Skip to main content

Thyroid hormone: sex-dependent role in nervous system regulation and disease

Abstract

Thyroid hormone (TH) regulates many functions including metabolism, cell differentiation, and nervous system development. Alteration of thyroid hormone level in the body can lead to nervous system-related problems linked to cognition, visual attention, visual processing, motor skills, language, and memory skills. TH has also been associated with neuropsychiatric disorders including schizophrenia, bipolar disorder, anxiety, and depression. Males and females display sex-specific differences in neuronal signaling. Steroid hormones including testosterone and estrogen are considered to be the prime regulators for programing the neuronal signaling in a male- and female-specific manner. However, other than steroid hormones, TH could also be one of the key signaling molecules to regulate different brain signaling in a male- and female-specific manner. Thyroid-related diseases and neurological diseases show sex-specific incidence; however, the molecular mechanisms behind this are not clear. Hence, it will be very beneficial to understand how TH acts in male and female brains and what are the critical genes and signaling networks. In this review, we have highlighted the role of TH in nervous system regulation and disease outcome and given special emphasis on its sex-specific role in male and female brains. A network model is also presented that provides critical information on TH-regulated genes, signaling, and disease.

Introduction

The thyroid gland is one of the earliest endocrine organs that can be observed at twenty paired somites stage in a developing human embryo [1]. Thyroid hormones (THs) are first detected in the human fetal circulation at 11–13 gestation weeks [2]. The thyroid is the only endocrine gland that can produce and store thyroid hormones (THs), triiodothyronine (T3) and thyroxine (T4). T4 is the major TH secreted by the thyroid gland, whereas T3 is the main biologically active form. TH plays crucial role in regulating different aspects of animal physiology. The major role played by TH is regulation of metabolism, cellular growth, and development [3, 4]. However, recent advances in medical and molecular fields have helped to further dissect its other important role and mechanisms of action. TH has been shown to regulate nervous system differentiation as it influences neurogenesis, neuronal migration, neuronal and glial differentiation, myelination, and synaptogenesis [5,6,7,8]. Insufficiency in TH can lead to problems in cognition, visual attention, visual processing, motor skills, language, and memory skills [9]. TH is also implicated in neuropsychiatric disorders such as schizophrenia, bipolar disorder, anxiety, and depression [10, 11]. However, the molecular mechanisms of TH-mediated regulation of neuronal cells in these disorders are largely unknown. Some of the neurological diseases including Alzheimer’s disease (AD), Parkinson’s disease, and depression show a clear sex-specific incidence [12]. Moreover, thyroid-stimulating hormone (TSH) level has been associated with increased risk of dementia [13], and TSH level in plasma has become a routine screening test for diagnosis of patients with suspected dementia [14]. Low and high TSH has been associated with an increased risk of developing AD in women [15]. This suggests that elucidation of TH regulation and mechanisms of action in both male and female brains could further help to understand neuronal differentiation as well as neurological disease pathogenesis.

TH production, transport, and mechanisms of action

THs are synthesized by the thyroid gland and circulated via blood, but tissue deiodinase enzymes play a critical role in regulating their levels inside the tissues [16]. There are three different types of iodothyronine deiodinase enzymes involved in TH regulation, namely DIO1, DIO2, and DIO3. DIO2 converts the pro-hormone, tetraiodothyronine or thyroxine (T4), into the biologically active form, triiodothyronine (T3), whereas deiodinase type 3 enzyme (DIO3) catalyzes the inactivation of T3 and T4. DIO1 can both activate and inactivate thyroid hormone and shows non-selectivity and high Km (requires a supraphysiological level of the substrate) for the conversion of T4 to T3 [17, 18].

Although T3 and T4 are lipophilic, they cannot cross the plasma membrane without the help of a transporter. There are different transporters including the monocarboxylate transporter (MCT) family (MCT8/SLC16A2 and MCT10/SLC16A10) and organic anion transporter polypeptide (OATP) family (SLCO1C1and OATP1C1) that are involved in TH transfer in and out of the cell [19]. In mice, the role of Mct8 is considered to be more relevant than Mct10 as Mct8 knockout mice showed altered tissue homeostasis and serum T3 and T4 levels compared to Mct10 knockout mice [20]. MCT8 gene inactivation in humans can lead to Allan-Herndon-Dudley syndrome, a condition where patients show severe neurological problems [21, 22]. Interestingly, Mct8 gene knockout in mice does not show severe phenotype as in humans and this could be due to the availability of T4 through Oatp1c1 transporter and its conversion to T3 at the cellular level [20].

TH action is mainly exerted by interaction with TH and thyroid hormone receptors (THRs) which are mainly of two types, THRα and THRβ. THR is a nuclear receptor that requires TH as a ligand to be activated. THR binds to thyroid hormone response element (TRE) on the gene promoter and generally forms hetero dimer with retinoid X receptor (RXR) [23]. The TRE sequence consists of two consensus half sites, AGGT/ACA, arranged either as direct repeat, palindrome, or inverted palindrome. The RXR binds to 5′ half site while THR binds 3′ half site [24].

In the absence of TH, corepressors associate with THR to inhibit gene transcription. The binding of TH to the THR facilitates conformational change of THR, dissociation of corepressors from THR, and recruitment of coactivators, and this, in turn, drives gene transcription [4, 25, 26] (Fig. 1). The two half sites are generally separated by four nucleotides (DR4); however, other combinations are also reported. Among THs, T3 has approximately 10-fold higher binding affinity to THRs compared to T4 [25].

Fig. 1
figure 1

Mechanism of TH intracellular action. The introduction of TH into the cell is mediated by transporters. T4 can be converted into T3 by the enzyme, DIO2. DIO1 is also involved in the conversion of T4 (not shown in the figure). T3 and T4 bind to the THRs mainly associated with RXR, and this binding leads to the conformational change of THRs followed by dissociation of corepressors and recruitment of coactivators. This leads to the expression of genes that are located downstream of TREs

In addition to genomic effects of TH, non-genomic (transcription-independent/TRE independent) signaling has also been reported [27,28,29,30,31]. Compared to the genomic, the non-genomic action is rapid which takes place within seconds or minutes [28, 32]. Initially, it was noted that T3 can bind to rat erythrocytes membranes and mitochondrial fractions from rat liver [33, 34]. Later non-genomic effects of TH were reported for production of ATP, consumption of oxygen, activation of Na+/H+ exchanger, and increase of intracellular pH [35, 36].

The non-genomic action was suggested to be important for maintaining cell homeostasis by regulating ion concentration and cytoskeleton; however, the presence of crosstalk between genomic and non-genomic activities of TH is also proposed, which implies that the TH molecular mechanisms of action is diverse and complex [29]. The non-genomic action can initiate either at the cell membrane or in the cytoplasm, but the molecular mechanisms are not understood properly [31]. The cell surface receptor is generated from the internal translation initiation site of THRα which then gets palmitoylated and associates with caveolin-containing plasma membrane domains [27]. It is also shown that TH can mediate non-genomic activity via surface receptors αVβ3 integrin. TH action via αVβ3 leads to activation of FGF2, HIF1α, COX2, THRA, THRB, ESR1, MMP9, NOS2, SREBP1, and CD74 genes while the expressions of CASP3, BBC3, PMAIP, and APAF1 are downregulated [30]. The non-genomic activity is considered to be stronger for T4; however, it is not certain whether T3 or T4 acts on αVβ3 to regulate these genes [30]. It is not reported if the non-genomic action of TH can facilitate sex-specific signaling. However, based on the documented roles, it can be suggested that non-genomic action could be involved in differential signaling in males and females. For instance, TH crosstalk between genomic and non-genomic actions has been indicated for immune regulation [29], and since the immune system of males and females show sharp contrast [37,38,39], a sex-specific effect of TH on immune system via non-genomic action can be expected. Non-genomic action of TH is also considered to be important for brain development as T4 has been shown to alter actin polymerization and neural migration [40]. It is also suggested that activation of protein kinase Akt and endothelial nitric oxide synthase via T3 non-genomic action in rat brain could contribute to neuroprotective effects of TH [28]. Further investigation of TH non-genomic action in brain development will help to understand molecular mechanisms of TH in sex-specific regulation of neuronal signaling.

The production and secretion of THs are regulated by hypothalamus-pituitary-thyroid (HPT) axis. The hypothalamus (medial region of the paraventricular nucleus) synthesizes thyrotropin-releasing hormone (TRH) that enters the pituitary portal circulation. In the anterior pituitary, TRH stimulates the release of thyroid-stimulating hormone (TSH). TSH then travels to the thyroid gland where it stimulates the thyroid gland to secrete TH. The TH released in the circulation can regulate the level of TRH and TSH in the blood by negative feedback loop [41, 42].

