strong>of</strong> thyrotropin <strong>and</strong> cAMP responses in<br />

human thyroid cells in primary culture. Mol Cell Endocrinol 95:13-21<br />

141. Colletta G, Cirafici AM, Di Carlo A 1989 Dual effect <strong>of</strong> transforming growth<br />

factor beta on rat thyroid cells: inhibition <strong>of</strong> thyrotropin-induced proliferation<br />

<strong>and</strong> reduction <strong>of</strong> thyroid-specific differentiation markers. Cancer Res 49:3457-<br />

3462<br />

142. Depoortere F, Pirson I, Bartek J, Dumont JE, Roger PP 2000 Transforming<br />

growth factor beta(1) selectively inhibits the cyclic AMP-dependent<br />

proliferation <strong>of</strong> primary thyroid epithelial cells by preventing the association <strong>of</strong><br />

cyclin D3-cdk4 with nuclear p27(kip1). Mol Biol Cell 11:1061-1076<br />

143. Grubeck-Loebenstein B, Buchan G, Sadeghi R, Kissonerghis M, Londei M,<br />

Turner M, Pirich K, Roka R, Niederle B, Kassal H 1989 Transforming growth<br />

factor beta regulates thyroid growth. Role in the <strong>pathogenesis</strong> <strong>of</strong> non<strong>toxic</strong><br />

<strong>goiter</strong>. J Clin Invest 83:764-770<br />

51


144. Tsushima T, Arai M, Saji M, Ohba Y, Murakami H, Ohmura E, Sato K,<br />

Shizume K 1988 Effects <strong>of</strong> transforming growth factor-beta on<br />

deoxyribonucleic acid synthesis <strong>and</strong> iodine metabolism in porcine thyroid cells<br />

in culture. Endocrinology 123:1187-1194<br />

145. Krohn K, Emmrich P, Ott N, Paschke R 1999 Increased thyroid epithelial cell<br />

proliferation in <strong>toxic</strong> thyroid nodules. Thyroid 9:241-246<br />

146. Eggo MC, King WJ, Black EG, Sheppard MC 1996 Functional human thyroid<br />

cells <strong>and</strong> their insulin-like growth factor-binding proteins: regulation by<br />

thyrotropin, cyclic 3',5' adenosine monophosphate, <strong>and</strong> growth factors. J Clin<br />

Endocrinol Metab 81:3056-3062<br />

147. Eszlinger M, Krohn K, Paschke R 2001 Complementary DNA expression array<br />

analysis suggests a lower expression <strong>of</strong> signal transduction proteins <strong>and</strong><br />

receptors in cold <strong>and</strong> hot thyroid nodules. J Clin Endocrinol Metab 86:4834-<br />

4842<br />

148. Derwahl M, Studer H 2001 Nodular <strong>goiter</strong> <strong>and</strong> <strong>goiter</strong> nodules: Where iodine<br />

deficiency falls short <strong>of</strong> explaining the facts. Exp Clin Endocrinol Diabetes<br />

109:250-260<br />

149. Studer H, Peter HJ, Gerber H 1989 Natural heterogeneity <strong>of</strong> thyroid cells: the<br />

basis for underst<strong>and</strong>ing thyroid function <strong>and</strong> nodular <strong>goiter</strong> growth. Endocr<br />

Rev 10:125-135<br />

150. Ponder BA 2001 Cancer genetics. Nature 411:336-341<br />

151. Albertson DG, Collins C, McCormick F, Gray JW 2003 Chromosome<br />

aberrations in solid tumors. Nat Genet 34:369-376<br />

152. Gimm O 2001 Thyroid cancer. Cancer Lett 163:143-156<br />

153. Esapa CT, Johnson SJ, Kendall-Taylor P, Lennard TW, Harris PE 1999<br />

Prevalence <strong>of</strong> Ras mutations in thyroid neoplasia. Clin Endocrinol (Oxf )<br />

50:529-535<br />

154. Bos JL 1989 ras oncogenes in human cancer: a review [published erratum<br />

appears in Cancer Res 1990 Feb 15;50(4):1352]. Cancer Res 49:4682-4689<br />

155. Wynford-Thomas D 1997 Origin <strong>and</strong> progression <strong>of</strong> thyroid epithelial tumours:<br />

cellular <strong>and</strong> molecular mechanisms. Horm Res 47:145-157<br />

156. Kim DS, McCabe CJ, Buchanan MA, Watkinson JC 2003 Oncogenes in thyroid<br />

cancer. Clin Otolaryngol 28:386-395<br />

157. Paschke R, Neumann S 2001 Sodium/iodide symporter mRNA expression in<br />

cold thyroid nodules. Exp Clin Endocrinol Diabetes 109:45-46<br />

158. Dohan O, Baloch Z, Banrevi Z, LiVolsi V, Carrasco N 2001 Rapid<br />

communication: predominant intracellular overexpression <strong>of</strong> the Na(+)/I(-)<br />

symporter (NIS) in a large sampling <strong>of</strong> thyroid cancer cases. J Clin Endocrinol<br />

Metab 86:2697-2700<br />

52


159. Dohan O, De L, V, Paroder V, Riedel C, Artani M, Reed M, Ginter CS,<br />

Carrasco N 2003 The sodium/iodide Symporter (NIS): characterization,<br />

regulation, <strong>and</strong> medical significance. Endocr Rev 24:48-77<br />

160. Neumann S, Schuchardt K, Reske A, Reske A, Emmrich P, Paschke R 2004<br />

Lack <strong>of</strong> correlation for sodium iodide symporter mRNA <strong>and</strong> protein expression<br />

<strong>and</strong> analysis <strong>of</strong> sodium iodide symporter promoter methylation in benign cold<br />

thyroid nodules. Thyroid 14:99-111<br />

161. Joba W, Spitzweg C, Schriever K, Heufelder AE 1999 Analysis <strong>of</strong> human<br />

sodium/iodide symporter, thyroid transcription factor-1, <strong>and</strong> paired-boxprotein-8<br />

gene expression in benign thyroid diseases. Thyroid 9:455-466<br />

162. Russo D, Bulotta S, Bruno R, Arturi F, Giannasio P, Derwahl M, Bidart JM,<br />

Schlumberger M, Filetti S 2001 Sodium/iodide symporter (NIS) <strong>and</strong> pendrin<br />

are expressed differently in hot <strong>and</strong> cold nodules <strong>of</strong> thyroid <strong>toxic</strong> <strong>multinodular</strong><br />

<strong>goiter</strong>. Eur J Endocrinol 145:591-597<br />

163. Lazar V, Bidart JM, Caillou B, Mahe C, Lacroix L, Filetti S, Schlumberger M<br />

1999 Expression <strong>of</strong> the Na+/I- symporter gene in human thyroid tumors: a<br />

comparison study with other thyroid-specific genes. J Clin Endocrinol Metab<br />

