Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 376(1): 74-83, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33127750

RESUMO

Acute respiratory distress syndrome (ARDS) is a severe, life-threatening form of respiratory failure characterized by pulmonary edema, inflammation, and hypoxemia due to reduced alveolar fluid clearance (AFC). Alveolar fluid clearance is required for recovery and effective gas exchange, and higher rates of AFC are associated with reduced mortality. Thyroid hormones play multiple roles in lung function, and L-3,5,3'-triiodothyronine (T3) has multiple effects on lung alveolar type II cells. T3 enhances AFC in normal adult rat lungs when administered intramuscularly and in normal or hypoxia-injured lungs when given intratracheally. The safety of a commercially available formulation of liothyronine sodium (synthetic T3) administered intratracheally was assessed in an Investigational New Drug Application-enabling toxicology study in healthy rats. Instillation of the commercial formulation of T3 without modification rapidly caused tracheal injury and often mortality. Intratracheal instillation of T3 that was reformulated and brought to a neutral pH at the maximum feasible dose of 2.73 µg T3 in 300 µl for 5 consecutive days had no clinically relevant T3-related adverse clinical, histopathologic, or clinical pathology findings. There were no unscheduled deaths that could be attributed to the reformulated T3 or control articles, no differences in the lung weights, and no macroscopic or microscopic findings considered to be related to treatment with T3. This preclinical safety study has paved the way for a phase I/II study to determine the safety and tolerability of a T3 formulation delivered into the lungs of patients with ARDS, including coronavirus disease 2019-associated ARDS, and to measure the effect on extravascular lung water in these patients. SIGNIFICANCE STATEMENT: There is growing interest in treating lung disease with thyroid hormone [triiodothyronine (T3)] in pulmonary edema and acute respiratory distress syndrome (ARDS). However, there is not any published experience on the impact of direct administration of T3 into the lung. An essential step is to determine the safety of multiple doses of T3 administered in a relevant animal species. This study enabled Food and Drug Administration approval of a phase I/II clinical trial of T3 instillation in patients with ARDS, including coronavirus disease 2019-associated ARDS (T3-ARDS ClinicalTrials.gov Identifier NCT04115514).


Assuntos
Instilação de Medicamentos , Pulmão/efeitos dos fármacos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Tri-Iodotironina/efeitos adversos , Animais , Avaliação Pré-Clínica de Medicamentos , Feminino , Intubação Intratraqueal/efeitos adversos , Intubação Intratraqueal/métodos , Masculino , Ratos , Ratos Sprague-Dawley , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/uso terapêutico
2.
J Clin Endocrinol Metab ; 105(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32614450

RESUMO

CONTEXT: It is well recognized that some hypothyroid patients on levothyroxine (LT4) remain symptomatic, but why patients are susceptible to this condition, why symptoms persist, and what is the role of combination therapy with LT4 and liothyronine (LT3), are questions that remain unclear. Here we explore evidence of abnormal thyroid hormone (TH) metabolism in LT4-treated patients, and offer a rationale for why some patients perceive LT4 therapy as a failure. EVIDENCE ACQUISITION: This review is based on a collection of primary and review literature gathered from a PubMed search of "hypothyroidism," "levothyroxine," "liothyronine," and "desiccated thyroid extract," among other keywords. PubMed searches were supplemented by Google Scholar and the authors' prior knowledge of the subject. EVIDENCE SYNTHESIS: In most LT4-treated patients, normalization of serum thyrotropin levels results in decreased serum T3/T4 ratio, with relatively lower serum T3 levels; in at least 15% of the cases, serum T3 levels are below normal. These changes can lead to a reduction in TH action, which would explain the slower rate of metabolism and elevated serum cholesterol levels. A small percentage of patients might also experience persistent symptoms of hypothyroidism, with impaired cognition and tiredness. We propose that such patients carry a key clinical factor, for example, specific genetic and/or immunologic makeup, that is well compensated while the thyroid function is normal but might become apparent when compounded with relatively lower serum T3 levels. CONCLUSIONS: After excluding other explanations, physicians should openly discuss and consider therapy with LT4 and LT3 with those hypothyroid patients who have persistent symptoms or metabolic abnormalities despite normalization of serum thyrotropin level. New clinical trials focused on symptomatic patients, genetic makeup, and comorbidities, with the statistical power to identify differences between monotherapy and combination therapy, are needed.


