Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Thyroid ; 32(2): 196-205, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34641706

RESUMO

Background: L-triiodothyronine (LT3) has been increasingly used in combination with levothyroxine in the treatment of hypothyroidism. A metal coordinated form of LT3, known as poly-zinc-liothyronine (PZL), avoided in rats the typical triiodothyronine (T3) peak seen after oral administration of LT3. Objectives: To evaluate in healthy volunteers (i) the pharmacokinetics (PK) of PZL-derived T3 after a single dose, (ii) the pharmacodynamics of PZL-derived T3, (iii) incidence of adverse events, and (iv) exploratory analysis of the sleep patterns after LT3, PZL, or placebo (PB) administration. Methods: Twelve healthy volunteers 18-50 years of age were recruited for a Phase 1, double-blind, randomized, single-dose PB-controlled, crossover study to compare PZL against LT3 or PB. Subjects were admitted three separate times to receive a randomly assigned capsule containing PB, 50 µg LT3, or 50 µg PZL, and were observed for 48 hours. A 2-week washout period separated each admission. Results: LT3-derived serum T3 levels exhibited the expected profile, with a Tmax at 2 hours and return to basal levels by 24-36 hours. PZL-derived serum T3 levels exhibited ∼30% lower Cmax that was 1 hour delayed and extended into a plateau that lasted up to 6 hours. This was followed by a lower but much longer plateau; by 24 hours serum T3 levels still exceeded ½ of Cmax. Thyrotropin levels were similarly reduced in both groups. Conclusion: PZL possesses the necessary properties to achieve a much improved T3 PK. PZL is on track to provide hypothyroid patients with stable levels of serum T3.


Assuntos
Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/farmacocinética , Zinco/administração & dosagem , Adolescente , Adulto , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Terapia de Reposição Hormonal , Humanos , Hipotireoidismo/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Adulto Jovem
2.
Thyroid ; 29(10): 1371-1379, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31364488

RESUMO

Background: L-triiodothyronine (LT3) is a substitute for levothyroxine (LT4) for thyroid cancer (TC) patients during the preparation for nuclear medicine procedures, and it is used in combination with LT4 in patients who do not respond to the standard treatment for hypothyroidism. This therapy is commonly done by using fixed doses, potentially resulting in supraphysiologic levels of triiodothyronine (T3). A good understanding of the LT3 pharmacokinetics (PK) is necessary to design combination treatment schemes that are able to maintain serum T3 levels within the reference range, but data on the PK of LT3 are conflicting. Here, we present a study designed to characterize the PK of LT3 in patients devoid of endogenous thyroid hormone production, and not receiving LT4 therapy. Methods: We performed an open-label, PK study in patients undergoing thyroid hormone withdrawal in preparation for nuclear medicine procedures for the evaluation and treatment of follicular-derived TC. LT3 was substituted for LT4 at a 1:3 mcg/mcg dosage ratio thrice daily for at least 30 days. PK of the last LT3 dose while at steady state and terminal elimination was assessed over 11 days. Thereafter, a PK study was performed following the nuclear medicine procedure in patients who volunteered for a second study. Results: Fourteen patients age 48.5 ± 16.0 years completed the last dose study and five completed the second PK study. PK analysis indicates a time to maximum serum concentration of 1.8 ± 0.32 hours and two distinct phases of linear elimination, with a fast distribution phase and slow elimination phases with half-lives of 2.3 ± 0.11 hours and 22.9 ± 7.7 hours, supporting a two-compartment model. PK modeling predicts that a twice-daily administration of low-dose LT3 (0.07 mcg/kg twice daily) in combination with LT4 can predictably increase the serum T3 concentration without significant peaks above the reference range. Conclusions: The PK of LT3 is well described by a two-compartment model that assumes elimination only from the sampling compartment, with a rapid distribution phase and a slow elimination phase. This information will contribute to design therapeutic strategies for LT3/LT4 combination therapies directed to maintain stable T3 serum levels.


Assuntos
Hipotireoidismo/tratamento farmacológico , Neoplasias da Glândula Tireoide/cirurgia , Tireoidectomia , Tiroxina/uso terapêutico , Tri-Iodotironina/farmacocinética , Adulto , Desprescrições , Feminino , Terapia de Reposição Hormonal , Humanos , Hipotireoidismo/etiologia , Masculino , Pessoa de Meia-Idade , Cintilografia , Tireotropina/sangue , Tri-Iodotironina/uso terapêutico
3.
Am J Cardiol ; 123(6): 905-912, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30638544

RESUMO

The aim of the study was to investigate whether TH replacement therapy is safe and impact infarct size, left ventricular (LV) volumes and function in patients with acute myocardial infarction (AMI) and low T3 syndrome (LT3S). Thirty-seven AMI/LT3S patients were randomly treated or untreated with liothyronine (T3) therapy (maximum dosage 15 mcg/m2/die) in addition to standardized treatment (T3-treated group, n = 19; untreated group, n = 18). TH and thyroxine (TSH) during hospital stay and at 1-month and 6 months were evaluated. At discharge and at 6 months LV volumes, ejection fraction, wall motion score index (WMSI) and infarct extent were measured by cardiac MR. T3-treated patients had a significant increase in fT3 (p = 0.003 and p <0.001) at discharge and 1-month. These patients had no signs or symptoms of hyperthyroidism or arrhythmias. At follow-up, there was a significant reduction in WMSI in both groups (T3-treated group: Δ = -0.12, p = 0.001; untreated group: Δ = -0.04, p = 0.04) and the difference value (discharge/follow-up) was significantly higher in T3-treated group than in untreated group (mean difference between groups = 0.08, 95% confidence interval [CI]: 0.01 to 0.15, p = 0.05). Also, stroke volume increased significantly in the T3-treated group (Δ = 3.4, 95% CI: 0.8 to 6, p <0.01) at follow-up. In conclusion, this is the first pilot experience in which T3 replacement therapy resulted safe and able to improve regional dysfunction in patients with STEMI/LT3S.


