9,189
Views
55
CrossRef citations to date
0
Altmetric
Review

Thyroid physiology and autoimmunity in pregnancy and after delivery

&
Pages 697-707 | Published online: 10 Jan 2014

Abstract

During pregnancy and after delivery, the maternal thyroid gland faces several metabolic, hemodynamic and immunologic changes. In this article we first summarize the current knowledge on the physiologic adaptation of the healthy thyroid to pregnancy, including variations of thyroid-stimulating hormone and free thyroid hormones, as well as variations of thyroid volume. Our second aim is to illustrate the background of thyroid autoimmunity in this period, which characteristically ameliorates during pregnancy and aggravates after delivery. Although rare during pregnancy, Graves’ disease is the most frequent cause of hyperthyroidism, while Hashimoto’s thyroiditis is the most frequent cause for hypothyroidism. Both types of thyroid dysfunction may lead to detrimental complications in mother and child and therefore timely recognition and treatment is essential. Postpartum autoimmunity most frequently exacerbates in the form of postpartum thyroiditis, which presents with diverse clinical presentations and may lead to permanent hypothyroidism.

Medscape: Continuing Medical Education Online

This activity has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education through the joint sponsorship of Medscape, LLC and Expert Reviews Ltd. Medscape, LLC is accredited by the ACCME to provide continuing medical education for physicians.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1 AMA PRA Category 1 Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test with a 70% minimum passing score and complete the evaluation at http://www.medscape.org/journal/expertimmunology; (4) view/print certificate.

Release date: 6 September 2011; Expiration date: 6 September 2012

Learning objectives

Upon completion of this activity, participants should be able to:

  • • Describe changes in thyroid hormones and thyroid volume in normal pregnancy

  • • Describe changes in thyroid autoimmunity during pregnancy

  • • Describe characteristics and complications of Graves’ disease and Hashimoto’s thyroiditis during pregnancy, as well as of postpartum thyroiditis

Financial & competing interests disclosure

EDITOR

Elisa Manzotti,Editorial Director, Future Science Group, London, UK

Disclosure:Elisa Manzotti has disclosed no relevant financial relationships.

CME AUTHOR

Laurie Barclay, MD,Freelance writer and reviewer, Medscape, LLC

Disclosure:Laurie Barclay, MD, has disclosed no relevant financial relationships.

AUTHORS

Simona Gaberšček, MD, PhD,Assistant Professor, Department of Nuclear Medicine, University Medical Centre Ljubljana, Zaloška 7, Ljubljana, Slovenia

Disclosure:Simona Gaberšček, MD, PhD, has disclosed no relevant financial relationships.

Katja Zaletel, MD, PhD,Assistant Professor, Department of Nuclear Medicine, University Medical Centre Ljubljana, Zaloška 7, Ljubljana, Slovenia

Disclosure:Katja Zaletel, MD, PhD, has disclosed no relevant financial relationships.

Thyroid function & size in pregnancy & after delivery

Iodine in pregnancy & after delivery

Iodine metabolism

Iodine metabolism in pregnancy is marked by several characteristics. Synthesis of thyroid hormones is increased by up to 50% due to estrogen-induced increase in thyroxine-binding globulin (TBG) concentration Citation[1,2]. Renal clearance of iodide increases owing to the higher glomerular filtration rate Citation[1,3]. Iodide and iodothyronines are transported from maternal circulation to the fetus Citation[1]. Fetal thyroid hormone production increases during the second half of gestation Citation[3] and after delivery, iodide is also transported into the breast milk.

Iodine supply

According to the Endocrine Society clinical practice guidelines, iodine intake before pregnancy should be 150 µg/day in order to maintain adequate intrathyroidal iodine stores. During pregnancy and lactation, the recommended iodine intake is 250 µg/day. There is no need to provide more than 500 µg/day. Women, living in countries with adequate and lasting universal salt iodization programs are not at risk of iodine deficiency Citation[4]. When evaluating adequacy of iodine supply in pregnant women, urinary iodine concentration, as a measure of iodine supply, should be seen in light of the fact that the volume of daily urine usually totals 1.5 l and that approximately 10% of iodine is not excreted via urine Citation[5].

Iodine deficiency causes several metabolic changes and goiter in mother and fetus Citation[1]. In mild iodine deficiency, lower levels of free thyroxine (fT4) and free triiodothyronine (fT3) and higher levels of thyroid-stimulating hormone (TSH), TBG and thyroglobulin (Tg) were observed in the second and third trimester of pregnancy when compared with the first trimester. In the third trimester, maternal thyroid volume was also larger Citation[6].

In continuation, we will primarily focus on thyroid function and size in healthy pregnant women living in areas with adequate iodine intake.

Maternal thyroid function

Transport proteins

Besides TBG, which is a major thyroid hormone transport protein, transthyretin and albumin are also important. The level of albumin, which has the lowest thyroxine (T4) affinity and enables a fast release of T4Citation[7], gradually decreases during pregnancy Citation[8]. TBG is an active carrier and has a possibility to switch between the high-affinity and the low-affinity form Citation[9]. TBG levels are the highest in the second and third trimester of pregnancy Citation[6] and the same holds true for thyroid-hormone binding ratio Citation[10] and thyroid-binding capacity Citation[11], which decreases as soon as 3–4 days after delivery. In pregnancy, TBG production in the liver is increased and the half-life of TBG is prolonged because of an estrogen-induced increase in sialylation of TBG Citation[1,12].

Variations of human chorionic gonadotropin

The placenta produces human chorionic gonadotropin (hCG) in the first week after conception and the level is the highest at week 10 before it begins to decrease Citation[3] and reach a plateau at week 20 Citation[2]. In twin pregnancies, peaks of hCG values were higher and of longer duration than in single pregnancies (6 weeks and 1 week, respectively) Citation[13]. The hCG and TSH molecules share similarities, as do the hCG and TSH receptors. Consequently, hCG weakly stimulates the thyroid gland. The TSH receptor stimulation depends on the amplitude and duration of the hCG peak. hCG may induce gestational hyperthyroidism, which occurs in 2–3% of pregnancies Citation[1].

Variations of TSH

Most authors agree that in the first trimester, TSH levels may be decreased in some women with otherwise healthy thyroid glands. Approximately 10% of women have TSH levels below normal and up to 10% of women have suppressed levels of TSH Citation[2,14]. In smokers, TSH levels in the first and third trimester were lower than in nonsmokers Citation[15].

During pregnancy, TSH levels increase and reach the highest value in the third trimester, irrespective of iodine supply Citation[6,10,16–18]. At 3–4 days after delivery, TSH levels were the highest Citation[11]. Higher TSH levels in the second half of pregnancy probably mirror hCG and free thyroid hormones levels, being lower in that period of pregnancy. A total of 4 months after delivery, TSH levels were lower than in the third trimester Citation[16]. Similarly, 1-year postpartum they were lower than in the second and third trimester of pregnancy Citation[19], except for one study, where the same TSH levels were established 1-year postpartum and during pregnancy Citation[20].

