Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Thyroid ; 34(2): 243-251, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38149585

RESUMO

Background: The importance of thyroid hormones (THs) for peripheral body temperature regulation has been long recognized, as medical conditions such as hyper- and hypothyroidism lead to alterations in body temperature and energy metabolism. In the past decade, the brain actions of THs and their respective nuclear receptors, thyroid hormone receptor α1 (TRα1) and thyroid hormone receptor beta (TRß), coordinating body temperature regulation have moved into focus. However, the exact roles of the individual TR isoforms and their precise neuroanatomical substrates remain poorly understood. Methods: Here we used mice expressing a mutant TRα1 (TRα1+m) as well as TRß knockouts to study body temperature regulation using radiotelemetry in conscious and freely moving animals at different ambient temperatures, including their response to oral 3,3',5-triiodothyronine (T3) treatment. Subsequently, we tested the effects of a dominant-negative TRα1 on body temperature after adeno-associated virus (AAV)-mediated expression in the hypothalamus, a region known to be involved in thermoregulation. Results: While TRß seems to play a negligible role in body temperature regulation, TRα1+m mice had lower body temperature, which was surprisingly not entirely normalized at 30°C, where defects in facultative thermogenesis or tail heat loss are eliminated as confounding factors. Only oral T3 treatment fully normalized the body temperature profile of TRα1+m mice, suggesting that the mutant TRα1 confers an altered central temperature set point in these mice. When we tested this hypothesis more directly by expressing the dominant-negative TRα1 selectively in the hypothalamus via AAV transfection, we observed a similarly reduced body temperature at room temperature and 30°C. Conclusion: Our data suggest that TRα1 signaling in the hypothalamus is important for maintaining body temperature. However, further studies are needed to dissect the precise neuroanatomical substrates and the downstream pathways mediating this effect.


Assuntos
Hipotireoidismo , Receptores dos Hormônios Tireóideos , Camundongos , Animais , Receptores dos Hormônios Tireóideos/metabolismo , Temperatura Corporal , Tri-Iodotironina/farmacologia , Tri-Iodotironina/metabolismo , Hipotireoidismo/genética , Hipotireoidismo/metabolismo , Hormônios Tireóideos , Hipotálamo , Receptores alfa dos Hormônios Tireóideos/genética , Receptores alfa dos Hormônios Tireóideos/metabolismo
2.
Nat Commun ; 14(1): 6742, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37875497

RESUMO

It is well established that maternal thyroid hormones play an important role for the developing fetus; however, the consequences of maternal hyperthyroidism for the offspring remain poorly understood. Here we show in mice that maternal 3,3',5-triiodothyronine (T3) treatment during pregnancy leads to improved glucose tolerance in the adult male offspring and hyperactivity of brown adipose tissue (BAT) thermogenesis in both sexes starting early after birth. The activated BAT provides advantages upon cold exposure, reducing the strain on other thermogenic organs like muscle. This maternal BAT programming requires intact maternal thyroid hormone receptor ß (TRß) signaling, as offspring of mothers lacking this receptor display the opposite phenotype. On the molecular level, we identify distinct T3 induced alterations in maternal serum metabolites, including choline, a key metabolite for healthy pregnancy. Taken together, our results connect maternal TRß activation to the fetal programming of a thermoregulatory phenotype in the offspring.


Assuntos
Tecido Adiposo Marrom , Receptores beta dos Hormônios Tireóideos , Gravidez , Feminino , Camundongos , Animais , Masculino , Tecido Adiposo Marrom/metabolismo , Receptores beta dos Hormônios Tireóideos/genética , Receptores beta dos Hormônios Tireóideos/metabolismo , Tri-Iodotironina/metabolismo , Termogênese/fisiologia , Hormônios Tireóideos/metabolismo
3.
iScience ; 26(10): 108064, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37822510