Assuntos
Terapia de Reposição Hormonal/métodos , Contração Miocárdica/efeitos dos fármacos , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Tri-Iodotironina/administração & dosagem , Remodelação Ventricular , Adulto , Idoso , Biomarcadores/sangue , Angiografia Coronária , Relação Dose-Resposta a Droga , Ecocardiografia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio com Supradesnível do Segmento ST/fisiopatologia , Hormônios Tireóideos/sangue , Resultado do Tratamento , Tri-Iodotironina/farmacocinética
4.
Thyroid ; 26(11): 1614-1622, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27349131

RESUMO

BACKGROUND: MicroSPECT/CT imaging was used to quantitatively evaluate how iodide uptake in the mouse thyroid is influenced by (i) route of iodine administration; (ii) injection of recombinant human thyrotropin (rhTSH); and (iii) low iodide diet (LID) in euthyroid and triiodothyronine (T3)-treated mice. METHODS: Pertechnetate (99mTcO4-) and 123I thyroid uptake in euthyroid and T3-treated animals fed either a normal-iodine diet (NID) or an LID, treated or not with rhTSH, and radiotracer administered intravenously, subcutaneously, intraperitoneally or by gavage, were assessed using microSPECT/CT imaging. Western blotting was performed to measure sodium/iodide symporter expression levels in the thyroid. RESULTS: Systemic administration of radioiodide resulted in a higher (2.35-fold in NID mice) accumulation of iodide in the thyroid than oral administration. Mice fed LID with systemic radioiodide administration showed a further two-fold increase in thyroid iodide uptake to yield a ∼5-fold increase in uptake compared to the standard NID/oral route. Although rhTSH injections stimulated thyroid activity in both euthyroid and T3-treated mice fed the NID, uptake levels for T3-treated mice remained low compared with those for the euthyroid mice. Combining LID and rhTSH in T3-treated mice resulted in a 2.8-fold higher uptake compared with NID/T3/rhTSH mice and helped restore thyroid activity to levels equivalent to those of euthyroid animals. CONCLUSIONS: Systemic radioiodide administration results in higher thyroidal iodide levels than oral administration, particularly in LID-fed mice. These data highlight the importance of LID, both in euthyroid and T3-treated, rhTSH-injected mice. Extrapolated to human patients, and in the context of clinical guidelines for the preparation of differentiated thyroid cancer patients, our data indicate that LID can potentiate the efficacy of rhTSH treatment in T3-treated patients.


Assuntos
Radioisótopos do Iodo/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Glândula Tireoide/diagnóstico por imagem , Tri-Iodotironina/farmacocinética , Administração Oral , Animais , Dieta/efeitos adversos , Feminino , Terapia de Reposição Hormonal/efeitos adversos , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Injeções Subcutâneas , Iodo/administração & dosagem , Iodo/efeitos adversos , Radioisótopos do Iodo/administração & dosagem , Radioisótopos do Iodo/metabolismo , Camundongos Endogâmicos C57BL , Compostos Radiofarmacêuticos/administração & dosagem , Compostos Radiofarmacêuticos/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacologia , Pertecnetato Tc 99m de Sódio/metabolismo , Pertecnetato Tc 99m de Sódio/farmacocinética , Glândula Tireoide/efeitos dos fármacos , Glândula Tireoide/metabolismo , Tireotropina/administração & dosagem , Tireotropina/efeitos adversos , Tireotropina/farmacologia , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/metabolismo
5.
Biofactors ; 42(6): 638-646, 2016 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-27248050