TH receptors (THRs) and distribution in the brain

TH mainly mediates its action by binding to the THRs in the cell cytoplasm. The THRs belong to the nuclear receptor superfamily, and there are two different types of THRs, THRα and THRβ. The protein sequence comparison using CLUSTLW showed that there is 62.3% similarity between THRα (451 aa) and THRβ (461 aa). Although they share structural and sequence similarities, mutation in one cannot fully compensate for the loss of another [31], and patients with mutations in either THRα or THRβ have strikingly different clinical phenotypes [43]. Patients and mutant mice for THRβ show large goiter and hearing impairment deregulation of HPT [43] axis while Thrα mutation in mice exhibits increased mortality, reduced fertility, and dwarfism [44]. In mammals, different TH receptor isoforms have been identified; for instance, in humans, 3 isoforms of THRα (THRα1, THRα2, and THRα3) and 3 isoforms of THRβ (THRβ1, THRβ2, and THRβ3) were found in NCBI database (Fig. 2). THRα expression is observed throughout the brain whereas THRβ is mainly expressed in the subcortical region of the brain [46]. TH regulation is critically regulated by the expression of THRα and THRβ.

Fig. 2
figure 2

Thyroid hormone receptors. In humans, there are three isoforms of THRα and three isoforms of THRβ. The human protein sequence was obtained from NCBI, and the protein domain was prepared using the protein illustrator software DOG 1.0 [45] (a). The DNA binding domain (DBD) is highly conserved among the receptors; however, the ligand binding domain (LBD) shows differences in sequence and length (b)

In mouse brain, Thrα and Thrβ expressions were observed in different cells including endothelial cells, microglia, astrocytes, oligodendrocytes, and neurons. In these cells, the expression of Thrα was higher than that of Thrβ with endothelial and microglia showing the lowest expression (Fig. 3).

Fig. 3
figure 3

Expression of genes involved in different brain cells. RNA sequencing data was obtained from a previous study [47] and analyzed using Partek Flow software. The expression of genes involved in TH regulation was analyzed for mouse brain cells including endothelial, microglia, astrocytes, newly formed oligodendrocytes (New Oligo), oligodendrocyte precursor cells (OPC), myelinating oligodendrocyte (Myl Oligo), and neurons. n = 2 for all cell types

TH regulation and action in the brain

The active T3 and T4 produced in the thyroid gland enter the blood circulation which then gets distributed to different body parts. TH uptake in the brain is a slower process compared to other organs and is tightly regulated. Both T4 and T3 can cross blood-brain barrier (BBB) and enter the brain. TH transporter solute carrier family 16 member 2 (SLC16A2/MCT-8) and solute carrier organic anion transporter family member 1C1 (OATP1C1/SLCO1C1) are both present in the endothelial cells of BBB [48]. OATP1C1 is a T4 transporter; however, MCT8 can transport both T3 and T4 [19]. From the endothelial cells at the blood-brain barrier, T4 is transported to the astrocytes via membrane transporter OATP1c1, and in the astrocyte, T4 gets converted to T3 by the DIO2 enzyme. The expression of Oatp1c1 was higher in the endothelial cells followed by astrocytes (Fig. 3). The expression of other transporters (Slc16a2/Mct8 and Slc16a10/Mct10) (Fig. 3) could also provide important clues on brain cell regulation by TH. In the brain, the released T4 is taken up by the astrocytes and gets converted into an active form triiodothyronine, T3 [49]. It is indicated that the development of certain parts of the brain is dependent on the expression of deiodinases that convert T4 to more active T3 [2].

There is differential expression of iodothyronine deiodinases in different brain regions [19]. Dio1 is mainly active in mice cerebellum [50] and mostly absent in other brain regions, making Dio2 and Dio3 the major iodothyronine deiodinases in the brain. Deiodinases are membrane-bound proteins [51]. DIO2 is mainly located in the endoplasmic reticulum, and its catalytic domain is exposed to the ER lumen, whereas DIO1 and DIO3 are located in the plasma membrane having a catalytic domain exposed to the cytosolic side [52]. DIO2 is mainly expressed in astrocytes and DIO3 being mostly expressed in neurons. Sonic hedgehog (SHH) is a common regulator of both deiodinases. SHH induces DIO3 mRNA expression whereas it degrades DIO2 at the protein level via ubiquitination by WD repeat and SOCS box-containing box 1 (WSB1) [53, 54].

In our analysis, we observed that DIO2 and DIO3 genes are expressed in both male and female brains. Although the expression of these two genes was high in female microglia (DIO2 1.7 fold, p value 0.05, and DIO3 1.4 fold, p value 0.02), it was not significant after FDR adjustment. Comparison of counts showed that DIO2 expression is higher than DIO3 expression in both mice (Fig. 4) and human brains (DIO2 mean count 350 and DIO3 mean count 38). It is indicated that the source of T3 in microglia is astrocytes [11]; however, the presence of DIO2 suggests that microglia can produce T3 locally. This data should be confirmed with DIO2 protein and T3 level analysis. In the developing rat brain, the expression of THRα is higher than that of THRβ [57]. Analysis of transcriptomic data of human fetal brain from a recent study [58] also showed that THRα (transcript mean count 5320) expression is higher than THRβ (transcript mean count 614) expression. Interestingly, it was shown that the negative outcome of hypothyroidism is not due to the lack of either Thrα or Thrβ, but due to decreased level of T3 in the circulation [59]. Using the knockout mouse model for Thrα or Thrβ, the authors analyzed gene expression in the cerebral cortex and the striatum and showed that individual knockout of either gene does not show marked differences and the two genes can largely compensate for each other’s loss [59]. The knockout mouse model for T3 transport, monocarboxylate transporter 8 (Mct8) and Dio2, provided important information on localized T3 synthesis [60]. Inactivation of Mct8 showed limited effect on cerebral cortex gene expression postnatally. The authors suggested that this could be due to upregulation of Dio2 and local increase of T3 as the double knockout of Dio2 showed similar effects as hypothyroidism [60].

Fig. 4
figure 4

Expression of TH receptors and TH-synthesizing genes. The RNA seq data was obtained from NCBI submitted by a previous study [55]. Raw data were aligned and normalized using Partek Flow software. Expression analysis suggests that Dio2 expression is high in mouse microglia from cortex while Dio3 expression is high in the hippocampus. Thrα expression was found to higher than Thrβ expression. n = 3 males and n = 4 females (a). We further analyzed RNA seq data from another study [56] and observed that the expression of Dio2, Dio3, Thrα, and Thrβ in the mouse hippocampus is not strikingly different for the different developmental stages, n= 5 (b)

Transcriptional repressor or activators can also regulate TH-dependent signaling. Nuclear receptor corepressor 1 (NCOR1) is identified as the key corepressor of TH-regulated genes in mice hepatic tissue [61]. In the same study, deletion of NCOR2 did not show significant changes in global TH signaling. However, the expression of NCOR1 during brain development is not well studied. T3-dependent transcription through TRE is abolished in mediator complex subunit 1 (MED1) null cells which suggest the possible role of MED1 as an activator for T3-dependent transcription [62]. Hence, MED1 shows the opposite activity of NCOR1 in TH-dependent gene expression.

T3 is critical for microglia development, and it can also induce microglial migration and phagocytosis. Microglia are immune cells of the brain that are involved in maintaining brain homeostasis and implicated in disease and injury [63]. Microglia are also involved in the regulation of neural functions and sexual behavior [64, 65]. The study by Guneykaya et al. show that the microglia in males are more frequent in specific brain areas and appear to have a higher potential to respond to stimuli [55]. It is also indicated that T3 can regulate morphological maturation of ameoboid microglial cells and limit their degeneration [66]. Decreased level of TH has been shown to reduce microglial processes in postnatal rats [67]. Given the importance of microglia in brain development, sexual behavior, and its regulation by TH, it can be speculated that brain sexual differentiation or sex-specific brain organization may be regulated by TH-mediated microglial functions.

Insufficient TH signaling could result in arborization of Purkinje cells, delay in neuronal migration, outgrowth of neuronal processes, myelination, and synaptogenesis [68, 69]. TH deficiency also leads to neuronal death and glial cell proliferation [70]. Low perinatal TH levels result in reduced dendritic complexity in Purkinje cells in the cerebellum [71]. This suggests an important role of TH for the cerebellar motor function, and dysregulation of TH in the perinatal phase can have long-term effects. TH in the early developmental phase also regulates GABAergic neuron morphology and connectivity via control of TrkB and mTOR signaling [72]. Synthesis of GABA from glutamate is regulated by glutamate decarboxylase 65 and 67 (GAD65 and GAD67). In vivo and in vitro data suggest TH regulates the expression of GAD enzymes in the brain [73] thereby regulating GABA production. TH deficiency in early rat developmental phase causes reduction in parvalbumin (PV)-positive neurons suggesting TH are also involved in early cortical circuit development [74].

TH in the brain regulates several pathways that contribute to structural aspects during development such as neurogenesis, cell migration, and myelination. TH is mainly involved in later events of neural development including neural migration or neuron-glia differentiation [19]. TH has been linked to adult neurogenesis [75], and it mainly occurs in two regions in the brain, namely the subventricular and subgranular zones, and is generally associated with cognitive deficits, psychiatric conditions, and depression [76]. TH administration stimulates neurogenesis in these two brain regions, whereas hypothyroidism inhibits neurogenesis [77, 78].