RESUMO

Agonists for thyroid hormone receptor ß (TRß) show promise in preclinical studies and clinical trials to improve non-alcoholic fatty liver disease. A recent study on human livers, however, revealed reduced TRß expression in non-alcoholic steatohepatitis (NASH), indicating a developing thyroid hormone resistance, which could constitute a major obstacle for those agonists. Using a rapid NASH paradigm combining choline-deficient high-fat diet and thermoneutrality, we confirm that TRß declines during disease progression in mice similar to humans. Contrary to expectations, mice lacking TRß showed less liver fibrosis, and NASH marker genes were not elevated. Conversely, increasing TRß expression in wild-type NASH mice using liver-targeted gene therapy did not improve histology, gene expression, or metabolic parameters, indicating that TRß receptor levels are of minor relevance for NASH development and progression in our model, and suggest that liver-rather than isoform-specificity might be more relevant for NASH treatment with thyroid hormone receptor agonists.

4.
Nat Commun ; 14(1): 3312, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37286550

RESUMO

Mutations in thyroid hormone receptor α1 (TRα1) cause Resistance to Thyroid Hormone α (RTHα), a disorder characterized by hypothyroidism in TRα1-expressing tissues including the heart. Surprisingly, we report that treatment of RTHα patients with thyroxine to overcome tissue hormone resistance does not elevate their heart rate. Cardiac telemetry in male, TRα1 mutant, mice indicates that such persistent bradycardia is caused by an intrinsic cardiac defect and not due to altered autonomic control. Transcriptomic analyses show preserved, thyroid hormone (T3)-dependent upregulation of pacemaker channels (Hcn2, Hcn4), but irreversibly reduced expression of several ion channel genes controlling heart rate. Exposure of TRα1 mutant male mice to higher maternal T3 concentrations in utero, restores altered expression and DNA methylation of ion channels, including Ryr2. Our findings indicate that target genes other than Hcn2 and Hcn4 mediate T3-induced tachycardia and suggest that treatment of RTHα patients with thyroxine in high dosage without concomitant tachycardia, is possible.


Assuntos
Síndrome da Resistência aos Hormônios Tireóideos , Tiroxina , Masculino , Animais , Camundongos , Tiroxina/uso terapêutico , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Hormônios Tireóideos , Receptores alfa dos Hormônios Tireóideos/genética , Receptores alfa dos Hormônios Tireóideos/metabolismo , Mutação , Taquicardia/genética
5.
Cell Rep ; 42(2): 112088, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36753417

RESUMO

The hypothalamic pituitary thyroid axis is a major regulator of many differentiation processes, including adipose tissue. However, it remains unclear whether and how thyroid hormone (TH) signaling contributes to preadipocyte commitment and differentiation into mature adipocytes. Here, we show a cell-autonomous effect of TH on the transcriptional regulation of zinc finger protein 423 (Zfp423), an early adipogenic determination factor, in murine adipose depots. Mechanistically, binding of the unliganded TH receptor to a negative TH responsive element within the Zfp423 promoter activates transcriptional activity that is reversed upon TH binding. Zfp423 upregulation is associated with increased GFP+ preadipocyte recruitment in stromal vascular fraction isolated from white fat of hypothyroid Zfp423GFP reporter mice. RNA sequencing identified Zfp423-driven gene programs that are modulated in response to TH during adipogenic differentiation. Collectively, we identified Zfp423 as a key molecule that integrates TH signaling into the regulation of adipose tissue plasticity.