Assuntos
Terapia de Reposição Hormonal , Hipotireoidismo/tratamento farmacológico , Medicina de Precisão , Tiroxina/administração & dosagem , Resistência a Medicamentos/efeitos dos fármacos , Quimioterapia Combinada , Terapia de Reposição Hormonal/métodos , Terapia de Reposição Hormonal/normas , Humanos , Hipotireoidismo/sangue , Hipotireoidismo/epidemiologia , Padrões de Prática Médica/normas , Padrões de Prática Médica/estatística & dados numéricos , Padrões de Prática Médica/tendências , Medicina de Precisão/métodos , Medicina de Precisão/tendências , Testes de Função Tireóidea , Tireotropina/sangue , Falha de Tratamento , Tri-Iodotironina/administração & dosagem
3.
Proc Natl Acad Sci U S A ; 116(26): 13116-13121, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31189592

RESUMO

Synthesis of triiodothyronine (T3) in the hypothalamus induces marked seasonal neuromorphology changes across taxa. How species-specific responses to T3 signaling in the CNS drive annual changes in body weight and energy balance remains uncharacterized. These experiments sequenced and annotated the Siberian hamster (Phodopus sungorus) genome, a model organism for seasonal physiology research, to facilitate the dissection of T3-dependent molecular mechanisms that govern predictable, robust, and long-term changes in body weight. Examination of the Phodopus genome, in combination with transcriptome sequencing of the hamster diencephalon under winter and summer conditions, and in vivo-targeted expression analyses confirmed that proopiomelanocortin (pomc) is a primary genomic target for the long-term T3-dependent regulation of body weight. Further in silico analyses of pomc promoter sequences revealed that thyroid hormone receptor 1ß-binding motif insertions have evolved in several genera of the Cricetidae family of rodents. Finally, experimental manipulation of food availability confirmed that hypothalamic pomc mRNA expression is dependent on longer-term photoperiod cues and is unresponsive to acute, short-term food availability. These observations suggest that species-specific responses to hypothalamic T3, driven in part by the receptor-binding motif insertions in some cricetid genomes, contribute critically to the long-term regulation of energy balance and the underlying physiological and behavioral adaptations associated with the seasonal organization of behavior.


Assuntos
Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Phodopus/fisiologia , Fotoperíodo , Pró-Opiomelanocortina/metabolismo , Aclimatação/fisiologia , Animais , Peso Corporal/fisiologia , Temperatura Baixa/efeitos adversos , Biologia Computacional , Regulação para Baixo , Ingestão de Alimentos/fisiologia , Evolução Molecular , Feminino , Privação de Alimentos/fisiologia , Perfilação da Expressão Gênica , Masculino , Anotação de Sequência Molecular , Neuropeptídeos/metabolismo , Pró-Opiomelanocortina/genética , Regiões Promotoras Genéticas/genética , Domínios e Motivos de Interação entre Proteínas/genética , Receptores dos Hormônios Tireóideos/metabolismo , Estações do Ano , Especificidade da Espécie , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/metabolismo , Aumento de Peso/efeitos dos fármacos , Aumento de Peso/fisiologia , Sequenciamento Completo do Genoma
4.
Artigo em Inglês | MEDLINE | ID: mdl-30553402

RESUMO

Supplementation with omega-3 fatty acids or thyroid hormone (T3) exhibit negative effects on inflammatory reactions in experimental animals. The aim of this work was to assess the hypothesis that docosahexaenoic acid (DHA) plus T3 co-administration enhances liver resolvin (Rv) levels as inflammation resolution mediators. Combined DHA (daily doses of 300 mg/kg for 3 consecutive days)-T3 (0.05 mg/kg at the fourth day) administration significantly increased the content of hepatic RvD1 and RvD2, without changes in that of RvE1 and RvE2, an effect that exhibits synergy when compared to the separate DHA and T3 treatments. Under these conditions, liver DHA levels increased by DHA administration were diminished when combined with T3 (p < 0.05), suggesting enhancement in resolvin D biosynthesis in extrahepatic tissues. It is concluded that co-administration of DHA and T3 rises the capacity of the liver for inflammation resolution by augmenting RvD1(2) availability, which represents an important protocol in hepatoprotection in the clinical setting.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Substâncias Protetoras/farmacologia , Tri-Iodotironina/farmacologia , Animais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácidos Docosa-Hexaenoicos/metabolismo , Sinergismo Farmacológico , Quimioterapia Combinada , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/metabolismo , Hepatite Animal/tratamento farmacológico , Masculino , Substâncias Protetoras/administração & dosagem , Ratos , Ratos Sprague-Dawley , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/efeitos adversos
5.
Food Funct ; 8(11): 3980-3988, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28990606

