Skip to main content

Risk assessment of transgender people: implementation of a demasculinizing–feminizing rodent model including the evaluation of thyroid homeostasis

A Correction to this article was published on 30 January 2024

This article has been updated

Abstract

Background

Individuals whose gender identity differs from the biological sex and the social norms are defined as transgender. Sometimes transgender undergo gender affirming hormone therapy, which lasts for the entire life making essential to evaluate its potential long-term effects. Moreover, transgender can represent a susceptible sub-group of population and specific attention is needed in risk assessment, including the development of targeted animal models. Aim of the study is the implementation of a rodent demasculinizing–feminizing model through the setting of appropriate dose of hormone therapy and the selection of specific biomarkers to evaluate the sex transition. Specific attention is paid to thyroid homeostasis due to the close link with reproductive functions. Four male adult rats/group were subcutaneously exposed to three doses plus control of β-estradiol valerate plus cyproterone acetate at: 0.045 + 0.2 (low), 0.09 + 0.2 (medium) and 0.18 + 0.2 (high) mg/dose, five times/week. The doses were selected considering the most recent recommendations for transgender woman. Sperm count, histopathological analysis (testis, liver, thyroid), testosterone, estradiol, triiodothyronine and thyroid-stimulating hormone serum levels and gene expression of sex dimorphic CYP450 were evaluated.

Results

The doses induced feminizing–demasculinizing effects: decreased testosterone serum levels at the corresponding cisgender, increased estradiol, impairment of male reproductive function and reversal of sex-specific CYP liver expression. However, the medium and high doses induced marked liver toxicity and the low dose is considered the best choice, also for long-term studies in risk assessment. The alterations of thyroid indicated follicular cell hypertrophy supported by increased thyroid-stimulating hormone serum levels at the higher doses.

Conclusions

The implementation of animal models that mimic the effects of gender affirming hormone therapy is essential for supporting clinical studies in transgender people and filling data gap in order to ensure an appropriate risk assessment and a more accurate, personalized care for transgender people.

Background

Gender identity is each person's sense of belonging to a gender or sex. It is an individual experience, where each person can identify her/his self as a woman, a man, both, neither, or anywhere along the gender spectrum. Gender identity can align or differ from the sex assigned at birth. Individuals whose gender identity differs from the biological sex are defined as transgender (TG), and they often undergo medical gender-affirming hormone therapy (HT) [1, 2]. The HT of TG woman consists of treatment with estrogens and antiandrogens at the same time: antiandrogens have the two important functions of lowering the testosterone (T) levels to the female range (< 50 ng/dL) and of reducing the dose of the estrogens to be used [3]. In fact, high estrogen doses suppress androgen production (via central feedback) but may be associated with strong adverse effects such as thrombosis [4]. Spironolactone in the US and cyproterone acetate (CPA) in EU are among the most popular adjunctive androgen-lowering/-inhibiting agents [5]. HT is a long-term treatment, which lasts for the entire life of the individual, so it is essential to study all possible side effects [6, 7]. In addition, like general population, TG people are exposed to environmental contaminants; [6, 8] indeed, for TG people, HT might represent an additional risk factor and the implementation of a specific animal model to address the hazard identification is of primary importance [6].

On that point, the recent in vivo study to implement a rodent model mimicking de-masculinizing feminizing (dMF) HT failed to define a reliable dose level of CPA in combination with estradiol (E2) valerate. The toxicological effects recorded during the treatment were considered not compatible with lifelong HT and they have been potentially attributable to CPA, that is known to suppress T in presence of marked hepatotoxicity [9]. In fact, the study showed that in rat 0.33–0.93 mg/dose of CPA (about 25–50 mg/dose in humans) caused hepatic steatosis, inflammation and sinusoidal dilatation [10], and all these effects are typical expression of CPA toxicity [11, 12]. Moreover, recent data provide indications that low-dose CPA treatment for TG women is as effective as high-dose treatment and possibly safer [13, 14].