TH can have effects on cell migration in different brain regions like the cerebellum, hippocampus, and cerebral cortex [76]. TH is responsible for formation of different layer patterns; this migration is achieved by regulation of genes RELN and PTGDS by TH [79, 80]. Hypothyroidism causes poor myelination; on the other hand, hyperthyroidism increases myelination [81,82,83]. Hyper and hypothyroidism show different sex-specific phenotype [84]. Behavioral activity including locomotor activity, water intake, motor coordination, and muscle strength showed sex-specific alteration in thyroid dysfunction mice in this study [84]. In order to understand TH role in brain development, different mutant animals have been studied. For instance, congenital hypothyroid mice (cog/cog mouse) with mutation in thyroglobulin (Tg) gene show significantly low cerebrum and cerebellar weight [85]. Mutation in the same gene (Tg) in rat (rdw rat) shows altered dopamine level in the substantia nigra and striatum, impaired motor coordination, retarded cerebellar morphogenesis, retarded migration of granule cells, and poor dendritic aborization of Purkinje cells [86]. Interestingly, the analyzed parameters in this study showed sex-specific differences. The motor coordination and balance measurement using the rotarod test on this rat model showed that rdw female and male rats, respectively, showed a 15% and 5% decrease in activity compared to wild type female and male rats. Among other parameters, rearing behavior in rdw rats were significantly decreased compared to female rats. The dopamine level in the substantia nigra was increased to around 1.6 fold in females while it increased 1.9 fold in males. On the other hand, dopamine in the striatum decreased by 1.5 fold in females while it decreased to 1.2 fold in males [86]. Mutation in Dual oxidase 2 (DUOX2) gene in humans leads to congenital hypothyroidism [87]. DUOX2 is involved in generation of hydrogen peroxide, which will then be utilized by thyroid peroxidase for iodine incorporation into thyroglobulin [88]. Mouse Duox2 mutant shows severe hypothyroidism and hearing impairment [88]. Models with Thrα or Thrβ mutation have also been studied to understand brain function; however, the phenotype of Thr mutant is different from hypothyroid models. Since the expression of Thrα is high in the brain compared to Thrβ (also observed in our study; Fig 3), severe phenotype is expected with Thrα mutation [89]. In addition, deletion of Thrα or Thrβ leads to different phenotypes as it has been shown that deletion of Thrα1 in mice reduces female sexual behavior while deletion of Thrβ increases it when stimulated with estrogen [90]. However, the impact of male sexual behavior was not evaluated in this study. Another study showed that deletion of Thrα1 in male mice altered exploratory behavior, decreased rearing behavior, and increased freezing behavior [91]. This behavior change was linked to altered hippocampal signaling [91].

Sex-specific effects of TH in the brain

Sex differences in the brain regulate not only reproductive functions but also cognitive abilities and susceptibility to neurological diseases. In mammals, gonadal steroid hormone surge during the fetal stage organizes the brain, and later during the adult stage, the second surge of gonadal hormone leads to behavioral activation. The classical model of brain sex differentiation suggests that the gonadal steroid hormones (androgens and estrogens) are the main drivers in establishing male and female neural networks [92, 93]. Although the role of steroid hormones is critical in organizing the brain in a male- and female-specific manner, the involvement of other key players including TH cannot be overruled.

The thyroid-related medical problems including hypothyroidism and hyperthyroidism are more common in females than males [94]. Transcriptomics analysis also revealed that aging-related changes in thyroid tissue are more common in females [95].

TH could have sex- and age-dependent effects as it has been shown that exposure of T4 to male mice results in activation of glial cells while that to female mice leads to deactivation [11]. Comparison of glial activation following exposure to T4 in young mice brain showed sex-specific effects. In males, T4 exposure activated glial cells while in females it deactivated them [11].

Critical information on sex-specific role of thyroid hormone came from songbird, zebra finch (Taeniopygia guttata). In this bird, the levels of T3 and T4 in the brain and plasma have been shown to be sex specific with male and female showing different peak periods [96].

The level of Dio2 mRNA was shown to increase at 21 days post-hatching (dph), and the level of T3 also started to increase after 21 dph in male brain [96]. In addition, Raymaekers et al. showed that the level of Dio2 is higher in the male song control nuclei [97]. The increase in Dio2 and T3 corresponds to the timing when zebra finch males learn to sing [96]. This suggests that TH is crucial for male typical brain development in zebra finches. Taken together, it can be suggested that TH could have sex-specific role in brain development.

In P0 (perinatal day 0) neonatal rats, Dio3 expression was transiently noted in regions involved in sexual differentiation in the brain. This expression was not observed in P10 rats, this suggests the role of Dio3 in early sexual differentiation in rodents [98]. There was sex-specific difference in Dio1 levels in mice. Expression of Dio1 in both pituitary and thyroid glands were higher in adult males compared to females [99]. However, there was no significant difference in TH levels between sexes in the same study.

SHH has been identified as a common regulator of both DIO2 and DIO3 [53, 54]. It induces DIO3 whereas degrades DIO2 and thereby plays an important role in maintaining balance of TH in intracellular context. SHH receptor patched1 (PTCH1) haplosufficiency shows sex-specific effect and female-specific reduction in hippocampus size and isocortical layer thickness [100]. It would be interesting to study whether SHH signaling can have sexually dimorphic effect in brain TH regulation.

Gould et al. noted in adult rats that females possessed more primary dendrites, whereas males showed more apical excrescences in CA3 pyramidal cells. TH treatment resulted in increased primary dendrites as well as apical excrescences in both sexes [101]. Sex difference was noted between serum TSH levels and depressive symptoms in cohort with normal serum T4 levels. Higher TSH level was correlated with higher prevalence of depressive symptoms in men whereas the opposite was noted for women [102].

To investigate whether thyroid signaling is differentially regulated in male and female brains, we analyzed different transcriptomics data that were available in the NCBI database deposited by previous studies [55, 56]. Analysis of male and female microglia from 1-, 2-, and 4-month-old mice did not show any strong differences. In all the three stages, the expression pattern showed a similar trend with higher expression of Dio2 compared to Dio3 and higher expression of Thrα than Thrβ (Fig. 4).

Analysis of transcriptomics data of microglia from the frontal cortex and the hippocampus of adult mice was also analyzed. In the frontal cortex, only 8 genes showed significant difference, and out of these, one gene, Dbp, is a T3-regulated gene [103]. Interestingly, the microglia from the hippocampus showed 1386 differentially regulated genes between males and females, and from this list, we extracted the T3-regulated genes (Fig. 5). Out of 23 regulated genes, 17 genes were high in males and 6 genes were high in females. The genes including THR, TH synthesis, and transport were not differentially regulated. Critical links are missing to understand the mechanisms of how TH can differentially regulate gene expression in male vs female brain.

Fig. 5
figure 5

THR response genes are differentially regulated in male and female microglia. RNA seq data was obtained from a previous study [55] and analyzed using Partek Flow software. The microglia from hippocampus showed sex-biased differences in the expression of TH-regulated genes. Fold change ± 1.5, p value 0.05, and FDR 0.05. n = 3 males and n = 4 females

Thyroid hormone in neurodegenerative and psychiatric diseases

Several neurological and psychiatric conditions are associated with TH dysregulation. Hypothyroidism during pregnancy increases risk of autism, cognitive impairment, and attention deficits [46]. On the other hand, hyperthyroidism is known to cause anxiety, hyperflexia, and irritability. Both hyper and hypothyroidism are associated with mood-related conditions, personality disorders, and dementia [76]. Hypothyroidism has been shown to induce interleukin 1 (IL-1)-mediated autophagy and neuronal apoptosis in postnatal rats that accounts for cognitive impairment [104].

Females are more frequently affected by AD than males [105]; the same is observed in thyroid dysfunction diseases [94]. It is intriguing whether there is an underlying correlation between TH and AD onset/pathogenesis. Intebi et al. studied some of the plasma markers in an AD cohort; however, they could not identify any change in circulating T3 and TSH levels between male and female AD patients [106]. However, in another study, female sex and thyroid dysfunction were correlated with AD endophenotype in the middle-aged population [107]. Further mechanistic understanding is needed to have a clear view on this aspect.

Low serum T4 and upregulated serum TSH levels showed correlation with brain amyloid beta levels and AD-specific brain alterations [108]. T3 administration in diabetic mice decreased glycemia, improved insulin sensitivity, and reduced GSK3B activation as well as tau protein load in hippocampus [109]. This is considered beneficial since hyperphosphorylated Tau (MAPT) accumulation and GSK3B activation are hallmarks of AD [110]. Apolipoprotein E (APOE) more specifically isoform APOE4 is associated with AD [111, 112]. A study in older Down syndrome (DS) patients having AD suggests that APOE2 might protect against hypothyroidism; however, APOE4 predispose towards the same [113]. This effect is only observed in females, and no such correlation was noted for males in the same study. It is concluded that APOE4 pathogenesis in AD patients is partially affected by thyroid function [113].