RESUMO

Liver preconditioning by a docosahexaenoic acid (DHA) and triiodothyronine (T3) combined protocol underlies peroxisome-proliferator activated receptor α (PPARα)-fibroblast growth factor 21 (FGF21) upregulation, the study of the regulatory mechanisms involved being the aim of this work. Combined DHA (daily doses of 300 mg kg-1 for 3 days)-T3 (0.05 mg kg-1 at the fourth day) administration elicited higher levels of liver DHA and serum T3, with enhanced hepatic nuclear/cytosolic PPARα ratios, upregulation of FGF21 and ß-Klotho expression, and a small reduction in that of FGF receptor 1 (FGFR1), compared with the respective controls. Concomitantly, the components of the FGF21 cascade extracellular-signal-regulated kinase 1/2 (ERK1/2), FGF receptor substrate 2α (FRS2α), cFos, ribosomal S6 kinase 1 (RSK1), liver kinase B1 (LKB1), and AMP-activated protein kinase (AMPK) were activated. The upregulation of liver PPARα-FGF21-AMPK signaling by the combined DHA-T3 protocol resulted in values significantly higher than those elicited by the addition of the data obtained for DHA and T3 alone. It is concluded that combined DHA-T3 supplementation achieves synergistic effects on liver PPARα-FGF21-AMPK signaling, which may result in significant metabolic changes associated with energy expenditure that are of importance in the treatment of obesity and other metabolic disorders.


Assuntos
Ácidos Docosa-Hexaenoicos/administração & dosagem , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Fígado/metabolismo , Doenças Metabólicas/tratamento farmacológico , Tri-Iodotironina/administração & dosagem , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Metabolismo Energético/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/genética , Glucuronidase/genética , Humanos , Proteínas Klotho , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/fisiopatologia , PPAR alfa/genética , PPAR alfa/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
6.
BMJ Case Rep ; 20162016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27908914

RESUMO

We describe the case of a young man with repeated hospital presentations for a variety of symptoms related to excessive bodybuilding and associated behaviours. He presented to our department (radiology) with right arm pain and loss of function. Ultrasound showed complete triceps rupture, rare in young patients and multiple cystic areas within the muscles of the arm. MRI revealed these to be multiple proteinaceous lesions within the muscle bellies and the possibility of self-innoculation was raised by the reporting radiologist. The patient subsequently admitted to injecting coconut oil to improve muscle contour lost secondary to injury. A review of his hospital presentations was then made and revealed further concerning practices performed by the patient to enhance his muscular appearance.


Assuntos
Braço/patologia , Imagem Corporal/psicologia , Comportamento Perigoso , Músculo Esquelético/patologia , Óleos de Plantas/administração & dosagem , Óleos de Plantas/efeitos adversos , Levantamento de Peso/psicologia , Adulto , Celulite (Flegmão)/induzido quimicamente , Óleo de Coco , Aconselhamento Diretivo , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/efeitos adversos , Injeções Intramusculares/efeitos adversos , Insulina/administração & dosagem , Insulina/efeitos adversos , Imageamento por Ressonância Magnética , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Atrofia Muscular/induzido quimicamente , Atrofia Muscular/patologia , Congêneres da Testosterona/administração & dosagem , Congêneres da Testosterona/efeitos adversos , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/efeitos adversos
7.
PLoS One ; 11(11): e0166152, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27832141

RESUMO

Zebrafish embryos are treated with anti-thyroidal compounds, such as phenylthiourea, to inhibit melanogenesis. However, the mechanism whereby the thyroidal system controls melanin synthesis has not been assessed in detail. In this work, we tested the effect of the administration of diets supplemented with T3 (500µg/g food) on the pigment pattern of adult zebrafish. Oral T3 induced a pronounced skin paling in both adult female and male zebrafish that was reversible upon cessation of treatment. The number of visible melanophores was significantly reduced in treated fish. Accordingly, treatment down-regulated expression of tyrosinase-related protein 1 in both sexes. We also found sexually dimorphic regulation of some melanogenic genes, such as Dct/Tyrp2 that was dramatically up-regulated in females after T3 treatment. Thus, we demonstrated that melanogenesis is reversibly inhibited by thyroid hormones in adult zebrafish and make the discovery of gender-specific differences in the response of melanogenic gene expression. Thus, fish gender is now shown to be an important variable that should be controlled in future studies of fish melanogenesis.


Assuntos
Melaninas/metabolismo , Melanóforos/efeitos dos fármacos , Tri-Iodotironina/farmacologia , Peixe-Zebra/metabolismo , Animais , Suplementos Nutricionais , Feminino , Expressão Gênica/efeitos dos fármacos , Masculino , Melanóforos/metabolismo , Oxirredutases/genética , Pigmentação/efeitos dos fármacos , Pigmentação/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/sangue , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
Endocr J ; 63(6): 563-7, 2016 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-27000530