Relationship between TSH & hCG

The lower TSH level in the first trimester mirrors the highest level of hCG in that period and a negative correlation between these levels is significant Citation[6]. A transient subnormal TSH in the first trimester was associated with hCG levels above 50,000 IU/l Citation[21]. At hCG concentrations above 400,000 IU/l, TSH was suppressed in 100% of women and fT4 increased in 80%. At hCG levels above 200,000 IU/l, 67% of women had TSH below 0.2 mU/l Citation[22]. From gestational weeks 11–18, only upper deciles of hCG were weakly associated with lower TSH values Citation[23].

Variations of free thyroid hormones

Even in areas with adequate iodine intake, many authors established pregnancy levels of fT4 and fT3 to be lower than in nonpregnant individuals. In the last months of pregnancy, fT4 levels were often below the reference interval Citation[24]. Total thyroxine slightly increased in the first trimester and decreased by approximately 30% to low normal values in the second and third trimester Citation[25]. During pregnancy, fT4 and fT3 were also decreased when compared with postpartum levels Citation[20]. fT4 was lower during pregnancy than 1-year postpartum Citation[19] and lower than those in the nonpregnant state Citation[26].

Several factors may influence the level of free thyroid hormones. Increased hCG at 11–13 weeks was associated with increased median values of fT4Citation[23]. Twin pregnancies with higher hCG values of longer duration frequently led to increased fT4 levels Citation[12]. In smokers, fT3 levels were higher than in nonsmokers and fT4 levels did not differ Citation[15]. Despite low iodine supply, Sudanese pregnant women had higher fT4 and lower TSH than Swedish pregnant women with adequate iodine intake, both in weeks 20–24 and 36–39 Citation[27]. The history of pre-pregnant iodine supply seems to be an important factor influencing thyroid response during pregnancy.

Several authors advocated for the use of gestational specific reference intervals Citation[18,24]. They have already been proposed for Swiss women Citation[18], Indian women Citation[17], Chinese women Citation[28], Danish women Citation[29] and others. In all, TSH increased during pregnancy, while fT4 and fT3 decreased.

Interpretations of free thyroid hormone variations

The decrease of fT4 and, less frequently, of fT3 in the second half of pregnancy remains an unresolved issue. This is not necessarily the reflection of maternal iodine deficiency Citation[5]. That assumption was confirmed by the following study, showing fT4 in the third trimester below the nonpregnant reference range in 64.5% of women and fT3 in 10.3%. The values returned to normal 4 months after delivery and were not associated with the urinary iodine concentration, which has been adequate and even lower after delivery than in the third trimester Citation[16].

Beside a decrease in fT4 and fT3, an increase in reverse T3 during pregnancy was found and a resemblance with the nonthyroidal illness was postulated Citation[30]. However, patients with that condition have frequently decreased fT3 and rarely decreased fT4Citation[31], whereas during pregnancy the fall of fT4 is more pronounced.

Variations of fT4 and fT3 are definitely pregnancy-related, since they increased just 3–4 days after delivery Citation[11]. Decreased fT4 might not be only a consequence of importantly increased TBG concentration, but also of its increased binding affinity. However, there are no reports on binding affinity of TBG during pregnancy. Besides, albumin levels decrease during pregnancy, diminishing the possibility of the quick release of the bound T4.

Methodological problems are another possible explanation for the observed variations. Negro et al. concluded that there are no optimal diagnostic tests for fT4 during pregnancy Citation[32]. Recently, several attempts have been undertaken to find the most appropriate method for the determination of thyroid hormones during pregnancy. Total T4 and fT4 index keep the inverse relationship with TSH during pregnancy and are possibly more reliable for the evaluation of thyroid function in pregnancy Citation[10]. With equilibrium dialysis and with nine immunoassays the levels of fT4 were in the lower part of the nonpregnant reference interval or below Citation[33]. With direct equilibrium dialysis – liquid chromatography/tandem mass spectrometry – fT4 and fT3 reference intervals decreased from week 14 to week 20 of pregnancy Citation[34]. The relationship between log(TSH) and fT4 was extremely weak for both the tandem mass spectrometry and the immunoassay Citation[35].

Maternal thyroid size

Factors influencing thyroid size

Thyroid size is influenced by different factors, including iodine supply, genetics, gender, age, TSH, anthropometric parameters, parity and smoking Citation[36]. Thyroid volume in individuals in the nonpregnant state increased with bodyweight, age Citation[37], BMI and total body water Citation[38]. Also during pregnancy, a positive correlation between thyroid volume and BMI was found Citation[39]. A negative correlation between thyroid volume and TSH Citation[38] was also confirmed in pregnant women Citation[39]. Thyroid volume was associated with family history of thyroid diseases and therefore with genetics Citation[40].

Iodine supply & thyroid volume

In an area with adequate iodine intake, thyroid volume did not change during pregnancy Citation[30]. In a group of women living in a marginally iodine-deficient area and taking 200 µg of iodide, thyroid volume did not change, whereas in the group of women taking 50 µg of iodide, thyroid volume slightly increased during pregnancy Citation[41]. In iodine sufficiency, thyroid volume did not differ between pregnant and nonpregnant women Citation[42]. Berghout and Wiersinga Citation[25] reviewed that in iodine-sufficient areas, thyroid volume did not increase during pregnancy. However, even in iodine sufficiency, there are reports on the increase of thyroid gland volume during pregnancy Citation[39,43,44]. The increase of thyroid volume during pregnancy was followed by the decrease after delivery and on the basis of this finding it was postulated that the increased vascularity may be the reason for the increase of thyroid volume Citation[20].

Hemodynamic changes

Weight gain during pregnancy is predominantly caused by the increase in total body water from 6 to 8 l, of which 4–6 l are extracellular Citation[45]. Pregnancy is therefore characterized by hypervolemia. In healthy adults, thyroid volume is positively correlated with total body water Citation[38]. Increased water volume increases body weight and BMI Citation[46]. Both factors may also increase thyroid volume in pregnancy. By color flow Doppler sonography, an increased intrathyroidal blood flow during pregnancy Citation[47] and a decrease of intrathyroidal blood flow in a year after delivery was found Citation[39]. Authors assumed that, to some extent, thyroid volume increased during pregnancy and decreased after delivery because of hemodynamic changes during pregnancy.