Assuntos
Adipócitos , Proteínas de Ligação a DNA , Animais , Camundongos , Adipócitos/metabolismo , Adipogenia/fisiologia , Tecido Adiposo/metabolismo , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Obesidade/metabolismo , Hormônios Tireóideos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Mol Metab ; 69: 101691, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36746332

RESUMO

OBJECTIVE: Snacking, i.e., the intake of small amounts of palatable food items, is a common behavior in modern societies, promoting overeating and obesity. Shifting food intake into the daily rest phase disrupts circadian rhythms and is also known to stimulate weight gain. We therefore hypothesized that chronic snacking in the inactive phase may promote body weight gain and that this effect is based on disruption of circadian clocks. METHODS: Male mice were fed a daily chocolate snack either during their rest or their active phase and body weight development and metabolic parameters were investigated. Snacking experiments were repeated in constant darkness and in clock-deficient mutant mice to examine the role of external and internal time cues in mediating the metabolic effects of snacking. RESULTS: Chronic snacking in the rest phase increased body weight gain and disrupted metabolic circadian rhythms in energy expenditure, body temperature, and locomotor activity. Additionally, these rest phase snacking mice assimilated more energy during the inactive phase. Body weight remained increased in rest phase snacking wildtype mice in constant darkness as well as in clock-deficient mutant mice under a regular light-dark cycle compared to mice snacking in the active phase. Weight gain effects were abolished in clock-deficient mice in constant darkness. CONCLUSIONS: Our data suggest that mistimed snacking increases energy resorption and promotes body weight gain. This effect requires a functional circadian clock at least under constant darkness conditions.


Assuntos
Lanches , Aumento de Peso , Camundongos , Animais , Masculino , Ritmo Circadiano , Obesidade , Peso Corporal
8.
Nat Metab ; 3(10): 1415-1431, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34675439

RESUMO

Current pharmacological therapies for treating obesity are of limited efficacy. Genetic ablation or loss of function of AMP-activated protein kinase alpha 1 (AMPKα1) in steroidogenic factor 1 (SF1) neurons of the ventromedial nucleus of the hypothalamus (VMH) induces feeding-independent resistance to obesity due to sympathetic activation of brown adipose tissue (BAT) thermogenesis. Here, we show that body weight of obese mice can be reduced by intravenous injection of small extracellular vesicles (sEVs) delivering a plasmid encoding an AMPKα1 dominant negative mutant (AMPKα1-DN) targeted to VMH-SF1 neurons. The beneficial effect of SF1-AMPKα1-DN-loaded sEVs is feeding-independent and involves sympathetic nerve activation and increased UCP1-dependent thermogenesis in BAT. Our results underscore the potential of sEVs to specifically target AMPK in hypothalamic neurons and introduce a broader strategy to manipulate body weight and reduce obesity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo Marrom/enzimologia , Vesículas Extracelulares/metabolismo , Hipotálamo/enzimologia , Obesidade/metabolismo , Animais , Metabolismo Energético , Camundongos , Termogênese , Redução de Peso
9.
Thyroid ; 31(9): 1424-1435, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34269617

RESUMO

Background: Maternal thyroid hormone (TH) plays an essential role for fetal development, especially for the cardiovascular system and its central control. However, the precise consequences of altered TH action during the different periods in pregnancy remain poorly understood. Methods: To address this question, we used mice heterozygous for a mutant thyroid hormone receptor α1 (TRα1) and wild-type controls that were born to wild-type mothers treated with 3,3',5-triiodothyronine (T3) during the first or the second half of pregnancy. We then phenotyped the offspring animals as adults by in vivo measurements and postmortem tissue analyses. Results: Maternal T3 treatment in either half of the pregnancy did not affect postnatal growth development. Serum thyroxine and hypophyseal thyrotropin subunit beta or deiodinase type II expression was also not affected in any group, only TRα1 mutant males exhibited a reduction in serum T3 levels after the treatment. Likewise, hepatic deiodinase type I was not altered, but serum selenium levels were reduced by the maternal treatment in wild-type offspring of both genders. Most interestingly, a significant increase in heart weight was found in adult wild-types born to mothers that received T3 during the first or second half of pregnancy, while TRα1 mutant males were protected from this effect. Moreover, we detected a significant increase in heart rate selectively in male mice that were exposed to elevated maternal T3 in the second half of the pregnancy. Conclusion: Taken together, our findings demonstrate that maternal TH is of particular relevance during the second half of pregnancy for establishing cardiac properties, with specific effects depending on TRα1 or gender. The data advocate routinely monitoring TH levels during pregnancy to avoid adverse cardiac effects in the offspring.