RESUMO

Prevention of ischemia-reperfusion liver injury is achieved by a combined omega-3 and thyroid hormone (T3 ) protocol, which may involve peroxisome-proliferator activated receptor-α (PPAR-α)-fibroblast growth factor 21 (FGF21) signaling supporting energy requirements. Combined docosahexaenoic acid (DHA; daily doses of 300 mg/kg for 3 days) plus 0.05 mg T3 /kg given to fed rats elicited higher hepatic DHA contents and serum T3 levels, increased PPAR-α mRNA and its DNA binding, with higher mRNA expression of the PPAR-α target genes for carnitine-palmitoyl transferase 1α, acyl-CoA oxidase, and 3-hydroxyl-3-methylglutaryl-CoA synthase 2, effects that were mimicked by 0.1 mg T3 /kg given alone or by the PPAR-α agonist WY-14632. Under these conditions, the mRNA expression of retinoic X receptor-α (RXR-α) is also increased, with concomitant elevation of the hepatic mRNA and protein FGF21 levels and those of serum FGF21. It is concluded that PPAR-α-FGF21 induction by DHA combined with T3 may involve ligand activation of PPAR-α by DHA and enhanced expression of PPAR-α by T3 , with consequent upregulation of the FGF21 that is controlled by PPAR-α. Considering the beneficial effects of PPAR-α-FGF21 signaling on carbohydrate and lipid metabolism, further investigations are required to clarify its potential therapeutic applications in human metabolic disorders. © 2016 BioFactors, 42(6):638-646, 2016.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fígado/metabolismo , PPAR alfa/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Tri-Iodotironina/farmacologia , Animais , Ácidos Docosa-Hexaenoicos/farmacocinética , Ácidos Docosa-Hexaenoicos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Quimioterapia Combinada , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , PPAR alfa/genética , Ratos Sprague-Dawley , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo , Transdução de Sinais , Ativação Transcricional , Tri-Iodotironina/farmacocinética , Tri-Iodotironina/uso terapêutico , Regulação para Cima
6.
Endocrinology ; 157(4): 1694-701, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26910310

RESUMO

Thyroid hormones (THs) are charged and iodinated amino acid derivatives that need to pass the cell membrane facilitated by thyroid hormone transmembrane transporters (THTT) to exert their biological function. The importance of functional THTT is affirmed by the devastating effects of mutations in the human monocarboxylate transporter (MCT) 8, leading to a severe form of psychomotor retardation. Modulation of THTT function by pharmacological or environmental compounds might disturb TH action on a tissue-specific level. Therefore, it is important to identify compounds with relevant environmental exposure and THTT-modulating activity. Based on a nonradioactive TH uptake assay, we performed a screening of 13 chemicals, suspicious for TH receptor interaction, to test their potential effects on THTT in MCT8-overexpressing MDCK1-cells. We identified silymarin, an extract of the milk thistle, to be a potent inhibitor of T3 uptake by MCT8. Because silymarin is a complex mixture of flavonolignan substances, we further tested its individual components and identified silychristin as the most effective one with an IC50 of approximately 100 nM. The measured IC50 value is at least 1 order of magnitude below those of other known THTT inhibitors. This finding was confirmed by T3 uptake in primary murine astrocytes expressing endogenous Mct8 but not in MCT10-overexpressing MDCK1-cells, indicating a remarkable specificity of the inhibitor toward MCT8. Because silymarin is a frequently used adjuvant therapeutic for hepatitis C infection and chronic liver disease, our observations raise questions regarding its safety with respect to unwanted effects on the TH axis.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Silybum marianum/química , Silimarina/farmacologia , Hormônios Tireóideos/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/metabolismo , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Cães , Relação Dose-Resposta a Droga , Células Madin Darby de Rim Canino , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos , Extratos Vegetais/farmacologia , Simportadores , Hormônios Tireóideos/farmacocinética , Tri-Iodotironina/metabolismo , Tri-Iodotironina/farmacocinética
7.
Apoptosis ; 21(2): 184-94, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26659365

RESUMO

Apoptosis is a key process associated with pathological cardiac remodelling in early-phase post-myocardial infarction. In this context, several studies have demonstrated an anti-apoptotic effect of thyroid hormones (TH). The aim of this study was to evaluate the effects of TH on the expression of proteins associated with the apoptotic process 14 days after infarction. Male Wistar rats (300-350 g) (n = 8/group) were divided into four groups: Sham-operated (SHAM), infarcted (AMI), sham-operated + TH (SHAMT) and infarcted + TH (AMIT). For 12 days, the animals received T3 and T4 [2 and 8 µg/(100 g day)] by gavage. After this, the rats were submitted to haemodynamic and echocardiographic analysis, and then were sacrificed and the heart tissue was collected for molecular analysis. Statistical analyses included two-way ANOVA with Student-Newman-Keuls post test. Ethics Committee number: 23262. TH administration prevented the loss of ventricular wall thickness and improved cardiac function in the infarcted rats 14 days after the injury. AMI rats presented an increase in the pro-apoptotic proteins p53 and JNK. The hormonal treatment prevented this increase in AMIT rats. In addition, TH administration decreased the Bax:Bcl-2 ratio in the infarcted rats. TH administration improved cardiac functional parameters, and decreased the expression of pro-apoptotic proteins 14 days after myocardial infarction.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Cardiotônicos/administração & dosagem , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Tiroxina/administração & dosagem , Tri-Iodotironina/administração & dosagem , Animais , Proteínas Reguladoras de Apoptose/genética , Cardiotônicos/farmacocinética , Avaliação Pré-Clínica de Medicamentos , Expressão Gênica , Peroxidação de Lipídeos , Masculino , Infarto do Miocárdio/metabolismo , Miocárdio/patologia , Oxirredução , Estresse Oxidativo , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Tiroxina/farmacocinética , Tri-Iodotironina/farmacocinética , Pressão Ventricular/efeitos dos fármacos
8.
Cell Physiol Biochem ; 35(2): 755-66, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25613909

