Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 6742, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37875497

ABSTRACT

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.


Subject(s)
Adipose Tissue, Brown , Thyroid Hormone Receptors beta , Pregnancy , Female , Mice , Animals , Male , Adipose Tissue, Brown/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Triiodothyronine/metabolism , Thermogenesis/physiology , Thyroid Hormones/metabolism
2.
Thromb Haemost ; 122(10): 1814-1826, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36075234

ABSTRACT

BACKGROUND: Smooth muscle cell (SMC) phenotype switching plays a central role during vascular remodeling. Growth factor receptors are negatively regulated by protein tyrosine phosphatases (PTPs), including its prototype PTP1B. Here, we examine how reduction of PTP1B in SMCs affects the vascular remodeling response to injury. METHODS: Mice with inducible PTP1B deletion in SMCs (SMC.PTP1B-KO) were generated by crossing mice expressing Cre.ERT2 recombinase under the Myh11 promoter with PTP1Bflox/flox mice and subjected to FeCl3 carotid artery injury. RESULTS: Genetic deletion of PTP1B in SMCs resulted in adventitia enlargement, perivascular SMA+ and PDGFRß+ myofibroblast expansion, and collagen accumulation following vascular injury. Lineage tracing confirmed the appearance of Myh11-Cre reporter cells in the remodeling adventitia, and SCA1+ CD45- vascular progenitor cells increased. Elevated mRNA expression of transforming growth factor ß (TGFß) signaling components or enzymes involved in extracellular matrix remodeling and TGFß liberation was seen in injured SMC.PTP1B-KO mouse carotid arteries, and mRNA transcript levels of contractile SMC marker genes were reduced already at baseline. Mechanistically, Cre recombinase (mice) or siRNA (cells)-mediated downregulation of PTP1B or inhibition of ERK1/2 signaling in SMCs resulted in nuclear accumulation of KLF4, a central transcriptional repressor of SMC differentiation, whereas phosphorylation and nuclear translocation of SMAD2 and SMAD3 were reduced. SMAD2 siRNA transfection increased protein levels of PDGFRß and MYH10 while reducing ERK1/2 phosphorylation, thus phenocopying genetic PTP1B deletion. CONCLUSION: Chronic reduction of PTP1B in SMCs promotes dedifferentiation, perivascular fibrosis, and adverse remodeling following vascular injury by mechanisms involving an ERK1/2 phosphorylation-driven shift from SMAD2 to KLF4-regulated gene transcription.


Subject(s)
Muscle, Smooth, Vascular , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Vascular System Injuries , Animals , Cells, Cultured , Fibrosis , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Recombinases/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
3.
J Am Heart Assoc ; 10(6): e018322, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33666096

ABSTRACT

Background Brain-derived neurotrophic factor (BDNF) is expressed in neuronal and nonneuronal cells and may affect vascular functions via its receptor, tropomyosin-related kinase B (TrkB). In this study, we determined the expression of BDNF in different perivascular adipose tissue (PVAT) depots of patients with established coronary atherosclerosis. Methods and Results Serum, vascular tissue, and PVAT surrounding the proximal aorta (C-PVAT) or internal mammary artery (IMA-PVAT) was obtained from 24 patients (79% men; mean age, 71.7±9.7 years; median body mass index, 27.4±4.8 kg/m2) with coronary atherosclerosis undergoing elective coronary artery bypass surgery. BDNF protein levels were significantly higher in C-PVAT compared with IMA-PVAT, independent of obesity, metabolic syndrome, or systemic biomarkers of inflammation. mRNA transcripts of TrkB, the BDNF receptor, were significantly reduced in aorta compared with IMA. Vessel wall TrkB immunosignals colocalized with cells expressing smooth muscle cell markers, and confocal microscopy and flow cytometry confirmed BDNF receptor expression in human aortic smooth muscle cells. Significantly elevated levels of protein tyrosine phosphatase 1B, a negative regulator of TrkB signaling in the brain, were also observed in C-PVAT. In vitro, inhibition of protein tyrosine phosphatase 1B blunted the effects of BDNF on smooth muscle cell proliferation, migration, differentiation, and collagen production, possibly by upregulation of low-affinity p75 neurotrophin receptors. Expression of nerve growth factor or its receptor tropomyosin-related kinase A did not differ between C-PVAT and IMA-PVAT. Conclusions Elevated expression of BDNF in parallel with local upregulation of negative regulators of neurotrophin signaling in perivascular fat and lower TrkB expression suggest that vascular BDNF signaling is reduced or lost in patients with coronary atherosclerosis.