Thyroid autoimmunity in pregnancy & after delivery

The role of pregnancy in triggering of thyroid autoimmunity

Immune adaptations in pregnancy

In order to tolerate the fetus during the intrauterine life, the mother’s immune system undergoes several adjustments. Both maternal systemic suppression and placental immune suppression are involved in preserving the pregnancy, being induced by significant hormonal changes. The key regulatory role is carried out by regulatory CD4+CD25+ T cells (Treg), being important not only in peripheral tolerance against both foreign and self-antigens, but also in fetal tolerance. Treg cells were shown to regulate both Th1-type activity, which leads to cellular immunity, and Th2‑type activity, being involved in humoral immunity Citation[48]. In pregnancy, the expansion of Treg cells is presumably provoked by fetal antigen presentation and estrogen-induced expression of several chemokines. They occur in early pregnancy and they increase rapidly during pregnancy, peaking in the second trimester Citation[49]. Treg cells, accumulated predominantly in decidual tissue and to a lesser extent in peripheral blood Citation[49], were shown to significantly suppress both Th1-type and Th2-type reactions against paternal/fetal alloantigens. However, Th2 clones seem to be less sensitive to this suppression than Th1 clones, leading to predomination of Th2 cells and cytokines over a Th1 cellular immune response, driving the cytokine balance away from the detrimental effects of Th1-cell activity, which may cause fetal loss Citation[50]. Therefore, the maintenance of pregnancy is enabled by proper balance of Th1/Th2 immunity, with a slight shift towards Th2 immunity. This physiological state of lowered immune responsiveness in pregnancy results in amelioration of some pre-existing autoimmune disorders, such as rheumatoid arthritis, multiple sclerosis or thyroid autoimmune disease Citation[51]. During the weeks immediately prior to delivery a clear decline in Treg cells occurs. After delivery, this imbalance in Treg cells and shift of cytokine profile away from Th2 to Th1 during the return to normal pre-pregnancy state may be reflected in exacerbation or aggravation of autoimmunity Citation[52,53].

In pregnancy and postpartum, different types of autoimmune thyroid disease may occur, including Graves’ disease (GD), Hashimoto’s thyroiditis (HT) and postpartum thyroiditis (PPT). Characteristically, thyroid autoantibodies decline during pregnancy, which might be explained by Treg-mediated suppression Citation[52]. After delivery, they return to the pre-pregnant values, frequently ending in postpartum exacerbation of thyroid autoimmunity Citation[54–57].

The role of fetal microchimerism

Fetal microchimerism refers to the phenomena of fetal cell leakage into the mother’s circulation through the placenta during pregnancy. The presence of chimeric male cells has been established in the peripheral blood and maternal tissues, including thyroid Citation[58], and they have been found circulating in mothers several years after delivery Citation[59]. In GD and HT, intrathyroidal fetal microchimeric cells were detected significantly more often than in nonautoimmune thyroid disease Citation[101]. However, large population-based studies found no association between parity and thyroid autoimmunity, arguing against a key role of fetal microchimerism Citation[60,61].

Female sex

Several large epidemiological studies confirmed the female predominance in thyroid autoimmunity, as they present with positive thyroid autoantibodies approximately two- to three-times more often than males Citation[62–64]. Estimation, based on the largest National Health and Nutrition Examination Survey (NHANES) III study, indicated that 17% of females were positive for thyroid peroxidase antibodies (TPOAb), while 15.2% were positive for Tg antibodies (TgAb). Additionally, the prevalence of antibodies was twice as high in white females compared with black females Citation[62].

Besides fetal microchimerism, higher genetic susceptibility for thyroid autoantibody production in females than in males has been reported in the study of Danish twins Citation[65]. X chromosome genes are essential in determining sex hormone levels, as well as in maintaining immune tolerance. Therefore, the alterations in X chromosome, including monosomy or structural abnormalities, and disturbances in X chromosome inactivation with consequent impaired thymic deletion of autoreactive cells, might contribute to the impaired immune response Citation[66].

Risk predisposing factors

Genes

Appropriate genetic background is needed to allow different endogenous and environmental influences to trigger thyroid autoimmunity. Initial observations of higher incidence of thyroid autoimmune disease in families have been recently confirmed by two reports, showing that risk for developing thyroid autoimmune disease was around 16-fold increased in children and siblings of the affected individuals Citation[67,68]. According to the estimation based on Danish twins, genetic influence seems to contribute 73% to thyroid autoantibody positivity Citation[65]. Until now, several putative genes have been identified. Among immune regulatory genes, HLA-DR gene, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) gene, CD40 gene, protein tyrosine phosphatase-22 (PTPN22) gene and CD25 gene have shown an association with thyroid autoimmune disease. Among thyroid-specific genes, major candidates are the gene for Tg and TSH receptor gene Citation[69]. Besides being involved in clinical disease, genetic susceptibility is crucial also for thyroid antibody production. Among putative genes, CTLA-4 was confirmed as a major locus for thyroid antibodies Citation[70], being associated with higher thyroid antibody levels in GD, HT and PPT Citation[71–73].

Iodine intake

The enhancing influence of iodine on thyroid autoimmunity has been confirmed by studies on experimental animal models and also by large observational studies of populations with different iodine intake. Among mechanisms, autoantigenic potency of highly iodinated Tg or iodine toxicity to thyrocytes have been proposed, but the precise mechanism is still unknown Citation[74]. In humans, the improvement of iodine prophylaxis lead to a three–fourfold increase in incidence of thyroid autoimmunity in a population with previously mild iodine deficiency Citation[75]. The prevalence of thyroid antibodies, estimated by large epidemiological studies, was up to 18% in the areas with sufficient iodine intake Citation[62], up to 25% in conditions of excessive iodine intake Citation[63], but only up to 13% in the circumstances of iodine deficiency Citation[64]. High iodine intake in pregnancy was associated with a higher risk of developing PPT Citation[76], but this observation was not supported by other studies showing that iodine supplementation in pregnancy and after delivery is safe even in TPOAb-positive females Citation[41,77].

Other risk factors

Other risk factors, although less frequent in pregnancy and postpartum, might contribute to thyroid autoimmunity in females in the reproductive period. Smokers are at risk for GD Citation[78] and at even greater risk for either development or deterioration of Graves’ orbitopathy Citation[79]. In HT, few early investigations implicated the association with smoking Citation[80], while a recent report indicated even negative relation of smoking with both TPOAb and TgAb as well as with hypothyroidism Citation[81]. Also, data regarding PPT are scarce, with only two studies implying the increased risk in association with smoking Citation[80,82]. Triggers such as stress, infections, environmental toxicants or immune-modulating drugs may contribute to thyroid autoimmunity in the reproductive period equally as in the general population Citation[83].

Thyroid autoimmune disease in pregnancy & after delivery

Graves’ disease

In females in the reproductive period, GD is the most frequent cause of hyperthyroidism, which occurs in the population with an estimated prevalence of approximately 1% Citation[62]. Among pregnant women the prevalence rate of overt hyperthyroidism is approximately 0.1–0.4% and GD accounts for 85–90% of all cases Citation[53]. In this type of thyroid autoimmunity, the humoral immune response predominates with the characteristic appearance of stimulating antibodies against TSH receptor (TRAbs), causing hyperthyroidism, goitre and nonthyroid manifestations, such as Graves’ orbitopathy or dermopathy.