RESUMO

In the last ten years a liquid formulation of liothyronine (L-T3) became available. To date, no studies on its systematic use have been reported. This study is aimed at assessing the reliability of liquid L-T3 in achieving target TSH in patients with differentiated thyroid cancers (DTC). Twenty-one high risk DTC patients in whom levothyroxine treatment up to 2.0 µg/kg/day did not suppress TSH levels (i.e. >0.1 mIU/L) were selected. Maintaining the same L-T4 dose, they started to assume liquid L-T3 at an initial fixed dose of 3.55 µg (5 drops). Further adjustments of L-T3 dose were tailored according to individual assessment. Initial serum TSH ranged from 0.8 to 12.0 mIU/L, when patients assumed high dose of L-T4 alone. Following the addition of a daily single dose of 3.55 µg L-T3, the target TSH was attained in five patients (23.8%). After increasing L-T3 dose up to a mean of 7.3±3.4 µg/day all patients reached target serum TSH (<0.1 mIU/L). The mean individual L-T3 dose was significantly correlated with the body weight and was 0.11±0.04 µg/kg/day (p=0.013). Mean L-T4:L-T3 ratio was 21:1. No patients showed skewed free-T3 or free-T4 values, neither experienced discomfort nor reported adverse events. Liquid L-T3 can be useful to achieve optimal TSH suppression in high risk DTC with not suppressed TSH on L-T4 alone. This formulation allows an individual tailoring of L-T3, minimizing risks of side effects as well as of overtreatment in these clinical conditions.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Tireotropina/sangue , Tri-Iodotironina/administração & dosagem , Adenocarcinoma/sangue , Adenocarcinoma/radioterapia , Adulto , Formas de Dosagem , Feminino , Objetivos , Terapia de Reposição Hormonal , Humanos , Radioisótopos do Iodo/uso terapêutico , Masculino , Pessoa de Meia-Idade , Neoplasias da Glândula Tireoide/sangue , Neoplasias da Glândula Tireoide/radioterapia
9.
Eur J Neurosci ; 43(2): 148-61, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26386265

RESUMO

Functional maturation of afferent synaptic connections to inner hair cells (IHCs) involves pruning of excess synapses formed during development, as well as the strengthening and survival of the retained synapses. These events take place during the thyroid hormone (TH)-critical period of cochlear development, which is in the perinatal period for mice and in the third trimester for humans. Here, we used the hypothyroid Snell dwarf mouse (Pit1(dw)) as a model to study the role of TH in afferent type I synaptic refinement and functional maturation. We observed defects in afferent synaptic pruning and delays in calcium channel clustering in the IHCs of Pit1(dw) mice. Nevertheless, calcium currents and capacitance reached near normal levels in Pit1(dw) IHCs by the age of onset of hearing, despite the excess number of retained synapses. We restored normal synaptic pruning in Pit1(dw) IHCs by supplementing with TH from postnatal day (P)3 to P8, establishing this window as being critical for TH action on this process. Afferent terminals of older Pit1(dw) IHCs showed evidence of excitotoxic damage accompanied by a concomitant reduction in the levels of the glial glutamate transporter, GLAST. Our results indicate that a lack of TH during a critical period of inner ear development causes defects in pruning and long-term homeostatic maintenance of afferent synapses.


Assuntos
Cóclea/crescimento & desenvolvimento , Células Ciliadas Auditivas Internas/fisiologia , Células Ciliadas Auditivas Internas/ultraestrutura , Sinapses/fisiologia , Sinapses/ultraestrutura , Tri-Iodotironina/fisiologia , Oxirredutases do Álcool , Animais , Canais de Cálcio Tipo L/metabolismo , Proteínas Correpressoras , Cóclea/efeitos dos fármacos , Cóclea/ultraestrutura , Proteínas de Ligação a DNA/metabolismo , Transportador 1 de Aminoácido Excitatório/metabolismo , Células Ciliadas Auditivas Internas/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/metabolismo , Sinapses/efeitos dos fármacos , Fator de Transcrição Pit-1/genética , Tri-Iodotironina/administração & dosagem
10.
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
11.
Am J Physiol Heart Circ Physiol ; 306(8): H1164-70, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24531815

RESUMO

Extracorporeal membrane oxygenation (ECMO) is frequently used in infants with postoperative cardiopulmonary failure. ECMO also suppresses circulating triiodothyronine (T3) levels and modifies myocardial metabolism. We assessed the hypothesis that T3 supplementation reverses ECMO-induced metabolic abnormalities in the immature heart. Twenty-two male Yorkshire pigs (age: 25-38 days) with ECMO received [2-(13)C]lactate, [2,4,6,8-(13)C4]octanoate (medium-chain fatty acid), and [U-(13)C]long-chain fatty acids as metabolic tracers either systemically (totally physiological intracoronary concentration) or directly into the coronary artery (high substrate concentration) for the last 60 min of each protocol. NMR analysis of left ventricular tissue determined the fractional contribution of these substrates to the tricarboxylic acid cycle. Fifty percent of the pigs in each group received intravenous T3 supplement (bolus at 0.6 µg/kg and then continuous infusion at 0.2 µg·kg(-1)·h(-1)) during ECMO. Under both substrate loading conditions, T3 significantly increased the fractional contribution of lactate with a marginal increase in the fractional contribution of octanoate. Both T3 and high substrate provision increased the myocardial energy status, as indexed by phosphocreatine concentration/ATP concentration. In conclusion, T3 supplementation promoted lactate metabolism to the tricarboxylic acid cycle during ECMO, suggesting that T3 releases the inhibition of pyruvate dehydrogenase. Manipulation of substrate utilization by T3 may be used therapeutically during ECMO to improve the resting energy state and facilitate weaning.