Subject(s)
Adipose Tissue/metabolism , Aorta, Thoracic/metabolism , Atherosclerosis/genetics , Brain-Derived Neurotrophic Factor/genetics , Coronary Artery Disease/genetics , Coronary Vessels/metabolism , Gene Expression Regulation , Aged , Aorta, Thoracic/diagnostic imaging , Atherosclerosis/diagnosis , Atherosclerosis/metabolism , Biomarkers/metabolism , Brain-Derived Neurotrophic Factor/biosynthesis , Cell Movement , Cell Proliferation , Coronary Artery Disease/diagnosis , Coronary Artery Disease/metabolism , Coronary Vessels/diagnostic imaging , Female , Flow Cytometry , Follow-Up Studies , Humans , Immunohistochemistry , Male , Microscopy, Confocal , RNA/genetics , RNA/metabolism , Retrospective Studies , Signal Transduction
4.
Article in English | MEDLINE | ID: mdl-33503577

ABSTRACT

3-iodothyronamine (3-T1AM) has been suggested as a novel chemical messenger and potent trace amine-associated receptor 1 ligand in the CNS that occurs naturally as endogenous metabolite of the thyroid hormones. Discrepancies and variations in 3-T1AM plasma and tissue concentrations have nonetheless caused controversy regarding the existence and biological role of 3-T1AM. These discussions are at least partially based on potential analytical artefacts caused by differential decay kinetics of 3-T1AM and the widely used deuterated quantification standard D4-T1AM. Here, we report a novel LC-MS/MS method for the quantification of 3-T1AM in biological specimens using stable isotope dilution with 13C6-T1AM, a new internal standard that showed pharmacodynamic properties comparable to endogenous 3-T1AM. The method detection limit (MDL) and method quantification limit (MQL) of 3-T1AM were 0.04 and 0.09 ng/g, respectively. The spike-recoveries of 3-T1AM were between 85.4% and 94.3%, with a coefficient of variation of 3.7-5.8%. The intra-day and inter-day variations of 3-T1AM were 8.45-11.2% and 3.58-5.73%, respectively. Endogenous 3-T1AM liver values in C57BL/6J mice were 2.20 ± 0.49 pmol/g with a detection frequency of 50%. Higher liver 3-T1AM values were found when C57BL/6J mice were treated with N-acetyl-3-iodothyronamine or O-acetyl-3-iodothyronamine. Overall, our new stable isotope dilution LC-MS/MS method improves both the sensitivity and selectivity compared with existing methods. The concomitant possibility to quantify additional thyroid hormones such as thyroxine, 3,5,3'-triiodo-L-thyronine, 3,3',5'-triiodo-L-thyronine, 3,3'-diiodo-L-thyronine, and 3,5-diiodo-L-thyronine further adds to the value of our novel method in exploring the natural occurrence and fate of 3-T1AM in biological tissues and fluids.


Subject(s)
Chromatography, High Pressure Liquid/methods , Liver/chemistry , Tandem Mass Spectrometry/methods , Thyronines/analysis , Animals , Female , Limit of Detection , Linear Models , Liver/metabolism , Mice , Mice, Inbred C57BL , Reproducibility of Results , Thyronines/pharmacokinetics
5.
Cell Rep ; 33(5): 108351, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33147454

ABSTRACT

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.


Subject(s)
Adipose Tissue, Brown/physiology , Glucose/metabolism , Thermogenesis/physiology , Animals , Energy Metabolism/physiology , Female , Homeostasis , Humans , Male , Metabolomics , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Physical Conditioning, Animal , Pregnancy , Temperature , Uncoupling Protein 1/deficiency , Uncoupling Protein 1/metabolism
6.
Eur Thyroid J ; 9(2): 57-66, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32257954

ABSTRACT

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.

7.
Thyroid ; 30(8): 1205-1216, 2020 08.
Article in English | MEDLINE | ID: mdl-32188347

ABSTRACT

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.