Owing to physiological immunosuppression during pregnancy, the development of GD or relapse of hyperthyroidism in this period is rare, usually emerging in the first trimester of pregnancy. In the second half of pregnancy even the gradual improvement of previously existing hyperthyroidism is frequently observed, being most probably the reflection of the stimulating TRAbs decrease in the second and third trimester Citation[56,57]. In the postpartum period, when the immunosuppression ceases, the increase of stimulating TRAbs Citation[57], together with relapse of GD, is frequently observed, usually between 4 and 8 months after delivery. In the recent study of patients in remission after antithyroid drug treatment, the recurrence of GD was determined in 84% of patients in the postpartum period compared with only 56% of patients not being pregnant Citation[84]. However, as indicated by one single study, the postpartum period itself has not been shown to be a major risk factor for the first onset of GD Citation[85].

Untreated or inadequately treated GD in pregnancy may lead to several detrimental complications. In mothers, hyperthyroidism has been associated with preeclampsia and with the increased risk of congestive heart failure and thyroid storm. In the pregnancy course, hyperthyroidism may increase the risk of miscarriage, stillbirth, preterm delivery and placental abruption. Fetal hyperthyroidism, which occurs in less than 0.01% of pregnancies Citation[86], may lead to tachycardia, fetal goitre, accelerated bone maturation, growth retardation, low birth weight and malformations. In the fetus, the excess of thyroid hormones may be the reflection of the mother’s thyroid hormones or the mother’s stimulating TRAbs crossing the placenta. Those antibodies have the impact on fetus only after the twelfth week of gestation, when the fetal thyroid starts to respond to the stimulation Citation[56,87]. In late pregnancy they represent a risk of neonatal hyperthyroidism, which occurs in up to 5% of newborns of mothers with GD. It usually persists for up to 12 weeks due to slow clearance of maternal antibodies, having a half-life of approximately 3 weeks Citation[56,87].

Hashimoto’s thyroiditis

With the estimated prevalence of 18% in the population, HT is probably one of the most prevalent autoimmune disorders in general. In women in the reproductive period, the prevalence of thyroid antibodies was approximately 10–15% and the prevalence was increasing with age Citation[62]. In contrast to GD, in HT the cell-mediated immune response predominates with consequent gradual destruction of thyroid tissue, which frequently leads to hypothyroidism. Although not playing a significant role in the pathogenesis of the disease, TPOAb and TgAb characteristically appear in the patients’ sera in more than 90% and in up to 80% of cases, respectively Citation[88]. In pregnancy, those antibodies were shown to decline gradually with the lowest values in the third trimester, while the increase was observed as soon as 6 weeks after delivery and returning to the pre-pregnant values 12 weeks after delivery Citation[54,55,89].

In HT, both hypothyroidism and thyroid autoantibodies have been implicated to be involved in pregnancy complications. Overt or subclinical hypothyroidism, occurring in approximately 2–4% of apparently healthy women, has been related to two–threefold increased risk of gestational hypertension, placental abruption, postpartum hemorrhage, preterm delivery or miscarriage. Besides increased risk of low birth weight, neonatal respiratory distress and fetal abnormalities, such as hydrocephalus and hypospadias, maternal hypothyroidism during pregnancy has also been demonstrated to affect neuropsychological development of the child Citation[2,90]. However, rapid and adequate correction of hypothyroidism with L-thyroxine therapy has been shown to improve obstetrical outcome Citation[2,4]. In euthyroid pregnant women, elevated thyroid autoantibodies have been associated with two- to four-fold increased risk of miscarriage and with up to threefold increased risk of preterm delivery, although the etiology remains unresolved. Those complications may be associated with underlying generalized immune imbalance, with subtle deficiency of thyroid hormones due to thyroid autoimmunity, or with older age of those females Citation[90,91].

Hypothyroidism may also lead to infertility, since menstrual irregularities, including oligomenorrhea, menorrhagia and ovulatory dysfunction may occur and their severity correlates with the elevation of serum TSH levels. Similarly, hypothyroidism may provoke in vitro fertilization failure in infertile females, while L-thyroxine replacement has been shown to improve embryo implantation rate and pregnancy outcome Citation[90,92]. However, the clinical importance of thyroid antibodies in infertility remains controversial and underlying pathogenic mechanisms of putative association still need to be clarified Citation[90].

Postpartum thyroiditis

Postpartum thyroiditis refers to thyroid dysfunction within the first year after delivery or miscarriage, when the known immunosuppressive effect of pregnancy disappears. The clinical disease may present with hyperthyroidism alone, only with hypothyroidism, or with hyperthyroidism followed by hypothyroidism. The prevalence varies significantly between studies from 1.1 to 21.1% Citation[93], with estimated pooled prevalence in the general population of approximately 8%, occurring up to six-times more often in females with elevated TPOAb and three-times more often in females with Type 1 diabetes Citation[94]. Therefore, in these two groups screening for thyroid dysfunction is recommended 3 and 6 months after delivery Citation[4]. Females positive for TPOAb in early pregnancy develop PPT in 40–60% of cases, while among patients with PPT 70% present with positive TPOAb, putting them at risk for developing a permanent thyroid dysfunction Citation[54].

The hyperthyroid phase of the disease is only transient, more frequently occurring in TPOAb-negative patients between 1 and 6 months after delivery and lasting 1–2 months. Hypothyroidism may occur with or without a previous hyperthyroid phase, more often in TPOAb-positive patients and between 3 and 8 months after delivery, being caused by destruction of thyroid tissue Citation[54,73]. It may be only transient, lasting 4–6 months and passing within 1 year after delivery or it may be permanent Citation[4]. A few earlier studies reported permanent hypothyroidism in up to 30% of PPT patients Citation[2], but a recent large prospective report demonstrated a significantly higher incidence of approximately 50%. The latter observation might be an overestimation, since owing to limited sampling only 6 and 12 months after delivery a considerable number of patients with transient hypothyroidism may have been missed Citation[95,96]. However, patients with transient hypothyroidism are also at risk for developing permanent hypothyroidism, which is established within 5–10 years after PPT in 20–60% of females Citation[94]. While in the hyperthyroid phase no specific antithyroid therapy is indicated, replacement therapy with L-thyroxine frequently needs to be started in hypothyroid patients Citation[4].

Expert commentary

In pregnancy and the postpartum period, several physiological changes develop, which are reflected in the healthy thyroid gland as well as in autoimmune thyroid disease .

Appropriate adaptation of the healthy thyroid gland to metabolic challenges during pregnancy and lactation is assured by adequate iodine supply. When dealing with thyroid biochemical tests, we should also keep in mind that levels of TSH and free thyroid hormones may vary outside the normal reference ranges both with adequate iodine supply and in the healthy pregnant state. TSH levels, being lower in the first trimester and increasing in the second and third trimester, probably mirror hCG levels. Additionally, higher TSH in the second half of pregnancy may be the reflection of lower free thyroid hormones in this period, although the reason for the latter observation has not yet been established. Several explanations have been proposed, including increased binding capacity for thyroid hormones or methodological problems. In order to reduce the interpretation difficulties in daily clinical practice, the use of gestational-specific reference intervals proposed by several authors seems plausible. Beside biochemical changes, thyroid volume changes in pregnancy remain an open issue. Despite contradictive reports, in our experience thyroid volume increases in pregnancy and decreases after delivery even with sufficient iodine intake, most probably being associated with anthropometric and hemodynamic changes.