Assuntos
Pressão Arterial/efeitos dos fármacos , Sistema Cardiovascular/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Tri-Iodotironina/farmacologia , Animais , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Metilação de DNA , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Masculino , Exposição Materna , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Gravidez , Fatores Sexuais , Receptores alfa dos Hormônios Tireóideos/genética
10.
Sci Rep ; 11(1): 4799, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637831

RESUMO

Since the discovery of functional brown adipose tissue (BAT) in adult humans, research on BAT gained a new popularity to combat obesity and related comorbidities. To date, however, methods to quantify BAT thermogenesis are often either highly invasive, require advanced equipment, are time consuming or of limited sensitivity. Here we present a simple yet highly effective and minimally invasive protocol for the Precise Unravelment of Non-shivering brown fat thermoKinetics (PUNK) in mice using infrared thermography in combination with Vaseline to brush up the fur between the shoulder blades. We also use physiological and molecular readouts including indirect calorimetry, qPCR and Western Blots to test our protocol. Our study demonstrates that Vaseline before thermography vastly advances the reproducibility and quality of BAT infrared pictures or videos, as it exposes the skin above the BAT for a direct line of sight for the infrared camera and thereby circumvents the well-known problems associated with shaving and anaesthesia. We subsequently validate that this approach does not affect physiological and molecular BAT function, but instead leads to more robust and less variable results when comparing for instance norepinephrine stimulation tests or knockout animals. Taken together, the PUNK protocol for BAT thermography quickly and effectively improves scientific outcomes of this method, and can be easily added to existing paradigms. Consequently, it safes money, time and experimental animals, thereby putting the 3R's principles of animal welfare into practice.


Assuntos
Tecido Adiposo Marrom/fisiologia , Termografia/métodos , Animais , Temperatura Corporal , Gorduras/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Termogênese
11.
Endocr Connect ; 10(2): R106-R115, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33491659

RESUMO

A normal thyroid status is crucial for body temperature homeostasis, as thyroid hormone regulates both heat loss and conservation as well as heat production in the thermogenic tissues. Brown adipose tissue (BAT) is the major site of non-shivering thermogenesis and an important target of thyroid hormone action. Thyroid hormone not only regulates the tissue's sensitivity to sympathetic stimulation by norepinephrine but also the expression of uncoupling protein 1, the key driver of BAT thermogenesis. Vice versa, sympathetic stimulation of BAT triggers the expression of deiodinase type II, an enzyme that enhances local thyroid hormone availability and signaling. This review summarizes the current knowledge on how thyroid hormone controls BAT thermogenesis, aiming to dissect the direct actions of the hormone in BAT and its indirect actions via the CNS, browning of white adipose tissue or heat loss over body surfaces. Of particular relevance is the apparent dose dependency of the observed effects, as we find that minor or moderate changes in thyroid hormone levels often have different effects as compared to high pharmacological doses. Moreover, we conclude that the more recent findings require a reevaluation of older studies, as key aspects such as heat loss or central BAT activation may not have received the necessary attention during the interpretation of these early findings. Finally, we provide a list of what we believe are the most relevant questions in the field that to date are still enigmatic and require further studies.

12.
Cell Rep ; 33(5): 108351, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33147454

RESUMO

Environmental temperature is a driving factor in evolution, and it is commonly assumed that metabolic adaptations to cold climates are the result of transgenerational selection. Here, we show in mice that even minor changes in maternal thermogenesis alter the metabolic phenotype already in the next generation. Male offspring of mothers genetically lacking brown adipose tissue (BAT) thermogenesis display increased lean mass and improved glucose tolerance as adults, while females are unaffected. The phenotype is replicated in offspring of mothers kept at thermoneutrality; conversely, mothers with higher gestational BAT thermogenesis produce male offspring with reduced lean mass and impaired glucose tolerance. Running-wheel exercise reverses the offspring's metabolic impairments, pointing to the muscle as target of these fetal programming effects. Our data demonstrate that gestational BAT activation negatively affects metabolic health of the male offspring; however, these unfavorable fetal programming effects may be negated by active lifestyle.