Assuntos
Ciclo do Ácido Cítrico/efeitos dos fármacos , Ciclo do Ácido Cítrico/fisiologia , Oxigenação por Membrana Extracorpórea , Miocárdio/metabolismo , Tri-Iodotironina/administração & dosagem , Trifosfato de Adenosina/análise , Animais , Caprilatos/metabolismo , Isótopos de Carbono , Metabolismo Energético , Ácido Láctico/sangue , Ácido Láctico/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Miocárdio/química , Consumo de Oxigênio , Fosfocreatina/análise , Complexo Piruvato Desidrogenase/metabolismo , Sus scrofa , Tri-Iodotironina/sangue
12.
Am J Ther ; 21(6): e211-4, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23567793

RESUMO

Thyrotoxic periodic paralysis (TPP) is a rare reversible cause of paralysis and cramping. TPP is usually precipitated by common causes of thyrotoxicosis such as Grave disease or multinodular goiter. TPP precipitated by exogenous triiodothyronine (T3) intake is an extremely rare occurrence with only 3 cases reported to date. We now report a 24-year-old healthy manual laborer who developed quadriparesis during a period of rest after heavy exertion and carbohydrate intake. He had severe hypokalemia (potassium level 1.9 mmole/L). Correction of his hypokalemia reversed the paralysis without rebound hyperkalemia. After a detailed history review, he reported that he had been consuming nutraceuticals containing T3 for 1 month to lose weight, and laboratory studies confirmed factitious T3 toxicosis. There was no evidence of renal or gastrointestinal potassium wasting. This episode of TPP was the first manifestation of thyrotoxicosis in this patient, and avoidance of T3 intake prevented more episodes.


Assuntos
Suplementos Nutricionais/efeitos adversos , Paralisia Periódica Hipopotassêmica/induzido quimicamente , Tri-Iodotironina/efeitos adversos , Redução de Peso/efeitos dos fármacos , Humanos , Masculino , Tri-Iodotironina/administração & dosagem , Adulto Jovem
13.
Endocrinology ; 153(1): 101-12, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22028444

RESUMO

Siberian hamsters display photoperiodically regulated annual cycles in body weight, appetite, and reproduction. Previous studies have revealed a profound up-regulation of type 3 deiodinase (DIO3) mRNA in the ventral ependyma of the hypothalamus associated with hypophagia and weight loss in short-day photoperiods. DIO3 reduces the local availability of T(3), so the aim of this study was to test the hypothesis that decreased hypothalamic T(3) availability underlies the short-day-induced catabolic state. The experimental approach was to determine whether a local increase in T(3) in the hypothalamus of hamsters exposed to short days could reverse the behavioral and physiological changes induced by this photoperiod. In study 1, microimplants releasing T(3) were placed bilaterally into the hypothalamus. This treatment rapidly induced a long-day phenotype including increased appetite and body weight within 3 wk of treatment and increased fat mass and testis size by the end of the 10-wk study period. In study 2, hypothalamic T(3) implants were placed into hamsters carrying abdominal radiotelemetry implants. Again body weight increased significantly, and the occurrence of winter torpor bouts was dramatically decreased to less than one bout per week, whereas sham-implanted hamsters entered torpor up to six times a week. Our findings demonstrate that increased central T(3) induces a long-day metabolic phenotype, but in neither study was the molt cycle affected, so we infer that we had not disrupted the initial detection of photoperiod. We conclude that hypothalamic thyroid hormone availability plays a key role in seasonal regulation of appetite, body weight, and torpor.


Assuntos
Hipotálamo/metabolismo , Phodopus/anatomia & histologia , Phodopus/fisiologia , Tri-Iodotironina/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Cricetinae , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Masculino , Muda/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Fotoperíodo , Precursores de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Estações do Ano , Hormônio Liberador de Tireotropina/genética , Tri-Iodotironina/administração & dosagem
14.
J Nutr Biochem ; 23(9): 1113-20, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22137030