In this context, the aim of the present study is the selection of the suitable dose to implement the dMF animal model based on the current data on human therapies. T serum level is measured to verify the achievement of the corresponding cisgender levels and specific functional and tissue biomarkers have been identified to characterize the model, in particular: sperm count, E2 serum levels, histopathological analysis of testis and liver, and the gene expression of sex-specific liver cytochrome P450 (CYP450) isoforms identified as biomarkers of liver demasculinization after two weeks of HT in the dMF rat model [10]. Since in TG individuals undergoing HT the endocrine system is overstimulated by the therapy, they may represent a sub-population more vulnerable and susceptible [6,7,8] than the general population to the effects of endocrine disrupters (EDs). EDs are defined as "an exogenous chemical, or mixture of chemicals, that can interfere with any aspect of hormone action". Several pesticides, fungicides, industrial chemicals, plasticizers, nonylphenols, metals, pharmaceutical agents and phytoestrogens are EDs and humans can be exposed by ingestion, inhalation and dermal uptake [15, 16]. Thus, EDs and HT can have mutual targets in the organism: for these reasons, the analysis of thyroid gland homeostasis in rats receiving different dosages of HT has been also included in this study. Indeed, it is known that the thyroid influences several systems such as reproductive, metabolic, nervous, etc. [17, 18], and currently no data are available on the potential impact of HT on thyroid and on the prevalence of thyroid diseases in TG population.

Methods

Ethical approval

The animal study was performed in accordance with the Directive 2010/63/EU, the Italian Legislative Decree n. 26 of 4 March 2014 and the Organisation for Economic Co-operation and Development Principles of Good Laboratory Practice. The study protocol was approved by the Italian Ministry of Health (authorization n° 806/2021-PR).

Experimental design

Sixteen young male Sprague–Dawley rats (8/9 weeks old) were purchased from Envigo (Italy). Upon arrival, animals were pair-housed under standard laboratory conditions (see [10] for further details). Pellet food and water were available ad libitum. Following two weeks of adaptation, male rats were randomly divided into four groups as follows:

  • Control group (C): sesame oil (vehicle)

  • Low dose (L): 0.045 mg E2 valerate + 0.2 CPA mg per dose

  • Medium dose (M): 0.09 mg E2 valerate + 0.2 CPA mg per dose

  • High dose (H): 0.18 mg E2 valerate + 0.2 CPA mg per dose

The drugs were administered by single subcutaneous injections (200 µL), five times a week for two weeks. The group size and the doses were calculated as reported in Tassinari et al., 2023 [10]. Briefly, the dose of E2 valerate (0.18 mg per day), after the suitable adaptation for rat, corresponds to the higher dose of 10 mg/day suggested by the main clinical guidelines for TG women [1, 19]; the dose of CPA (0.2 mg per dose) was selected considering the recent HT recommendations for TG woman, and correspond to a daily dose of 10 mg [2, 5].

During the experiment, rats were monitored twice a day (at 9:00 a.m. and 4:00 p.m.) for general health conditions. Body weight (bw) and food consumption were recorded two times a week. Twenty-four hours after the last treatment, rats were anaesthetized with a gaseous solution of isoflurane and blood samples were collected by intracardiac puncture for measuring serum hormones [T, E2, thyroid stimulating hormone (TSH) and Thyroxine (T4)]. Subsequently, animals were sacrificed by CO2 inhalation, and necropsy and gross pathology were performed. Liver and testes were excised and weighted to evaluate absolute and relative (organ weight/body weight*100) weight. Right epididymis was sampled and used for sperm count analysis. For histopathological analysis, liver and thyroid were fixed in 10% buffered formalin whereas, to better preserve tissue morphology, testes were fixed in Bouin’s solution. A lobe of liver was flash frozen in liquid nitrogen and stored at – 80 °C for gene expression analysis.

Sperm count

Right caudae epididymides were excised, rinsed with Dulbecco's Modified Eagle Medium (DMEM) medium (Gibco Rodano (MI), Italy), transferred on a Petri dish containing 1 mL DMEM medium and minced with scissors. Epididymal pieces were fluxed through a Pasteur pipette to facilitate sperm extrusion. Sperm suspension was filtered through a 200-mesh nylon to remove tissue fragments and diluted up to 10 mL. Sperm were counted by a Neubauer chamber under a light microscopy (Nikon Eclipse Ts2) [10].