84:3228-3234<br />

164. Knauf JA, Kuroda H, Basu S, Fagin JA 2003 RET/PTC-induced<br />

dedifferentiation <strong>of</strong> thyroid cells is mediated through Y1062 signaling through<br />

SHC-RAS-MAP kinase. Oncogene 22:4406-4412<br />

165. Trapasso F, Iuliano R, Chiefari E, Arturi F, Stella A, Filetti S, Fusco A, Russo<br />

D 1999 Iodide symporter gene expression in normal <strong>and</strong> transformed rat<br />

thyroid cells. Eur J Endocrinol 140:447-451<br />

166. Tong Q, Ryu KY, Jhiang SM 1997 Promoter characterization <strong>of</strong> the rat Na+/I-<br />

symporter gene. Biochem Biophys Res Commun 239:34-41<br />

167. Spitzweg C, Morris JC 2002 The sodium iodide symporter: its<br />

pathophysiological <strong>and</strong> therapeutic implications. Clin Endocrinol (Oxf) 57:559-<br />

574<br />

168. Paschke R, Neumann S. 2001 Clinical, epidemiologic <strong>and</strong> molecular evidence<br />

for a genetic etiology <strong>of</strong> a subset <strong>of</strong> <strong>euthyroid</strong> <strong>goiter</strong>s. In: Duntas LH (ed)<br />

Bronchocele Goiter 2000. Goitrogenesis upon the advent <strong>of</strong> the new<br />

millenium.BETA Medical Publishers Ltd., Athens43-49<br />

169. Tonacchera M, Viacava P, Agretti P, de Marco G, Perri A, di Cosmo C, De<br />

Servi M, Miccoli P, Lippi F, Naccarato AG, Pinchera A, Chiovato L, Vitti P<br />

2002 Benign nonfunctioning thyroid adenomas are characterized by a defective<br />

targeting to cell membrane or a reduced expression <strong>of</strong> the sodium iodide<br />

symporter protein. J Clin Endocrinol Metab 87:352-357<br />

170. Dunn JT, Dunn AD 2001 Update on intrathyroidal iodine metabolism. Thyroid<br />

11:407-414<br />

53


171. Caillou B, Dupuy C, Lacroix L, Nocera M, Talbot M, Ohayon R, Deme D,<br />

Bidart JM, Schlumberger M, Virion A 2001 Expression <strong>of</strong> reduced<br />

nicotinamide adenine dinucleotide phosphate oxidase (ThoX, LNOX, Duox)<br />

genes <strong>and</strong> proteins in human thyroid tissues. J Clin Endocrinol Metab 86:3351-<br />

3358<br />

172. De Vijlder JJ 2003 Primary congenital hypothyroidism: defects in iodine<br />

pathways. Eur J Endocrinol 149:247-256<br />

173. Gärtner R, Dugrillon A, Bechtner G 1990 Evidence that thyroid growth<br />

autoregulation is mediated by an iodolactone. Acta Med Austriaca 17 Suppl<br />

1:24-6:24-26<br />

174. Pisarev MA, Krawiec L, Juvenal GJ, Bocanera LV, Pregliasco LB, Sartorio G,<br />

Chester HA 1994 Studies on the <strong>goiter</strong> inhibiting action <strong>of</strong> iodolactones. Eur J<br />

Pharmacol 258:33-37<br />

175. Krohn K, Paschke R 2001 Loss <strong>of</strong> heterozygocity at the thyroid peroxidase gene<br />

locus in solitary cold thyroid nodules. Thyroid 11:741-747<br />

176. Yuen ST, Davies H, Chan TL, Ho JW, Bignell GR, Cox C, Stephens P, Edkins<br />

S, Tsui WW, Chan AS, Futreal PA, Stratton MR, Wooster R, Leung SY 2002<br />

Similarity <strong>of</strong> the phenotypic patterns associated with BRAF <strong>and</strong> KRAS<br />

mutations in colorectal neoplasia. Cancer Res 62:6451-6455<br />

177. Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE, Fagin JA 2003<br />

High prevalence <strong>of</strong> BRAF mutations in thyroid cancer: genetic evidence for<br />

constitutive activation <strong>of</strong> the RET/PTC-RAS-BRAF signaling pathway in<br />

papillary thyroid carcinoma. Cancer Res 63:1454-1457<br />

178. Krohn K, Paschke R 2004 BRAF Mutations Are Not an Alternative<br />

Explanation for the <strong>Molecular</strong> Etiology <strong>of</strong> ras -Mutation Negative Cold<br />

Thyroid Nodules. Thyroid 14:359-361<br />

179. Xing M, Vasko V, Tallini G, Larin A, Wu G, Udelsman R, Ringel MD,<br />

Ladenson PW, Sidransky D 2004 BRAF T1796A transversion mutation in<br />

various thyroid neoplasms. J Clin Endocrinol Metab 89:1365-1368<br />

180. Puxeddu E, Moretti S, Elisei R, Romei C, Pascucci R, Martinelli M, Marino C,<br />

Avenia N, Rossi ED, Fadda G, Cavaliere A, Ribacchi R, Falorni A, Pontecorvi<br />

A, Pacini F, Pinchera A, Santeusanio F 2004 BRAF(V599E) mutation is the<br />

leading genetic event in adult sporadic papillary thyroid carcinomas. J Clin<br />

Endocrinol Metab 89:2414-2420<br />

181. Soares P, Trovisco V, Rocha AS, Lima J, Castro P, Preto A, Maximo V, Botelho<br />

T, Seruca R, Sobrinho-Simoes M 2003 BRAF mutations <strong>and</strong> RET/PTC<br />

rearrangements are alternative events in the etio<strong>pathogenesis</strong> <strong>of</strong> PTC.<br />

Oncogene 22:4578-4580<br />

182. Esapa C, Foster S, Johnson S, Jameson JL, Kendall-Taylor P, Harris PE 1997<br />

G protein <strong>and</strong> thyrotropin receptor mutations in thyroid neoplasia. J Clin<br />

Endocrinol Metab 82:493-496<br />

54


183. Ringel MD, Saji M, Schwindinger WF, Segev D, Zeiger MA, Levine MA 1998<br />

Absence <strong>of</strong> activating mutations <strong>of</strong> the genes encoding the alpha-subunits <strong>of</strong><br />

G11 <strong>and</strong> Gq in thyroid neoplasia. J Clin Endocrinol Metab 83:554-559<br />

184. Vanvooren V, Allgeier A, Nguyen M, Massart C, Parma J, Dumont JE, Van<br />

S<strong>and</strong>e J 2001 Mutation analysis <strong>of</strong> the Epac--Rap1 signaling pathway in cold<br />

thyroid follicular adenomas. Eur J Endocrinol 144:605-610<br />

185. Barden CB, Shister KW, Zhu B, Guiter G, Greenblatt DY, Zeiger MA, Fahey<br />

TJ, III 2003 Classification <strong>of</strong> follicular thyroid tumors by molecular signature:<br />

results <strong>of</strong> gene pr<strong>of</strong>iling. Clin Cancer Res 9:1792-1800<br />

186. Huang Y, Prasad M, Lemon WJ, Hampel H, Wright FA, Kornacker K, LiVolsi<br />

V, Frankel W, Kloos RT, Eng C, Pellegata NS, de la CA 2001 Gene expression<br />

in papillary thyroid carcinoma reveals highly consistent pr<strong>of</strong>iles. Proc Natl<br />

Acad Sci U S A 98:15044-15049<br />

187. Aldred MA, Ginn-Pease ME, Morrison CD, Popkie AP, Gimm O, Hoang-Vu C,<br />

Krause U, Dralle H, Jhiang SM, Plass C, Eng C 2003 Caveolin-1 <strong>and</strong> caveolin-<br />

2,together with three bone morphogenetic protein-related genes, may encode<br />

novel tumor suppressors down-regulated in sporadic follicular thyroid<br />

carcinogenesis. Cancer Res 63:2864-2871<br />

188. Xu J, Moatamed F, Caldwell JS, Walker JR, Kraiem Z, Taki K, Brent GA,<br />

Hershman JM 2003 Enhanced expression <strong>of</strong> nicotinamide N-methyltransferase<br />

in human papillary thyroid carcinoma cells. J Clin Endocrinol Metab 88:4990-<br />

4996<br />

189. Krohn K, Stricker I, Emmrich P, Paschke R 2003 Cold thyroid nodules show a<br />

marked increase <strong>of</strong> proliferation markers. Thyroid 13:569-576<br />

190. Stoler DL, Datta RV, Charles MA, Block AW, Brenner BM, Sieczka EM, Hicks<br />

WL, Jr., Loree TR, Anderson GR 2002 Genomic instability measurement in the<br />

diagnosis <strong>of</strong> thyroid neoplasms. Head Neck 24:290-295<br />

191. Dobosz T, Lukienczuk T, Sasiadek M, Kuczy inverted question mA, Jankowska<br />

E, Blin N 2000 Microsatellite instability in thyroid papillary carcinoma <strong>and</strong><br />

<strong>multinodular</strong> hyperplasia. Oncology 58:305-310<br />

192. Lang W, Georgii A, Stauch G, Kienzle E 1980 The differentiation <strong>of</strong> atypical<br />

adenomas <strong>and</strong> encapsulated follicular carcinomas in the thyroid gl<strong>and</strong>.<br />

Virchows Arch A Pathol Anat Histol 385:125-141<br />

193. Bol S, Belge G, Thode B, Bartnitzke S, Bullerdiek J 1999 Structural<br />

abnormalities <strong>of</strong> chromosome 2 in benign thyroid tumors. Three new cases <strong>and</strong><br />

review <strong>of</strong> the literature. Cancer Genet Cytogenet 114:75-77<br />

194. Rippe V, Drieschner N, Meiboom M, Murua EH, Bonk U, Belge G, Bullerdiek J<br />

2003 Identification <strong>of</strong> a gene rearranged by 2p21 aberrations in thyroid<br />

adenomas. Oncogene 22:6111-6114<br />

55


195. Ward LS, Brenta G, Medvedovic M, Fagin JA 1998 Studies <strong>of</strong> allelic loss in<br />

thyroid tumors reveal major differences in chromosomal instability between<br />

papillary <strong>and</strong> follicular carcinomas. J Clin Endocrinol Metab 83:525-530<br />

196. Kroll TG, Sarraf P, Pecciarini L, Chen CJ, Mueller E, Spiegelman BM,<br />

Fletcher JA 2000 PAX8-PPARgamma1 fusion oncogene in human thyroid<br />

carcinoma. Science 289:1357-1360<br />

197. Marques AR, Espadinha C, Catarino AL, Moniz S, Pereira T, Sobrinho LG,<br />

Leite V 2002 Expression <strong>of</strong> PAX8-PPAR gamma 1 rearrangements in both<br />

follicular thyroid carcinomas <strong>and</strong> adenomas. J Clin Endocrinol Metab 87:3947-<br />

3952<br />

198. Nikiforova MN, Lynch RA, Biddinger PW, Alex<strong>and</strong>er EK, Dorn GW, Tallini<br />

G, Kroll TG, Nikiforov YE 2003 RAS point mutations <strong>and</strong> PAX8-PPAR<br />

gamma rearrangement in thyroid tumors: evidence for distinct molecular<br />

pathways in thyroid follicular carcinoma. J Clin Endocrinol Metab 88:2318-<br />

2326<br />

199. Dwight T, Thoppe SR, Foukakis T, Lui WO, Wallin G, Hoog A, Frisk T,<br />

Larsson C, Zedenius J 2003 Involvement <strong>of</strong> the PAX8/peroxisome proliferatoractivated<br />

receptor gamma rearrangement in follicular thyroid tumors. J Clin<br />

Endocrinol Metab 88:4440-4445<br />

200. Nikiforova MN, Biddinger PW, Caudill CM, Kroll TG, Nikiforov YE 2002<br />

PAX8-PPARgamma rearrangement in thyroid tumors: RT-PCR <strong>and</strong><br />

immunohistochemical analyses. Am J Surg Pathol 26:1016-1023<br />

201. Wiersinga WM 1992 Determinants <strong>of</strong> outcome in sporadic non<strong>toxic</strong> <strong>goiter</strong>.<br />

Thyroidology 4:41-43<br />

202. V<strong>and</strong>erpump MPJ, Tunbridge WMG 1996 The epidemiology <strong>of</strong> thyroid<br />

diseases. In: Braverman LE, Utiger R (eds) The Thyroid.Lippincott,<br />

Philadelphia474-482<br />

203. Denef JF, Haumont S, Cornette C, Beckers C 1981 Correlated functional <strong>and</strong><br />

morphometric study <strong>of</strong> thyroid hyperplasia induced by iodine deficiency.<br />

Endocrinology 108:2352-2358<br />

204. Van Middlesworth L 1986 T-2 mycotoxin intensifies iodine deficiency in mice<br />

fed low iodine diet. Endocrinology 118:583-586<br />

205. Raspe E, Dumont JE 1995 Tonic modulation <strong>of</strong> dog thyrocyte H2O2 generation<br />

<strong>and</strong> I- uptake by thyrotropin through the cyclic adenosine 3',5'-monophosphate<br />

cascade. Endocrinology 136:965-973<br />

206. Wiseman H, Halliwell B 1996 Damage to DNA by reactive oxygen <strong>and</strong> nitrogen<br />

species: role in inflammatory disease <strong>and</strong> progression to cancer. Biochem J<br />

313:17-29<br />

207. Abs R, Stevenaert A, Beckers A 1994 Autonomously functioning thyroid<br />

nodules in a patient with a thyrotropin-secreting pituitary adenoma: possible<br />

cause--effect relationship. Eur J Endocrinol 131:355-358<br />

56


208. Studer H, Huber G, Derwahl M, Frey P 1989 [The transformation <strong>of</strong> Basedow's<br />

struma into nodular <strong>goiter</strong>: a reason for recurrence <strong>of</strong> hyperthyroidism].<br />

Schweiz Med Wochenschr 119:203-208<br />

209. Cheung NW, Boyages SC 1997 The thyroid gl<strong>and</strong> in acromegaly: an<br />

ultrasonographic study. Clin Endocrinol (Oxf ) 46:545-549<br />

210. Dumont JE, Ermans AM, Maenhaut C, Coppee F, Stanbury JB 1995 Large<br />

goitre as a maladaptation to iodine deficiency. Clin Endocrinol (Oxf ) 43:1-10<br />

211. Hadjidakis S, Koutras DA, Daikos G 1964 Endemic goitre in greece: Family<br />

studies. J Med Genet 82:82-87<br />

212. Malamos B, Koutras DA, Kostamis P, Kralios AC, Rigopoulos G, Zerefos N<br />

1966 Endemic <strong>goiter</strong> in Greece: epidemiologic <strong>and</strong> genetic studies. J Clin<br />

Endocrinol Metab 26:688-695<br />

213. Malamos B, Koutras DA, Kostamis P 1967 Endemic goitre in greece: A study <strong>of</strong><br />

379 twin pairs. J Med Genet 4:16-18<br />

214. Tajtakova M, Langer P, Gonsorcikova V, Bohov, Hancinova D 1998<br />

Recognition <strong>of</strong> a subgroup <strong>of</strong> adolescents with rapidly growing thyroids under<br />

iodine-replete conditions: seven year follow-up. Eur J Endocrinol 138:674-680<br />

215. Abuye C, Omwega AM, Imungi JK 1999 Familial tendency <strong>and</strong> dietary<br />

association <strong>of</strong> goitre in Gamo-G<strong>of</strong>a, Ethiopia. East Afr Med J 76:447-451<br />

216. Langer P, Tajtakova M, Bohov P, Klimes I 1999 Possible role <strong>of</strong> genetic factors<br />

in thyroid growth rate <strong>and</strong> in the assessment <strong>of</strong> upper limit <strong>of</strong> normal thyroid<br />

volume in iodine-replete adolescents. Thyroid 9:557-562<br />

217. Doufas AG, Mastorakos G, Chatziioannou S, Tseleni-Balafouta S, Piperingos<br />

G, Boukis MA, Mantzos E, Caraiskos CS, Mantzos J, Alevizaki M, Koutras DA<br />

1999 The predominant form <strong>of</strong> non-<strong>toxic</strong> <strong>goiter</strong> in Greece is now autoimmune<br />

thyroiditis. Eur J Endocrinol 140:505-511<br />

218. Heimann P 1966 Familial incidence <strong>of</strong> thyroid disease <strong>and</strong> anamnestic<br />

incidence <strong>of</strong> pubertal struma in 449 consecutive struma patients. Acta Med<br />

Sc<strong>and</strong> 179:113-119<br />

219. Greig WR, Boyle JA, Duncan A, Nicol J, Gray MJ, Buchanan WW, McGirr<br />

EM 1967 Genetic <strong>and</strong> non-genetic factors in simple goitre formation: Evidence<br />

from a twin study. Q J Med 142:175-188<br />

220. Brix TH, Kyvik KO, Hegedus L 1999 Major role <strong>of</strong> genes in the etiology <strong>of</strong><br />

simple <strong>goiter</strong> in females: a population-based twin study. J Clin Endocrinol<br />

Metab 84:3071-3075<br />

221. Medeiros-Neto G, Targovnik H, Knobel M, Propato F, Varela V, Alkmin M,<br />

Barbosa S, Wajchenberg BL 1989 Qualitative <strong>and</strong> quantitative defects <strong>of</strong><br />

thyroglobulin resulting in congenital <strong>goiter</strong>. Absence <strong>of</strong> gross gene deletion <strong>of</strong><br />

coding sequences in the TG gene structure. J Endocrinol Invest 12:805-813<br />

57


222. Targovnik HM, Cochaux P, Corach D, Vassart G 1992 Identification <strong>of</strong> a minor<br />

Tg mRNA transcript in RNA from normal <strong>and</strong> goitrous thyroids. Mol Cell<br />

Endocrinol 84:R23-R26<br />

223. Targovnik HM, Medeiros-Neto G, Varela V, Cochaux P, Wajchenberg BL,<br />

Vassart G 1993 A nonsense mutation causes human hereditary congenital<br />

<strong>goiter</strong> with preferential production <strong>of</strong> a 171-nucleotide-deleted thyroglobulin<br />

ribonucleic acid messenger. J Clin Endocrinol Metab 77:210-215<br />

224. Targovnik HM, Frechtel GD, Mendive FM, Vono J, Cochaux P, Vassart G,<br />

Medeiros-Neto G 1998 Evidence for the segregation <strong>of</strong> three different mutated<br />

alleles <strong>of</strong> the thyroglobulin gene in a Brazilian family with congenital <strong>goiter</strong> <strong>and</strong><br />

hypothyroidism. Thyroid 8:291-297<br />

225. Ieiri T, Cochaux P, Targovnik HM, Suzuki M, Shimoda S, Perret J, Vassart G<br />

1991 A 3' splice site mutation in the thyroglobulin gene responsible for<br />

congenital <strong>goiter</strong> with hypothyroidism. J Clin Invest 88:1901-1905<br />

226. Hishinuma A, Takamatsu J, Ohyama Y, Yokozawa T, Kanno Y, Kuma K,<br />

Yoshida S, Matsuura N, Ieiri T 1999 Two novel cysteine substitutions (C1263R<br />

<strong>and</strong> C1995S) <strong>of</strong> thyroglobulin cause a defect in intracellular transport <strong>of</strong><br />

thyroglobulin in patients with congenital <strong>goiter</strong> <strong>and</strong> the variant type <strong>of</strong><br />

adenomatous <strong>goiter</strong>. J Clin Endocrinol Metab 84:1438-1444<br />

227. van de Graaf SA, Cammenga M, Ponne NJ, Veenboer GJ, Gons MH, Orgiazzi<br />

J, De Vijlder JJ, Ris-Stalpers C 1999 The screening for mutations in the<br />

thyroglobulin cDNA from six patients with congenital hypothyroidism.<br />

Biochimie 81:425-432<br />

228. Targovnik HM, Rivolta CM, Mendive FM, Moya CM, Vono J, Medeiros-Neto<br />

G 2001 Congenital <strong>goiter</strong> with hypothyroidism caused by a 5' splice site<br />

mutation in the thyroglobulin gene. Thyroid 11:685-690<br />

229. Hishinuma A, Kasai K, Masawa N, Kanno Y, Arimura M, Shimoda SI, Ieiri T<br />

1998 Missense mutation (C1263R) in the thyroglobulin gene causes congenital<br />

<strong>goiter</strong> with mild hypothyroidism by impaired intracellular transport. Endocr J<br />

45:315-327<br />

230. Yoshida S, Takamatsu J, Kuma K, Murakami Y, Sakane S, Katayama S,<br />

Tarutani O, Ohsawa N 1996 A variant <strong>of</strong> adenomatous <strong>goiter</strong> with<br />

characteristic histology <strong>and</strong> possible hereditary thyroglobulin abnormality. J<br />

Clin Endocrinol Metab 81:1961-1966<br />

231. Kim PS, Hossain SA, Park YN, Lee I, Yoo SE, Arvan P 1998 A single amino<br />

acid change in the acetylcholinesterase-like domain <strong>of</strong> thyroglobulin causes<br />

congenital <strong>goiter</strong> with hypothyroidism in the cog/cog mouse: a model <strong>of</strong> human<br />

endoplasmic reticulum storage diseases. Proc Natl Acad Sci U S A 95:9909-9913<br />

232. Corral J, Martin C, Perez R, Sanchez I, Mories MT, San Millan JL, Miralles<br />

JM, Gonzalez-Sarmiento R 1993 Thyroglobulin gene point mutation associated<br />

with non-endemic simple goitre. Lancet 341:462-464<br />

58


233. Perez-Centeno C, Gonzalez-Sarmiento R, Mories MT, Corrales JJ, Miralles-<br />

Garcia JM 1996 Thyroglobulin exon 10 gene point mutation in a patient with<br />

endemic <strong>goiter</strong>. Thyroid 6:423-427<br />

234. Gonzalez-Sarmiento R, Corral J, Mories MT, Corrales JJ, Miguel-Velado E,<br />

Miralles-Garcia JM 2001 Monoallelic deletion in the 5' region <strong>of</strong> the<br />

thyroglobulin gene as a cause <strong>of</strong> sporadic nonendemic simple <strong>goiter</strong>. Thyroid<br />

11:789-793<br />

235. Abramowicz MJ, Targovnik HM, Varela V, Cochaux P, Krawiec L, Pisarev<br />

MA, Propato FV, Juvenal G, Chester HA, Vassart G 1992 Identification <strong>of</strong> a<br />

mutation in the coding sequence <strong>of</strong> the human thyroid peroxidase gene causing<br />

congenital <strong>goiter</strong>. J Clin Invest 90:1200-1204<br />

236. Bakker B, Bikker H, Vulsma T, de R<strong>and</strong>amie JS, Wiedijk BM, De Vijlder JJ<br />

2000 Two decades <strong>of</strong> screening for congenital hypothyroidism in The<br />

Netherl<strong>and</strong>s: TPO gene mutations in total iodide organification defects (an<br />

update) [In Process Citation]. J Clin Endocrinol Metab 85:3708-3712<br />

237. Bikker H, Baas F, De Vijlder JJ 1997 <strong>Molecular</strong> analysis <strong>of</strong> mutated thyroid<br />

peroxidase detected in patients with total iodide organification defects. J Clin<br />

Endocrinol Metab 82:649-653<br />

238. Bikker H, Waelkens JJ, Bravenboer B, De Vijlder JJ 1996 Congenital<br />

hypothyroidism caused by a premature termination signal in exon 10 <strong>of</strong> the<br />

human thyroid peroxidase gene. J Clin Endocrinol Metab 81:2076-2079<br />

239. Bikker H, Vulsma T, Baas F, De Vijlder JJ 1995 Identification <strong>of</strong> five novel<br />

inactivating mutations in the human thyroid peroxidase gene by denaturing<br />

gradient gel electrophoresis. Hum Mutat 6:9-16<br />

240. Bikker H, den Hartog MT, Baas F, Gons MH, Vulsma T, De Vijlder JJ 1994 A<br />

20-basepair duplication in the human thyroid peroxidase gene results in a total<br />

iodide organification defect <strong>and</strong> congenital hypothyroidism. J Clin Endocrinol<br />

Metab 79:248-252<br />

241. Hagen GA, Niepomniszcze H, Haibach H, Bigazzi M, Hati R, Rapoport B,<br />

Jimenez C, DeGroot LJ, Frawley TF 1971 Peroxidase deficiency in familial<br />

<strong>goiter</strong> with iodide organification defect. N Engl J Med 285:1394-1398<br />

242. Pommier J, Tourniaire J, Deme D, Chalendar D, Bornet H, Nunez J 1974 A<br />

defective thyroid peroxidase solubilized from a familial <strong>goiter</strong> with iodine<br />

organification defect. J Clin Endocrinol Metab 39:69-80<br />

243. Pannain S, Weiss RE, Jackson CE, Dian D, Beck JC, Sheffield VC, Cox N,<br />

Refet<strong>of</strong>f S 1999 Two different mutations in the thyroid peroxidase gene <strong>of</strong> a<br />

large inbred Amish kindred: power <strong>and</strong> limits <strong>of</strong> homozygosity mapping. J Clin<br />

Endocrinol Metab 84:1061-1071<br />

244. Niu DM, Hwang B, Chu YK, Liao CJ, Wang PL, Lin CY 2002 High prevalence<br />

<strong>of</strong> a novel mutation (2268 insT) <strong>of</strong> the thyroid peroxidase gene in Taiwanese<br />

patients with total iodide organification defect, <strong>and</strong> evidence for a founder<br />

effect. J Clin Endocrinol Metab 87:4208-4212<br />

59


245. Smanik PA, Liu Q, Furminger TL, Ryu K, Xing S, Mazzaferri EL, Jhiang SM<br />

1996 Cloning <strong>of</strong> the human sodium iodide symporter. Biochem Biophys Res<br />

Communications 226:339-345<br />

246. Shen DH, Kloos RT, Mazzaferri EL, Jhian SM 2001 Sodium iodide symporter<br />

in health <strong>and</strong> disease. Thyroid 11:415-425<br />

247. Kosugi S, Bhayana S, Dean HJ 1999 A novel mutation in the sodium/iodide<br />

symporter gene in the largest family with iodide transport defect. J Clin<br />

Endocrinol Metab 84:3248-3253<br />

248. Kotani T, Umeki K, Yamamoto I, Maesaka H, Tachibana K, Ohtaki S 1999 A<br />

novel mutation in the human thyroid peroxidase gene resulting in a total iodide<br />

organification defect. J Endocrinol 160:267-273<br />

249. Fujiwara H, Tatsumi K, Miki K, Harada T, Miyai K, Takai S-I, Amino N 1997<br />

Congenital hypothyroidism caused by a mutation in the Na+/I-symporter.<br />

Nature Genetics 16:124-125<br />

250. Kosugi S, Inoue S, Matsuda A, Jhiang SM 1998 Novel, missense <strong>and</strong> loss-<strong>of</strong>function<br />

mutations in the sodium/iodide symporter gene causing iodide<br />

transport defect in three Japanese patients. J Clin Endocrinol Metab 83:3373-<br />

3376<br />

251. Levy O, Ginter CS, De la Vieja A, Levy D, Carrasco N 1998 Identification <strong>of</strong> a<br />

structural requirement for thyroid Na+/I- symporter (NIS) function from<br />

analysis <strong>of</strong> a mutation that causes human congenital hypothyroidism. FEBS<br />

Lett 429:36-40<br />

252. Fujiwara H, Tatsumi K, Miki K, Harada T, Okada S, Nose O, Kodama S,<br />

Amino N 1998 Recurrent T354P mutation <strong>of</strong> the Na+/I- symporter in patients<br />

with iodide transport defect. J Clin Endocrinol Metab 83:2940-2943<br />

253. Matsuda A, Kosugi S 1997 A homozygous missense mutation <strong>of</strong> the sodium<br />

/iodide symporter gene causing iodide transport defect. J Clin Endocrinol<br />

Metab 82:3966-3971<br />

254. Pohlenz J, Rosenthal IM, Weiss RE, Jhiang SM, Burant C, Refet<strong>of</strong>f S 1998<br />

Congenital hypothyroidism due to mutations in the sodium/iodide symporter.<br />

Identification <strong>of</strong> a nonsense mutation producing a downstream cryptic 3' splice<br />

site. J Clin Invest 101:1028-1035<br />

255. Pohlenz J, Refet<strong>of</strong>f S 1999 Mutations in the sodium/iodide symporter (NIS)<br />

gene as a cause for iodide transport defects <strong>and</strong> congenital hypothyroidism.<br />

Biochimie 81:469-476<br />

256. Reardon W, Trembath RC 1996 Pendred syndrome. J Med Genet 33:1037-1040<br />

257. Kopp P 1999 Pendred's syndrome: identification <strong>of</strong> the genetic defect a century<br />

after its recognition. Thyroid 9:65-69<br />

60


258. Medeiros-Neto G, Stanbury JB 1994 Pendred`s Syndrom: association <strong>of</strong><br />

congenital deafness with sporadic <strong>goiter</strong>. In: Medeiros-Neto G, Stanbury JB<br />

(eds) Inherited disorders <strong>of</strong> the thyroid system. Boca Raton:CRC Press81-105<br />

259. Illum P, Kiaer HW, Hvidberg-Hansen J, Sondergaard G 1972 Fifteen cases <strong>of</strong><br />

Pendred's syndrome. Congenital deafness <strong>and</strong> sporadic <strong>goiter</strong>. Arch<br />

Otolaryngol 96:297-304<br />

260. Everett LA, Glaser B, Beck JC, Idol JR, Buchs A, Heyman M, Adawi F, Hazani<br />

E, Nassir E, Baxevanis AD, Sheffield VC, Green ED 1997 Pendred syndrome is<br />

caused by mutations in a putative sulphate transporter gene (PDS). Nat Genet<br />

17:411-422<br />

261. Kopp P, Arseven OK, Sabacan L, Kotlar T, Dupuis J, Cavaliere H, Santos CL,<br />

Jameson JL, Medeiros-Neto G 1999 Phenocopies for deafness <strong>and</strong> <strong>goiter</strong><br />

development in a large inbred Brazilian kindred with Pendred's syndrome<br />

associated with a novel mutation in the PDS gene. J Clin Endocrinol Metab<br />

84:336-341<br />

262. Van Hauwe P, Everett LA, Coucke P, Scott DA, Kraft ML, Ris-Stalpers C,<br />

Bolder C, Otten B, De Vijlder JJ, Dietrich NL, Ramesh A, Srisailapathy SC,<br />

Parving A, Cremers CW, Willems PJ, Smith RJ, Green ED, Van Camp G 1998<br />

Two frequent missense mutations in Pendred syndrome. Hum Mol Genet<br />

7:1099-1104<br />

263. Campbell C, Cucci RA, Prasad S, Green GE, Edeal JB, Galer CE, Karniski LP,<br />

Sheffield VC, Smith RJ 2001 Pendred syndrome, DFNB4, <strong>and</strong> PDS/SLC26A4<br />

identification <strong>of</strong> eight novel mutations <strong>and</strong> possible genotype-phenotype<br />

correlations. Hum Mutat 17:403-411<br />

264. de Roux N, Misrahi M, Chatelain N, Gross B, Milgrom E 1996 Microsatellites<br />

<strong>and</strong> PCR primers for genetic studies <strong>and</strong> genomic sequencing <strong>of</strong> the human<br />

TSH receptor gene. Mol Cell Endocrinol 117:253-256<br />

265. Gabriel EM, Bergert ER, Grant CS, van Heerden JA, Thompson GB, Morris<br />

JC 1999 Germline polymorphism <strong>of</strong> codon 727 <strong>of</strong> human thyroid-stimulating<br />

hormone receptor is associated with <strong>toxic</strong> <strong>multinodular</strong> <strong>goiter</strong>. J Clin<br />

Endocrinol Metab 84:3328-3335<br />

266. Nogueira CR, Kopp P, Arseven OK, Santos CL, Jameson JL, Medeiros-Neto G<br />

1999 Thyrotropin receptor mutations in hyperfunctioning thyroid adenomas<br />

from Brazil. Thyroid 9:1063-1068<br />

267. Mühlberg T, Herrmann K, Joba W, Kirchberger M, Heberling HJ, Heufelder<br />

AE 2000 Lack <strong>of</strong> association <strong>of</strong> nonautoimmune hyperfunctioning thyroid<br />

disorders <strong>and</strong> a germline polymorphism <strong>of</strong> codon 727 <strong>of</strong> the human thyrotropin<br />

receptor in a European Caucasian population. J Clin Endocrinol Metab<br />

85:2640-2643<br />

268. Peeters RP, van Toor H, Klootwijk W, de Rijke YB, Kuiper GG, Uitterlinden<br />

AG, Visser TJ 2003 Polymorphisms in thyroid hormone pathway genes are<br />

associated with plasma TSH <strong>and</strong> iodothyronine levels in healthy subjects. J Clin<br />

Endocrinol Metab 88:2880-2888<br />

61


269. Bignell GR, Canzian F, Shayeghi M, Stark M, Shugart YY, Biggs P, Mangion J,<br />

Hamoudi R, Rosenblatt J, Buu P, Sun S, St<strong>of</strong>fer SS, Goldgar DE, Romeo G,<br />

Houlston RS, Narod SA, Stratton MR, Foulkes WD 1997 Familial non<strong>toxic</strong><br />

<strong>multinodular</strong> thyroid <strong>goiter</strong> locus maps to chromosome 14q but does not<br />

account for familial nonmedullary thyroid cancer. Am J Hum Genet 61:1123-<br />

1130<br />

270. Neumann S, Willgerodt H, Ackermann F, Reske A, Jung M, Reis A, Paschke R<br />

1999 Linkage <strong>of</strong> familial <strong>euthyroid</strong> <strong>goiter</strong> to the <strong>multinodular</strong> <strong>goiter</strong>-1 locus <strong>and</strong><br />

exclusion <strong>of</strong> the c<strong>and</strong>idate genes thyroglobulin, thyroperoxidase, <strong>and</strong> Na+/I-<br />

symporter [In Process Citation]. J Clin Endocrinol Metab 84:3750-3756<br />

271. Capon F, Tacconelli A, Giardina E, Sciacchitano S, Bruno R, Tassi V,<br />

Trischitta V, Filetti S, Dallapiccola B, Novelli G 2000 Mapping a dominant<br />

form <strong>of</strong> <strong>multinodular</strong> <strong>goiter</strong> to chromosome Xp22. Am J Hum Genet 67:1004-<br />

1007<br />

272. Neumann S, Bayer Y, Reske A, Tajtakova M, Langer P, Paschke R 2003<br />

Further indications for genetic heterogeneity <strong>of</strong> <strong>euthyroid</strong> familial <strong>goiter</strong>. J Mol<br />

Med 81:736-745<br />

273. Bayer Y, Neumann S., Meyer B, Rüschendorf F, Reske A., Brix TH, Hegedus L,<br />

Langer P, Nürnberg P, Paschke R 2004 Genome-wide linkage analysis reveals<br />

evidence for four new susceptibility loci for familial <strong>euthyroid</strong> <strong>goiter</strong>. J Clin<br />

Endocrinol Metab in press:<br />

274. Gharib H, James EM, Charboneau JW, Naessens JM, Offord KP, Gorman CA<br />

1987 Suppressive therapy with levothyroxine for solitary thyroid nodules. A<br />

double-blind controlled clinical study. N Engl J Med 317:70-75<br />

275. Cheung PS, Lee JM, Boey JH 1989 Thyroxine suppressive therapy <strong>of</strong> benign<br />

solitary thyroid nodules: a prospective r<strong>and</strong>omized study. World J Surg<br />

13:818-821<br />

276. Reverter JL, Lucas A, Salinas I, Audi L, Foz M, Sanmarti A 1992 Suppressive<br />

therapy with levothyroxine for solitary thyroid nodules. Clin Endocrinol (Oxf)<br />

36:25-28<br />

277. Mainini E, Martinelli I, Mor<strong>and</strong>i G, Villa S, Stefani I, Mazzi C 1995<br />

Levothyroxine suppressive therapy for solitary thyroid nodule. J Endocrinol<br />

Invest 18:796-799<br />

278. Lima N, Knobel M, Cavaliere H, Sztejnsznajd C, Tomimori E, Medeiros-Neto<br />

G 1997 Levothyroxine suppressive therapy is partially effective in treating<br />

patients with benign, solid thyroid nodules <strong>and</strong> <strong>multinodular</strong> <strong>goiter</strong>s. Thyroid<br />

7:691-697<br />

279. Zelmanovitz F, Genro S, Gross JL 1998 Suppressive therapy with<br />

levothyroxine for solitary thyroid nodules: a double-blind controlled clinical<br />

study <strong>and</strong> cumulative meta-analyses. J Clin Endocrinol Metab 83:3881-3885<br />

280. Papini E, Guglielmi R, Bianchini A, Crescenzi A, Taccogna S, Nardi F, Panunzi<br />

C, Rinaldi R, Toscano V, Pacella CM 2002 Risk <strong>of</strong> malignancy in nonpalpable<br />

62


thyroid nodules: predictive value <strong>of</strong> ultrasound <strong>and</strong> color-Doppler features. J<br />

Clin Endocrinol Metab 87:1941-1946<br />

281. Brauer VF, Hentschel B, Paschke R 2003 [Euthyroid thyroid nodules. Aims,<br />

results <strong>and</strong> perspectives concerning drug therapy]. Dtsch Med Wochenschr<br />

128:2381-2387<br />

282. Carrillo JF, Frias-Mendivil M, Ochoa-Carrillo FJ, Ibarra M 2000 Accuracy <strong>of</strong><br />

fine-needle aspiration biopsy <strong>of</strong> the thyroid combined with an evaluation <strong>of</strong><br />

clinical <strong>and</strong> radiologic factors. Otolaryngol Head Neck Surg 122:917-921<br />

283. Cappelli C, Agosti B, Tironi A, Morassi ML, Pelizzari G, Cumetti D, Cerudelli<br />

B 2002 [Prevalence <strong>and</strong> aggressiveness <strong>of</strong> thyroid carcinoma with diameter less<br />

than one centimetre in iodine deficiency areas]. Minerva Endocrinol 27:65-71<br />

284. Vierhapper H, Raber W, Bieglmayer C, Kaserer K, Weinhausl A, Niederle B<br />

1997 Routine measurement <strong>of</strong> plasma calcitonin in nodular thyroid diseases. J<br />

Clin Endocrinol Metab 82:1589-1593<br />

285. Daniels GH 1996 Thyroid nodules <strong>and</strong> nodular thyroids: a clinical overview.<br />

Compr Ther 22:239-250<br />

286. Richter B, Neises G, Clar C 2002 Pharmacotherapy for thyroid nodules. A<br />

systematic review <strong>and</strong> meta-analysis. Endocrinol Metab Clin North Am 31:699-<br />

722<br />

287. Uzzan B, Campos J, Cucherat M, Nony P, Boissel JP, Perret GY 1996 Effects<br />

on bone mass <strong>of</strong> long term treatment with thyroid hormones: a meta-analysis. J<br />

Clin Endocrinol Metab 81:4278-4289<br />

288. Sawin CT, Geller A, Wolf PA, Belanger AJ, Baker E, Bacharach P, Wilson PW,<br />

Benjamin EJ, D'Agostino RB 1994 Low serum thyrotropin concentrations as a<br />

risk factor for atrial fibrillation in older persons. N Engl J Med 331:1249-1252<br />

289. Grussendorf M 1996 [Therapy <strong>of</strong> <strong>euthyroid</strong> iron deficiency <strong>goiter</strong>. Effectiveness<br />

<strong>of</strong> a combination <strong>of</strong> L-thyroxine <strong>and</strong> 150 micrograms iodine in comparison with<br />

mono-L-thyroxine]. Med Klin (Munich) 91:489-493<br />

290. Klemenz B, Forster G, Wieler H, Kahaly G, Kaiser KP, Hansen C, Willkomm<br />

P, Ruhlmann J 1998 [Combination therapy <strong>of</strong> endemic <strong>goiter</strong> with two different<br />

thyroxine/iodine combinations]. Nuklearmedizin 37:101-106<br />

291. Saller B, Hoermann R, Ritter MM, Morell R, Kreisig T, Mann K 1991 Course<br />

<strong>of</strong> thyroid iodine concentration during treatment <strong>of</strong> endemic goitre with iodine<br />

<strong>and</strong> a combination <strong>of</strong> iodine <strong>and</strong> levothyroxine. Acta Endocrinol (Copenh)<br />

125:662-667<br />

292. Oertel YC 2002 A pathologist trying to help endocrinologists to interpret<br />

cytopathology reports from thyroid aspirates. J Clin Endocrinol Metab<br />

87:1459-1461<br />

63


293. Belfiore A, La Rosa GL, Padova G, Sava L, Ippolito O, Vigneri R 1987 The<br />

frequency <strong>of</strong> cold thyroid nodules <strong>and</strong> thyroid malignancies in patients from an<br />

iodine-deficient area. Cancer 60:3096-3102<br />

294. La Rosa GL, Lupo L, Giuffrida D, Gullo D, Vigneri R, Belfiore A 1995<br />

Levothyroxine <strong>and</strong> potassium iodide are both effective in treating benign<br />

solitary solid cold nodules <strong>of</strong> the thyroid. Ann Intern Med 122:1-8<br />

295. Ridgway EC 1998 Medical treatment <strong>of</strong> benign thyroid nodules: have we<br />

defined a benefit? Ann Intern Med 128:403-405<br />

296. Kahaly GJ, Dietlein M 2002 Cost estimation <strong>of</strong> thyroid disorders in Germany.<br />

Thyroid 12:909-914<br />

297. Gharib H 1997 Changing concepts in the diagnosis <strong>and</strong> management <strong>of</strong> thyroid<br />

nodules. Endocrinol Metab Clin North Am 26:777-800<br />

298. Castro MR, Gharib H 2000 Thyroid nodules <strong>and</strong> cancer. When to wait <strong>and</strong><br />

watch, when to refer. Postgrad Med 107:113-20, 123<br />

299. Wiersinga WM 1995 Is repeated fine-needle aspiration cytology indicated in<br />

(benign) thyroid nodules? Eur J Endocrinol 132:661-662<br />

300. Erdogan MF, Kamel N, Aras D, Akdogan A, Baskal N, Erdogan G 1998 Value<br />

<strong>of</strong> re-aspirations in benign nodular thyroid disease. Thyroid 8:1087-1090<br />

301. Hamburger JI 1994 Diagnosis <strong>of</strong> thyroid nodules by fine needle biopsy: use <strong>and</strong><br />

abuse. J Clin Endocrinol Metab 79:335-339<br />

302. Baloch ZW, LiVolsi VA 2002 Follicular-patterned lesions <strong>of</strong> the thyroid: the<br />

bane <strong>of</strong> the pathologist. Am J Clin Pathol 117:143-150<br />

303. Bartolazzi A, Gasbarri A, Papotti M, Bussolati G, Lucante T, Khan A, Inohara<br />

H, Mar<strong>and</strong>ino F, Orl<strong>and</strong>i F, Nardi F, Vecchione A, Tecce R, Larsson O 2001<br />

Application <strong>of</strong> an immunodiagnostic method for improving preoperative<br />

diagnosis <strong>of</strong> nodular thyroid lesions. Lancet 357:1644-1650<br />

304. de Micco C, Ruf J, Chrestian MA, Gros N, Henry JF, Carayon P 1991<br />

Immunohistochemical study <strong>of</strong> thyroid peroxidase in normal, hyperplastic, <strong>and</strong><br />

neoplastic human thyroid tissues. Cancer 67:3036-3041<br />

305. Fagin JA 2002 Perspective: lessons learned from molecular genetic studies <strong>of</strong><br />

thyroid cancer--insights into <strong>pathogenesis</strong> <strong>and</strong> tumor-specific therapeutic<br />

targets. Endocrinology 143:2025-2028<br />

306. Korstanje R, Paigen B 2002 From QTL to gene: the harvest begins. Nat Genet<br />

31:235-236<br />

307. Neumann S., Schuchardt K, Reske A., Reske A., Emmrich P, Paschke R 2004<br />

Lack <strong>of</strong> correlation for sodium iodide symporter mRNA <strong>and</strong> protein expression<br />

<strong>and</strong> analysis <strong>of</strong> NIS promoter methylation in benign cold thyroid nodules.<br />

Thyroid in press:<br />

64


lig<strong>and</strong><br />

TGF β<br />

TGF β<br />

type III<br />

TGFβR<br />

type I/II<br />

TGFβR<br />

receptors<br />

Smad 1<br />

Smad 2<br />

receptor<br />

Smads<br />

Smad 3<br />

Smad 5<br />

Smad 4<br />

coregulatory<br />

Smads<br />

inhibitory-<br />

Smads<br />

Smad 7<br />

Smad 6<br />

P300<br />

inhibition <strong>of</strong><br />

thyroid cell<br />

differentiation <strong>and</strong><br />

cell proliferation


impaired<br />

hormone<br />

Synthesis<br />

H2O2 ?<br />

free radicals ?<br />

hypertrophy<br />

hyperplasia<br />

proliferation<br />

mutagenesis<br />

single cells with<br />

somatic mutations<br />

adaptation by more<br />

efficient iodine<br />

clearence, trapping,<br />

metabolism<br />

<strong>and</strong> increased gene<br />

expression<br />

<strong>goiter</strong> with cell<br />

clones containing a<br />

somatic mutation<br />

.<br />

.<br />

.<br />

.<br />

.<br />

.<br />

.<br />

.<br />

expansion <strong>of</strong> cell<br />

clones<br />

with advantageous<br />

mutations leading to<br />

hot (or cold) nodules


a)<br />

D3S4545<br />

D3S1259<br />

D3S3038<br />

D3S1266<br />

D3S2432<br />

D3S1768<br />

D3S2409<br />

b)<br />

2.0<br />

1.0<br />

0<br />

NPL score<br />

1.0<br />

2.0<br />

3.0<br />

16 cM interval

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!