RESUMO

Several preconditioning strategies are used to prevent ischemia-reperfusion (IR) liver injury, a deleterious condition associated with tissue resection, transplantation or trauma. Although thyroid hormone (T3) administration exerts significant protection against liver IR injury in the rat, its clinical application is controversial due to possible adverse effects. Considering that prevention of liver IR injury has also been achieved by n-3 polyunsaturated fatty acid (n-3 PUFA) supplementation to rats, we studied the effect of n-3 PUFA dietary supplementation plus a lower dose of T3 against IR injury. Male Sprague-Dawley rats receiving fish oil (300 mg/kg) for 3 days followed by a single intraperitoneal dose of 0.05 mg T3/kg were subjected to 1 h of ischemia followed by 20 h of reperfusion. Parameters of liver injury (serum transaminases, histology) and oxidative stress (liver contents of GSH and oxidized proteins) were correlated with fatty acid composition, NF-κB activity, and tumor necrosis factor-α (TNF-α) and haptoglobin expression. IR significantly modified liver histology; enhanced serum transaminases, TNF-α response or liver oxidative stress; and decreased liver NF-κB activity and haptoglobin expression. Although IR injury was not prevented by either n-3 PUFA supplementation or T3 administration, substantial decrease in liver injury and oxidative stress was achieved by the combined protocol, which also led to increased liver n-3 PUFA content and decreased n-6/n-3 PUFA ratios, with recovery of NF-κB activity and TNF-α and haptoglobin expression. Prevention of liver IR injury achieved by a combined protocol of T3 and n-3 PUFA supplementation may represent a novel noninvasive preconditioning strategy with potential clinical application.


Assuntos
Antioxidantes/uso terapêutico , Suplementos Nutricionais , Óleos de Peixe/uso terapêutico , Interações Alimento-Droga , Fígado/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Tri-Iodotironina/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/sangue , Anti-Inflamatórios não Esteroides/uso terapêutico , Antioxidantes/administração & dosagem , Antioxidantes/análise , Ácidos Graxos Ômega-3/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Haptoglobinas/genética , Haptoglobinas/metabolismo , Insuficiência Hepática/etiologia , Insuficiência Hepática/prevenção & controle , Injeções Intraperitoneais , Fígado/metabolismo , Fígado/patologia , Fígado/fisiopatologia , Masculino , NF-kappa B/genética , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
15.
Homeopathy ; 100(4): 220-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21962196

RESUMO

BACKGROUND: Ultra High Dilutions (UHD) are diluted beyond the Avogadro limit with dynamization (dilution with succussion). The process of anuran amphibian metamorphosis is controlled by thyroid hormones, including the resorption of the tadpole tail. METHODS: A randomized and blinded study was performed to investigate the influence of triiodothyronine (T3) 5·10(-24)M (10cH) on apoptosis induced by T3 100 nM in Rana catesbeiana tadpoles' tail tips, in vitro. Explants were randomized to three groups: control: no T3 in pharmacological or UHD dose; test: T3 100 nM and challenged with T3 10cH (UHD); positive control: T3 100 nM, treated with unsuccussed ethanol. The apoptotic index and the area of explants of test and control groups at the first and final day of the experiment were compared by t-test. RESULTS: There was no difference in tail tip area between test and control groups, but a significantly higher (p<0.01) index of apoptosis in explants of the test group. CONCLUSION: This data suggest that T3 10cH modifies the effect of T3 at pharmacological dose, opening new perspectives for further studies and investigation of the dose-effect curve.


Assuntos
Apoptose/efeitos dos fármacos , Modelos Biológicos , Tri-Iodotironina/administração & dosagem , Animais , Homeopatia , Metamorfose Biológica , Rana catesbeiana , Distribuição Aleatória , Método Simples-Cego , Soluções , Cauda , Tri-Iodotironina/química
16.
J Clin Endocrinol Metab ; 94(10): 4003-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19602558

RESUMO

CONTEXT: Although acquired abnormalities of thyroid hormone metabolism are common, inherited defects in humans involving the synthesis of selenoproteins, including iodothyronine deiodinases, have been described in only one recent publication. OBJECTIVE: We report the study of a novel selenocysteine insertion sequence-binding protein 2 (SBP2) gene mutation (R128X) and its clinical and molecular characterization. SUBJECTS AND METHODS: A family of African origin was studied. The proband presented with growth retardation, low serum selenium level, and thyroid test abnormalities consisting of high serum total and free T(4) concentrations associated with low T(3), high rT(3), and normal TSH. The entire coding region of the SBP2 gene was sequenced and minigenes constructed to explain the nature of the defect. RESULTS: The proband was homozygous for a nonsense gene mutation that produces an early stop codon (R128X). Both parents and a sister were heterozygous but showed no growth or thyroid test abnormalities. Despite the severity of the defect, the patient had a relatively mild phenotype, similar to that associated with partial SBP2 deficiency. In vitro analysis showed that the mutant minigene synthesized SBP2 from at least three downstream ATGs capable of generating molecules containing the essential functional domains. Treatment with l-T(3) accelerated the growth velocity and advanced the bone age. CONCLUSIONS: We identified a novel SBP2 gene mutation producing an early arrest in the synthesis of a full-length molecule. The demonstration that SBP2 isoforms containing all functional domains could be synthesized from three downstream ATGs explains the relatively mild phenotype caused by this defect.


