18 research outputs found

    Thyroid Hormones Regulate Selenoprotein Expression and Selenium Status in Mice

    Get PDF
    Impaired expression of selenium-containing proteins leads to perturbed thyroid hormone (TH) levels, indicating the central importance of selenium for TH homeostasis. Moreover, critically ill patients with declining serum selenium develop a syndrome of low circulating TH and a central downregulation of the hypothalamus-pituitary-thyroid axis. This prompted us to test the reciprocal effect, i.e., if TH status would also regulate selenoprotein expression and selenium levels. To investigate the TH dependency of selenium metabolism, we analyzed mice expressing a mutant TH receptor α1 (TRα1+m) that confers a receptor-mediated hypothyroidism. Serum selenium was reduced in these animals, which was a direct consequence of the mutant TRα1 and not related to their metabolic alterations. Accordingly, hyperthyroidism, genetically caused by the inactivation of TRβ or by oral TH treatment of adult mice, increased serum selenium levels in TRα1+m and controls, thus demonstrating a novel and specific role for TRα1 in selenium metabolism. Furthermore, TH affected the mRNA levels for several enzymes involved in selenoprotein biosynthesis as well as serum selenoprotein P concentrations and the expression of other antioxidative selenoproteins. Taken together, our results show that TH positively affects the serum selenium status and regulates the expression of several selenoproteins. This demonstrates that selenium and TH metabolism are interconnected through a feed-forward regulation, which can in part explain the rapid parallel downregulation of both systems in critical illness

    3-Iodothyronamine Activates a Set of Membrane Proteins in Murine Hypothalamic Cell Lines

    Get PDF
    3-Iodothyronamine (3-T1AM) is an endogenous thyroid hormone metabolite. The profound pharmacological effects of 3-T1AM on energy metabolism and thermal homeostasis have raised interest to elucidate its signaling properties in tissues that pertain to metabolic regulation and thermogenesis. Previous studies identified G protein-coupled receptors (GPCRs) and transient receptor potential channels (TRPs) as targets of 3-T1AM in different cell types. These two superfamilies of membrane proteins are largely expressed in tissue which influences energy balance and metabolism. As the first indication that 3-T1AM virtually modulates the function of the neurons in hypothalamus, we observed that intraperitoneal administration of 50 mg/kg bodyweight of 3-T1AM significantly increased the c-FOS activation in the paraventricular nucleus (PVN) of C57BL/6 mice. To elucidate the underlying mechanism behind this 3-T1AM-induced signalosome, we used three different murine hypothalamic cell lines, which are all known to express PVN markers, GT1-7, mHypoE-N39 (N39) and mHypoE-N41 (N41). Various aminergic GPCRs, which are the known targets of 3-T1AM, as well as numerous members of TRP channel superfamily, are expressed in these cell lines. Effects of 3-T1AM on activation of GPCRs were tested for the two major signaling pathways, the action of Gαs/adenylyl cyclase and Gi/o. Here, we demonstrated that this thyroid hormone metabolite has no significant effect on Gi/o signaling and only a minor effect on the Gαs/adenylyl cyclase pathway, despite the expression of known GPCR targets of 3-T1AM. Next, to test for other potential mechanisms involved in 3-T1AM-induced c-FOS activation in PVN, we evaluated the effect of 3-T1AM on the intracellular Ca2+ concentration and whole-cell currents. The fluorescence-optic measurements showed a significant increase of intracellular Ca2+ concentration in the three cell lines in the presence of 10 μM 3-T1AM. Furthermore, this thyroid hormone metabolite led to an increase of whole-cell currents in N41 cells. Interestingly, the TRPM8 selective inhibitor (10 μM AMTB) reduced the 3-T1AM stimulatory effects on cytosolic Ca2+ and whole-cell currents. Our results suggest that the profound pharmacological effects of 3-T1AM on selected brain nuclei of murine hypothalamus, which are known to be involved in energy metabolism and thermoregulation, might be partially attributable to TRP channel activation in hypothalamic cells