Cerebrospinal fluid (CSF) T3 levels were found to be higher in hippocampal sclerosis (HS) but at a normal level in AD [114]. HS-associated SNP rs73069071 was associated with mRNA expression levels of astrocyte TH transporter SLCO1C1 [114]. Mutations in TH transporters like MCT8 (SLC16A2), and OATP1C1 (SLCO1C1) cause juvenile neurodegeneration and brain developmental disease, Allan-Herndon-Dudley syndrome. oatp1c1 (slco1c1) knockout zebrafish also showed a similar phenotype [115, 116]. The function of TH in the context of myelination has been implicated in neurological disorders including multiple sclerosis (MS) to the extent that TH benefits MS by augmenting myelination [117, 118]. TH is often associated with antioxidant activities, and dysfunction of TH could increase reactive oxygen species (ROS) and, hence, oxidative stress which increases neurodegenerative mechanism in the brain [119]. T3 treatment showed neuroprotection in traumatic brain injury murine model [120]. This suggests that decreased TH level could predispose individual to ROS-mediated brain damage, and this, in turn, could aggravate the neurodegenerative outcome.

DIO1 polymorphism is associated with serum TH level and temporal lobe atrophy in the elderly population [121]. Thr92Ala-DIO2 has been associated with increased risk for AD in various cohorts [122].

Although there is no direct correlation between Parkinson’s disease (PD) and TH, there are reports explaining the commonalities between Parkinsonism and thyroid dysfunction. In particular PD patients suffering from hypothyroidism, hormone therapy proved to be helpful in reducing Parkinson’s bradykinesia and hypomimia [123]. On the other hand hyperthyroidism increases tremor in PD cases, which can be managed by anti-thyroid treatment [124].

Crystalline mu (CRYM) is a regulator of T3 transportation [125]. It has been reported that CRYM expression in the striatum is reduced in Huntington’s disease (HD) mouse model and overexpression of CRYM reduced mutant Htt-mediated neurotoxicity [126]. This could be an important mechanism linking decreased TSH and T3 levels observed in HD patients [127].

Many studies have associated thyroid status with cognition, mood, and behavior. Thyroid dysfunction can lead to psychiatric changes without other symptoms of the disorder to the extent that hypothyroidism can be falsely presented as psychosis in older women [128]. Thyroid dysfunction is also noted in patients with schizophrenia spectrum disorders, bipolar disorder, and major depressive disorder [129]. Higher T3 and T4 and lower TSH levels were observed in schizophrenic patients [130]. The T3 levels in schizophrenics correlated significantly with plasma malondialdehyde and total plasma peroxides (TPP), which suggest higher TPP could contribute to better thyroid homeostasis in schizophrenia through regulation of free radicals and oxidative stress [130]. A strong correlation has been noted between anti-psychotic drug lithium and higher TSH and T4 and lower T3 levels in bipolar disorder patients [131]. The increased volume of thyroid gland following lithium treatment was also noted in the same study. Hypothyroidism is a common effect of long-term lithium treatment [132]. Thyroid dysfunction is more common in females than males, and this contributes to increased difficulties in diagnosis and treatment of mood disorders like bipolar disease [133]. Hypothyroidism is also noted in women with postpartum depression [134]. Presence of anti-thyroid auto-antibodies correlated with higher occurrence of panic disorder and major depressive disorder in a cohort of celiac disease [135]. Interestingly, higher serum TSH levels correlate with lower depressive symptoms in individuals with normal serum TH levels [102].

Thyroid-related diseases show sex-specific and age-dependent incidences with females showing 5–20 times higher susceptibility than males [136]. The underlying molecular mechanism is not clear but the difference in sex steroid milieu could be a critical determining factor. During menopause and andropause, the level of estrogen in females and testosterone in males drops down [137]. In males, low serum testosterone was associated with depression [138], low memory and cognitive skills [139], and risk of AD [140]. On the other, menopause in females decreases cognition and increases the chances for AD [141]. However, there are conflicting data whether hormone replacement therapy in women can prevent neurological diseases [141, 142]. This suggests that the neurological disease outcome in elderly people is multifactorial where TH could also play a crucial role. In aged men and women, the thyroid function decreases [143] and consequently it alters brain function [144] and increases AD risk in women [15]. In menopausal women with altered thyroid function, neurological problems including depression and anxiety are common [145]. Since the prevalence of thyroid-related and neurological diseases increase and steroid hormone level decreases with age, a positive correlation between sex steroid, thyroid dysfunction, and neurological diseases could be expected.

Overall, there are many reports and indications that TH status is critical in several neurodegenerative and psychiatric diseases. However, the molecular mechanisms of the pathogenesis are not well elucidated. In Fig. 6, we have tried to assemble the known biomolecules and pathways associated with TH regulation and signaling. Mechanistic studies are required to have a better understanding of the involvement of TH in neurodegenerative and psychiatric diseases.

Fig. 6
figure 6

Thyroid hormone-related molecular mechanism in the brain: TH-related molecular causal relationships were extracted from the articles listed in the reference section of this manuscript, and the manually curated network was built on systems biology graphical notation (SBGN) [146] using Pathvisio 3.3.0 network editor [147]. Only major causal (cause-effect) relationships were used for the network; correlative relationships were not included in this model

Conclusion and future perspectives

TH regulates critical biological processes including brain differentiation. TH has been shown to regulate brain differentiation, and any alteration in level could lead to various nervous system-related problems. The common neurological problems associated with TH are cognition, visual attention, visual processing, motor skills, language, and memory skills. TH shows sex-specific effects in brain cell differentiation which could lead to differential organization of neural circuits. TH-related problems are also on the rise with females showing higher incidence. Our study suggests that there is clear sex-specific effects and regulation of TH in male and female brains. The sex-specific role of TH has started to emerge; however, critical links are missing to fully understand the molecular mechanisms. Understanding of TH sex-specific effects could further help to advance the diagnostic as well as the therapeutic field.

Availability of data and materials

Not applicable

Abbreviations

AD:

Alzheimer’s disease

APOE:

Apolipoprotein E

BBB:

Blood-brain barrier

CRYM:

Crystalline mu

CSF:

Cerebrospinal fluid

DIO:

Iodothyronine deiodinase

DS:

Down’s syndrome

HPT:

Hypothalamus-pituitary-thyroid axis

HD:

Huntington’s disease

MCT8:

Solute carrier family 16 member 2 (SLC16A2)

OATP1C1:

Solute carrier organic anion transporter family member 1C1 (SLCO1C1)

PD:

Parkinson’s disease

ROS:

Reactive oxygen species

RXR:

Retinoid X receptor

TH:

Thyroid hormone

THR:

Thyroid hormone receptor

TRH:

Thyrotropin-releasing hormone

TSH:

Thyroid-stimulating hormone

T4:

Thyroxine

T3:

Triiodothyronine

TPP:

Total plasma peroxides

SHH:

Sonic hedgehog

References

  1. Davis CL. Description of a human embryo having twenty paired somites; 1923.

    Google Scholar 

  2. Stathatos N. Thyroid physiology. Med Clin North Am. 2012;96(2):165–73.

    Article  CAS  PubMed  Google Scholar 

  3. Mullur R, Liu YY, Brent GA. Thyroid hormone regulation of metabolism. Physiological Rev. 2014;94(2):355–82.

    Article  CAS  Google Scholar 

  4. Brent GA. Mechanisms of thyroid hormone action. J Clin Invest. 2012;122(9):3035–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schoonover CM, Seibel MM, Jolson DM, Stack MJ, Rahman RJ, Jones SA, Mariash CN, Anderson GW. Thyroid hormone regulates oligodendrocyte accumulation in developing rat brain white matter tracts. Endocrinology. 2004;145(11):5013–20.

    Article  CAS  PubMed  Google Scholar 

  6. Martinez-Galan JR, Pedraza P, Santacana M, Escobar del Ray F, Morreale de Escobar G, Ruiz-Marcos A. Early effects of iodine deficiency on radial glial cells of the hippocampus of the rat fetus. A model of neurological cretinism. J Clin Invest. 1997;99(11):2701–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lima FR, Goncalves N, Gomes FC, de Freitas MS, Moura Neto V. Thyroid hormone action on astroglial cells from distinct brain regions during development. Int J Dev Neurosci. 1998;16(1):19–27.

    Article  CAS  PubMed  Google Scholar 

  8. Manzano J, Bernal J, Morte B. Influence of thyroid hormones on maturation of rat cerebellar astrocytes. Int J Dev Neurosci. 2007;25(3):171–9.

    Article  CAS  PubMed  Google Scholar 

  9. Williams GR. Neurodevelopmental and neurophysiological actions of thyroid hormone. J Neuroendocrinol. 2008;20(6):784–94.

    Article  CAS  PubMed  Google Scholar 

  10. Nandi-Munshi D, Taplin CE. Thyroid-related neurological disorders and complications in children. Pediatr Neurol. 2015;52(4):373–82.

    Article  PubMed  Google Scholar 

  11. Noda M. Thyroid hormone in the CNS: contribution of neuron-glia interaction. Vitam Horm. 2018;106:313–31.

    Article  CAS  PubMed  Google Scholar 

  12. Podcasy JL, Epperson CN. Considering sex and gender in Alzheimer disease and other dementias. Dialogues Clin Neurosci. 2016;18(4):437–46.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Ganguli M, Burmeister LA, Seaberg EC, Belle S, DeKosky ST. Association between dementia and elevated TSH: a community-based study. Biol Psychiatry. 1996;40(8):714–25.

    Article  CAS  PubMed  Google Scholar 

  14. Knopman DS, DeKosky ST, Cummings JL, Chui H, Corey-Bloom J, Relkin N, Small GW, Miller B, Stevens JC. Practice parameter: diagnosis of dementia (an evidence-based review). Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2001;56(9):1143–53.

    Article  CAS  PubMed  Google Scholar 

  15. Tan ZS, Beiser A, Vasan RS, Au R, Auerbach S, Kiel DP, Wolf PA, Seshadri S. Thyroid function and the risk of Alzheimer disease: the Framingham study. Arch Intern Med. 2008;168(14):1514–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rousset B, Dupuy C, Miot F, Dumont J. Chapter 2 thyroid hormone synthesis and secretion. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, et al., editors. Endotext. South Dartmouth; 2000.

  17. Gereben B, McAninch EA, Ribeiro MO, Bianco AC. Scope and limitations of iodothyronine deiodinases in hypothyroidism. Nat Rev Endocrinol. 2015;11(11):642–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Darras VM, Van Herck SL. Iodothyronine deiodinase structure and function: from ascidians to humans. J Endocrinol. 2012;215(2):189–206.

    Article  CAS  PubMed  Google Scholar 

  19. Bernal J. Thyroid hormones in brain development and function. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dungan K, Grossman A, Hershman JM, Hofland HJ, Kaltsas G, et al., editors. Endotext: South Dartmouth; 2000.

    Google Scholar 

  20. Muller J, Mayerl S, Visser TJ, Darras VM, Boelen A, Frappart L, Mariotta L, Verrey F, Heuer H. Tissue-specific alterations in thyroid hormone homeostasis in combined Mct10 and Mct8 deficiency. Endocrinology. 2014;155(1):315–25.

    Article  PubMed  Google Scholar 

  21. Dumitrescu AM, Liao XH, Best TB, Brockmann K, Refetoff S. A novel syndrome combining thyroid and neurological abnormalities is associated with mutations in a monocarboxylate transporter gene. Am J Hum Genet. 2004;74(1):168–75.

    Article  CAS  PubMed  Google Scholar 

  22. Friesema ECH, Grueters A, Biebermann H, Krude H, von Moers A, Reeser M, Barrett TG, Mancilla EE, Svensson J, Kester MHA, et al. Association between mutations in a thyroid hormone transporter and severe X-linked psychomotor retardation. Lancet. 2004;364(9443):1435–7.

    Article  CAS  PubMed  Google Scholar 

  23. Phan TQ, Jow MM, Privalsky ML. DNA recognition by thyroid hormone and retinoic acid receptors: 3,4,5 rule modified. Mol Cell Endocrinol. 2010;319(1–2):88–98.

    Article  CAS  PubMed  Google Scholar 

  24. Chatonnet F, Guyot R, Benoit G, Flamant F. Genome-wide analysis of thyroid hormone receptors shared and specific functions in neural cells. Proc Natl Acad Sci U S A. 2013;110(8):E766–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sinha R, Yen PM. Cellular action of thyroid hormone. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, Dungan K, Grossman A, Hershman JM, Kaltsas G, Koch C, Kopp P, et al., editors. Endotext. South Dartmouth; 2000.

  26. Sinha RA, Singh BK, Yen PM. Thyroid hormone regulation of hepatic lipid and carbohydrate metabolism. Trends Endocrinol Metab. 2014;25(10):538–45.

    Article  CAS  PubMed  Google Scholar 

  27. Kalyanaraman H, Schwappacher R, Joshua JS, Zhuang SH, Scott BT, Klos M, Casteel DE, Frangos JA, Dillmann W, Boss GR, et al. Nongenomic thyroid hormone signaling occurs through a plasma membrane-localized receptor. Sci Signal. 2014;7(326).

  28. Hiroi Y, Kim HH, Ying H, Furuya F, Huang Z, Simoncini T, Noma K, Ueki K, Nguyen NH, Scanlan TS, et al. Rapid nongenomic actions of thyroid hormone. Proc Natl Acad Sci U S A. 2006;103(38):14104–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Incerpi S, Davis PJ, Pedersen JZ, Lanni A. Nongenomic actions of thyroid hormones. In: Belfiore A, LeRoith D, editors. Principles of endocrinology and hormone action. Cham: Springer International Publishing; 2016. p. 1–26.

    Google Scholar 

  30. Davis PJ, Goglia F, Leonard JL. Nongenomic actions of thyroid hormone. Nat Rev Endocrinol. 2016;12(2):111–21.

    Article  CAS  PubMed  Google Scholar 

  31. Cheng SY, Leonard JL, Davis PJ. Molecular aspects of thyroid hormone actions. Endocr Rev. 2010;31(2):139–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sterling K, Brenner MA, Sakurada T. Rapid effect of triiodothyronine on the mitochondrial pathway in rat liver in vivo. Science. 1980;210(4467):340–2.

    Article  CAS  PubMed  Google Scholar 

  33. Botta JA, de Mendoza D, Morero RD, Farias RN. High affinity L-triiodothyronine binding sites on washed rat erythrocyte membranes. J Biol Chem. 1983;258(11):6690–2.

    Article  CAS  PubMed  Google Scholar 

  34. Sterling K, Lazarus JH, Milch PO, Sakurada T, Brenner MA. Mitochondrial thyroid hormone receptor: localization and physiological significance. Science. 1978;201(4361):1126–9.

    Article  CAS  PubMed  Google Scholar 

  35. D'Arezzo S, Incerpi S, Davis FB, Acconcia F, Marino M, Farias RN, Davis PJ. Rapid nongenomic effects of 3,5,3′-triiodo-L-thyronine on the intracellular pH of L-6 myoblasts are mediated by intracellular calcium mobilization and kinase pathways. Endocrinology. 2004;145(12):5694–703.

    Article  CAS  PubMed  Google Scholar 

  36. Incerpi S, Luly P, De Vito P, Farias RN. Short-term effects of thyroid hormones on the Na/H antiport in L-6 myoblasts: high molecular specificity for 3,3′,5-triiodo-L-thyronine. Endocrinology. 1999;140(2):683–9.

    Article  CAS  PubMed  Google Scholar 

  37. Schurz H, Salie M, Tromp G, Hoal EG, Kinnear CJ, Moller M. The X chromosome and sex-specific effects in infectious disease susceptibility. Hum Genomics. 2019;13.

  38. Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. 2016;16(10):626–38.

    Article  CAS  PubMed  Google Scholar 

  39. Pradhan A, Olsson PE. Sex differences in severity and mortality from COVID-19: are males more vulnerable? Biol Sex Differ. 2020;11(1):53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Leonard JL. Non-genomic actions of thyroid hormone in brain development. Steroids. 2008;73(9–10):1008–12.

    Article  CAS  PubMed  Google Scholar 

  41. Nillni EA. Regulation of the hypothalamic thyrotropin releasing hormone (TRH) neuron by neuronal and peripheral inputs. Front Neuroendocrinol. 2010;31(2):134–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Smith SM, Vale WW. The role of the hypothalamic-pituitary-adrenal axis in neuroendocrine responses to stress. Dialogues Clin Neurosci. 2006;8(4):383–95.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Ortiga-Carvalho TM, Sidhaye AR, Wondisford FE. Thyroid hormone receptors and resistance to thyroid hormone disorders. Nat Rev Endocrinol. 2014;10(10):582–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kaneshige M, Suzuki H, Kaneshige K, Cheng J, Wimbrow H, Barlow C, Willingham MC, Cheng S. A targeted dominant negative mutation of the thyroid hormone alpha 1 receptor causes increased mortality, infertility, and dwarfism in mice. Proc Natl Acad Sci U S A. 2001;98(26):15095–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ren J, Wen LP, Gao XJ, Jin CJ, Xue Y, Yao XB. DOG 1.0: illustrator of protein domain structures. Cell Res. 2009;19(2):271–3.

    Article  CAS  PubMed  Google Scholar 

  46. Batista G, Hensch TK. Critical period regulation by thyroid hormones: potential mechanisms and sex-specific aspects. Front Mol Neurosci. 2019;12:77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang Y, Chen KN, Sloan SA, Bennett ML, Scholze AR, O'Keeffe S, Phatnani HP, Guarnieri P, Caneda C, Ruderisch N, et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci. 2014;34(36):11929–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Roberts LM, Woodford K, Zhou M, Black DS, Haggerty JE, Tate EH, Grindstaff KK, Mengesha W, Raman C, Zerangue N. Expression of the thyroid hormone transporters monocarboxylate transporter-8 (SLC16A2) and organic ion transporter-14 (SLCO1C1) at the blood-brain barrier. Endocrinology. 2008;149(12):6251–61.

    Article  CAS  PubMed  Google Scholar 

  49. Prezioso G, Giannini C, Chiarelli F. Effect of thyroid hormones on neurons and neurodevelopment. Horm Res Paediatr. 2018;90(2):73–81.

    Article  CAS  PubMed  Google Scholar 

  50. Barez-Lopez S, Bosch-Garcia D, Gomez-Andres D, Pulido-Valdeolivas I, Montero-Pedrazuela A, Obregon MJ, Guadano-Ferraz A. Abnormal motor phenotype at adult stages in mice lacking type 2 deiodinase. PLoS One. 2014;9(8):e103857.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Bianco AC, Larsen PR. Cellular and structural biology of the deiodinases. Thyroid. 2005;15(8):777–86.

    Article  CAS  PubMed  Google Scholar 

  52. Baqui M, Botero D, Gereben B, Curcio C, Harney JW, Salvatore D, Sorimachi K, Larsen PR, Bianco AC. Human type 3 iodothyronine selenodeiodinase is located in the plasma membrane and undergoes rapid internalization to endosomes. J Biol Chem. 2003;278(2):1206–11.

    Article  CAS  PubMed  Google Scholar 

  53. Dentice M, Bandyopadhyay A, Gereben B, Callebaut I, Christoffolete MA, Kim BW, Nissim S, Mornon JP, Zavacki AM, Zeold A, et al. The hedgehog-inducible ubiquitin ligase subunit WSB-1 modulates thyroid hormone activation and PTHrP secretion in the developing growth plate. Nat Cell Biol. 2005;7(7):698–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dentice M, Luongo C, Huang S, Ambrosio R, Elefante A, Mirebeau-Prunier D, Zavacki AM, Fenzi G, Grachtchouk M, Hutchin M, et al. Sonic hedgehog-induced type 3 deiodinase blocks thyroid hormone action enhancing proliferation of normal and malignant keratinocytes. Proc Natl Acad Sci U S A. 2007;104(36):14466–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Guneykaya D, Ivanov A, Hernandez DP, Haage V, Wojtas B, Meyer N, Maricos M, Jordan P, Buonfiglioli A, Gielniewski B, et al. Transcriptional and translational differences of microglia from male and female brains. Cell Rep. 2018;24(10):2773–83 e2776.

    Article  CAS  PubMed  Google Scholar 

  56. Bundy JL, Vied C, Nowakowski RS. Sex differences in the molecular signature of the developing mouse hippocampus. BMC Genomics. 2017;18(1):237.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Oppenheimer JH, Schwartz HL. Molecular basis of thyroid hormone-dependent brain development. Endocr Rev. 1997;18(4):462–75.

    CAS  PubMed  Google Scholar 

  58. O'Brien H, Hannon E, Jeffries AR, Davies W, Hill MJ, Anney R, O'Donovan M, Mill J, Bray NJB. Sex differences in gene expression in the human fetal brain; 2019.

    Google Scholar 

  59. Gil-Ibanez P, Morte B, Bernal J. Role of thyroid hormone receptor subtypes alpha and beta on gene expression in the cerebral cortex and striatum of postnatal mice. Endocrinology. 2013;154(5):1940–7.

    Article  PubMed  Google Scholar 

  60. Morte B, Ceballos A, Diez D, Grijota-Martinez C, Dumitrescu AM, Di Cosmo C, Galton VA, Refetoff S, Bernal J. Thyroid hormone-regulated mouse cerebral cortex genes are differentially dependent on the source of the hormone: a study in monocarboxylate transporter-8-and deiodinase-2-deficient mice. Endocrinology. 2010;151(5):2381–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Shimizu H, Astapova I, Ye F, Bilban M, Cohen RN, Hollenberg AN. NCoR1 and SMRT play unique roles in thyroid hormone action in vivo. Mol Cell Biol. 2015;35(3):555–65.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Park SW, Li G, Lin YP, Barrero MJ, Ge K, Roeder RG, Wei LN. Thyroid hormone-induced juxtaposition of regulatory elements/factors and chromatin remodeling of Crabp1 dependent on MED1/TRAP220. Mol Cell. 2005;19(5):643–53.

    Article  CAS  PubMed  Google Scholar 

  63. Li Q, Barres BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol. 2018;18(4):225–42.

    Article  CAS  PubMed  Google Scholar 

  64. Lenz KM, McCarthy MM. A starring role for microglia in brain sex differences. Neuroscientist. 2015;21(3):306–21.

    Article  CAS  PubMed  Google Scholar 

  65. Schafer DP, Lehrman EK, Stevens B. The “quad-partite” synapse: microglia-synapse interactions in the developing and mature CNS. Glia. 2013;61(1):24–36.

    Article  PubMed  Google Scholar 

  66. Mallat M, Lima FRS, Gervais A, Colin C, Neto VM. New insights into the role of thyroid hormone in the CNS: the microglial track. Mol Psychiatry. 2002;7(1):7–8.

    Article  CAS  PubMed  Google Scholar 

  67. Lima FR, Gervais A, Colin C, Izembart M, Neto VM, Mallat M. Regulation of microglial development: a novel role for thyroid hormone. J Neurosci. 2001;21(6):2028–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Hashimoto K, Curty FH, Borges PP, Lee CE, Abel ED, Elmquist JK, Cohen RN, Wondisford FE. An unliganded thyroid hormone receptor causes severe neurological dysfunction. Proc Natl Acad Sci U S A. 2001;98(7):3998–4003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Venero C, Guadaño-Ferraz A, Herrero AI, Nordström K, Manzano J, de Escobar GM, Bernal J, BJG V. Development. Anxiety, memory impairment, and locomotor dysfunction caused by a mutant thyroid hormone receptor α1 can be ameliorated by T3 treatment. Genes Dev. 2005;19(18):2152–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nunez J, Celi FS, Ng L, Forrest D. Multigenic control of thyroid hormone functions in the nervous system. Mol Cell Endocrinol. 2008;287(1–2):1–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Nicholson JL, Altman J. The effects of early hypo- and hyperthyroidism on the development of the rat cerebellar cortex. II. Synaptogenesis in the molecular layer. Brain Res. 1972;44(1):25–36.

    Article  CAS  PubMed  Google Scholar 

  72. Westerholz S, de Lima AD, Voigt T. Thyroid hormone-dependent development of early cortical networks: temporal specificity and the contribution of trkB and mTOR pathways. Front Cell Neurosci. 2013;7:121.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Wiens SC, Trudeau VL. Thyroid hormone and gamma-aminobutyric acid (GABA) interactions in neuroendocrine systems. Comp Biochem Physiol A Mol Integr Physiol. 2006;144(3):332–44.

    Article  PubMed  CAS  Google Scholar 

  74. Berbel P, Marco P, Cerezo JR, DeFelipe J. Distribution of parvalbumin immunoreactivity in the neocortex of hypothyroid adult rats. Neurosci Lett. 1996;204(1–2):65–8.

    Article  CAS  PubMed  Google Scholar 

  75. Remaud S, Gothie JD, Morvan-Dubois G, Demeneix BA. Thyroid hormone signaling and adult neurogenesis in mammals. Front Endocrinol. 2014;5:62.

    Article  Google Scholar 

  76. Bernal J. Thyroid hormones in brain development and function; 2000.

    Google Scholar 

  77. Ambrogini P, Cuppini R, Ferri P, Mancini C, Ciaroni S, Voci A, Gerdoni E, Gallo G. Thyroid hormones affect neurogenesis in the dentate gyrus of adult rat. Neuroendocrinology. 2005;81(4):244–53.

    Article  CAS  PubMed  Google Scholar 

  78. Desouza LA, Ladiwala U, Daniel SM, Agashe S, Vaidya RA, Vaidya VA. Thyroid hormone regulates hippocampal neurogenesis in the adult rat brain. Mol Cell Neurosci. 2005;29(3):414–26.

    Article  CAS  PubMed  Google Scholar 

  79. Alvarez-Dolado M, Gonzalez-Moreno M, Valencia A, Zenke M, Bernal J, Munoz A. Identification of a mammalian homologue of the fungal Tom70 mitochondrial precursor protein import receptor as a thyroid hormone-regulated gene in specific brain regions. J Neurochem. 1999;73(6):2240–9.

    Article  CAS  PubMed  Google Scholar 

  80. Garcia-Fernandez LF, Rausell E, Urade Y, Hayaishi O, Bernal J, Munoz A. Hypothyroidism alters the expression of prostaglandin D2 synthase/beta trace in specific areas of the developing rat brain. Eur J Neurosci. 1997;9(8):1566–73.

    Article  CAS  PubMed  Google Scholar 

  81. Balazs R, Brooksbank BW, Davison AN, Eayrs JT, Wilson DA. The effect of neonatal thyroidectomy on myelination in the rat brain. Brain Res. 1969;15(1):219–32.

    Article  CAS  PubMed  Google Scholar 

  82. Malone MJ, Rosman NP, Szoke M, Davis D. Myelination of brain in experimental hypothyroidism. An electron-microscopic and biochemical study of purified myelin isolates. J Neurol Sci. 1975;26(1):1–11.

    Article  CAS  PubMed  Google Scholar 

  83. Adamo AM, Aloise PA, Soto EF, Pasquini JM. Neonatal hyperthyroidism in the rat produces an increase in the activity of microperoxisomal marker enzymes coincident with biochemical signs of accelerated myelination. J Neurosci Res. 1990;25(3):353–9.

    Article  CAS  PubMed  Google Scholar 

  84. Rakov H, Engels K, Hones GS, Strucksberg KH, Moeller LC, Kohrle J, Zwanziger D, Fuhrer D. Sex-specific phenotypes of hyperthyroidism and hypothyroidism in mice. Biol Sex Diff. 2016;7(1):36.

    Article  CAS  Google Scholar 

  85. Sugisaki T, Beamer WG, Noguchi T. Microcephalic cerebrum with hypomyelination in the congenital goiter mouse (cog). Neurochem Res. 1992;17(10):1037–40.

    Article  CAS  PubMed  Google Scholar 

  86. Shimokawa N, Yousefi B, Morioka S, Yamaguchi S, Ohsawa A, Hayashi H, Azuma A, Mizuno H, Kasagi M, Masuda H, et al. Altered cerebellum development and dopamine distribution in a rat genetic model with congenital hypothyroidism. J Neuroendocrinol. 2014;26(3):164–75.

    Article  CAS  PubMed  Google Scholar 

  87. Vigone MC, Fugazzola L, Zamproni I, Passoni A, Di Candia S, Chiumello G, Persani L, Weber G. Persistent mild hypothyroidism associated with novel sequence variants of the DUOX2 gene in two siblings. Hum Mutat. 2005;26(4):395.

    Article  PubMed  Google Scholar 

  88. Johnson KR, Marden CC, Ward-Bailey P, Gagnon LH, Bronson RT, Donahue LR. Congenital hypothyroidism, dwarfism, and hearing impairment caused by a missense mutation in the mouse dual oxidase 2 gene, Duox2. Mol Endocrinol. 2007;21(7):1593–602.

    Article  CAS  PubMed  Google Scholar 

  89. Koibuchi N. Animal models to study thyroid hormone action in cerebellum. Cerebellum. 2009;8(2):89–97.

    Article  CAS  PubMed  Google Scholar 

  90. Dellovade TL, Chan J, Vennstrom B, Forrest D, Pfaff DW. The two thyroid hormone receptor genes have opposite effects on estrogen-stimulated sex behaviors. Nat Neurosci. 2000;3(5):472–5.

    Article  CAS  PubMed  Google Scholar 

  91. Guadaño-Ferraz A, Benavides-Piccione A, Venero C, Lancha C, Vennström B, Sandi C, DeFelipe J, Bernal J. Lack of thyroid hormone receptor alpha1 is associated with selective alterations in behavior and hippocampal circuits. Mol Psychiatry. 2003;8(26):30–8.

    Article  PubMed  CAS  Google Scholar 

  92. Phoenix CH, Goy RW, Gerall AA, Young WC. Organizing action of prenatally administered testosterone propionate on the tissues mediating mating behavior in the female guinea pig. Endocrinology. 1959;65:369–82.

    Article  CAS  PubMed  Google Scholar 

  93. Arnold AP. The organizational-activational hypothesis as the foundation for a unified theory of sexual differentiation of all mammalian tissues. Horm Behav. 2009;55(5):570–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Morganti S, Ceda GP, Saccani M, Milli B, Ugolotti D, Prampolini R, Maggio M, Valenti G, Ceresini G. Thyroid disease in the elderly: sex-related differences in clinical expression. J Endocrinol Investig. 2005;28(11 Suppl Proceedings):101–4.

    CAS  Google Scholar 

  95. Cho BA, Yoo SK, Song YS, Kim SJ, Lee KE, Shong M, Park YJ, Seo JS. Transcriptome network analysis reveals aging-related mitochondrial and proteasomal dysfunction and immune activation in human thyroid. Thyroid. 2018;28(5):656–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Yamaguchi S, Hayase S, Aoki N, Takehara A, Ishigohoka J, Matsushima T, Wada K, Homma KJ. Sex differences in brain thyroid hormone levels during early post-hatching development in zebra finch (Taeniopygia guttata). PLoS One. 2017;12(1):e0169643.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Raymaekers SR, Verbeure W, Ter Haar SM, Cornil CA, Balthazart J, Darras VM. A dynamic, sex-specific expression pattern of genes regulating thyroid hormone action in the developing zebra finch song control system. Gen Comp Endocrinol. 2017;240:91–102.

    Article  CAS  PubMed  Google Scholar 

  98. Escamez MJ, Guadano-Ferraz A, Cuadrado A, Bernal J. Type 3 iodothyronine deiodinase is selectively expressed in areas related to sexual differentiation in the newborn rat brain. Endocrinology. 1999;140(11):5443–6.

    Article  CAS  PubMed  Google Scholar 

  99. Marassi MP, Fortunato RS, da Silva AC, Pereira VS, Carvalho DP, Rosenthal D, da Costa VM. Sexual dimorphism in thyroid function and type 1 iodothyronine deiodinase activity in pre-pubertal and adult rats. J Endocrinol. 2007;192(1):121–30.

    Article  CAS  PubMed  Google Scholar 

  100. Jackson TW, Bendfeldt GA, Beam KA, Rock KD, Belcher SM. Heterozygous mutation of sonic hedgehog receptor (Ptch1) drives cerebellar overgrowth and sex-specifically alters hippocampal and cortical layer structure, activity, and social behavior in female mice. Neurotoxicol Teratol. 2020;78:106866.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Gould E, Westlind-Danielsson A, Frankfurt M, McEwen BS. Sex differences and thyroid hormone sensitivity of hippocampal pyramidal cells. J Neurosci. 1990;10(3):996–1003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Lee S, Oh SS, Park EC, Jang SI. Sex differences in the association between thyroid-stimulating hormone levels and depressive symptoms among the general population with normal free T4 levels. J Affect Disord. 2019;249:151–8.

    Article  CAS  PubMed  Google Scholar 

  103. Morte B, Gil-Ibanez P, Bernal J. Regulation of gene expression by thyroid hormone in primary astrocytes: factors influencing the genomic response. Endocrinology. 2018;159(5):2083–92.

    Article  CAS  PubMed  Google Scholar 

  104. Mishra J, Vishwakarma J, Malik R, Gupta K, Pandey R, Maurya SK, Garg A, Shukla M, Chattopadhyay N, Bandyopadhyay S. Hypothyroidism induces interleukin-1-dependent autophagy mechanism as a key mediator of hippocampal neuronal apoptosis and cognitive decline in postnatal rats. Mol Neurobiol. 2020.

  105. Beam CR, Kaneshiro C, Jang JY, Reynolds CA, Pedersen NL, Gatz M. Differences between women and men in incidence rates of dementia and Alzheimer’s disease. JAD. 2018;64(4):1077–83.

    Article  PubMed  Google Scholar 

  106. Intebi AD, Garau L, Brusco I, Pagano M, Gaillard RC, Spinedi E. Alzheimer’s disease patients display gender dimorphism in circulating anorectic adipokines. Neuroimmunomodulation. 2002;10(6):351–8.

    Article  CAS  PubMed  Google Scholar 

  107. Rahman A, Schelbaum E, Hoffman K, Diaz I, Hristov H, Andrews R, Jett S, Jackson H, Lee A, Sarva H, et al. Sex-driven modifiers of Alzheimer risk: a multimodality brain imaging study. Neurology. 2020;95(2):e166–78.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Choi HJ, Byun MS, Yi D, Sohn BK, Lee JH, Lee JY, Kim YK, Lee DY. Associations of thyroid hormone serum levels with in-vivo Alzheimer’s disease pathologies. Alzheimers Res Ther. 2017;9(1):64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Prieto-Almeida F, Panveloski-Costa AC, Crunfli F, da Silva TS, Nunes MT, Torrao ADS. Thyroid hormone improves insulin signaling and reduces the activation of neurodegenerative pathway in the hippocampus of diabetic adult male rats. Life Sci. 2018;192:253–8.

    Article  CAS  PubMed  Google Scholar 

  110. Congdon EE, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease. Nat Rev Neurol. 2018;14(7):399–415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Uddin MS, Kabir MT, Al Mamun A, Abdel-Daim MM, Barreto GE, Ashraf GM. APOE and Alzheimer’s disease: evidence mounts that targeting APOE4 may combat Alzheimer’s pathogenesis. Mol Neurobiol. 2019;56(4):2450–65.

    Article  CAS  PubMed  Google Scholar 

  112. Munoz SS, Garner B, Ooi L. Understanding the role of ApoE fragments in Alzheimer’s disease. Neurochem Res. 2019;44(6):1297–305.

    Article  CAS  PubMed  Google Scholar 

  113. Percy ME, Potyomkina Z, Dalton AJ, Fedor B, Mehta P, Andrews DF, Mazzulli T, Murk L, Warren AC, Wallace RA, et al. Relation between apolipoprotein E genotype, hepatitis B virus status, and thyroid status in a sample of older persons with Down syndrome. Am J Med Genet A. 2003;120A(2):191–8.

    Article  PubMed  Google Scholar 

  114. Nelson PT, Katsumata Y, Nho K, Artiushin SC, Jicha GA, Wang WX, Abner EL, Saykin AJ, Kukull WA, Fardo DW. Genomics and CSF analyses implicate thyroid hormone in hippocampal sclerosis of aging. Acta Neuropathol. 2016;132(6):841–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Admati I, Wasserman-Bartov T, Tovin A, Rozenblat R, Blitz E, Zada D, Lerer-Goldshtein T, Appelbaum L. Neural alterations and hyperactivity of the hypothalamic-pituitary-thyroid axis in Oatp1c1 deficiency. Thyroid. 2020;30(1):161–74.

    Article  CAS  PubMed  Google Scholar 

  116. Stromme P, Groeneweg S, Lima de Souza EC, Zevenbergen C, Torgersbraten A, Holmgren A, Gurcan E, Meima ME, Peeters RP, Visser WE, et al. Mutated thyroid hormone transporter OATP1C1 associates with severe brain hypometabolism and juvenile neurodegeneration. Thyroid. 2018;28(11):1406–15.

    Article  CAS  PubMed  Google Scholar 

  117. Hartley MD, Banerji T, Tagge IJ, Kirkemo LL, Chaudhary P, Calkins E, Galipeau D, Shokat MD, DeBell MJ, Van Leuven S, et al. Myelin repair stimulated by CNS-selective thyroid hormone action. JCI Insight. 2019;4(8).

  118. Zhang M, Ma Z, Qin H, Yao Z. Thyroid hormone potentially benefits multiple sclerosis via facilitating remyelination. Mol Neurobiol. 2016;53(7):4406–16.

    Article  CAS  PubMed  Google Scholar 

  119. Villanueva I, Alva-Sanchez C, Pacheco-Rosado J. The role of thyroid hormones as inductors of oxidative stress and neurodegeneration. Oxidative Med Cell Longev. 2013;2013:218145.

    Article  CAS  Google Scholar 

  120. Crupi R, Paterniti I, Campolo M, Di Paola R, Cuzzocrea S, Esposito E. Exogenous T3 administration provides neuroprotection in a murine model of traumatic brain injury. Pharmacol Res. 2013;70(1):80–9.

    Article  CAS  PubMed  Google Scholar 

  121. de Jong FJ, Peeters RP, den Heijer T, van der Deure WM, Hofman A, Uitterlinden AG, Visser TJ, Breteler MM. The association of polymorphisms in the type 1 and 2 deiodinase genes with circulating thyroid hormone parameters and atrophy of the medial temporal lobe. J Clin Endocrinol Metab. 2007;92(2):636–40.

    Article  PubMed  CAS  Google Scholar 

  122. Bianco AC, Kim BS. Pathophysiological relevance of deiodinase polymorphism. Curr Opin Endocrinol Diabetes Obes. 2018;25(5):341–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Garcia-Moreno JM, Chacon J. Hypothyroidism concealed by Parkinson’s disease. Rev Neurol. 2002;35(8):741–2.

    CAS  PubMed  Google Scholar 

  124. Kim HT, Edwards MJ, Lakshmi Narsimhan R, Bhatia KP. Hyperthyroidism exaggerating parkinsonian tremor: a clinical lesson. Parkinsonism Relat Disord. 2005;11(5):331–2.

    Article  PubMed  Google Scholar 

  125. Takeshige K, Sekido T, Kitahara J, Ohkubo Y, Hiwatashi D, Ishii H, Nishio S, Takeda T, Komatsu M, Suzuki S. Cytosolic T3-binding protein modulates dynamic alteration of T3-mediated gene expression in cells. Endocr J. 2014;61(6):561–70.

    Article  CAS  PubMed  Google Scholar 

  126. Francelle L, Galvan L, Gaillard MC, Guillermier M, Houitte D, Bonvento G, Petit F, Jan C, Dufour N, Hantraye P, et al. Loss of the thyroid hormone-binding protein Crym renders striatal neurons more vulnerable to mutant huntingtin in Huntington’s disease. Hum Mol Genet. 2015;24(6):1563–73.

    Article  CAS  PubMed  Google Scholar 

  127. Saleh N, Moutereau S, Durr A, Krystkowiak P, Azulay JP, Tranchant C, Broussolle E, Morin F, Bachoud-Levi AC, Maison P. Neuroendocrine disturbances in Huntington’s disease. PLoS One. 2009;4(3):e4962.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Heinrich TW, Grahm G. Hypothyroidism presenting as psychosis: myxedema madness revisited. Prim Care Companion J Clin Psychiatry. 2003;5(6):260–6.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Radhakrishnan R, Calvin S, Singh JK, Thomas B, Srinivasan K. Thyroid dysfunction in major psychiatric disorders in a hospital based sample. Indian J Med Res. 2013;138(6):888–93.

    PubMed  PubMed Central  Google Scholar 

  130. Akiibinu MO, Ogundahunsi OA, Ogunyemi EO. Inter-relationship of plasma markers of oxidative stress and thyroid hormones in schizophrenics. BMC Res Notes. 2012;5:169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Kraszewska A, Ziemnicka K, Jonczyk-Potoczna K, Sowinski J, Rybakowski JK. Thyroid structure and function in long-term lithium-treated and lithium-naive bipolar patients. Hum Psychopharmacol. 2019;34(4):e2708.

    PubMed  Google Scholar 

  132. Kraszewska A, Abramowicz M, Chlopocka-Wozniak M, Sowinski J, Rybakowski J. The effect of lithium on thyroid function in patients with bipolar disorder. Psychiatr Pol. 2014;48(3):417–28.

    Article  PubMed  Google Scholar 

  133. Bauer M, Glenn T, Pilhatsch M, Pfennig A, Whybrow PC. Gender differences in thyroid system function: relevance to bipolar disorder and its treatment. Bipolar Disord. 2014;16(1):58–71.

    Article  PubMed  Google Scholar 

  134. Harris B. Postpartum depression and thyroid antibody status. Thyroid. 1999;9(7):699–703.

    Article  CAS  PubMed  Google Scholar 

  135. Carta MG, Hardoy MC, Boi MF, Mariotti S, Carpiniello B, Usai P. Association between panic disorder, major depressive disorder and celiac disease: a possible role of thyroid autoimmunity. J Psychosom Res. 2002;53(3):789–93.

    Article  PubMed  Google Scholar 

  136. Gietka-Czernel M. The thyroid gland in postmenopausal women: physiology and diseases. Prz Menopauzalny. 2017;16(2):33–7.

    PubMed  PubMed Central  Google Scholar 

  137. Decaroli MC, Rochira V. Aging and sex hormones in males. Virulence. 2017;8(5):545–70.

    Article  CAS  PubMed  Google Scholar 

  138. Ford AH, Yeap BB, Flicker L, Hankey GJ, Chubb SA, Handelsman DJ, Golledge J, Almeida OP. Prospective longitudinal study of testosterone and incident depression in older men: the health in men study. Psychoneuroendocrinology. 2016;64:57–65.

    Article  CAS  PubMed  Google Scholar 

  139. Moffat SD, Zonderman AB, Metter EJ, Blackman MR, Harman SM, Resnick SM. Longitudinal assessment of serum free testosterone concentration predicts memory performance and cognitive status in elderly men. J Clin Endocrinol Metab. 2002;87(11):5001–7.

    Article  CAS  PubMed  Google Scholar 

  140. Zonderman AB. Predicting Alzheimer’s disease in the Baltimore longitudinal study of aging. J Geriatr Psychiatry Neurol. 2005;18(4):192–5.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Henderson VW. Cognition and cognitive aging. Climacteric. 2007;10(Suppl 2):88–91.

    Article  PubMed  Google Scholar 

  142. Henderson VW. The neurology of menopause. Neurologist. 2006;12(3):149–59.

    Article  PubMed  Google Scholar 

  143. Barbesino G. Thyroid function changes in the elderly and their relationship to cardiovascular health: a mini-review. Gerontology. 2019;65(1):1–8.

    Article  PubMed  Google Scholar 

  144. Shin YW, Choi YM, Kim HS, Kim DJ, Jo HJ, O'Donnell BF, Jang EK, Kim TY, Shong YK, Hong JP, et al. Diminished quality of life and increased brain functional connectivity in patients with hypothyroidism after total thyroidectomy. Thyroid. 2016;26(5):641–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Gulseren S, Gulseren L, Hekimsoy Z, Cetinay P, Ozen C, Tokatlioglu B. Depression, anxiety, health-related quality of life, and disability in patients with overt and subclinical thyroid dysfunction. Arch Med Res. 2006;37(1):133–9.

    Article  PubMed  Google Scholar 

  146. Le Novere N, Hucka M, Mi H, Moodie S, Schreiber F, Sorokin A, Demir E, Wegner K, Aladjem MI, Wimalaratne SM, et al. The systems biology graphical notation. Nat Biotechnol. 2009;27(8):735–41.

    Article  PubMed  CAS  Google Scholar 

  147. Kutmon M, van Iersel MP, Bohler A, Kelder T, Nunes N, Pico AR, Evelo CT. PathVisio 3: an extendable pathway analysis toolbox. PLoS Comput Biol. 2015;11(2):e1004085.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank the funding agency and Örebro University for supporting this study. We would also like to thank Professor Nicholas Bray (Cardiff University) for providing the human fetal brain data.

Funding

This study was financed by O.E and Edla Johanssons Scientific Foundation and Örebro University. Open Access funding provided by Örebro University.

Author information

Authors and Affiliations

Authors

Contributions

SB and AP did the manuscript writing and editing and data analysis. AP is responsible for the funding acquisition. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Ajay Pradhan.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable.

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.

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

Baksi, S., Pradhan, A. Thyroid hormone: sex-dependent role in nervous system regulation and disease. Biol Sex Differ 12, 25 (2021). https://doi.org/10.1186/s13293-021-00367-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13293-021-00367-2

Keywords