Assuntos
População Negra/genética , Estatura/genética , Mutação , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Selênio/deficiência , Hormônios Tireóideos/sangue , Tri-Iodotironina/uso terapêutico , Arginina , Criança , Códon/genética , Gana , Humanos , Masculino , Fenótipo , Reação em Cadeia da Polimerase , Biossíntese de Proteínas , Testes de Função Tireóidea , Tireotropina/sangue , Tri-Iodotironina/administração & dosagem
17.
Pediatrics ; 124(2): e258-68, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19581264

RESUMO

BACKGROUND: Transiently low levels of thyroid hormones occur in approximately 50% of neonates born 24-28 weeks' gestation and are associated with higher rates of cerebral palsy and cognitive impairment. Raising hormone levels shows promise for improving neurodevelopmental outcome. OBJECTIVE: To identify whether any of 4 thyroid hormone supplementation regimens could raise T(4) and FT(4) without suppressing TSH (biochemical euthyroidism). METHODS: Eligible subjects had gestational ages between 24 07 and 2767 weeks and were randomized <24 hours of birth to one of six study arms (n = 20-27 per arm): placebo (vehicle: 5% dextrose), potassium iodide (30 microg/kg/d) and continuous or bolus daily infusions of either 4 or 8 microg/kg/d of T(4) for 42 days. T(4) was accompanied by 1 microg/kg/d T(3) during the first 14 postnatal days and infused with 1 mg/mL albumin to prevent adherence to plastic tubing. RESULTS: FT(4) was elevated in the first 7 days in all hormone-treated subjects; however, only the continuous 8 microg/kg/d treatment arm showed a significant elevation in all treatment epochs (P < .002 versus all other groups). TT(4) remained elevated in the first 7 days in all hormone-treated subjects (P < .05 versus placebo or iodine arms). After 14 days, both 8 microg/kg/d arms as well as the continuous 4 microg/kg/d arm produced a sustained elevation of the mean and median TT(4), >7 microg/dL (90 nM/L; P < .002 versus placebo). The least suppression of THS was achieved in the 4 microg/kg/d T(4) continuous infusion arm. Although not pre-hypothesized, the duration of mechanical ventilation was significantly lower in the continuous 4 microg/kg/d T(4) arm and in the 8 microg/kg/d T(4) bolus arm (P < .05 versus remaining arms). ROP was significantly lower in the combined 4 thyroid hormone treatment arms than in the combined placebo and iodine arms (P < .04). NEC was higher in the combined 8 microg/kg/d arms (P < .05 versus other arms). CONCLUSIONS: Elevation of TT(4) with only modest suppression of TSH was associated with trends suggesting clinical benefits using a continuous supplement of low-dose thyroid hormone (4 microg/kg/d) for 42 days. Future trials will be needed to assess the long-term neurodevelopmental effects of such supplementation.


Assuntos
Recém-Nascido de Peso Extremamente Baixo ao Nascer , Doenças do Prematuro/tratamento farmacológico , Tiroxina/administração & dosagem , Tiroxina/deficiência , Tri-Iodotironina/administração & dosagem , Administração Oral , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Seguimentos , Humanos , Hidrocortisona/sangue , Recém-Nascido , Doenças do Prematuro/sangue , Doenças do Prematuro/mortalidade , Infusões Intravenosas , Iodo/administração & dosagem , Masculino , Taxa de Sobrevida , Testes de Função Tireóidea , Tireotropina/sangue , Tiroxina/sangue , Tri-Iodotironina/sangue
18.
Regul Pept ; 151(1-3): 164-9, 2008 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-18708095

RESUMO

Thyroid hormone regulates food intake. We previously reported that rats with triiodothyronine (T3)-induced thyrotoxicosis display hyperphagia associated with suppressed circulating leptin levels, increased hypothalamic neuropeptide Y (NPY) mRNA and decreased hypothalamic pro-opiomelanocortin (POMC) mRNA. AMP-activated kinase (AMPK) is a serine/threonine protein kinase that is activated when cellular energy is depleted. We hypothesized that T3 causes an increase in hypothalamic AMPK activity, which in turn contributes to the development of T3-induced hyperphagia. Rats that were given s.c. injections of T3 (4.5 nmol/kg) had increased food intake 2 h later without alterations in NPY and POMC mRNA levels, but with increased hypothalamic phosphorylated AMPK (169%) and phosphorylated acetyl-CoA carboxylase (194%). To determine the more chronic effects of T3, rats were given 6 daily s.c. injection of T3 or the vehicle. Food intake was significantly increased. Multiple T3 injections increased hypothalamic phosphorylated AMPK (278%) and phosphorylated acetyl-CoA carboxylase (335%) compared to the controls. Intracerebroventricular administration of compound C, an AMPK inhibitor, blocked the food intake induced by a single or multiple injections of T3. Taken together, these results suggest that enhanced hypothalamic AMPK phosphorylation contributes to T3-induced hyperphagia. Hypothalamic AMPK plays an important role in the regulation of food intake and body weight.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/enzimologia , Tri-Iodotironina/farmacologia , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Acetil-CoA Carboxilase/metabolismo , Adiposidade/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Masculino , Neuropeptídeo Y/genética , Fosforilação , Pró-Opiomelanocortina/genética , Inibidores de Proteínas Quinases/administração & dosagem , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Tri-Iodotironina/administração & dosagem
19.
Biol Psychiatry ; 63(12): 1127-34, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18206856