Assuntos
Tecido Adiposo Marrom/fisiologia , Glucose/metabolismo , Termogênese/fisiologia , Animais , Metabolismo Energético/fisiologia , Feminino , Homeostase , Humanos , Masculino , Metabolômica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Condicionamento Físico Animal , Gravidez , Temperatura , Proteína Desacopladora 1/deficiência , Proteína Desacopladora 1/metabolismo
13.
Sci Rep ; 10(1): 20203, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33214601

RESUMO

Brown adipose tissue (BAT) thermogenesis is considered a potential target for treatment of obesity and diabetes. In vitro data suggest dopamine receptor signaling as a promising approach; however, the biological relevance of dopamine receptors in the direct activation of BAT thermogenesis in vivo remains unclear. We investigated BAT thermogenesis in vivo in mice using peripheral administration of D1-agonist SKF38393 or D2-agonist Sumanirole, infrared thermography, and in-depth molecular analyses of potential target tissues; and ex vivo in BAT explants to identify direct effects on key thermogenic markers. Acute in vivo treatment with the D1- or D2-agonist caused a short spike or brief decrease in BAT temperature, respectively. However, repeated daily administration did not induce lasting effects on BAT thermogenesis. Likewise, neither agonist directly affected Ucp1 or Dio2 mRNA expression in BAT explants. Taken together, the investigated agonists do not seem to exert lasting and physiologically relevant effects on BAT thermogenesis after peripheral administration, demonstrating that D1- and D2-receptors in iBAT are unlikely to constitute targets for obesity treatment via BAT activation.


Assuntos
Tecido Adiposo Marrom/efeitos dos fármacos , Agonistas de Dopamina/farmacologia , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D2/agonistas , Termogênese/efeitos dos fármacos , 2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/farmacologia , Tecido Adiposo Marrom/metabolismo , Animais , Benzimidazóis/farmacologia , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Termografia
14.
Eur Thyroid J ; 9(2): 57-66, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32257954

RESUMO

INTRODUCTION: Injection of 3-iodothyronamine into experimental animals profoundly affects their metabolism and body temperature. As 3-iodothyronamine is rapidly acetylated in vivo after injection, it was hypothesized that the metabolites N- or O-acetyl-3-iodothyronamines could constitute the active hormones. METHODS: Adult male mice were injected once daily with one of the metabolites (5 mg/kg body weight intraperitoneally dissolved in 60% DMSO in PBS) or solvent. Metabolism was monitored by indirect calorimetry, body temperature by infrared thermography, and body composition by nuclear magnetic resonance analysis. Signaling activities in brown fat or liver were assessed by studying target gene transcription by qPCR including uncoupling protein 1 or deiodinase type 1 or 2, and Western blot. RESULTS: The markers of metabolism, body composition, or temperature tested were similar in the mice injected with solvent and those injected with one of the acetylated 3-iodothyronamines. CONCLUSIONS: In our experimental setup, N- and O-acetyl-3-iodothyronamine do not constitute compounds contributing to the metabolic or temperature effects described for 3-iodothyronamine. The acetylation of 3-iodothyronamine observed in vivo may thus rather serve degradation and elimination purposes.

15.
Thyroid ; 30(8): 1205-1216, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32188347

RESUMO

Background: Tachycardia, cardiac hypertrophy, and elevated body temperature are major signs of systemic hyperthyroidism, which are considered to reflect the excessive thyroid hormone (TH) action in the respective peripheral tissues. However, recent observations indicate that the central actions of TH also contribute substantially to cardiovascular regulation and thermogenesis. Methods: In this study, we dissect the individual contributions of peripheral TH action versus the central effects in body temperature regulation and cardiovascular functions by taking advantage of mice lacking the TH transporters monocarboxylate transporter 8 (MCT8) and organic anion transporting polypeptide 1C1 (OATP1C1) (M/O double knock-out [dko]), which exhibit elevated serum triiodothyronine (T3) levels while their brain is in a profoundly hypothyroid state. We compared these animals with wild-type (WT) mice that were treated orally with T3 to achieve similarly elevated serum T3 levels, but are centrally hyperthyroid. For the studies, we used radiotelemetry, infrared thermography, gene expression profiling, Western blot analyses, and enzyme linked immunosorbent assays (ELISA) assays. Results: Our analyses revealed mild hyperthermia and cardiac hypertrophy in T3-treated WT mice but not in M/O dko animals, suggesting that central actions of TH are required for these hyperthyroid phenotypes. Although the average heart rate was unaffected in either model, the M/O dko exhibited an altered heart rate frequency distribution with tachycardic bursts in active periods and bradycardic episodes during resting time, demonstrating that the stabilization of heart rate by the autonomic nervous system can be impaired in centrally hypothyroid animals. Conclusions: Our studies unravel distinct phenotypical traits of hyperthyroidism that depend on an intact central nervous system, and provide valuable insight into the cardiovascular pathology of the Allan-Herndon-Dudley syndrome, a condition caused by the lack of MCT8 in humans.


Assuntos
Cardiomegalia/metabolismo , Febre/metabolismo , Frequência Cardíaca , Hipotireoidismo/complicações , Hormônios Tireóideos/metabolismo , Animais , Cardiomegalia/prevenção & controle , Cruzamentos Genéticos , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Glicogênio/metabolismo , Lipólise , Fígado/metabolismo , Masculino , Retardo Mental Ligado ao Cromossomo X/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos/genética , Hipotonia Muscular/metabolismo , Atrofia Muscular/metabolismo , Fenótipo , Telemetria , Termogênese , Termografia , Fatores de Tempo , Tri-Iodotironina/sangue
16.
Cell Rep ; 27(11): 3385-3400.e3, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31189119

RESUMO

Regulation of body temperature critically depends on thyroid hormone (TH). Recent studies revealed that TH induces browning of white adipose tissue, possibly contributing to the observed hyperthermia in hyperthyroid patients and potentially providing metabolic benefits. Here, we show that browning by TH requires TH-receptor ß and occurs independently of the sympathetic nervous system. The beige fat, however, lacks sufficient adrenergic stimulation and is not metabolically activated despite high levels of uncoupling protein 1 (UCP1). Studies at different environmental temperatures reveal that TH instead causes hyperthermia by actions in skeletal muscle combined with a central body temperature set-point elevation. Consequently, the metabolic and thermogenic effects of systemic hyperthyroidism were maintained in UCP1 knockout mice, demonstrating that neither beige nor brown fat contributes to the TH-induced hyperthermia and elevated glucose consumption, and underlining that the mere presence of UCP1 is insufficient to draw conclusions on the therapeutic potential of browning agents.


Assuntos
Tecido Adiposo Bege/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Glucose/metabolismo , Termogênese , Hormônios Tireóideos/metabolismo , Tecido Adiposo Bege/fisiologia , Tecido Adiposo Marrom/fisiologia , Tecido Adiposo Branco/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
17.
J Mol Endocrinol ; 62(3): 91-99, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30608905