RESUMO

BACKGROUND: Thyroid hormones (THs) are key regulators of cardiac physiology as well as modulators of different cellular signals including the sphingomyelin/ceramide pathway. The objective of this study was to examine the effect of hyperthyroidism on the metabolism of sphingolipids in the muscle heart. METHODS: Male Wistar rats were treated for 10 days with triiodothyronine (T3) at a dose of 50µg/100g of body weight. Animals were then anaesthetized and samples of the left ventricle were excised. RESULTS: We have demonstrated that prolonged, in vivo, T3 treatment increased the content of sphinganine (SFA), sphingosine (SFO), ceramide (CER) and sphingomyelin (SM), but decreased the level of sphingosine-1-phosphate (S1P) in cardiac muscle. Accordingly, the changes in sphingolipids content were accompanied by a lesser activity of neutral sphingomyelinase and without significant changes in ceramidases activity. Hyperthyroidism also induced activation of AMP-activated protein kinase (AMPK) with subsequently increased expression of mitochondrial proteins: cytochrome c oxidase IV (COX IV), ß-hydroxyacyl-CoA dehydrogenase (ß-HAD), carnityne palmitoyltransferase I (CPT I) and nuclear peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α). CONCLUSIONS: We conclude that prolonged T3 treatment increases sphingolipids metabolism which is reflected by higher concentration of SFA and CER in heart muscle. Furthermore, hyperthyroidism-induced increase in heart sphingomyelin (SM) concentration might be one of the mechanisms underlying maintenance of CER at relatively low level by its conversion to SM together with decreased S1P content.


Assuntos
Ceramidas/metabolismo , Hipertireoidismo/metabolismo , Miocárdio/metabolismo , Esfingomielinas/metabolismo , Tri-Iodotironina/administração & dosagem , Animais , Ceramidases/metabolismo , Modelos Animais de Doenças , Hipertireoidismo/induzido quimicamente , Hipertireoidismo/enzimologia , Lisofosfolipídeos/metabolismo , Masculino , Ratos , Ratos Wistar , Esfingomielina Fosfodiesterase/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Tri-Iodotironina/farmacocinética
9.
Ther Drug Monit ; 37(1): 110-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24977379

RESUMO

BACKGROUND: As changes in thyroid stimulating hormone (TSH), thyroid hormones, and vital signs after administration of a single dose of liothyronine have typically only been documented for 24 hours, we documented these parameters more than 96 hours. METHODS: Blood samples were obtained for 4 days after administration of 50-mcg liothyronine to 12 healthy euthyroid participants. Concentrations of total and free triiodothyronine, free and total thyroxine, and TSH were measured. Vital signs were documented. RESULTS: Triiodothyronine concentrations peaked at 2.5 hours after liothyronine administration. Heart rate (HR) increased by 5 hours after liothyronine administration, subsequently reaching a value higher than baseline (P = 0.009). Suppression of TSH concentrations began at 2 hours. The nadir TSH value at 12 hours was significantly different from baseline (P < 0.001) and remained lower than the baseline value for 2-3 days. CONCLUSIONS: A single dose of liothyronine has both short-term and long-term effects. There is clearly a different lag time between the serum concentrations of triiodothyronine and its effects on the heart and pituitary, respectively. The increase in serum triiodothyronine concentration occurred within hours and was then followed by an increase in HR. The increased HR was transient and was followed by a reduction in TSH concentration. The suppression of TSH was delayed but was more sustained. Thus, sustained TSH reduction beyond 24 hours was achieved by a single dose of liothyronine that produced only brief increases in serum triiodothyronine levels and transient increases in HR.


Assuntos
Tri-Iodotironina/farmacologia , Tri-Iodotironina/farmacocinética , Adolescente , Adulto , Temperatura Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Feminino , Meia-Vida , Frequência Cardíaca/efeitos dos fármacos , Humanos , Masculino , Tireotropina/sangue , Tiroxina/sangue , Adulto Jovem
10.
Int J Pharm Compd ; 16(5): 376-80, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23072197

RESUMO

Effective thyroid replacement therapy may be elusive to some patients, and compounding pharmacists have an opportunity to deliver more effective therapy. Goodman & Gilman's The Pharmacological Basis of Therapeutics 12th edition states that the body usually secretes T4:T3 in an 11:1 ratio but cautions against pursuing combined thyroid replacement due to the short halflife of T3 that necessitates multiple daily dosing; no commercial availability and lack of benefit were shown in trials. Commercial combinations of T4/T3 such as Armour Thyroid and Nature-Throid have a 4.22:1 T4:T3 ratio. Applying the same concept as bioidentical hormone replacement therapy, compounding pharmacists can deliver an 11:1 ratio using a commercial T4 product and taking into account oral bioavailability of each entity. The short half-life of T3 can be remedied by taking the patient's daily T3 dose and dividing it into two slow-release capsules to be dosed every 12 hours.


Assuntos
Terapia de Reposição Hormonal , Hipotireoidismo/tratamento farmacológico , Tiroxina/administração & dosagem , Tri-Iodotironina/administração & dosagem , Disponibilidade Biológica , Humanos , Hipotireoidismo/sangue , Tireotropina/sangue , Tiroxina/farmacocinética , Resultado do Tratamento , Tri-Iodotironina/farmacocinética
11.
Thyroid ; 22(10): 1069-75, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22947347

RESUMO

BACKGROUND: The effectiveness of short-term 3,5,3'-triiodothyroacetic acid (TRIAC) therapy for the treatment of hyperthyroidism caused by thyroid hormone resistance (RTH) has been documented. Here, we report a 3-year course of TRIAC therapy in an RTH boy, with a quantitative evaluation of the therapeutic effects and pharmacological study of TRIAC. PATIENT FINDINGS: The gene encoding the thyroid hormone receptor beta (THRB) of the patient carries a P453T mutation. During treatment with up to 3.0 mg TRIAC per day, reduction in the thyroid volume, resolution of supraventricular arrhythmia, and decrease in thyroid-stimulating hormone (TSH) and free-thyroxine (FT4) levels were achieved. In addition, attention-deficit hyperactivity disorder (ADHD) symptoms improved, with a concomitant decline in the ADHD Rating Scale score. SUMMARY: A TRIAC pharmacokinetic study, conducted using triiodothyronine level as a surrogate for TRIAC level, demonstrated that TRIAC disappears from the circulation rapidly and has a shorter duration of TSH secretion inhibitory effect in the RTH patient compared to that in the control subject. Studies of TSH and FT4 levels over a period of 3 years indicated that the TRIAC effect is dose dependent. CONCLUSIONS: TRIAC was effective and safe in ameliorating the effects of hyperthyroidism and ADHD symptoms in a child with known genetic RTH. Further, it was demonstrated that TRIAC has a short half-life and functions dose dependently.


Assuntos
Hipertireoidismo/tratamento farmacológico , Síndrome da Resistência aos Hormônios Tireóideos/tratamento farmacológico , Tri-Iodotironina/análogos & derivados , Adolescente , Adulto , Complexos Atriais Prematuros/tratamento farmacológico , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Criança , Pré-Escolar , Feminino , Meia-Vida , Humanos , Masculino , Receptores beta dos Hormônios Tireóideos/genética , Tireotropina/sangue , Tiroxina/sangue , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/sangue , Tri-Iodotironina/farmacocinética , Tri-Iodotironina/uso terapêutico
12.
J. physiol. biochem ; 68(3): 345-351, sept. 2012.
Artigo em Inglês | IBECS | ID: ibc-122323

RESUMO

Thyroid diseases are one of the most common metabolic disorders in the human population. In this work, we present data concerning changes in the activity and kinetic parameters of several enzymes associated with both anabolic (glucose-6-phosphate dehydrogenase-G6PDH, EC 1.1.1.49; 6-phosphogluconate dehydrogenase-6PGDH, EC 1.1.1.44; malic enzyme-ME, EC 1.1.1.40; and isocitrate dehydrogenase-IDH, EC 1.1.1.42) and catabolic (NAD-dependent malate dehydrogenase-NAD-MDH, EC 1.1.1.37; and lactate dehydrogenase-LDH, EC 1.1.1.27) processes under conditions of hypothyroidism and T3 treatment. Hypothyroidism was induced in rats by the surgical removal of the thyroid gland. T3-treated rats were injected by T3 (0.5 mg T3/kg body weight daily during 10 days). We have found that T3 treatment caused an increase of NAD-MDH activity as well as heart hypertrophy whereas the activity of LDH increased in the direction of pyruvate reduction. Moreover, we observed increased activity of both enzymes in the liver. These results confirm earlier observation concerning the relevance of oxidative metabolism in the heart under T3 treatment. Hypothyroidism resulted in changes in the LDH activity in the heart whereas NAD-MDH activity did not change. Moreover, our data show that T3 treatment caused an increase of G6PDH, 6PGDH, and ME activities in the liver. We also observed a decrease of IDH activity in both organs, whereas hypothyroidism caused the opposite effect. This data indicate that either deficiency or excess of thyroid hormones can prove to be particularly dangerous for the physiology of the heart muscle by disturbing bioenergetic and anabolic processes (AU)


Assuntos
Animais , Ratos , Hipotireoidismo/tratamento farmacológico , Tri-Iodotironina/farmacocinética , Fígado/enzimologia , Coração/fisiopatologia , Miocárdio/enzimologia
13.
J Clin Endocrinol Metab ; 97(1): E100-5, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22031512

RESUMO

CONTEXT: Tyrosine kinase inhibitors (TKI) are used for the treatment of various cancers. Case reports and clinical trials have reported abnormal thyroid function tests (TFT) after treatment with sunitinib, imatinib, sorafenib, dasatinib, and nilotinib. An increased requirement for levothyroxine was reported in thyroidectomized patients during TKI treatment. OBJECTIVE: We hypothesized that abnormal TFT are compatible with inhibition of thyroid hormone (TH) transporters and subsequently reduced pituitary-TH feedback. Monocarboxylate transporter 8 (MCT8) is a TH transmembrane transporter in brain, pituitary, and other organs. MCT8 mutation leads to abnormal TFT in patients and respective mouse models. We tested whether TKI are able to inhibit MCT8-mediated TH uptake into cells. DESIGN: Madin-Darby-canine kidney (MDCK1) cells stably expressing human MCT8 were exposed in vitro to TKI at increasing concentrations, and MCT8-mediated [(125)I]T(3) uptake and efflux were measured. The mode of inhibition was determined. RESULTS: TKI exposure dose-dependently inhibited MCT8-dependent T(3) and T(4) uptake. IC(50) values for sunitinib, imatinib, dasatinib, and bosutinib ranged from 13-38 µm, i.e. similar to the Michaelis-Menten constant K(m) for T(3) and T(4), 4 and 8 µm, respectively. Kinetic experiments revealed a noncompetitive mode of inhibition for all TKI tested. CONCLUSIONS: Partial inhibition by TKI of pituitary or hypothalamic TH feedback may increase TSH or increase the levothyroxine requirement of thyroidectomized patients. It is still possible that other mechanisms contribute to TKI-mediated impairments of TFT, e.g. altered metabolism of TH. Bosutinib was not previously reported to alter TFT.


Assuntos
Transportadores de Ácidos Monocarboxílicos/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Tri-Iodotironina/metabolismo , Animais , Benzamidas , Ligação Competitiva , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Ensaios Clínicos como Assunto , Cães , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Mesilato de Imatinib , Indóis/farmacologia , Radioisótopos do Iodo/farmacocinética , Radioisótopos do Iodo/urina , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Piperazinas/farmacologia , Proteínas Tirosina Quinases/metabolismo , Pirimidinas/farmacologia , Pirróis/farmacologia , Sunitinibe , Simportadores , Tiroxina/metabolismo , Tiroxina/farmacocinética , Tiroxina/urina , Transfecção , Tri-Iodotironina/farmacocinética , Tri-Iodotironina/urina
14.
Food Chem Toxicol ; 49(9): 2068-73, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21624425

RESUMO

Maternal nicotine exposure leads to neonatal hypothyroidism that can be returned to euthyroidism after nicotine withdrawal. Here, we examined the transfer of iodine through milk, deiodinase activities (D1 and D2), and serum T3, T4 and TSH in rat offspring after maternal exposure to nicotine. One day after birth, a minipump was implanted to dams releasing nicotine (NIC), 6 mg/kg/day for 13 days or vehicle saline. Animals were killed at the day 15 and 21 of lactation. At day 15, NIC-treated dams showed decreased T4 and mammary 2h-radioiodine uptake (RAIU) and increase of TSH, thyroid 2h-RAIU, liver D1 and mammary D2. At the cessation of NIC-exposure, pups displayed decreased T3, T4 and thyroid 2h-RAIU and increased TSH. At weaning (21-postnatal day), NIC-treated dams recovered their T4 and TSH, but increased deiodinase level in the liver and mammary gland. Milk T3 content in NIC-treated dams was higher at both day 15 and 21, and thyroid function was recovered at the day 21. Thus, thyroid function was affected by nicotine in both mothers and pups, suggesting a primary hypothyroidism. After nicotine withdrawal, pups recovered thyroid function probably due to the increased lactational transfer of T3 in relation with increased mammary gland deiodinase activities.


Assuntos
Animais Recém-Nascidos , Hipotireoidismo/induzido quimicamente , Exposição Materna , Leite , Nicotina/efeitos adversos , Tri-Iodotironina/farmacocinética , Animais , Feminino , Lactação , Nicotina/administração & dosagem , Ratos , Ratos Wistar , Síndrome de Abstinência a Substâncias , Tireotropina/sangue
15.
Endocrinol. nutr. (Ed. impr.) ; 58(4): 185-196, abr. 2011. ilus
Artigo em Espanhol | IBECS | ID: ibc-94156

RESUMO

La resistencia a hormonas tiroideas es un grupo de sindromes de causa genetica caracterizados por la disminucion de la sensibilidad tisular a estas hormonas. En la actualidad se distinguen tres formas, en los que la resistencia a la accion hormonal se debe, respectivamente, a mutaciones del gen que codifica el receptor nuclear de T3 TRbeta, a alteraciones en el transporte celular de T4 y T3, y a defectos en la conversion de T4 en T3 mediada por desyodasas. En esta revision se hace una exposicion resumida y actualizada de cada una de estas tres formas de resistencia y se discuten los mecanismos patogonicos y aproximaciones clinicas (AU)


Thyroid hormone resistance syndromes are a group of genetic conditions characterized by decreased tissue sensitivity to thyroid hormones. Three syndromes, in which resistance to hormone action is respectively due to mutations in the gene encoding for thyroid hormone receptor TR¦Â, impaired T4 and T3 transport, and impaired conversion of T4 to T3 mediated by deiodinases. An updated review of each of these forms of resistance is provided, and their pathogenetic mechanisms and clinical approaches are discussed (AU)


Assuntos
Humanos , /fisiopatologia , Hipotireoidismo/fisiopatologia , Tri-Iodotironina/farmacocinética , Mutação , Tiroxina/farmacocinética , Proteínas de Membrana Transportadoras/farmacocinética , Receptores Citoplasmáticos e Nucleares/análise
16.
Mol Cell Endocrinol ; 337(1-2): 96-100, 2011 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-21315799

RESUMO

Thyroid hormone (TH) transporter proteins mediate transport of TH across the plasma membrane, thereby facilitating its intracellular bioavailability. As only a few transporters have been identified which are relatively specific for TH, including monocarboxylate transporter (MCT) 8 and MCT10, the need for identification of novel specific TH transporters is obvious. A possible strategy to identify TH transporters is their modification with a ligand-derived affinity-label and subsequent identification by mass spectrometry. Previously, N-bromoacetyl (BrAc)-iodothyronines have been reported as useful affinity-labels for human (h) MCT8. In the present study we reinvestigated possible BrAc[(125)I]T3-labeling of hMCT8 and hMCT10. The present study demonstrates that hMCT8 and hMCT10 both facilitate BrAc[(125)I]T3 transport, but are not labeled by BrAc[(125)I]T3. We provide evidence that human protein disulfide isomerase, which molecular mass is similar to hMCT8, is labeled by BrAc[(125)I]T3. In addition, differential inhibitory effects were observed of iodothyronines derivatives with different side chains on T3 transport by hMCT8 and hMCT10. In conclusion, we demonstrated that not hMCT8 and hMCT10, but human protein disulfide isomerase, is labeled by BrAc[(125)I]T3. The usefulness of BrAc[(125)I]T3 as a tool for the identification of novel TH transporters remains to be explored.


Assuntos
Marcadores de Afinidade/farmacologia , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Recombinantes/metabolismo , Tri-Iodotironina/análogos & derivados , Marcadores de Afinidade/farmacocinética , Animais , Células COS , Chlorocebus aethiops , Humanos , Iodeto Peroxidase/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Ratos , Simportadores , Tri-Iodotironina/farmacocinética , Tri-Iodotironina/farmacologia
17.
Ann Rheum Dis ; 70(1): 164-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20724312

RESUMO

OBJECTIVE: To study whether common genetic variants of the genes involved in the complex regulatory mechanism determining the intracellular bio-availability of T3 influence osteoarthritis onset. METHODS: In total 17 genetic variants within the genes encoding WD40-repeat/SOCS-box protein 1, ubiquitin specific protease 33, thyroid hormone receptor α, deiodinase, iodothyronine, type III (DIO3) and Indian hedgehog were measured and associated with osteoarthritis in a meta-analyses in European populations from the UK, The Netherlands, Greece and Spain containing a total of 3252 osteoarthritis cases and 2132 controls. RESULTS: The minor allele of the DIO3 variant rs945006 showed suggestive evidence for protective association in the overall meta-analyses, which was supported by individual osteoarthritis studies and osteoarthritis subtypes. The association appeared most significant in cases with knee and/or hip with an allelic OR of 0.81 (95% CI 0.70 to 0.930) with a nominal p value of 0.004 and a permutation-based corrected p value for multiple testing of 0.039. CONCLUSION: The findings suggest that the DIO3 gene modulates osteoarthritis disease risk; however, additional studies are necessary to replicate our findings. To elucidate the molecular mechanisms focus should be on the local adaptation to T3 availability either during the endochondral ossification process or during ageing of the articular cartilage.


Assuntos
Iodeto Peroxidase/genética , Osteoartrite/genética , Tri-Iodotironina/farmacocinética , Idoso , Disponibilidade Biológica , Feminino , Frequência do Gene , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Osteoartrite/metabolismo , Polimorfismo de Nucleotídeo Único
18.
J Clin Endocrinol Metab ; 95(10): 4762-70, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660035

RESUMO

CONTEXT: Abnormal placentation in human pregnancy is associated with intrauterine fetal growth restriction (IUGR). Our group has previously reported the association between severe IUGR, lower fetal circulating concentrations of thyroid hormones (THs), and altered expression of TH receptors and TH transporters within human placental villi. We postulate that altered TH bioavailability to trophoblasts may contribute to the pathogenesis of IUGR. DESIGN AND OBJECTIVE: Cytotrophoblasts were isolated from normal and IUGR human placentae to compare their responsiveness to T(3) and their capability for T(3) transport. RESULTS: Compared with normal cytotrophoblasts, the viability of IUGR cytotrophoblasts (assessed by methyltetrazoleum assay) was significantly reduced (P < 0.001), whereas apoptosis (assessed using caspase 3/7 activity and M30 immunoreactivity) was significantly increased after T(3) treatment for 48 h (P < 0.001 and P < 0.01, respectively). The secretion of human chorionic gonadotropin was significantly increased by IUGR cytotrophoblasts compared with normal cytotrophoblasts (P < 0.001), independently of T(3) treatment. Net transport of [(125)I]T(3) was 20% higher by IUGR cytotrophoblasts compared with normal cytotrophoblasts (P < 0.001), and this was accompanied by a 2-fold increase in the protein expression of the TH transporter, monocarboxylate transporter 8, as assessed by Western immunoblotting (P < 0.01). CONCLUSIONS: IUGR cytotrophoblasts demonstrate altered responsiveness to T(3) with significant effects on cell survival and apoptosis compared with normal cytotrophoblasts. Increased monocarboxylate transporter 8 expression and intracellular T(3) accumulation may contribute to the altered T(3) responsiveness of IUGR cytotrophoblasts.