RESUMO

BACKGROUND: Low cerebral bioenergetic metabolism has been reported in subjects with major depressive disorder (MDD). Thyroid hormones have been shown to increase brain bioenergetic metabolism. We assessed whether changes in brain bioenergetics measured with phosphorus magnetic resonance spectroscopy ((31)P MRS) correlate with treatment outcome during augmentation treatment with triiodothyronine (T3) in MDD. METHODS: Nineteen subjects meeting DSM-IV criteria for MDD who had previously failed to respond to selective serotonin reuptake inhibitor (SSRI) antidepressant drugs received open label and prospective augmentation treatment with T3 for 4 weeks. We obtained (31)P MRS spectra before and after treatment from all MDD subjects and baseline (31)P MRS from nine normal control subjects matched for age and gender. RESULTS: At baseline, depressed subjects had lower intracellular Mg(2+) compared with control subjects. Seven MDD subjects (38.9%) were treatment responders (>or= 50% improvement). Total nucleoside triphosphate (NTP), which primarily represents adenosine triphosphate (ATP), increased significantly in MDD subjects responding to T3 augmentation compared with treatment nonresponders. Phosphocreatine, which has a buffer role for ATP, decreased in treatment responders compared with nonresponders. CONCLUSIONS: The antidepressant effect of thyroid hormone (T3) augmentation of SSRIs is correlated with significant changes in the brain bioenergetic metabolism. This seems to be a re-normalization of brain bioenergetics in treatment responders. Further studies will determine whether these findings can be generalized to other antidepressant treatments.


Assuntos
Encéfalo/efeitos dos fármacos , Transtorno Depressivo Maior/tratamento farmacológico , Metabolismo Energético/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Tri-Iodotironina/administração & dosagem , Trifosfato de Adenosina/metabolismo , Adulto , Antidepressivos/uso terapêutico , Encéfalo/fisiopatologia , Transtorno Depressivo Maior/fisiopatologia , Quimioterapia Combinada , Metabolismo Energético/fisiologia , Feminino , Humanos , Magnésio/metabolismo , Masculino , Pessoa de Meia-Idade , Inventário de Personalidade , Fosfocreatina/metabolismo , Estudos Prospectivos , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Tireotropina/sangue , Resultado do Tratamento
20.
Endocrinology ; 147(9): 4036-43, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16709608

RESUMO

Mutations of the X-linked thyroid hormone (TH) transporter (monocarboxylate transporter, MCT8) produce in humans unusual abnormalities of thyroid function characterized by high serum T3 and low T4 and rT3. The mechanism of these changes remains obscure and raises questions regarding the regulation of intracellular availability and metabolism of TH. To study the pathophysiology of MCT8 deficiency, we generated Mct8 knockout mice. Male mice deficient in Mct8 (Mct8(-/y)) replicate the thyroid abnormalities observed in affected men. TH deprivation and replacement with L-T3 showed that suppression of TSH required higher serum levels T3 in Mct8(-/y) than wild-type (WT) littermates, indicating hypothalamus and/or thyrotroph resistance to T3. Furthermore, T4 is required to maintain the high serum T3 level because the latter was not different between the two genotypes during administration of T3. Mct8(-/y) mice have 2.3-fold higher T3 content in liver associated with 6.1- and 3.1-fold increase in deiodinase 1 mRNA and enzymatic activity, respectively. The relative T3 excess in liver of Mct8(-/y) mice produced a decrease in serum cholesterol (79 +/- 18 vs. 137 +/- 38 mg/dl in WT) and an increase in alkaline phosphatase (107 +/- 23 vs. 58 +/- 3 U/liter in WT) levels. In contrast, T3 content in cerebrum was 1.8-fold lower in Mct8(-/y) mice, associated with a 1.6- and 10.6-fold increase in D2 mRNA and enzymatic activity, respectively, as previously observed in TH-deprived WT mice. We conclude that cell-specific differences in intracellular TH content due to differences in contribution of the various TH transporters are responsible for the unusual clinical presentation of this defect, in contrast to TH deficiency.


Assuntos
Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/fisiologia , Hormônios Tireóideos/metabolismo , Fosfatase Alcalina/sangue , Animais , Química Encefálica , Colesterol/sangue , Modelos Animais de Doenças , Resistência a Medicamentos , Feminino , Humanos , Hipotálamo/efeitos dos fármacos , Cinética , Fígado/química , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos , Mutação , Fenótipo , Simportadores , Tireotropina/fisiologia , Tiroxina/administração & dosagem , Tiroxina/sangue , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/análise , Tri-Iodotironina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA