Skip to main content
Erschienen in: Reproductive Biology and Endocrinology 1/2016

Open Access 01.12.2016 | Research

What affects functional ovarian reserve, thyroid function or thyroid autoimmunity?

verfasst von: Andrea Weghofer, David H. Barad, Sarah Darmon, Vitaly A. Kushnir, Norbert Gleicher

Erschienen in: Reproductive Biology and Endocrinology | Ausgabe 1/2016

Abstract

Background

Thyroid dysfunction is the most common autoimmune endocrine disorder in women of reproductive age, and is associated with menstrual irregularities, anovulation and infertility. Whether it is thyroid function or thyroid autoimmunity that affects functional ovarian reserve (FOR, i.e., the small growing ovarian follicle pool) reflected in anti-Müllerian hormone (AMH) has, however, remained under dispute.

Methods

We investigated in 225 infertile women whether thyroid function, after adjustment for thyroid autoimmunity, affects FOR within what is considered normal thyroid function (TSH, 0.4–4.5μIU/mL) by assessing AMH levels in reference to TSH levels, stratified for TSH < or ≥3.0μIU/mL. Thyroid autoimmunity was defined by presence of anti-thyroid peroxidase, −thyroglobulin and/or -thyroid receptor antibodies.

Results

Mean age of studied women was 38.4 ± 5.0 years; their mean AMH was 1.3 ± 2.0 ng/mL and mean TSH 1.8 ± 0.9 μIU/mL. Thyroid autoimmunity was present in 11.1 % of patients. Women with TSH <3.0μIU/mL presented with significantly higher AMH compared to those with TSH ≥3.0μIU/Ml (P = 0.03). This difference remained significant after adjustment for thyroid autoimmunity as well as age (P = 0.02).

Conclusions

Even after adjustment for thyroid autoimmunity and age, TSH <3.0μIU/mL in euthyroid infertility patients is associated with significantly better FOR (higher AMH) than TSH ≥3.0μIU/mL. This observation suggests a direct beneficial effect of lower TSH levels on follicular recruitment, and warrants investigations of thyroxin supplementation in infertile women with TSH levels ≥3.0μIU/mL in attempts to improve FOR.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Study concept: A.W., D.H.B., V.A.K., N.G.; Study execution: All authors; Data analysis and statistical evaluation; A.W., H.B., S.D.; Manuscript preparation: A.W, N.G.; Final manuscript approval: All authors; Study supervision: N.G.; A.W.

Background

Thyroid dysfunction is the most common endocrine disorder in women of reproductive age. Overt and subclinical hypothyroidism may cause menstrual irregularities and anovulation [1], and has been associated with female infertility [2]. These observations have led to the commonly adopted clinical practice of supplementing women trying to conceive with thyroxin if their TSH levels are ≥2.5 μIU/mL [3]. Investigations of this TSH cut off resulted, however, in conflicting results [46]: Reh et al., reported comparable pregnancy and delivery rates in euthyroid in vitro fertilitzation (IVF) patients with TSH cut offs of 2.5 μIU/mL and 4.5 μIU/mL [7], while Murto et al. identified TSH <2.5 μIU/mL and anti-Müllerian hormone (AMH) ≥10pmol/L (1.4 ng/ml) as significant predictors of live births in women with unexplained infertility [8].
Based on such diverging findings, some suggested that a TSH 2.5 μIU/mL may be too low to impair reproductive function [9]; others suggested that, as estrogen concentrations rise during follicular development, more thyroxin is bound, leaving less free thyroid hormone available for clinical utilization. As a result, TSH levels increase [10, 11].
During controlled ovarian hyperstimulation (COH), TSH increases 50–80 % from cycle start to ovulation induction with human chorionic gonadotropin (hCG), though most patients will around ovulation still experience TSH concentrations < 2.5 μIU/mL [9, 12]. Those with initial TSH above 3.0 μIU/mL at cycle start will, however, likely experience significant clinical hypothyroidism.
How abnormal thyroid function affects female fertility remains unknown. Reported associations with menstrual irregularities and anovulation in hypothyroidism suggest that thyroid dysfunction might impair follicular growth and maturation [13]. Assuming this to be the case, TSH levels should influence AMH concentrations, independent of thyroid autoimmunity and female age. A recent report by Busnelli et al. supports such an assumption by demonstrating increasing TSH levels during COH, and a trend toward lower AMH concentrations in women with TSH levels ≥ 2.5μIU/mL seeking fertility treatments [12].
AMH is produced by small growing follicles, from primary up to small antral follicles and, therefore, reflects what is called functional ovarian reserve (FOR) [14]. If thyroid function affects follicular growth and development, higher AMH concentrations should be observed in women with lower TSH levels independent of thyroid autoimmunity and female age. This study was designed to investigate this hypothesis.

Methods

Study population

This study investigated 225 infertile women who between July 2009 and March 2014 underwent an initial work up prior to fertility treatments at the Center for Human Reproduction (CHR) in New York. Only women with normal TSH levels (i.e., TSH 0.4–4.5μIU/mL) were eligible for enrollment. As AMH levels decrease under oral contraceptives (OC) and during pregnancy [15, 16], pregnancy and OC usage up to three months prior to testing served as exclusion criteria.

Laboratory assays

As part of routine pre-IVF evaluations, AMH, TSH and thyroid antibody status, i.e. anti-thyroid peroxidase (TPO), −thyroglobulin (TG) and –thyroid receptor antibodies are assessed. TSH assessments were made in blood serum, using standard commercial third generation electrochemiluminescence immunoassay, with reference values of 0.4–4.5 μIU/mL as normal range. This normal TSH range was subdivided into low- and high- normal ranges, based on TSH levels < or ≥ 3.0 μIU/mL. AMH levels were assessed in blood serum by a 2nd generation Beckman Coulter assay, with values being log-transformed to account for skew among study subjects. Thyroid autoantibodies were assessed in blood serum, utilizing electrochemiluminescence immunoassay.

Ovarian stimulation

COH for IVF was strictly standardized, with only two ovarian stimulation protocols in use: Women with normal FOR (NFOR) under age 38 were stimulated in long agonist cycles with 150–300 IU of gonadotropins daily; while women above age 38 and with low FOR (LFOR) for age (defined as outside of the 95 % CI for age of FSH and/or AMH) were stimulated in a microdose agonist cycle with 450–600 IU of gonadotropins daily. Both study groups involved similar distributions of both stimulation protocols.

Statistics

Low-normal and high-normal TSH was compared using a two-sampled t-test for continuous variables and a two-sided Fisher exact test for categorical variables. Since there was a statistical difference in AMH and thyroid autoimmunity between TSH classifications, a multiple logistic regression model was used, with AMH and thyroid autoimmunity predicting TSH classification. This model passed the Hosmer-Lemeshow goodness-of-fit test.
Analyses were conducted using SPSS 22.0 under the supervision of the center’s senior medical statistician (SD). A P-value <0.05 was considered statistically significant.

Institutional review board

The presented data only involved retrospective review of medical records and data retrieval from an anonymized research database. Patients at CHR sign an informed consent at initial consultation, which allows for such reviews if the patient’s medical record remains confidential and her identity protected. These conditions were met in this case, allowing for expedited review and approval by the Institutional Review Board (IRB) of The Center for Human Reproduction, New York, NY.

Results

Patient characteristics are presented in Table 1. Mean age for study participants was 38.4 ± 5.0 years. Mean AMH levels were 1.3 ± 2.0 ng/mL, mean TSH levels were 1.8 ± 0.9 μIU/mL. Body mass index (BMI) and ethnicity were comparable between TSH groups (p = 0.58 and p = 0.49). Thyroid autoimmunity was present in 11.1 % of all patients. TPO antibodies were positive in 11.1 % of patients and TG antibodies in 1.8 % of women. There were no patients with thyroid receptor antibodies. Patients with low-normal and high-normal TSH levels were of comparable age (i.e. 38.3 ± 5.1 vs. 38.9 ± 4.4 years, P = 0.67). Women with high-normal TSH levels presented with thyroid autoimmunity in 26.9 %, while thyroid autoimmunity was present in only 9.0 % of women with low-normal TSH levels (P = 0.01). Thyroid autoimmunity was mainly attributed to TPO antibodies (Table 1).
Table 1
Patient characteristics of 225 women who underwent work up for in vitro fertilization
 
All (n = 225)
TSH < 3μIU/mL (n = 199)
TSH ≥ 3μIU/mL (n = 26)
P-value
Female age (years)
38.4 ± 5.0
38.3 ± 5.1
38.9 ± 4.4
0.67
TSH (μIU/mL)
1.8 ± 0.9
1.6 ± 0.6
3.5 ± 0.5
 
Thyroid autoimmunity (%)
11.1 %
9.0 %
26.9 %
0.01
Thyroid peroxidase antibodies (%)
11.1 %
9.0 %
26.9 %
0.02
Thyroglobulin antibodies (%)
1.8 %
1.0 %
0.8 %
0.10
AMH (ng/mL)
1.3 ± 2.0
1.4 ± 2.0
0.8 ± 1.8
0.02
 Positive Thyroid autoimmunity
0.9 ± 1.3
1.1 ± 1.5
0.5 ± 0.6
0.45
 Negative Thyroid autoimmunity
1.4 ± 2.1
1.4 ± 2.1
0.9 ± 2.1
0.04
Values are presented as means ± standard deviation
Women with TSH <3.0 μIU/mL presented with significantly higher mean AMH levels (1.4 ± 2.0 ng/mL) than those with TSH ≥3.0 μIU/mL (0.8 ± 1.8 ng/mL; P = 0.02). Those findings remained significant when the analysis was adjusted for presence or absence of autoimmunity and age (P = 0.02) (Table 2).
Table 2
Logistic regression model for the probability of TSH ≥ 3 μIU/mL in 225 euthyroid infertile women
 
Estimate
OR
p-value
Thyroid autoimmunity
1.23
3.41
0.02
log (AMH)
−0.40
0.67
0.03
Not surprisingly, thyroid autoimmunity was found to be a significant predictor of TSH classification. The estimated odds of a patient with positive thyroid autoimmunity exhibiting a high-normal TSH was 3.14 times that of a patient with negative thyroid autoimmunity and the same AMH level. Conversely, for a single unit log increase in AMH, odds of having a high-normal TSH level decreased by 33 % after controlling for thyroid autoimmunity.

Discussion

Whether and, if so how, thyroid function affects FOR has remained unresolved. We in this study attempted to pierce out how thyroid function, thyroid autoimmunity and, possibly age, may interphase in answering this question. In this context it is important to note that this study was performed in women within euthyroid range to exclude secondary effects of cofounders that may be associated with abnormal thyroid function, like for example hyperprolactinemia.
Consequently, we here, even within normal thyroid function levels, are able to report a significant association of TSH levels with FOR, as assessed by AMH. What is generally considered sub-clinical hypothyroidism, reflected in TSH levels of ≥3.0μIU/mL, therefore, already appears to exert negative effects on FOR, resulting in significantly lower AMH levels. That this effect is thyroid function-dependent and not a consequence of thyroid autoimmunity was demonstrated in the study by the complete absence of any effects of adjustment for thyroid autoimmunity on the significance of the findings. Significance was also not affected by age, thus suggesting that here observed association of thyroid function with FOR holds at all ages. However, women included in our study had a mean age of 38.4 ± 5 years. It would therefore be interesting to see whether the lack of association can be confirmed in a study group of young women.
Here reported data are supported by a very recent publication by Kuroda et al., who also reported a clear statistical association between elevated TSH levels and decreases in AMH concentrations [17]. This Japanese study, however, investigated this association in infertile as well as fertile women, and unrestricted to the normal TSH range. Combined, both studies, thus, suggest that hypothyroidism already from very mild stages on in all women negatively affects FOR.
These findings explain why thyroid hormone supplementation has been reported to improve pregnancy potential in euthyroid women with high-normal TSH levels [12]. They, however, also contribute to the ongoing controversy in the medical literature whether thyroid function or thyroid autoimmunity are more important in affecting female reproduction [18]. Here presented data suggest that thyroid function appears to have the upper hand - at least when it comes to effects on FOR. One may, however, argue that thyroid autoimmunity has an impact on thyroid function, as tissue damage results in TSH level increases in women with thyroid autoantibodies [19]. Karmisholt et al. demonstrated highly significant associations between TPO-autoantibodies and TSH increase. They also reported that a considerable number of TPO positive women showed declining thyroid function over one year while none of the study subjects presented with improved TSH levels [20].
FOR is defined as the cohort of small growing follicles, consistently recruited from a woman’s primordial follicle pool. Though the follicular recruitment process in the human experience is not fully understood yet, the number of follicles recruited within a given time unit are believed to directly relate to the size of a woman’s primordial follicle pool [21]. As the granulosa cells of small growing follicles produce AMH, this hormone, except at extreme ages, is increasingly considered the most accurate marker of the growing follicle pool and, therefore, of ovarian function [14]. In general, higher AMH concentrations are associated with larger oocyte yields and improved pregnancy potential [22].
Our here reported results are indirectly also supported by previous reports on negative impact of hypothyroidism on prolactin, gonadotropin releasing hormone and sex steroid levels [23]. Moreover, the human ovary contains specific binding sites for thyroxin, which suggests direct effects of thyroid hormone [24].
Here reported data raise the intriguing possibility that thyroxin supplementation prior to fertility treatments in women with even only high-normal TSH levels (i.e., sub-clinical hypothyroidism) and certainly in women with overt hypothyroidism, may enhance follicular recruitment and/or growth at small growing follicle stages. This, as an automatic consequence, would lead to larger oocyte and embryos yields with in vitro fertilization (IVF) and, therefore, as one would assume, also to higher cumulative pregnancy rates.
A report by Revelli et al. a number of years ago, however, confirmed these assumptions only partially by demonstrating larger oocyte yields after levothyroxin supplementation in euthyroid fertility patients with thyroid autoimmunity; but pregnancy rates improved only when thyroxin supplementation was combined with acetylsalicylic acid and prednisolone treatments [25].
If confirmed, Revelli’s data in combination with here reported outcomes would suggest that hypothyroidism negatively affects female fertility by reducing FOR, which may be correctable by thyroxin supplementation. Frequently associated autoimmunity and/or abnormalities in inflammatory pathways, however, negatively affect implantation potential or even increase miscarriage risks, both in the literature strongly associated with thyroid disease [21]. Unfortunately, the number of patients who demonstrated thyroid autoimmunity in this study was too small to assess effects of autoimmunity outright on clinical pregnancy and live birth rates. A meta-analysis by Toulis et al., however, supports these conclusions by confirming higher miscarriage rates in euthyroid IVF patients with thyroid autoimmunity [26].
Not everybody agrees, however, with the conclusion that hypothyroidism is associated with poor FOR. Based on a very recent cross-sectional study in Belgium, Polyzos et al. reported that neither functional hypothyroid nor thyroid autoimmunity in their patient population were statistically associated with FOR [27].
Their findings are, however, not only difficult to integrate with here reported findings but also with above cited reports in the literature. Moreover, they, themselves, note that their study design did not preclude significant patient selection biases. In addition, their study failed to statistically adjust outcomes for cofounders. A 2012 meta-analysis, in contrast, supports the contention that supplementation with L-thyroxin improves clinical pregnancy outcomes in association with IVF in women with subclinical hypothyroidism and/or thyroid autoimmunity [28].
Magri et al. in the same year concluded that IVF outcomes were negatively influenced by autoimmune thyroid disease but that keeping TSH <2.5mIU/mL may negate such effects [29]. A Turkish group actually reported increased AMH levels in association with Hashimoto’s thyroiditis, and, therefore, suggested that this autoimmune thyroiditis and polycystic ovary syndrome (PCOS) may share a common etiology [30]. As previously noted, Japanese colleagues also very recently reported an association between elevated TSH and low AMH levels [17], Saglam et al., in contrast, after age-adjustment reported a strong statistical association (P = 0.008) between autoimmune thyroid disease and AMH levels [31]. Finally, Magri et al. in a very recent follow up study to above cited 2012 study, not surprisingly reported that the likelihood of poor response to ovarian hyperstimulation with gonadotropins (i.e., of poor FOR) was high in women with low AMH but, apparently, not related to autoimmune thyroid disease; when FOR was considered “good,” autoimmune thyroid disease was, however, associated with decreased response to ovarian stimulation [32]. This, of course, is also an anticipated finding because once FOR falls below a minimal threshold, further effects of thyroid autoimmunity can no longer be expected to be visible.
A principal reason why the here presented study was limited to patients in euthyroid range was, indeed, to avoid cofounders that, unrecognized, may affect study results. Our hypothesis was that in this normal range the respective influences of thyroid function and autoimmunity might be the easiest to dissect since secondary effects on ovarian function, like at very low levels of FOR in Magri’s study [32], would be less likely.
The clarity and statistical power of here reported results, their consistency after adjustments for autoimmunity and age appear to support our hypothesis. Further research on the subject, however, appears urgently needed, considering the very obvious difficulties in differentiating effect of thyroid function and autoimmunity on FOR independently.
Such research, however, as this and above cited studies suggest, has to consider the likelihood that thyroid function and thyroid autoimmunity affect female fertility via different effects and/or pathways. Studies, therefore, have to be designed accordingly, by statistically controlling for distinctively contributing factors, such as FOR, thyroid autoimmunity and age; but also with attention paid to how ovaries are being stimulated, and what other potential cofounders may be present. Considering the multiple cofounders that may play a role, only a multicenter study, involving much larger patient cohorts than are usually available to single IVF centers will, therefore, be able to resolve matters.

Conclusion

The present study offers solid statistical evidence that thyroid function, at least in euthyroid range, plays a more important role in affecting FOR than thyroid autoimmunity. This finding further suggests that, even within what generally is considered normal euthyroid TSH range, close attention should be paid in infertile women to TSH values.

Acknowledgements

None

Data availability statement

All data are available from CHR’s data depositor by contacting Ms. Jolanta Tapper at jtapper@thechr.com.

Funding

This work was supported by grants from the Foundation for Reproductive Medicine and intramural support from the Center for Human Reproduction (CHR) – New York. The funders had no role in study design, data collection, and analysis, decision to publish or preparation of the manuscript.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Study concept: A.W., D.H.B., V.A.K., N.G.; Study execution: All authors; Data analysis and statistical evaluation; A.W., H.B., S.D.; Manuscript preparation: A.W, N.G.; Final manuscript approval: All authors; Study supervision: N.G.; A.W.
Literatur
1.
Zurück zum Zitat Krassas GE, Pontikides N, Kaltsas T, Papadopoulou P, Paunkovic J, Paunkovic N, Duntas LH. Disturbances of menstruation in hypothyroidism. Clin Endocrinol (Oxf). 1999;50:655–9.CrossRef Krassas GE, Pontikides N, Kaltsas T, Papadopoulou P, Paunkovic J, Paunkovic N, Duntas LH. Disturbances of menstruation in hypothyroidism. Clin Endocrinol (Oxf). 1999;50:655–9.CrossRef
2.
Zurück zum Zitat Poppe K, Velkeniers B. Female infertility and the thyroid. Best Pract Res Clin Endocrinol Metab. 2004;18:153–65.CrossRefPubMed Poppe K, Velkeniers B. Female infertility and the thyroid. Best Pract Res Clin Endocrinol Metab. 2004;18:153–65.CrossRefPubMed
3.
Zurück zum Zitat Mintziori G, Anagnostis P, Toulis KA, Goulis DG. Thyroid diseases and female reproduction. Minerva Med. 2012;103:47–62.PubMed Mintziori G, Anagnostis P, Toulis KA, Goulis DG. Thyroid diseases and female reproduction. Minerva Med. 2012;103:47–62.PubMed
4.
Zurück zum Zitat Reid SM, Middleton P, Cossich MC, Crowther CA, Bain E. Interventions for clinical and subclinical hypothyroidism pre-pregnancy and during pregnancy. Cochrane Database Syst Rev. 2013;5:CD007752.PubMed Reid SM, Middleton P, Cossich MC, Crowther CA, Bain E. Interventions for clinical and subclinical hypothyroidism pre-pregnancy and during pregnancy. Cochrane Database Syst Rev. 2013;5:CD007752.PubMed
5.
Zurück zum Zitat Negro R, Schwartz A, Gismondi R, Tinelli A, Mangieri T, Stagnaro-Green A. Thyroid antibody positivity in the first trimester of pregnancy is associated with negative pregnancy outcomes. J Clin Endocrinol Metab. 2011;96:E920–924.CrossRefPubMed Negro R, Schwartz A, Gismondi R, Tinelli A, Mangieri T, Stagnaro-Green A. Thyroid antibody positivity in the first trimester of pregnancy is associated with negative pregnancy outcomes. J Clin Endocrinol Metab. 2011;96:E920–924.CrossRefPubMed
6.
Zurück zum Zitat Fumarola A, Grani G, Romanzi D, Del Sordo M, Bianchini M, Aragona A, Tranquilli D, Aragona C. Thyroid function in infertile patients undergoing assisted reproduction. Am J Reprod Immunol. 2013;70:336–41.CrossRefPubMed Fumarola A, Grani G, Romanzi D, Del Sordo M, Bianchini M, Aragona A, Tranquilli D, Aragona C. Thyroid function in infertile patients undergoing assisted reproduction. Am J Reprod Immunol. 2013;70:336–41.CrossRefPubMed
7.
Zurück zum Zitat Reh A, Grifo J, Danoff A. What is a normal thyroid-stimulating hormone (TSH) level? Effects of stricter TSH thresholds on pregnancy outcomes after in vitro fertilization. Fertil Steril. 2010;94:2920–2.CrossRefPubMed Reh A, Grifo J, Danoff A. What is a normal thyroid-stimulating hormone (TSH) level? Effects of stricter TSH thresholds on pregnancy outcomes after in vitro fertilization. Fertil Steril. 2010;94:2920–2.CrossRefPubMed
8.
Zurück zum Zitat Murto T, Bjuresten K, Landgren BM, Stavreus-Evers A. Predictive value of hormonal parameters for live birth in women with unexplained infertility and male infertility. Reprod Biol Endocrinol. 2013;11:61.CrossRefPubMedPubMedCentral Murto T, Bjuresten K, Landgren BM, Stavreus-Evers A. Predictive value of hormonal parameters for live birth in women with unexplained infertility and male infertility. Reprod Biol Endocrinol. 2013;11:61.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Benaglia L, Busnelli A, Somigliana E, Leonardi M, Vannucchi G, De Leo S, Fugazzola L, Ragni G, Fedele L. Incidence of elevation of serum thyroid-stimulating hormone during controlled ovarian hyperstimulation for in vitro fertilization. Eur J Obstet Gynecol Reprod Biol. 2014;173:53–7.CrossRefPubMed Benaglia L, Busnelli A, Somigliana E, Leonardi M, Vannucchi G, De Leo S, Fugazzola L, Ragni G, Fedele L. Incidence of elevation of serum thyroid-stimulating hormone during controlled ovarian hyperstimulation for in vitro fertilization. Eur J Obstet Gynecol Reprod Biol. 2014;173:53–7.CrossRefPubMed
10.
Zurück zum Zitat Stuckey BG, Yeap D, Turner SR. Thyroxine replacement during super-ovulation for in vitro fertilization: a potential gap in management? Fertil Steril. 2010;93:2414 e2411-2413.CrossRef Stuckey BG, Yeap D, Turner SR. Thyroxine replacement during super-ovulation for in vitro fertilization: a potential gap in management? Fertil Steril. 2010;93:2414 e2411-2413.CrossRef
11.
Zurück zum Zitat Scoccia B, Demir H, Kang Y, Fierro MA, Winston NJ. In vitro fertilization pregnancy rates in levothyroxine-treated women with hypothyroidism compared to women without thyroid dysfunction disorders. Thyroid. 2012;22:631–6.CrossRefPubMedPubMedCentral Scoccia B, Demir H, Kang Y, Fierro MA, Winston NJ. In vitro fertilization pregnancy rates in levothyroxine-treated women with hypothyroidism compared to women without thyroid dysfunction disorders. Thyroid. 2012;22:631–6.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Busnelli A, Somigliana E, Benaglia L, Sarais V, Ragni G, Fedele L. Thyroid axis dysregulation during in vitro fertilization in hypothyroid-treated patients. Thyroid. 2014;24:1650–5.CrossRefPubMed Busnelli A, Somigliana E, Benaglia L, Sarais V, Ragni G, Fedele L. Thyroid axis dysregulation during in vitro fertilization in hypothyroid-treated patients. Thyroid. 2014;24:1650–5.CrossRefPubMed
13.
Zurück zum Zitat Krassas GE, Doumas A, Kaltsas T, Halkias A, Pontikides N. Somatostatin receptor scintigraphy before and after treatment with somatostatin analogues in patients with thyroid eye disease. Thyroid. 1999;9:47–52.CrossRefPubMed Krassas GE, Doumas A, Kaltsas T, Halkias A, Pontikides N. Somatostatin receptor scintigraphy before and after treatment with somatostatin analogues in patients with thyroid eye disease. Thyroid. 1999;9:47–52.CrossRefPubMed
14.
Zurück zum Zitat Broer SL, Broekmans FJ, Laven JS, Fauser BC. Anti-Mullerian hormone: ovarian reserve testing and its potential clinical implications. Hum Reprod Update. 2014;20:688–701.CrossRefPubMed Broer SL, Broekmans FJ, Laven JS, Fauser BC. Anti-Mullerian hormone: ovarian reserve testing and its potential clinical implications. Hum Reprod Update. 2014;20:688–701.CrossRefPubMed
15.
Zurück zum Zitat Weghofer A, Dietrich W, Ortner I, Bieglmayer C, Barad D, Gleicher N. Anti-Mullerian hormone levels decline under hormonal suppression: a prospective analysis in fertile women after delivery. Reprod Biol Endocrinol. 2011;9:98.CrossRefPubMedPubMedCentral Weghofer A, Dietrich W, Ortner I, Bieglmayer C, Barad D, Gleicher N. Anti-Mullerian hormone levels decline under hormonal suppression: a prospective analysis in fertile women after delivery. Reprod Biol Endocrinol. 2011;9:98.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat van den Berg MH, van Dulmen-den Broeder E, Overbeek A, Twisk JW, Schats R, van Leeuwen FE, Kaspers GJ, Lambalk CB. Comparison of ovarian function markers in users of hormonal contraceptives during the hormone-free interval and subsequent natural early follicular phases. Hum Reprod. 2010;25:1520–7.CrossRefPubMed van den Berg MH, van Dulmen-den Broeder E, Overbeek A, Twisk JW, Schats R, van Leeuwen FE, Kaspers GJ, Lambalk CB. Comparison of ovarian function markers in users of hormonal contraceptives during the hormone-free interval and subsequent natural early follicular phases. Hum Reprod. 2010;25:1520–7.CrossRefPubMed
17.
Zurück zum Zitat Kuroda K, Uchida T, Nagai S, Ozaki R, Yamaguchi T, Sato Y, Brosens JJ, Takeda S. Elevated serum thyroid-stimulating hormone is associated with decreased anti-Mullerian hormone in infertile women of reproductive age. J Assist Reprod Genet. 2015;32:243–7.CrossRefPubMedPubMedCentral Kuroda K, Uchida T, Nagai S, Ozaki R, Yamaguchi T, Sato Y, Brosens JJ, Takeda S. Elevated serum thyroid-stimulating hormone is associated with decreased anti-Mullerian hormone in infertile women of reproductive age. J Assist Reprod Genet. 2015;32:243–7.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Dosiou C, Barnes J, Schwartz A, Negro R, Crapo L, Stagnaro-Green A. Cost-effectiveness of universal and risk-based screening for autoimmune thyroid disease in pregnant women. J Clin Endocrinol Metab. 2012;97:1536–46.CrossRefPubMed Dosiou C, Barnes J, Schwartz A, Negro R, Crapo L, Stagnaro-Green A. Cost-effectiveness of universal and risk-based screening for autoimmune thyroid disease in pregnant women. J Clin Endocrinol Metab. 2012;97:1536–46.CrossRefPubMed
19.
Zurück zum Zitat Xue H, Yang Y, Zhang Y, Song S, Zhang L, Ma L, Brosens JJ, Takeda S. Macrophage migration inhibitory factor interacting with Th17 cells may be involved in the pathogenesis of autoimmune damage in Hashimoto’s thyroiditis. Mediators Inflamm. 2015;2015:621072. Xue H, Yang Y, Zhang Y, Song S, Zhang L, Ma L, Brosens JJ, Takeda S. Macrophage migration inhibitory factor interacting with Th17 cells may be involved in the pathogenesis of autoimmune damage in Hashimoto’s thyroiditis. Mediators Inflamm. 2015;2015:621072.
20.
Zurück zum Zitat Karmisholt J, Laurberg P. Serum TSH and serum thyroid peroxidase antibody fluctuate in parallel and high urinary iodine excretion predicts subsequent thyroid failure in a 1-year study of patients with untreated subclinical hypothyroidism. Eur J Endocrinol. 2008;158:209–15.CrossRefPubMed Karmisholt J, Laurberg P. Serum TSH and serum thyroid peroxidase antibody fluctuate in parallel and high urinary iodine excretion predicts subsequent thyroid failure in a 1-year study of patients with untreated subclinical hypothyroidism. Eur J Endocrinol. 2008;158:209–15.CrossRefPubMed
21.
Zurück zum Zitat Gleicher N, Kim A, Weghofer A, Kushnir VA, Shohat-Tal A, Lazzaroni E, Lee HJ, Barad DH. Hypoandrogenism in association with diminished functional ovarian reserve. Hum Reprod. 2013;28:1084–91.CrossRefPubMed Gleicher N, Kim A, Weghofer A, Kushnir VA, Shohat-Tal A, Lazzaroni E, Lee HJ, Barad DH. Hypoandrogenism in association with diminished functional ovarian reserve. Hum Reprod. 2013;28:1084–91.CrossRefPubMed
22.
Zurück zum Zitat La Marca A, Nelson SM, Sighinolfi G, Manno M, Baraldi E, Roli L, Xella S, Marsella T, Tagliasacchi D, D'Amico R, Volpe A. Anti-Mullerian hormone-based prediction model for a live birth in assisted reproduction. Reprod Biomed Online. 2011;22:341–9. La Marca A, Nelson SM, Sighinolfi G, Manno M, Baraldi E, Roli L, Xella S, Marsella T, Tagliasacchi D, D'Amico R, Volpe A. Anti-Mullerian hormone-based prediction model for a live birth in assisted reproduction. Reprod Biomed Online. 2011;22:341–9.
23.
Zurück zum Zitat Dittrich R, Beckmann MW, Oppelt PG, Hoffmann I, Lotz L, Kuwert T, Mueller A. Thyroid hormone receptors and reproduction. J Reprod Immunol. 2011;90:58–66.CrossRefPubMed Dittrich R, Beckmann MW, Oppelt PG, Hoffmann I, Lotz L, Kuwert T, Mueller A. Thyroid hormone receptors and reproduction. J Reprod Immunol. 2011;90:58–66.CrossRefPubMed
24.
Zurück zum Zitat Krassas GE, Poppe K, Glinoer D. Thyroid function and human reproductive health. Endocr Rev. 2010;31:702–55.CrossRefPubMed Krassas GE, Poppe K, Glinoer D. Thyroid function and human reproductive health. Endocr Rev. 2010;31:702–55.CrossRefPubMed
25.
Zurück zum Zitat Revelli A, Casano S, Piane LD, Grassi G, Gennarelli G, Guidetti D, Massobrio M. A retrospective study on IVF outcome in euthyroid patients with anti-thyroid antibodies: effects of levothyroxine, acetyl-salicylic acid and prednisolone adjuvant treatments. Reprod Biol Endocrinol. 2009;7:137.CrossRefPubMedPubMedCentral Revelli A, Casano S, Piane LD, Grassi G, Gennarelli G, Guidetti D, Massobrio M. A retrospective study on IVF outcome in euthyroid patients with anti-thyroid antibodies: effects of levothyroxine, acetyl-salicylic acid and prednisolone adjuvant treatments. Reprod Biol Endocrinol. 2009;7:137.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Toulis KA, Goulis DG, Venetis CA, Kolibianakis EM, Negro R, Tarlatzis BC, Papadimas I. Risk of spontaneous miscarriage in euthyroid women with thyroid autoimmunity undergoing IVF: a meta-analysis. Eur J Endocrinol. 2010;162:643–52.CrossRefPubMed Toulis KA, Goulis DG, Venetis CA, Kolibianakis EM, Negro R, Tarlatzis BC, Papadimas I. Risk of spontaneous miscarriage in euthyroid women with thyroid autoimmunity undergoing IVF: a meta-analysis. Eur J Endocrinol. 2010;162:643–52.CrossRefPubMed
27.
Zurück zum Zitat Polyzos NP, Sakkas E, Vaiarelli A, Poppe K, Camus M, Tournaye H. Thyroid autoimmunity, hypothyroidism and ovarian reserve: a cross-sectional study of 5000 women based on age-specific AMH values. Hum Reprod. 2015;30:1690–6.CrossRefPubMed Polyzos NP, Sakkas E, Vaiarelli A, Poppe K, Camus M, Tournaye H. Thyroid autoimmunity, hypothyroidism and ovarian reserve: a cross-sectional study of 5000 women based on age-specific AMH values. Hum Reprod. 2015;30:1690–6.CrossRefPubMed
28.
Zurück zum Zitat Velkeniers B, Van Meerhaeghe A, Poppe K, Unuane D, Tournaye H, Haentjens P. Levothyroxine treatment and pregnancy outcome in women with subclinical hypothyroidism undergoing assisted reproduction technologies: systematic review and meta-analysis of RCTs. Hum Reprod Update. 2013;19:251–8.CrossRefPubMed Velkeniers B, Van Meerhaeghe A, Poppe K, Unuane D, Tournaye H, Haentjens P. Levothyroxine treatment and pregnancy outcome in women with subclinical hypothyroidism undergoing assisted reproduction technologies: systematic review and meta-analysis of RCTs. Hum Reprod Update. 2013;19:251–8.CrossRefPubMed
29.
Zurück zum Zitat Magri F, Capelli V, Gaiti M, Brambilla E, Montesion L, Rotondi M, Spinillo A, Nappi RE, Chiovato L. Impaired outcome of controlled ovarian hyperstimulation in women with thyroid autoimmune disease. Thyroid. 2013;23:1312–8.CrossRefPubMed Magri F, Capelli V, Gaiti M, Brambilla E, Montesion L, Rotondi M, Spinillo A, Nappi RE, Chiovato L. Impaired outcome of controlled ovarian hyperstimulation in women with thyroid autoimmune disease. Thyroid. 2013;23:1312–8.CrossRefPubMed
30.
Zurück zum Zitat Tuten A, Hatipoglu E, Oncul M, Imamoglu M, Acikgoz AS, Yilmaz N, Ozcil MD, Kaya B, Misirlioglu AM, Sahmay S. Evaluation of ovarian reserve in Hashimoto’s thyroiditis. Gynecol Endocrinol. 2014;30:708–11.CrossRefPubMed Tuten A, Hatipoglu E, Oncul M, Imamoglu M, Acikgoz AS, Yilmaz N, Ozcil MD, Kaya B, Misirlioglu AM, Sahmay S. Evaluation of ovarian reserve in Hashimoto’s thyroiditis. Gynecol Endocrinol. 2014;30:708–11.CrossRefPubMed
31.
Zurück zum Zitat Saglam F, Onal ED, Ersoy R, Koca C, Ergin M, Erel O, Cakir B. Anti-Mullerian hormone as a marker of premature ovarian aging in autoimmune thyroid disease. Gynecol Endocrinol. 2015;31:165–8.CrossRefPubMed Saglam F, Onal ED, Ersoy R, Koca C, Ergin M, Erel O, Cakir B. Anti-Mullerian hormone as a marker of premature ovarian aging in autoimmune thyroid disease. Gynecol Endocrinol. 2015;31:165–8.CrossRefPubMed
32.
Zurück zum Zitat Magri F, Schena L, Capelli V, Gaiti M, Zerbini F, Brambilla E, Rotondi M, De Amici M, Spinillo A, Nappi RE, Chiovato L. Anti-Mullerian hormone as a predictor of ovarian reserve in ART protocols: the hidden role of thyroid autoimmunity. Reprod Biol Endocrinol. 2015;13:106.CrossRefPubMedPubMedCentral Magri F, Schena L, Capelli V, Gaiti M, Zerbini F, Brambilla E, Rotondi M, De Amici M, Spinillo A, Nappi RE, Chiovato L. Anti-Mullerian hormone as a predictor of ovarian reserve in ART protocols: the hidden role of thyroid autoimmunity. Reprod Biol Endocrinol. 2015;13:106.CrossRefPubMedPubMedCentral
Metadaten
Titel
What affects functional ovarian reserve, thyroid function or thyroid autoimmunity?
verfasst von
Andrea Weghofer
David H. Barad
Sarah Darmon
Vitaly A. Kushnir
Norbert Gleicher
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Reproductive Biology and Endocrinology / Ausgabe 1/2016
Elektronische ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-016-0162-0

Weitere Artikel der Ausgabe 1/2016

Reproductive Biology and Endocrinology 1/2016 Zur Ausgabe

Alter der Mutter beeinflusst Risiko für kongenitale Anomalie

28.05.2024 Kinder- und Jugendgynäkologie Nachrichten

Welchen Einfluss das Alter ihrer Mutter auf das Risiko hat, dass Kinder mit nicht chromosomal bedingter Malformation zur Welt kommen, hat eine ungarische Studie untersucht. Sie zeigt: Nicht nur fortgeschrittenes Alter ist riskant.

Fehlerkultur in der Medizin – Offenheit zählt!

28.05.2024 Fehlerkultur Podcast

Darüber reden und aus Fehlern lernen, sollte das Motto in der Medizin lauten. Und zwar nicht nur im Sinne der Patientensicherheit. Eine negative Fehlerkultur kann auch die Behandelnden ernsthaft krank machen, warnt Prof. Dr. Reinhard Strametz. Ein Plädoyer und ein Leitfaden für den offenen Umgang mit kritischen Ereignissen in Medizin und Pflege.

Mammakarzinom: Brustdichte beeinflusst rezidivfreies Überleben

26.05.2024 Mammakarzinom Nachrichten

Frauen, die zum Zeitpunkt der Brustkrebsdiagnose eine hohe mammografische Brustdichte aufweisen, haben ein erhöhtes Risiko für ein baldiges Rezidiv, legen neue Daten nahe.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.