Blood collection and biochemical evaluation of hormones

Shortly before the sacrifice, blood was collected by intracardiac puncture with stratification across groups to reduce the potential impact of circadian rhythm and pulsatility. Blood samples were left to coagulate at room temperature for 1 h, centrifuged for 15 min at 2000 rpm twice in a cooled bench-top centrifuge (Microlite Microfuge, Thermo Electron Corporation) and stored at -80 °C until use. Serum levels of all hormones were measured in the same analytical section by the following commercial ELISA kits of the same lot(s):

  • E2 Rat kit (RTC009R—BioVendor Brno, Czech Republic), LOD 2.5 pg/ml

  • T Mouse/Rat kit (RTC001R—BioVendor Brno, Czech Republic), LOD 2.5 pg/ml

  • TSH Rat Kit (ELK2283—ELK Biotechnology, China), LOD 0.071 ng/mL

  • T4 Rat Kit (ELK8716—ELK Biotechnology, China), LOD 1.42 ng/mL

Each kit provided a standard solution of the hormone and serial dilutions were prepared to derive a standard curve and define the range of linearity of each test. For all the analyses, the manufacturer’s instructions were followed. Each sample was assessed in duplicate and the absorbance was read at 450 nm on a VICTOR3 Multilabel reader (Perkin Elmer, USA). The unknown hormone concentrations in samples were derived using the standard curve of each hormone and the software GraphPad Prism 5.0 (GraphPad Software Inc.).

Histological and histomorphometrical analysis

After fixation, liver, testes and thyroid were dehydrated in a graded series of alcohol baths and embedded in paraffin by the tissue processor (Shandon Excelsior ES, Thermo Scientific). The 5-μm-thick histological sections were prepared using the Microm HM 325 (Thermo Scientific) and stained with hematoxylin/eosin for the examination under a light microscopy (Nikon Microphot FX) [20]. The scoring of the lesions was semi-quantitative, using a 5-point grading scale (0 to 4), taking into consideration the severity of the changes based on the criteria explained by Shackelford et al. [21] and summarized in Table 1.

Table 1 Scores based on distribution of tissue lesions

The quantitative histomorphometrical analysis were performed on testes and thyroid by means of an image analysis system (Nis-Elements BR) applied to an optical microscope (Nikon Microphot FX). Testis tubular diameters, the relative area of the seminiferous tubules and the lumen were measured in 20 randomly selected tubules (10 × objective) [10]. Thyroid morphometrical analysis were performed, according to Rasinger et al. [22] as follows: follicular density (ratio between number of follicles and a predetermined area, 10 × lens); indirect follicular cell height (mean ratio of follicle and colloid area in five randomly selected follicles/sample 40 × lens); the mean ratio of follicular epithelium areas and number of nuclei (in the same follicle to evaluate follicular maturation); direct follicular cell height (mean of five cell height in five randomly selected follicles/sample, 64 × lens).

Gene expression analysis

Gene expression was performed on liver according to Tassinari et al., 2023 [10]. Briefly, total RNA content was extracted by liver samples with the Norgen kit (Norgen Biotek Corp. Thorold, Canada) according to the manufacturer’s instructions, RNA quantity assessed by Nabi Nano Spectrophotometer (MicroDigital Co. Ltd., Seoul, Republic of Korea) and RNA integrity (A260/A280 ≥ 1.8) evaluated by 1% agarose gel electrophoresis. RNA (1 µg) was reverse-transcribed to cDNA using the Tetro cDNA Synthesis Kit (Quantace, Mumbai, India) according to the manufacturer’s instructions. Specific primers for Cyp2c11, Cyp3a18, Cyp2c12, Cyp2c6, and glyceraldehyde-3-phosphate dehydrogenase (Gapdh), as housekeeping gene, were designed using the Primer-BLAST web application and purchased from Metabion (Metabion International AG, Germany). The Excel TaqTM Fast Q-PCR Master Mix SYBR (SMOBIO Technology Inc., Hsinchu City, Taiwan) was used to perform real-time PCR assays, running reactions on a Bioer LineGene 9600 (Bioer, Hangzhou, China). Experiments were performed in duplicate on 96-well PCR plates. Threshold cycles were calculated by the LineGen9620 software (Bioer). Data are expressed as ΔΔCt ± SD values for each target gene with control samples as calibrator and Gapdh [10].

Data analysis

Data management and enter were performed using Microsoft Excel 2013 and analysed using the software JMP 10 (SAS Institute Inc., Cary, NC, USA). Graphics were designed using the GraphPad Prism 5.0 software (GraphPad Software Inc., San Diego, CA, USA). Body weight, food consumption, absolute e relative organ weight, hormone serum levels and tissue morphometrical and gene expression data were presented as mean ± standard deviation and a non-parametric Kruskal–Wallis analysis was performed followed by post-hoc pairwise comparisons (Mann–Whitney test). Histological semi-quantitative data were analysed by 2-way Fisher Exact Test to assess significant differences with respect to control group including samples assigned to a category without reference to severity gradations (total finding incidence). The Cochran-Armitage Trend Test was used to evaluate a dose–response trend. Differences among groups were considered significant if the P-value was < 0.05.