RESUMO

It is well established that thyroid hormones are required for cardiovascular functions; however, the molecular mechanisms remain incompletely understood, especially the individual contributions of genomic and non-genomic signalling pathways. In this study, we dissected how thyroid hormones modulate aortic contractility. To test the immediate effects of thyroid hormones on vasocontractility, we used a wire myograph to record the contractile response of dissected mouse aortas to the adrenergic agonist phenylephrine in the presence of different doses of T3 (3,3',5-triiodothyronine). Interestingly, we observed reduced vasoconstriction under low and high T3 concentrations, indicating an inversed U-shaped curve with maximal constrictive capacity at euthyroid conditions. We then tested for possible genomic actions of thyroid hormones on vasocontractility by treating mice for 4 days with 1 mg/L thyroxine in drinking water. The study revealed that in contrast to the non-genomic actions the aortas of these animals were hyperresponsive to the contractile stimulus, an effect not observed in endogenously hyperthyroid TRß knockout mice. To identify targets of genomic thyroid hormone action, we analysed aortic gene expression by microarray, revealing several altered genes including the well-known thyroid hormone target gene hairless. Taken together, the findings demonstrate that thyroid hormones regulate aortic tone through genomic and non-genomic actions, although genomic actions seem to prevail in vivo. Moreover, we identified several novel thyroid hormone target genes that could provide a better understanding of the molecular changes occurring in the hyperthyroid aorta.


Assuntos
Aorta/efeitos dos fármacos , Hipertireoidismo/sangue , Receptores beta dos Hormônios Tireóideos/metabolismo , Hormônios Tireóideos/sangue , Agonistas Adrenérgicos/farmacologia , Animais , Hipertireoidismo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fenilefrina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Receptores beta dos Hormônios Tireóideos/genética , Tri-Iodotironina/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/genética
19.
Front Horm Res ; 49: 48-66, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29895010

RESUMO

Cardiometabolic risk factors like abdominal obesity, hyperglycemia, low high-density lipoprotein (HDL) cholesterol, elevated triglycerides, and hypertension are defined as metabolic syndrome (MetS), which represents one of the most frequent endocrine disorders particularly in a society with increasing weight problems. As more and more evidence is accumulated that thyroid hormones affect components of the MetS, the present review aims to summarize the rapidly expanding knowledge on the pathophysiological interaction between thyroid hormone status and MetS. The review is based on a PubMed search for combinations of thyroid hormone action and MetS, blood pressure, hypertension, hyperlipidemia, cholesterol, HDL cholesterol, glucose, diabetes mellitus, body weight, or visceral fat. A special focus was given for manuscripts published after 2000 but we included seminal papers published before year 2000 as well.


Assuntos
Síndrome Metabólica/metabolismo , Doenças da Glândula Tireoide/metabolismo , Humanos , Síndrome Metabólica/etiologia , Doenças da Glândula Tireoide/complicações
20.
Sci Rep ; 8(1): 5872, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29651131

RESUMO

Mutations in mitochondrial DNA (mtDNA) lead to heteroplasmy, i.e., the intracellular coexistence of wild-type and mutant mtDNA strands, which impact a wide spectrum of diseases but also physiological processes, including endurance exercise performance in athletes. However, the phenotypic consequences of limited levels of naturally arising heteroplasmy have not been experimentally studied to date. We hence generated a conplastic mouse strain carrying the mitochondrial genome of an AKR/J mouse strain (B6-mtAKR) in a C57BL/6 J nuclear genomic background, leading to >20% heteroplasmy in the origin of light-strand DNA replication (OriL). These conplastic mice demonstrate a shorter lifespan as well as dysregulation of multiple metabolic pathways, culminating in impaired glucose metabolism, compared to that of wild-type C57BL/6 J mice carrying lower levels of heteroplasmy. Our results indicate that physiologically relevant differences in mtDNA heteroplasmy levels at a single, functionally important site impair the metabolic health and lifespan in mice.


Assuntos
Replicação do DNA/genética , DNA Mitocondrial/genética , Longevidade/genética , Mitocôndrias/genética , Animais , Glucose/genética , Glucose/metabolismo , Humanos , Redes e Vias Metabólicas/genética , Camundongos , Mitocôndrias/patologia , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...