    Determinants of selenium status in healthy adults

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Selenium (Se) status in non-deficient subjects is typically assessed by the Se contents of plasma/serum. That pool comprises two functional, specific selenoprotein components and at least one non-functional, non-specific components which respond differently to changes in Se intake. A more informative means of characterizing Se status in non-deficient individuals is needed.</p> <p>Methods</p> <p>Multiple biomarkers of Se status (plasma Se, serum selenoprotein P [SEPP1], plasma glutathione peroxidase activity [GPX3], buccal cell Se, urinary Se) were evaluated in relation to selenoprotein genotypes (GPX1, GPX3, SEPP1, SEP15), dietary Se intake, and parameters of single-carbon metabolism in a cohort of healthy, non-Se-deficient men (n = 106) and women (n = 155).</p> <p>Conclusions</p> <p>Plasma Se concentration was 142.0 ± 23.5 ng/ml, with GPX3 and serum-derived SEPP1 calculated to comprise 20% and 34%, respectively, of that total. The balance, comprised of non-specific components, accounted for virtually all of the interindividual variation in total plasma Se. Buccal cell Se was associated with age and plasma homocysteine (hCys), but not plasma Se. SEPP1 showed a quadratic relationship with body mass index, peaking at BMI 25-30. Urinary Se was greater in women than men, and was associated with metabolic body weight (kg<sup>0.75</sup>), plasma folate, vitamin B<sub>12 </sub>and hCys (negatively). One <it>GPX1 </it>genotype (679T/T) was associated with significantly lower plasma Se levels than other allelic variants. Selenium intake, estimated from food frequency questionnaires, did not predict Se status as indicated by any biomarker. These results show that genotype, methyl-group status and BMI contribute to variation in Se biomarkers in Se-adequate individuals.</p

    3-Iodothyronamine Activates a Set of Membrane Proteins in Murine Hypothalamic Cell Lines

    No full text
    3-Iodothyronamine (3-T1AM) is an endogenous thyroid hormone metabolite. The profound pharmacological effects of 3-T1AM on energy metabolism and thermal homeostasis have raised interest to elucidate its signaling properties in tissues that pertain to metabolic regulation and thermogenesis. Previous studies identified G protein-coupled receptors (GPCRs) and transient receptor potential channels (TRPs) as targets of 3-T1AM in different cell types. These two superfamilies of membrane proteins are largely expressed in tissue which influences energy balance and metabolism. As the first indication that 3-T1AM virtually modulates the function of the neurons in hypothalamus, we observed that intraperitoneal administration of 50 mg/kg bodyweight of 3-T1AM significantly increased the c-FOS activation in the paraventricular nucleus (PVN) of C57BL/6 mice. To elucidate the underlying mechanism behind this 3-T1AM-induced signalosome, we used three different murine hypothalamic cell lines, which are all known to express PVN markers, GT1-7, mHypoE-N39 (N39) and mHypoE-N41 (N41). Various aminergic GPCRs, which are the known targets of 3-T1AM, as well as numerous members of TRP channel superfamily, are expressed in these cell lines. Effects of 3-T1AM on activation of GPCRs were tested for the two major signaling pathways, the action of Gαs/adenylyl cyclase and Gi/o. Here, we demonstrated that this thyroid hormone metabolite has no significant effect on Gi/o signaling and only a minor effect on the Gαs/adenylyl cyclase pathway, despite the expression of known GPCR targets of 3-T1AM. Next, to test for other potential mechanisms involved in 3-T1AM-induced c-FOS activation in PVN, we evaluated the effect of 3-T1AM on the intracellular Ca2+ concentration and whole-cell currents. The fluorescence-optic measurements showed a significant increase of intracellular Ca2+ concentration in the three cell lines in the presence of 10 μM 3-T1AM. Furthermore, this thyroid hormone metabolite led to an increase of whole-cell currents in N41 cells. Interestingly, the TRPM8 selective inhibitor (10 μM AMTB) reduced the 3-T1AM stimulatory effects on cytosolic Ca2+ and whole-cell currents. Our results suggest that the profound pharmacological effects of 3-T1AM on selected brain nuclei of murine hypothalamus, which are known to be involved in energy metabolism and thermoregulation, might be partially attributable to TRP channel activation in hypothalamic cells

    Effects of maternal or postnatal hyperthyroidism on serum selenium.

    No full text
    <p>Serum levels of selenium in wild-type (wt, white bars) and TRα1+m mutants (+m, black bars) born by hyperthyroid TRβ−/− mothers (high maternal TH), with inactivation of TRβ, which causes endogenous postnatal hyperthyroidism (high endogenous TH), or a combination of both (high endogenous, high maternal TH). (***: p<0.001 for T3 treatment, 2-way ANOVA, n = 5 per group).</p

    Selenium levels in wild-type and TRα1+m mutant mice with or without T3 treatment.

    No full text
    <p>A) Analysis of selenium concentration in serum, urine, liver and kidney of wild-type mice (wt, white bars) and animals heterozygous for a mutant TRα1 (+m, black bars), untreated or treated with supraphysiological doses of thyroid hormone (T3-treated). (##: p<0.01 for genotype, **: p<0.01 for T3 treatment, ***: p<0.001 for T3 treatment, 2-way ANOVA, n = 10 for serum, n = 5 for tissues and urine per group). B) Serum levels of selenium in wild-type (wt, white bars) and TRα1+m mice (+m, black bars) at thermoneutrality (30C). (#: p<0.05 for genotype, p = 0.39 for environmental temperature).</p

    Hepatic, renal and serum glutathione-peroxidase activity and serum Sepp concentrations in wild-type and TRα1+m mice with or without T3 treatment.

    No full text
    <p>A) Enzymatic activity of glutathione peroxidase (Gpx) in liver and kidney of untreated and TH treated (T3-treated) wild-type (wt, white bars) and TRα1+m mice (+m, black bars). The activity is normalized against the protein content of the sample. (**: p<0.01 for T3 treatment, ##: p<0.01 for genotype, 2-way ANOVA, n = 5 for each group). B) Enzymatic activity of serum glutathione peroxidase 3 (Serum Gpx) and concentrations of selenium protein P (Serum Sepp) levels in serum of untreated and TH treated (T3-treated) wild-type (wt, white bars) and TRα1+m mice (+m, black bars). (#: p<0.05 for genotype, **: p<0.01 for T3 treatment, 2-way ANOVA).</p

    Hepatic and renal gene expression in wild-type and TRα1+m mice with or without T3 treatment.

    No full text
    <p>A) Expression profiling of genes involved in selenium metabolism in livers of untreated and TH treated (T3-treated) wild-type (wt, white bars) and TRα1+m mice (+m, black bars). The expression is normalized against the housekeeping gene HPRT. Sepp: selenoprotein P, SecS: selenocysteine t-RNA synthase, Sebp1: selenium binding protein 1, Pstk: phosphoseryl-tRNA kinase, EFsec: selenocysteine-specific elongation factor, Sephs2: selenophosphate-synthetase 2, mScLy: selenocysteine lyase, GPx1: glutathione peroxidase 1, Dio1: deiodinase type I, SelW/SelH: selenoprotein W or H. B) Expression profiling of genes involved in selenium metabolism in kidneys of untreated and TH treated (T3-treated) wild-type (wt, white bars) and TRα1+m mice (+m, black bars). The expression is normalized against the housekeeping gene HPRT. Dio1: deiodinases type I, Sepp: selenoprotein P, Gpx1: glutathione peroxidase 1, Gpx3: glutathione peroxidase 3, SelW/SelH: selenoprotein W or H. (###: p<0.001 for T3 treatment of the wild-type, *: p<0.05 for T3 treatment, **: p<0.01 for T3 treatment, ***: p<0.001 for T3 treatment, 2-way ANOVA with Bonferroni post hoc test, n = 5 for each group).</p

    3,5-Diiodo-L-Thyronine (3,5-T2) Exerts Thyromimetic Effects on Hypothalamus-Pituitary-Thyroid Axis, Body Composition, and Energy Metabolism in Male Diet-Induced Obese Mice

    No full text
    Effective and safe antiobesity drugs are still needed in face of the obesity pandemic worldwide. Recent interventions in rodents revealed 3,5-diiodo-L-thyronine (3,5-T2) as a metabolically active iodothyronine affecting energy and lipid metabolism without thyromimetic side effects typically associated with T3 administration. Accordingly, 3,5-T2 has been proposed as a potential hypolipidemic agent for treatment of obesity and hepatic steatosis. In contrast to other observations, our experiments revealed dose-dependent thyromimetic effects of 3,5-T2 akin to those of T3 in diet-induced obese male C57BL/6J mice. 3,5-T2 treatment exerted a negative feedback regulation on the hypothalamus-pituitary-thyroid axis, similar to T3. This is demonstrated by decreased expression of genes responsive to thyroid hormones (TH) in pituitary resulting in a suppressed thyroid function with lower T4 and T3 concentrations in serum and liver of 3,5-T2-treated mice. Analyses of hepatic TH target genes involved in lipid metabolism revealed T3-like changes in gene expression and increased type I-deiodinase activity after application of 3,5-T2 (2.5 μg/g body weight). Reduced hepatic triglyceride and serum cholesterol concentrations reflected enhanced lipid metabolism. Desired increased metabolic rate and reduction of different fat depots were, however, compromised by increased food intake preventing significant body weight loss. Moreover, enlarged heart weights indicate potential cardiac side effects of 3,5-T2 beyond hepatic thyromimetic actions. Altogether, the observed thyromimetic effects of 3,5-T2 in several mouse TH target tissues raise concern about indiscriminate administration of 3,5-T2 as powerful natural hormone for the treatment of hyperlipidemia and pandemic obesity
    corecore