Results

General toxicity, body weight and food consumption

No death or adverse clinical effects have been recorded. The bw at treatment days 6, 9, 13, 15, bw gain and food consumption were significantly decreased in all treatment groups in comparison to C (Fig. 1).

Fig. 1
figure 1

General toxicity data. General toxicity data of male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week for 2 weeks: C: 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Panel A Body weight (bw) at treatment days 6, 9, 13, 15. Panel B bw gain. Panel C food consumption. Data are presented as mean ± standard deviation. Statistical significance: * p < 0.05 Mann–Whitney test

Reproductive endpoints

Testis absolute weight was significantly decreased in all treatment groups in comparison to C; no differences in relative weight were seen among  treatment and C groups (Table 2). Sperm count was dose-dependently decreased, statistically significant in all treatment groups (Fig. 2). T serum levels were significantly decreased and E2 serum levels were statistically increased in all treatment groups in comparison to C group (Fig. 3).

Table 2 Absolute and relative weight of testes and liver
Fig. 2
figure 2

Sperm count. Sperm count of male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week for 2 weeks C: 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Panel A sperm count. Data are presented as mean ± standard deviation. Statistical significance: * p < 0.05 Mann–Whitney test. Panel B Light microscopic photos of sperm count on Neubauer chamber (original magnification 10 × ; area of 1/16 mm2)

Fig. 3
figure 3

Reproductive hormone serum levels. Reproductive hormone serum levels of male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week for 2 weeks: Control (C): 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Panel A Testosterone. Panel B Estradiol. Data are presented as mean ± standard deviation. Statistical significance: * p < 0.05 Mann–Whitney test

Histopathological analysis showed dose-dependent, significant increase of tubule degeneration in testes with depletion of germ cells (L, 1/4 sample: grade 1; M, 2/4 samples; grade 1; H, 2/4 samples:grade 1 plus 2/4 samples: grade 2) and statistically significant reduction of tubule lumen area in the H group compared to the C group (Table 3 and Fig. 4).

Table 3 Histopathological data
Fig. 4
figure 4

Testis histophatological features. Testis tubule degeneration with depletion of germ cells in male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week for 2 weeks: Control (C): 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Bar 10 μm (original magnification 10 × ; haematoxylin and eosin stain)

Liver endpoints

Liver absolute weight was significantly decreased in all treatment groups in comparison to C, relative weight was unchanged (Table 2).

Histopathological evaluation showed statistically significant increase of sinusoidal dilatation in M (1/4 samples: grade 1 plus 3/4 samples: grade 3) and H (3/4 samples: grade 1 plus 1/4 samples: grade 3) groups and of hepatocyte vacuolization in M (2/4 samples: grade 1 plus 2/4 samples: grade 2) group compared to C group (Table 3; Fig. 5).

Fig. 5
figure 5

Liver histophatological features. Liver sinusoidal dilatation (S) and hepatocyte vacuolization (V) in male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week, for 2 weeks: Control (C): 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Bar 10 μm (original magnification 20 × ; haematoxylin and eosin stain)

Gene expression analysis indicate that Cyp2c11 (male specific isoform) was down-regulated in H group and Cyp3a18 (male predominant isoform) was significantly down-regulated in all treatment groups (Fig. 6; Panels A and B). Cyp2c12 (female specific isoform) was up-regulated in all treatment groups, statistically significant in L and M (Fig. 6; Panel C), the Cyp2c6 (female predominant isoform) was significantly up-regulated in the M group in comparison to C (Fig. 6, Panel D).

Fig. 6
figure 6

Gene expression. Gene expression analysis of sex specific CYP450 isoforms by real-time PCR in liver of male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week for 2 weeks: Control (C): 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Panel A Cyp2c11; Panel B Cyp3a18; Panel C Cyp2c12; Panel D Cyp2c6. Data are presented as mean ± standard deviation. Statistical significance: * p < 0.05 Mann–Whitney test

Thyroid endpoint

TSH serum levels was significantly increased in M and H (p = 0.06) treatment groups in comparison to C. No treatment-related alterations were observed in T4 serum levels (Fig. 7).

Fig. 7
figure 7

Thyroid biomarker serum levels. Serum levels of thyroid biomarkers of male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week for 2 weeks: Control (C): 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Panel A Thyroid-stimulating hormone (TSH). Panel B thyroxine (T4). Data are presented as mean ± standard deviation. Statistical significance: * p < 0.05 Mann–Whitney test

Histopathological analysis indicated follicular cell hypertrophy with central follicles tightly packed and smaller than normal in all treatment groups (Fig. 8). Follicular density was increased in all treatment groups with an increase of follicular number. The follicle dimension was decreased with reduction of both follicle and colloid areas in all treatment groups compared to the C. The epithelium cell height and area of follicles were unaffected (Table 3).

Fig. 8
figure 8

Thyroid histophatological features. Thyroid hypertrophy in male rats subcutaneously treated with estradiol valerate plus cyproterone acetate, five times a week for 2 weeks: Control (C): 0—sesame oil, Low (L) 0.045 + 0.2, Medium (M) 0.09 + 0.2 and High (H) 0.18 + 0.2 mg. Bar 10 μm (original magnification 10 × ; haematoxylin and eosin stain)

Discussion

TG people often undertake a gender affirming path, which includes lifelong HT. Indeed, although HT has shown to have positive physical and psychological effects on the transitioning individual, scarce data are still available about its long-term outcomes. For this reason, it is crucial to implement an animal model that allows to explore in detail mechanisms and pathways linked to such aspects. Moreover, considering the potential different susceptibility and vulnerability of TG people to environmental contaminants, the animal model can represent a key tool for risk assessment purposes [6], and to obtain information supporting clinical studies e.g., concerning bone health [7] or filling data gap e.g., on thyroid homeostasis.

In a previous study aimed at setting the dMF animal model, although several endpoints were positively correlated with the switching towards demasculinization, the doses of E2 valerate plus CPA appeared to be too high, causing marked toxicity effects, and consequently the model could not be implemented and/or used in long term studies [10].

In the present experiment, as expected, after two weeks of feminizing HT, the male rats showed reduction of bw and food consumption in all treatment groups. The available in vivo studies described similar effects on bw in rats with up to 30 days of E2 plus CPA administration [10, 23]. Indeed, the lack of weight gain could reflect the estrogenic inhibition of eating [24] which can explain both the reduction of food intake and of bw observed in all treatment groups. Interestingly, in TG woman, a bw increase consisting in gain of body fat and decline in lean body mass was observed after the beginning of HT [25], while the body mass index becomes stable following 3 to 6 years [26]. Moreover, T levels decreased and E2 increased in all the treatment groups; in particular, the T levels of 0.08 to 0.26 ng/ml fell into the range of female serum levels [10]. In TG woman upon the initiation of E2 and CPA treatment, the serum concentration of E2 rises whereas T drops, approximating the female physiological sex hormone range [27].

Regarding the reproductive system, testis weight and sperm count were significantly reduced in all treatment groups, together with dose dependent histopathological pictures of tubule degeneration (C = 0%; L = 20%; M = 50%; H = 100%) which become significant at the highest dose. In the previous study, with higher daily doses of E2 valerate and CPA, the HT impaired the testicular architecture and the different testicular cellular types in all the samples at all dose levels [10]. Indeed, the effects recorded are consistent with the antiandrogenic activity of CPA (increased E2) [28] and of estrogen (tubular degeneration) [29]. The data of the present study, although substantially confirming the previous results, are less marked [10]; in addition, they indicated that the reproductive function was not completely altered in male rats after two weeks of HT. Concerning TG women, in literature scarce data are available about the effects of HT on testis morphology and spermatogenesis; the existing papers showed lacking and variable results, and the number of patients is very limited. An interesting study performed in 2019 on seventy-two adult TG women subjected to HT for > 1 year before orchiectomy, showed that the 80% of TG women had still germ cells and that spermatogenesis was preserved in approximately 40% of the patients; thus, the data suggested that duration of HT did not affect the degree of preservation of germ cells or the spermatogenesis, as could happen starting the hormonal treatment at a younger age. In humans, the volume of testes can be used as a marker to predict the presence of preserved spermatogenesis [30].

It has been already discussed that the liver plays a pivotal role in the sex hormone metabolism, e.g., producing carrier proteins that are important in reproduction and development [31]. In fact, in mammals, liver shows the highest degree of sexual dimorphism, with 72% of the genes expressed in a sexually differentiated manner, while in other organs the degree of sex dependent variability ranges between 14 and 60% [32]. In the previous study, the selected doses induced marked histopathological effects, in particular sinusoidal dilatation and hepatocyte vacuolation [10]. In this study, although similar signs of hepatotoxicity were still evident, they were milder and limited to M and H groups. On the other hand, despite the reduction of doses, the CYP gene expression showed once again to be valid biomarker for evaluating the success of HT. In fact, similarly to the previous data [10], signs of demasculinization in the expression of sex-specific CYPs were evident up to the lowest dose of E2 valerate (0.045 mg) and CPA (0.2 mg).

In addition, considering the complexity of endocrine system that controls body processes and functions and the well-known link between thyroid and gonads through the hypothalamic-pituitary gonadal axis [33], mainly in females but evident also in males [34, 35], it appeared of interest to investigate the potential impact of HT on thyroid homeostasis. At present, this is the first in vivo study that explores the potential impact of HT on thyroid. The results showed early signs of follicular cell hypertrophy—identified by quantitative histomorphometrical measurement on follicles—supported by increased TSH serum levels in M and H groups [36]. It is important to note that, in rodents, increased serum TSH levels and resultant follicular cell hypertrophy/hyperplasia are typical hormonal and histopathological findings attributable to compounds altering thyroid function [37]. Indeed, a higher number of animals might have highlighted significant alterations of TSH; nevertheless, the results obtained lead to hypothesize a decrease in thyroid activity. At present, no data are available to indicate the potential influence of gender affirming HT on thyroid homeostasis in TG woman and men. Recent research estimated an increased prevalence of almost 9% of thyroid disease in the TG population, however, due to an assessment bias, the data may be underestimated [18].

In this respect, the inclusion of thyroid homeostasis in the animal model of gender affirming HT can represent a valuable biomarker to explore and complete the data gaps on TG people health.

Conclusions

Among the doses selected for this study, the lowest corresponding to the administration of 0.045 mg E2 valerate plus 0.2 mg CPA can be considered suitable for a long-term HT administration. In fact, after two weeks of administration, it: (i) allows the achievement of T serum levels in the range of the corresponding cisgender, and (ii) increases E2 serum levels and depresses the male reproductive function, without overt signs of toxicity; interestingly, these last are sex-transition biomarkers described also in TG women undergone gender-affirming HT [19]. Moreover, such dose induced liver demasculinization in the expression of sex-specific CYPs in the absence of tissue damage, proving the reliability of sex-dimorphic liver genes as biomarkers of sex transition in animal model [10].

Actually, no studies focused on the effect of HT on the thyroid of TG women are available, so it is essential to deepen further this aspect, in order to ensure accurate and personalized care for TG people.

Change history

Abbreviations

TG:

Transgender

HT:

Hormone therapy

T:

Testosterone

CPA:

Cyproterone acetate

dMF:

De-masculinizing feminizing

EDs:

Endocrine disrupters

E2:

Estradiol

CYP450:

Cytochrome P450

C:

Control group

L:

Low dose

M:

Medium dose

H:

High dose

BW:

Body weight

DMEM:

Dulbecco's Modified Eagle Medium

TSH:

Thyroid stimulating hormone

T4:

Thyroxine

References

  1. T’Sjoen G, Arcelus J, De Vries ALC, Fisher AD, Nieder TO, Özer M, et al. European society for sexual medicine position statement “assessment and hormonal management in adolescent and adult trans people, with attention for sexual function and satisfaction.” J Sex Med. 2020;17(4):570–84.

    Article  CAS  PubMed  Google Scholar 

  2. Fisher AD, Senofonte G, Cocchetti C, Guercio G, Lingiardi V, Meriggiola MC, et al. SIGIS-SIAMS-SIE position statement of gender affirming hormonal treatment in transgender and non-binary people. J Endocrinol Invest. 2022;45(3):657–73.

    Article  CAS  PubMed  Google Scholar 

  3. Tomlins L. Prescribing for transgender patients. Aust Prescr. 2019;42(1):10–3.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Safer JD. Research gaps in medical treatment of transgender/nonbinary people. J Clin Invest. 2021;131(4):e142029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kuijpers SME, Wiepjes CM, Conemans EB, Fisher AD, T’Sjoen G, den Heijer M. Toward a lowest effective dose of cyproterone acetate in trans women: results from the ENIGI study. J Clin Endocrinol Metab. 2021;106(10):e3936–45.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Tassinari R, Maranghi F. Rodent model of gender-affirming hormone therapies as specific tool for identifying susceptibility and vulnerability of transgender people and future applications for risk assessment. Int J Environ Res Public Health. 2021;18(23):12640.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Venkatesh VS, Nie T, Zajac JD, Grossmann M, Davey RA. The utility of preclinical models in understanding the bone health of transgender individuals undergoing gender-affirming hormone therapy. Curr Osteoporos Rep. 2023;14:1–7.

    Google Scholar 

  8. King DE. The inclusion of sex and gender beyond the binary in toxicology. Front Toxicol. 2022;4:929219.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Burinkul S, Panyakhamlerd K, Suwan A, Tuntiviriyapun P, Wainipitapong S. Anti-androgenic effects comparison between cyproterone acetate and spironolactone in transgender women: a randomized controlled Trial. J Sex Med. 2021;18(7):1299–307.

    Article  CAS  PubMed  Google Scholar 

  10. Tassinari R, Tammaro A, Lori G, Tait S, Martinelli A, Cancemi L, et al. Risk assessment of transgender people: development of rodent models mimicking gender-affirming hormone therapies and identification of sex-dimorphic liver genes as novel biomarkers of sex transition. Cells. 2023;12(3):474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Abenavoli L, Milic N, Beaugrand M. Severe hepatitis induced by cyproterone acetate: role of corticosteroids. A case report. Ann Hepatol. 2013;12(1):152–5.

    Article  PubMed  Google Scholar 

  12. Bessone F, Lucena MI, Roma MG, Stephens C, Medina-Cáliz I, Frider B, et al. Cyproterone acetate induces a wide spectrum of acute liver damage including corticosteroid-responsive hepatitis: report of 22 cases. Liver Int. 2016;36(2):302–10.

    Article  CAS  PubMed  Google Scholar 

  13. Even Zohar N, Sofer Y, Yaish I, Serebro M, Tordjman K, Greenman Y. Low-dose cyproterone acetate treatment for transgender women. J Sex Med. 2021;18(7):1292–8.

    Article  CAS  PubMed  Google Scholar 

  14. Glintborg D, T’Sjoen G, Ravn P, Andersen MS. Management of endocrine disease: optimal feminizing hormone treatment in transgender people. Eur J Endocrinol. 2021;185(2):R49–63.

    Article  PubMed  Google Scholar 

  15. Tassinari V, Smeriglio A, Stillittano V, Trombetta D, Zilli R, Tassinari R, et al. Endometriosis treatment: role of natural polyphenols as anti-inflammatory agents. Nutrients. 2023;15(13):2967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Yilmaz B, Terekeci H, Sandal S, Kelestimur F. Endocrine disrupting chemicals: exposure, effects on human health, mechanism of action, models for testing and strategies for prevention. Rev Endocr Metab Disord. 2020;21(1):127–47.

    Article  CAS  PubMed  Google Scholar 

  17. Frolov A, Polcaro L, Lawson C, Tan Y, Martin JR III. A Rare case of 83-year-old transgender female: can thyroid hormone deficiency be involved in transgenderism and gender dysphoria? Adv Sex Med. 2020;10(2):23–40.

    Article  Google Scholar 

  18. Christensen JD, Davidge-Pitts C, Castro MR, Caraballo P. Characterization of thyroid disease prevalence among transgender and gender-diverse patients. J Endocr Soc. 2021;5(Supplement_1):A837–8.

    Article  PubMed Central  Google Scholar 

  19. T’Sjoen G, Arcelus J, Gooren L, Klink DT, Tangpricha V. Endocrinology of transgender medicine. Endocr Rev. 2019;40(1):97–117.

    Article  PubMed  Google Scholar 

  20. Tassinari V, De Gennaro V, La Sala G, Marazziti D, Bolasco G, Aguanno S, De Angelis L, Naro F, Pellegrini M. Atrophy, oxidative switching and ultrastructural defects in skeletal muscle of the ataxia telangiectasia mouse model. J Cell Sci. 2019;132(5):223008.

    Article  Google Scholar 

  21. Shackelford C, Long G, Wolf J, Okerberg C, Herbert R. Qualitative and quantitative analysis of nonneoplastic lesions in toxicology studies. Toxicol Pathol. 2002;30(1):93–6.

    Article  PubMed  Google Scholar 

  22. Rasinger JD, Carroll TS, Maranghi F, Tassinari R, Moracci G, Altieri I, et al. Low dose exposure to HBCD, CB-153 or TCDD induces histopathological and hormonal effects and changes in brain protein and gene expression in juvenile female BALB/c mice. Reprod Toxicol. 2018;80:105–16.

    Article  CAS  PubMed  Google Scholar 

  23. Gómez Á, Cerdán S, Pérez-Laso C, Ortega E, Pásaro E, Fernández R, et al. Effects of adult male rat feminization treatments on brain morphology and metabolomic profile. Horm Behav. 2020;125:104839.

    Article  PubMed  Google Scholar 

  24. Asarian L, Geary N. Modulation of appetite by gonadal steroid hormones. Philos Trans R Soc Lond B Biol Sci. 2006;361(1471):1251–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Klaver M, Dekker MJ, de Mutsert R, Twisk JW, den Heijer M. Cross-sex hormone therapy in transgender persons affects total body weight, body fat and lean body mass: a meta-analysis. Andrologia. 2017;49(5):e12660.

    Article  Google Scholar 

  26. Suppakitjanusant P, Ji Y, Stevenson MO, Chantrapanichkul P, Sineath RC, Goodman M, et al. Effects of gender affirming hormone therapy on body mass index in transgender individuals: a longitudinal cohort study. J Clin Transl Endocrinol. 2020;21:100230.

    PubMed  PubMed Central  Google Scholar 

  27. Collet S, Gieles NC, Wiepjes CM, Heijboer AC, Reyns T, Fiers T, et al. Changes in serum testosterone and adrenal androgen levels in transgender women with and without gonadectomy. J Clin Endocrinol Metab. 2023;108(2):331–8.

    Article  PubMed  Google Scholar 

  28. O’Connor JC, Frame SR, Ladics GS. Evaluation of a 15-day screening assay using intact male rats for identifying antiandrogens. Toxicol Sci. 2002;69(1):92–108.

    Article  CAS  PubMed  Google Scholar 

  29. Hossaini A, Dalgaard M, Vinggaard AM, Pakarinen P, Larsen JJ. Male reproductive effects of octylphenol and estradiol in Fischer and Wistar rats. Reprod Toxicol. 2003;17(5):607–15.

    Article  CAS  PubMed  Google Scholar 

  30. Jiang DD, Swenson E, Mason M, Turner KR, Dugi DD, Hedges JC, et al. Effects of estrogen on spermatogenesis in transgender women. Urology. 2019;132:117–22.

    Article  PubMed  Google Scholar 

  31. Kasarinaite A, Sinton M, Saunders PTK, Hay DC. The influence of sex hormones in liver function and disease. Cells. 2023;12(12):1604.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Maggi A, Della TS. Sex, metabolism and health. Mol Metab. 2018;15:3–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Brown EDL, Obeng-Gyasi B, Hall JE, Shekhar S. The thyroid hormone axis and female reproduction. Int J Mol Sci. 2023;24(12):9815.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. De Vincentis S, Monzani ML, Brigante G. Crosstalk between gonadotropins and thyroid axis. Minerva Ginecol. 2018;70(5):609–20.

    Article  PubMed  Google Scholar 

  35. Morenas R, Singh D, Hellstrom WJ. Thyroid disorders and male sexual dysfunction. Int J Impot Res. 2023;26:1–6.

    Google Scholar 

  36. Pirahanchi Y, Tariq MA, Jialal I. Physiology, Thyroid. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023.

  37. Huisinga M, Bertrand L, Chamanza R, Damiani I, Engelhardt J, Francke S, et al. Adversity considerations for thyroid follicular cell hypertrophy and hyperplasia in nonclinical toxicity studies: results from the 6th ESTP international expert workshop. Toxicol Pathol. 2020;48(8):920–38.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors wish to thank Prof. Arefeh Raoui for the precious comments and suggestions. Dr. Elena Monaco and Mr. P. Frassanito for their valuable technical contribution to the present work.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, RT and FM; methodology, RT and FM; validation, RT and FM; formal analysis, RT; investigation, RT, AT, GL, AM, LC; data curation, RT, AT and GL; writing—original draft preparation, AT, RT and FM; writing—review and editing, RT, AT, GL and FM; visualization, RT and AT; supervision, RT; project administration, RT and FM. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Roberta Tassinari.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The original online version of this article was revised: The order of the first name and family name of each author has been exchanged.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tammaro, A., Lori, G., Martinelli, A. et al. Risk assessment of transgender people: implementation of a demasculinizing–feminizing rodent model including the evaluation of thyroid homeostasis. Biol Direct 19, 5 (2024). https://doi.org/10.1186/s13062-023-00450-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13062-023-00450-1

Keywords