Assuntos
Retardo do Crescimento Fetal/patologia , Tri-Iodotironina/metabolismo , Tri-Iodotironina/farmacologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Adulto , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Apoptose/efeitos dos fármacos , Transporte Biológico/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Gonadotropina Coriônica/metabolismo , Feminino , Retardo do Crescimento Fetal/metabolismo , Humanos , Masculino , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Placenta/metabolismo , Placenta/patologia , Gravidez , Simportadores , Tri-Iodotironina/farmacocinética , Trofoblastos/patologia , Adulto Jovem
19.
Clin Endocrinol (Oxf) ; 72(5): 709-15, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20447070

RESUMO

CONTEXT: The substitution of liothyronine (L-T3) for levothyroxine (L-T4) is commonly employed during thyroid hormone (TH) withdrawal in preparation for diagnostic and therapeutic interventions on thyroid cancer patients. Presently, only limited data are available on the L-T3 for L-T4 therapeutic substitution. Objective To characterize the pharmcodynamic equivalence of L-T3 and L-T4. DESIGN: Randomized, double-blind, cross-over intervention study. SETTING: NIH clinical center. PATIENTS: Ten thyroidectomized patients. INTERVENTIONS: Study participants were treated with L-T3 or L-T4 with a target TSH >or= 0.5

Assuntos
Terapia de Reposição Hormonal , Hipotireoidismo/tratamento farmacológico , Tiroxina/uso terapêutico , Tri-Iodotironina/uso terapêutico , Área Sob a Curva , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Hipotireoidismo/sangue , Hipotireoidismo/cirurgia , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Equivalência Terapêutica , Testes de Função Tireóidea , Tireoidectomia , Tireotropina/sangue , Tiroxina/farmacocinética , Resultado do Tratamento , Tri-Iodotironina/farmacocinética
20.
Reprod Toxicol ; 27(3-4): 387-399, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19429409

RESUMO

Perfluorooctanesulfonate (PFOS), a persistent and accumulative compound, is widely distributed in humans and wildlife. Human exposure can occur early in development, as evidenced by the detection of PFOS in umbilical cord blood and breast milk. As part of a developmental neurotoxicology study for which developmental endpoints, including those related to the developing nervous system, have been reported separately, groups of 25 pregnant Sprague Dawley rats were given daily oral doses of either vehicle control or potassium PFOS (K(+)PFOS) at 0.1, 0.3, and 1.0mg/kg-d from gestation day (GD) 0 (day positive for mating) through postnatal day (PND) 20. An additional 10 pregnant females per treatment group were treated through GD 19 and sacrificed on GD 20 in order to obtain maternal and fetal serum and tissue samples at the end of gestation. The present paper reports the results of samples of serum, liver, brain, and thyroid glands taken at various times to evaluate: (1) serum, liver, and brain PFOS concentrations by LC-MS/MS to establish the relationship between PFOS concentrations and study outcomes; (2) serum thyrotropin (TSH) concentrations by RIA; (3) thyroid follicular cell proliferation index by Ki-67 immunohistochemical staining; (4) thyroid follicle epithelial cell height and colloidal area by histomorphometric analysis; (5) selected liver mRNA transcripts by quantitative RT-PCR. PFOS concentrations in dam and pup serum, liver, and brain increased across treatment groups in approximate proportion to the proportional increases in maternal K(+)PFOS dose, and sex differences in PFOS concentrations were not apparent in pups on PND 21. In pups from K(+)PFOS maternal dose groups on PND 72, serum PFOS had decreased to about 3 and 11% of PND 21 concentrations in males and females, respectively, and liver PFOS had decreased to about 17% of PND 21 concentrations in both sexes. Liver PFOS concentrations were approximately 0.6-0.8 times serum PFOS in GD 20 fetuses, and increased to about 2-4 times serum concentrations on PND 4 and 21. GD 20 fetal and PND 4 pup brain PFOS concentrations were approximately 33% of the corresponding serum concentrations, dropping to approximately 10% by PND 21, in contrast to dam brain PFOS concentrations, which were approximately 4-9% of serum PFOS concentrations. Compared to controls, Cyp2b2 mRNA was increased (2.8-fold) in the 1.0mg/kg-d treatment-group dams on GD 20. In male pups on PND 21, Cyp4A1, ACoA, and Cyp2b2 were increased 2.1-, 1.5-, and 1.8-fold, respectively, and Cyp7A1 was decreased 3.5-fold. Serum TSH and thyroid follicular morphology were not altered by K(+)PFOS treatment. The mean number of proliferating thyroid follicular cells was increased 2.1-fold over control in GD 20 female fetuses from 1.0mg/kg-d-treated dams, yet the highest individual count was similar to that of controls (116 versus 113 in controls).


Assuntos
Ácidos Alcanossulfônicos/farmacocinética , Poluentes Ambientais/farmacocinética , Fluorocarbonos/farmacocinética , Expressão Gênica/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Hormônios Tireóideos/farmacocinética , Ácidos Alcanossulfônicos/administração & dosagem , Ácidos Alcanossulfônicos/sangue , Ácidos Alcanossulfônicos/farmacologia , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Poluentes Ambientais/administração & dosagem , Poluentes Ambientais/sangue , Poluentes Ambientais/farmacologia , Feminino , Fluorocarbonos/administração & dosagem , Fluorocarbonos/sangue , Fluorocarbonos/farmacologia , Idade Gestacional , Lactação , Masculino , Exposição Materna , Gravidez , Ratos , Ratos Sprague-Dawley , Hormônios Tireóideos/sangue , Tiroxina/sangue , Tiroxina/farmacocinética , Tri-Iodotironina/sangue , Tri-Iodotironina/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...