Pregnancy represents a challenge not only for thyroid function and size, but also for the immune system. It is important to consider the high prevalence of thyroid autoimmune disease in the reproductive period, being significantly influenced by the modification of immune response in pregnancy and after delivery. Given this view, the understanding of the pathophysiology of this condition is essential for the management of thyroid disorders in this period. Although immunotolerance in pregnancy frequently leads to amelioration of thyroid autoimmunity, the recognition and treatment of thyroid dysfunction is of crucial importance to avoid detrimental complications in mother and child. On the other hand, the exacerbation of thyroid autoimmunity after delivery, when the immune responsiveness increases, is expected and therefore screening for thyroid dysfunction in women with known thyroid disease in this period is required. Nevertheless, the warranty for the universal screening through TSH or even antibody testing in pregnancy remains an unsettled question. By the current evidence, case finding by TSH measurement is recommended in women at risk, including those with thyroid disease or family history of thyroid disease, those with symptoms and signs, women who other autoimmune disorders, like Type I diabetes, women who are infertile or with a history of miscarriage, and those with a history of neck irradiation Citation[4].

Five-year view

It is likely that within the next 5 years more accurate measurements of free thyroid hormones and implementation of gestational-specific reference intervals will enable realistic estimation of thyroid function in pregnancy. Grounds for low free thyroid hormones in that period will probably be experimentally elucidated. Further progress in the evaluation of thyroid volume changes in pregnancy is expected, based upon physiological characteristics of pregnancy with the emphasis on hemodynamic changes. With regards to thyroid autoimmunity during pregnancy and postpartum, a immense progress has been achieved in the understanding of pathophysiological mechanisms in the past decade. In the near future, further evidence will hopefully enable better recognition of women at risk in order to avoid the harmful effects of unrecognized thyroid dysfunction for mother and child.

Table 1. Thyroid physiology and autoimmunity in pregnancy and after delivery.

Key issues

  • • In normal pregnancy, thyroid-stimulating hormone levels may decrease in the first trimester due to human chorionic gonadotropin action, while the low level of free thyroxine in the third trimester remain a matter of debate.

  • • Thyroid volume may increase during pregnancy even in areas with adequate iodine supply, which does not represent an important clinical problem. However, in our opinion, such an observation and novel explanations for this occurrence represent a valuable contribution to the better understanding of thyroid physiology in pregnancy.

  • • Pregnancy itself causes significant variation in immune tolerance, which may trigger thyroid autoimmunity in susceptible individuals with appropriate genetic background, excessive iodine intake and other environmental risk factors.

  • • Graves’ disease rarely occurs in pregnancy and frequently relapses between 4 and 8 months after delivery. If untreated, several detrimental consequences may develop in the mother, fetus and in the pregnancy course.

  • • Hashimoto’s thyroiditis, present in approximately 10–15% of females in the reproductive period, may cause hypothyroidism, associated with several complications for the mother, fetus and the pregnancy course.

  • • Postpartum thyroiditis, occurring in 8% of females in the first year after delivery, usually presents with transient thyroid dysfunction and less frequently ends with permanent hypothyroidism.

References

  • Glinoer D. The regulation of thyroid function in pregnancy: pathways of endocrine adaptation from physiology to pathology. Endocr. Rev.18(3), 404–433 (1997).
  • Kennedy RL, Malabu UH, Jarrod G, Nigam P, Kannan K, Rane A. Thyroid function and pregnancy: before, during and beyond. J. Obstet. Gynaecol.30(8), 774–783 (2010).
  • Burrow GN, Fisher DA, Larsen PR. Maternal and fetal thyroid function. N. Engl. J. Med.331(16), 1072–1078 (1994).
  • Abalovich M, Amino N, Barbour LA et al. Management of thyroid dysfunction during pregnancy and postpartum: an Endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab.92(Suppl. 8), S1–S47 (2007).
  • Laurberg P, Andersen S, Bjarnadóttir RI et al. Evaluating iodine deficiency in pregnant women and young infants – complex physiology with a risk of misinterpretation. Public Health Nutr.10(12A), 1547–1552 (2009).
  • Glinoer D, De Nayer P, Bourdoux P et al. Regulation of maternal thyroid during pregnancy. J. Clin. Endocrinol. Metab.71(2), 276–287 (1990).
  • Schussler GC. The thyroxine-binding proteins. Thyroid10(2), 141–149 (2000).
  • Larsson A, Palm M, Hansson LO, Axelsson O. Reference values for clinical chemistry tests during normal pregnancy. BJOG115(7), 874–881 (2008).
  • Zhou A, Wei Z, Read RJ, Carrell RW. Structural mechanism for the carriage and release of thyroxine in the blood. Proc. Natl Acad. Sci. USA103(36), 13321–13326 (2006).
  • Lee RH, Spencer CA, Mestman JH et al. Free T4 immunoassays are flawed during pregnancy. Am. J. Obstet. Gynecol.200(3), 260 e1–e6 (2009).
  • Kurioka H, Takahashi K, Miyazaki K. Maternal thyroid function during pregnancy and puerperal period. Endocr. J.52(5), 587–591 (2005).
  • Ain KB, Mori Y, Refetoff S. Reduced clearance rate of thyroxine-binding globulin (TBG) with increased sialylation: a mechanism for estrogen-induced elevation of serum TBG concentration. J. Clin. Endocrinol. Metab.65(4), 689–696 (1987).
  • Grün JP, Meuris S, De Nayer P, Glinoer D. The thyrotropic role of human chorionic gonadotropin (hCG) in the early stages of twin (vs. single) pregnancy. Clin. Endocrinol. (Oxf.).46(6), 719–725 (1997).
  • Springer D, Limanova Z, Zima T. Reference intervals in evaluation of maternal thyroid function during the first trimester of pregnancy. Eur. J. Endocrinol.160(5), 791–797 (2009).
  • Shields B, Hill A, Bilous M et al. Cigarette smoking during pregnancy is associated with alterations in maternal and fetal thyroid function. J. Clin. Endocrinol. Metab.94(2), 570–574 (2009).
  • Fister P. Gaberscek S, Zaletel K, Krhin B, Hojker S, Gersak K. Thyroid function in the third trimester of pregnancy and after delivery in an area of adequate iodine intake. Int. J. Gynaecol. Obstet.112(1), 52–55 (2011).
  • Marwaha RK, Chopra S, Gopalakrishnan S et al. Establishment of reference range for thyroid hormones in normal pregnant Indian women. BJOG115(5), 602–606 (2008).
  • Stricker R, Echenard M, Eberhart R et al. Evaluation of maternal thyroid function during pregnancy: the importance of using gestational age-specific reference intervals. Eur. J. Endocrinol.157(4), 509–514 (2007).
  • Soldin OP, Tractenberg RE, Hollowell JG, Jonklaas J, Janicic N, Soldin SJ. Trimester-specific changes in maternal thyroid hormone, thyrotropin, and thyroglobulin concentrations during gestation: trends and associations across trimesters in iodine sufficiency. Thyroid14(12), 1084–1090 (2004).
  • Rasmussen NG, Hornnes PJ, Hegedus L. Ultrasonographically determined thyroid size in pregnancy and postpartum: the goitrogenic effect of pregnancy. Am. J. Obstet. Gynecol.160(5 Pt 1), 1216–1220 (1989).
  • Glinoer D, De Nayer P, Robyn C, Lejeune B, Kinthaert J, Meuris S. Serum levels of intact human chorionic gonadotropin (hCG) and its free α and β subunits, in relation to maternal thyroid stimulation during normal pregnancy. J. Endocrinol. Invest.16(11), 881–888 (1993).
  • Lockwood CM, Grenache DG, Gronowski AM. Serum human chorionic gonadotropin concentrations greater than 400,000 IU/l are invariably associated with suppressed serum thyrotropin concentrations. Thyroid19(8), 863–868 (2009).
  • Haddow JE, McClain MR, Lambert-Messerlian G et al. Variability in thyroid-stimulating hormone suppression by human chorionic gonadotropin during earyl pregnancy. J. Clin. Endocrinol. Metab.93(9), 3341–3347 (2008).
  • Sapin R, D’Herbomez M. Free thyroxine measured by equilibrium dialysis and nine immunoassays in sera with various serum thyroxine-binding capacities. Clin. Chem.49(9), 1531–1535 (2003).
  • Berghout A, Wiersinga W. Thyroid size and thyroid function during pregnancy: an analysis. Eur. J. Endocrinol.138(5), 536–542 (1998).
  • Yu B, Wang QW, Huang RP et al. Establishment of self-sequential longitudinal reference intervals of maternal thyroid function during pregnancy. Exp. Biol. Med. (Maywood).235(10), 1212–1215 (2010).
  • Elnagar B, Eltom A, Wide L et al. Iodine status, thyroid function and pregnancy: study of Swedish and Sudanese women. Eur. J. Clin. Nutr.52(5), 351–355 (1998).
  • Yan YQ, Dong ZL, Wang FR et al. Trimester- and method-specific reference intervals for thyroid tests in pregnant Chinese women: methodology, euthyroid definition and iodine status can influence the setting of reference intervals. Clin. Endocrinol (Oxf.)74(2), 262–269 (2011).
  • Boas M, Forman JL, Juul A et al. Narrow intra-individual variation of maternal thyroid function in pregnancy based on a longitudinal study on 132 women. Eur. J. Endocrinol.161(6), 903–910 (2009).
  • Berghout A, Endert E, Ross A, Hogerzeil HV, Smits NJ, Wiersinga WM. Thyroid function and thyroid size in normal pregnant women living in an iodine replete area. Clin. Endocrinol.41(3), 375–379 (1994).
  • Wiersinga WM. Nonthyroidal illness. In: The Thyroid. Braverman LE, Utiger RD (Eds). Lippincott, Philadelphia, PA, USA, 246–263 (2005).
  • Negro R, Soldin P, Obregon MJ, Stagnaro-Green A. Hypothyroxinemia and pregnancy. Endocr. Prac.17, 1–24 (2011).
  • Sapin R, D’Herbomez M, Schlienger JL. Free thyroxine measured with equilibrium dialysis and nine immunoassays decrease in late pregnancy. Clin. Lab.50(9–10), 581–584 (2004).
  • Yue B, Rockwood AL, Sandrock T, La’ulu SL, Kushnir MM, Meikle AW. Free thyroid hormones in serum by direct equilibrium dialysis and online solid-phase extraction-liquid chromatography/tandem mass spectrometry. Clin. Chem.54(4), 642–651 (2008).
  • Jonklaas J, Kahric-Janicic N, Soldin OP, Soldin SJ. Correlations of free thyroid hormones measured by tandem mass spectrometry and immunoassay with thyroid-stimulating hormone across 4 patient populations. Clin. Chem.55(7), 1380–1388 (2009).
  • Hansen PS, Brix TH, Bennesbaek FN, Bonnema SJ, Kyvik KO, Hegedüs L. Genetic and environmental causes of individual differences in thyroid size: a study of healthy Danish twins. J. Clin. Endocrinol. Metab.89(5), 2071–2077 (2004).
  • Hegedüs L. Thyroid size determined by ultrasound. Influence of physiological factors and non-thyroidal disease. Dan. Med. Bull.37(3), 249–263 (1990).
  • Gómez JM, Maravall FJ, Gómez N, Gumà A, Soler J. Determinants of thyroid volume as measured by ultrasonography in healthy adults randomly selected. Clin. Endocrinol.53(5), 629–634 (2000).
  • Fister P, Gaberšček S, Zaletel K, Krhin B, Geršak K, Hojker S. Thyroid volume changes during pregnancy and after delivery in an iodine-sufficient Republic of Slovenia. Eur. J. Obstet. Gynecol. Reprod. Biol.45(1), 45–48 (2009).
  • Soares R, Vanacor R, Manica D et al. Thyroid volume in associated with family history of thyroid disease in pregnant women with adequate iodine intake: a cross-sectional study in southern Brazil. J. Endocrinol. Invest.31(7), 614–617 (2008).
  • Antonangeli L, Maccherini D, Cavaliere R et al. Comparison of two different doses of iodide in the prevention of gestational goiter in marginal iodine deficiency: a longitudinal study. Eur. J. Endocrinol.147(1), 29–34 (2002).
  • Azizi F, Aminorroya A, Hedayati M, Rezvanian H, Amini M, Mirmiran P. Urinary iodine excretion in pregnant women residing in areas with adequate iodine intake. Pub. Health Nutr.6(1), 95–98 (2003).
  • Nelson M, Wickus GG, Caplan RH et al. Thyroid gland size in pregnancy. An ultrasound and clinical study. J. Reprod. Med.32(12), 888–890 (1987).
  • Brander A, Kivisaari L. Ultrasonography of the thyroid gland during pregnancy. J. Clin. Ultrasound17(6), 403–406 (1989).
  • Davison JM, Lindheimer MD. Volume homeostasis and osmoregualtion in human pregnancy. Baillieres Clin. Endocrinol. Metab.3(2), 451–472 (1989).
  • Schrier RW. Body water homeostasis: clinical disorders of urinary dilution and concentration. J. Am. Soc. Nephrol.17(7), 1820–1832 (2006).
  • Fister P, Gaberšček S, Zaletel, Krhin B, Geršak K, Hojker S. Thyroid volume and intrathyroidal blood flow increase during pregnancy. Clin. Endocrinol. (Oxf.)65(6), 828–829 (2006).
  • Saito S, Nakashima A, Shima T, Ito M. Th1/Th2/Th17 and regulatory T-cell paradigm in pregnancy. Am. J. Reprod. Immunol.63(6), 601–610 (2010).
  • Guerin LR, Prins JR, Robertson SA. Regulatory T-cells and immune tolerance in pregnancy: a new target for infertility treatment? Hum. Reprod. Update15(5), 517–535 (2009).
  • Mjösberg J, Berg G, Ernerudh J, Ekerfelt C. CD4+ CD25+ regulatory T cells in human pregnancy: development of a Treg-MLC-ELISPOT suppression assay and indications of paternal specific Tregs. Immunology120(4), 456–466 (2007).
  • Adams Waldorf KM, Nelson JL. Autoimmune disease during pregnancy and the microchimerism legacy of pregnancy. Immunol. Invest.37(5), 631–644 (2008).
  • Weetman AP. Immunity, thyroid function and pregnancy: molecular mechanisms. Nat. Rev. Endocrinol.6(6), 311–331 (2010).
  • Galofre JC, Davies TF. Autoimmune thyroid disease in pregnancy: a review. J. Womens Health18(11), 1847–1856 (2009).
  • Kuijpens JL, Pop VJ, Vader HL, Drexhage HA, Wiersinga WM. Prediction of post partum thyroid dysfunction: can it be improved? Eur. J. Endocrinol.139(1), 36–43 (1998).
  • Smyth PP, Wijeyaratne CN, Kaluarachi WN et al. Sequential studies on thyroid antibodies during pregnancy. Thyroid15(5), 474–477 (2005).
  • Chan GW, Mandel SJ. Therapy insight: mangement of Graves’ disease during pregnancy. Nat. Clin. Prac. Endocrinol. Metab.3(6), 470–478 (2007).
  • Amino N, Izumi Y, Hidaka Y et al. No increase of blocking type anti-thyrotropin receptor antibodies during pregnancy in patients with Graves’ disease. J. Clin. Endocrinol. Metab.88(12), 5871–5874 (2003).
  • Koopmans M, Kremer Hovinga IC, Baelda HJ et al. Chimerism occurs in thyroid, lung, skin and lymph nodes of women with sons. J. Reprod. Immunol.78(1), 68–75 (2008).
  • Bianchi DW, Zickwolf GK, Weil GJ, Sylvester S, DeMaria MA. Male fetal progenitor cells persist in maternal blood for as long as 27 years postpartum. Proc. Natl Acad. Sci. USA93(2), 705–708 (1996).
  • Bülow Pedersen I, Laurberg P, Knudsen N et al. Lack of association between thyroid autoantibodies and parity in a population study argues against microchimerism as a trigger of thyroid autoimmunity. Eur. J. Endocrinol.154(1), 39–45 (2006).
  • Friedrich N, Schwarz S, Thonack J, John U, Wallaschofski H, Völzke H. Association between parity and autoimmune thyroiditis in a general female population. Autoimmunity41(2), 174–180 (2008).
  • Hollowell JG, Staehling NW, Flanders WD et al. Serum TSH, T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J. Clin. Endocrinol. Metab.87(2), 489–499 (2002).
  • Kasagi K, Takahashi N, Inoue G, Honda T, Kawachi Y, Izumi Y. Thyroid function in Japanese adults as assessed by a general health checkup system in relation with thyroid-related antibodies and other clinical parameters. Thyroid19(9), 937–944 (2009).
  • Aghini-Lombardi F, Antonangeli L, Martino E et al. The spectrum of thyroid disorders in an iodine-deficient community: the Pescopagano survey. J. Clin. Endocrinol. Metab.84(2), 561–566 (1999).
  • Hansen PS, Brix TH, Iachine I, Kyvik KO, Hegedüs L. The relative importance of genetic and environmental effects for the early stages of thyroid autoimmunity: a study of healthy Danish twins. Eur. J. Endocrinol.154(1), 29–38 (2006).
  • Invernizzi P, Pasini S, Selmi C, Gershwin ME, Podda M. Female predominance and X chromosome defects in autoimmune diseases. J. Autoimmun.33(1), 12–16 (2009).
  • Dittmar M, Libich C, Brenzel T, Kahaly GJ. Increased familial clustering of autoimmune thyroid diseases. Horm. Metab. Res.43(3), 200–204 (2011).
  • Villanueva R, Greenberg DA, Davies TF, Tomer Y. Sibling recurrence risk in autoimmune thyroid disease. Thyroid13(8), 761–764 (2003).
  • Tomer Y. Genetic susceptibility to autoimmune thyroid disease: past, present, and future. Thyroid20(7), 715–725 (2010).
  • Ban Y, Greenberg DA, Davies TF, Jacobson E, Concepcion E, Tomer Y. Linkage analysis of thyroid antibody production: evidence for shared susceptibility to clinical autoimmune thyroid disease. J. Clin. Endocrinol. Metab.93(9), 3589–3596 (2008).
  • Zaletel K, Krhin B, Gaberšček S, Pirnat E, Hojker S. The influence of the exon 1 polymorphism of the cytotoxic T lymphocyte antigen 4 gene on thyroid antibody production in patients with newly diagnosed Graves’ disease. Thyroid12(5), 373–376 (2002).
  • Zaletel K, Krhin B, Gaberšček S, Hojker S. Thyroid autoantibody production is influenced by exon 1 and promoter CTLA-4 polymorphisms in patients with Hashimoto’s thyroiditis. Int. J. Immunogenet.33(2), 87–91 (2006).
  • Zaletel K, Krhin B, Gaberšček S, Biček A, Pajič T, Hojker S. Association of CT60 cytotoxic T lymphocyte antigen-4 gene polymorphism with thyroid autoantibody production in patients with Hashimoto’s and postpartum thyroiditis. Clin. Exp. Immunol.161(1), 41–47 (2010).
  • Fountoulakis S, Philippou G, Tsatsoulis A. The role of iodine in the evolution of thyroid disease in Greece: from endemic goiter to thyroid autoimmunity. Hormones (Athens)6(1), 25–35 (2007).
  • Heydarian P, Ordookhani A, Azizi F. Goiter rate, serum thyrotropin, thyroid autoantibodies and urinary iodine concentration in Tehranian adults before and after national salt iodization. J. Endocrinol. Invest.30(5), 404–410 (2007).
  • Guan H, Li C, Li Y et al. High iodine intake is a risk factor of post-partum thyroiditis: result of a survey from Shenyang, China. J. Endocrinol. Invest.28(10), 876–881 (2005).
  • Nøhr SB, Jørgensen A, Pedersen KM, Laurberg P. Postpartum thyroid dysfunction in pregnant thyroid peroxidase antibody-positive women living in an area with mild to moderate iodine deficiency: is iodine supplementation safe? J. Clin. Endocrinol. Metab.85(9), 3191–3198 (2000).
  • Holm IA, Manson JE, Michels KB, Alexander EK, Willett WC, Utiger RD. Smoking and other lifestyle factors and the risk of Graves’ hyperthyroidism. Arch. Intern. Med.165(14), 1606–1611 (2005).
  • Stan MN, Bahn RS. Risk factors for development or deterioration of Graves’ ophthalmopathy. Thyroid20(7), 777–783 (2010).
  • Vestergaard P. Smoking and thyroid disorders – a meta-analysis. Eur. J. Endocrinol.146(2), 153–161 (2002).
  • Pedersen IB, Laurberg P, Knudsen N et al. Smoking is negatively associated with the presence of thyroglobulin autoantibody and to a lesser degree with thyroid peroxidase autoantibody in serum: a population study. Eur. J. Endocrinol.158(3), 367–373 (2008).
  • Galanti MR, Cnattingius S, Granath F, Ekbom-Schnell A, Ekbom A. Smoking and environmental iodine as risk factors for thyroiditis among parous women. Eur. J. Epidemiol.22(7), 467–472 (2007).
  • Prummel MF, Strieder T, Wiersinga WM. The environment and autoimmune thyroid diseases. Eur. J. Endocrinol.150(5), 605–618 (2004).
  • Rotondi M, Cappelli C, Pirali B et al. The effect of pregnancy on subsequent relapse from Graves’ disease after a successful course of antithyroid drug therapy. J. Clin. Endocrinol. Metab.93(10), 3985–3988 (2008).
  • Rotondi M, Pirali B, Lodigiani S et al. The post partum period and the onset of Graves’ disease: an overestimated risk factor. Eur. J. Endocrinol.159(2), 161–165 (2008).
  • Nachum Z, Rakover Y, Weiner E, Shalev E. Graves’ disease in pregnancy: prospective evaluation of a selective invasive treatment protocol. Am. J. Obstet. Gynecol.189(1), 159–165 (2003).
  • Laurberg P, Bournaud C, Karmisholt J, Orgiazzi J. Eur. J. Endocrinol.160(1), 1–8 (2009).
  • Zaletel K. Determinants of thyroid autoantibody production in Hashimoto’s thyroiditis. Expert Rev. Clin. Immunol.3(2), 217–223 (2007).
  • Feki M, Omar S, Menif O et al. Thyroid disorders in pregnancy: frequency and association with selected diseases and obstetrical complications in Tunisian women. Clin. Biochem.41(12), 927–931 (2008).
  • Poppe K, Velkeniers B, Glinoer D. The role of thyroid autoimmunity in fertility and pregnancy. Nat. Clin. Prac. Endocrinol. Metab.4(7), 394–405 (2008).
  • Stagnaro-Green A. Maternal thyroid disease and preterm delivery. J. Clin. Endocrinol. Metab.94(1), 21–25 (2009).
  • Kim CH, Ahn JW, Kang SP, Kim SH, Chae HD, Kang BM. Effect of levothyroxine treatment on in vitro fertilization and pregnancy outcome in infertile women with subclinical hypothyroidism undergoing in vitro fertilization/intracytoplasmic sperm injection. Fertil. Steril.95(5), 1650–1654 (2011).
  • Muller AF, Drexhage HA, Berghout A. Postpartum thyroiditis and autoimmune thyroiditis in women of childbearing age: recent insights and consequences for antenatal and postnatal care. Endocr. Rev.22(5), 605–630 (2001).
  • Nicholson WK, Robinson KA, Smallridge RC, Ladenson PW, Powe NR. Prevalence of postpartum thyroid dysfunction: a quantitative review. Thyroid16(6), 573–582 (2006).
  • Stagnaro-Green A, Schwartz A, Gismondi R, Tinelli A, Mangieri T, Negro R. High rate of persistent hypothyroidism in a large-scale prospective study of postpartum thyroiditis in Southern Italy. J. Clin. Endocrinol. Metab.96(3), 652–657 (2011).
  • Lazarus JH. The continuing saga of postpartum thyroiditis. J. Clin. Endocrinol. Metab.96(3), 614–616 (2011).

Website

Thyroid physiology and autoimmunity in pregnancy and after delivery

To obtain credit, you should first read the journal article. After reading the article, you should be able to answer the following, related, multiple-choice questions. To complete the questions (with a minimum 70% passing score) and earn continuing medical education (CME) credit, please go to http://www.medscape.org/journal/expertimmunology. Credit cannot be obtained for tests completed on paper, although you may use the worksheet below to keep a record of your answers. You must be a registered user on Medscape.org. If you are not registered on Medscape.org, please click on the New Users: Free Registration link on the left hand side of the website to register. Only one answer is correct for each question. Once you successfully answer all post-test questions you will be able to view and/or print your certificate. For questions regarding the content of this activity, contact the accredited provider, [email protected]. For technical assistance, contact [email protected]. American Medical Association’s Physician’s Recognition Award (AMA PRA) credits are accepted in the US as evidence of participation in CME activities. For further information on this award, please refer to http://www.ama-assn.org/ama/pub/category/2922.html. The AMA has determined that physicians not licensed in the US who participate in this CME activity are eligible for AMA PRA Category 1 Credits™. Through agreements that the AMA has made with agencies in some countries, AMA PRA credit may be acceptable as evidence of participation in CME activities. If you are not licensed in the US, please complete the questions online, print the AMA PRA CME credit certificate and present it to your national medical association for review.

Activity Evaluation

1. A 25-year-old woman presents for her first prenatal visit. Based on the review by Drs. Gaberšček and Zaletel, which of the following thyroid changes are you most likely to expect during her pregnancy?

  • A Thyroid-stimulating hormone (TSH) levels may increase in the first trimester

  • B Free T4 (fT4) levels may be low in the first trimester

  • C Thyroid volume may decrease

  • D Synthesis of thyroid hormones increases by up to 50% because of estrogen-induced increase in thyroxine-binding globulin (TBG) concentration

2. Based on the review by Drs. Gaberšček and Zaletel, which of the following changes in immune tolerance would you most likely expect in the patient described in question 1?

  • A Regulatory CD4+ CD25+ T cells (Treg) rapidly decrease during pregnancy

  • B As pregnancy progresses, T-helper 1 (Th1) cellular immune response predominates over Th2 cells and cytokines

  • C Lowered immune responsiveness in pregnancy may result in amelioration of pre-existing rheumatoid arthritis, multiple sclerosis, thyroid autoimmune disease, or other autoimmune disorders

  • D During the last few weeks of pregnancy there is a sudden increase in Treg cells

3. Based on the above review by Drs. Gaberšček and Zaletel, which of the following statements about characteristics and complications of thyroid disorders during pregnancy is most likely correct?

  • A Graves’ disease is common during pregnancy

  • B Hashimoto’s thyroiditis is the most frequent cause of hyperthyroidism during pregnancy

  • C Postpartum thyroiditis occurs in about 8% of women in the first year after delivery

  • D Postpartum thyroiditis usually results in permanent hypothyroidism

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.