Subject(s)
Cardiomegaly/metabolism , Fever/metabolism , Heart Rate , Hypothyroidism/complications , Thyroid Hormones/metabolism , Animals , Cardiomegaly/prevention & control , Crosses, Genetic , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Glycogen/metabolism , Lipolysis , Liver/metabolism , Male , Mental Retardation, X-Linked/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/metabolism , Muscular Atrophy/metabolism , Phenotype , Telemetry , Thermogenesis , Thermography , Time Factors , Triiodothyronine/blood
8.
J Mol Endocrinol ; 62(3): 91-99, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30608905

ABSTRACT

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.


Subject(s)
Aorta/drug effects , Hyperthyroidism/blood , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormones/blood , Adrenergic Agonists/pharmacology , Animals , Hyperthyroidism/metabolism , Male , Mice , Mice, Knockout , Phenylephrine/pharmacology , Signal Transduction/drug effects , Thyroid Hormone Receptors beta/genetics , Triiodothyronine/pharmacology , Vasoconstriction/drug effects , Vasoconstriction/genetics
9.
Int J Mol Sci ; 21(1)2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31906225

ABSTRACT

Cardiovascular risk factors may act by modulating the composition and function of the adventitia. Here we examine how age affects perivascular adipose tissue (PVAT) and its paracrine activities during neointima formation. Aortic tissue and PVAT or primary aortic smooth muscle cells from male C57BL/6JRj mice aged 52 weeks ("middle-aged") were compared to tissue or cells from mice aged 16 weeks ("adult"). Vascular injury was induced at the carotid artery using 10% ferric chloride. Carotid arteries from the middle-aged mice exhibited smooth muscle de-differentiation and elevated senescence marker expression, and vascular injury further aggravated media and adventitia thickening. Perivascular transplantation of PVAT had no effect on these parameters, but age-independently reduced neointima formation and lumen stenosis. Quantitative PCR analysis revealed a blunted increase in senescence-associated proinflammatory changes in perivascular tissue compared to visceral adipose tissue and higher expression of mediators attenuating neointima formation. Elevated levels of protein inhibitor of activated STAT1 (PIAS1) and lower expression of STAT1- or NFκB-regulated genes involved in adipocyte differentiation, inflammation, and apoptosis/senescence were present in mouse PVAT, whereas PIAS1 was reduced in the PVAT of patients with atherosclerotic vessel disease. Our findings suggest that age affects adipose tissue and its paracrine vascular activities in a depot-specific manner. PIAS1 may mediate the age-independent vasculoprotective effects of perivascular fat.


Subject(s)
Adipose Tissue/metabolism , Aging/metabolism , Carotid Arteries/metabolism , Carotid Artery Diseases/metabolism , Carotid Artery Injuries/metabolism , Neointima/metabolism , Paracrine Communication , Adipose Tissue/pathology , Aging/genetics , Aging/pathology , Animals , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Humans , Mice , Mice, Mutant Strains , Neointima/genetics , Neointima/pathology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
10.
Nutr Diabetes ; 8(1): 9, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29549244

ABSTRACT

Stimulation of thermogenic pathways appears to be a promising approach to find new ways of tackling metabolic diseases like obesity and diabetes mellitus type 2. Thermogenic, weight reducing and insulin sensitizing effects of phosphodiesterase 5 (PDE 5) inhibitors have recently been postulated, suggesting that modulators of endogenous cGMP signaling have the therapeutic potential to treat metabolic disorders. However, most studies have been performed in vitro or in animals that were not glucose intolerant. We, thus, aimed to test the metabolic effects of the PDE 5 inhibitor sildenafil by treating diet-induced obese (DIO) mice orally for 8 days. Surprisingly, our results revealed no changes in body temperature, brown adipose tissue (BAT) thermogenesis and gene expression in BAT and inguinal white adipose tissue (iWAT), thus excluding a thermogenic or 'browning' effect of sildenafil in preexisting obesity. In contrast, sildenafil-treated DIO mice displayed changes in liver metabolism and glucose homeostasis resulting in impaired glucose tolerance (P < 0.05), demonstrating for the first time an unfavorable metabolic effect of increased hepatic cGMP signaling in obesity. As sildenafil is commonly prescribed to treat pulmonary arterial hypertension and erectile dysfunction in diabetic and/or obese patients, follow up studies are urgently required to re-evaluate the drug safety.


Subject(s)
Adipose Tissue/drug effects , Blood Glucose/metabolism , Glucose Intolerance/chemically induced , Liver/drug effects , Obesity/metabolism , Sildenafil Citrate/adverse effects , Thermogenesis/drug effects , Adipose Tissue/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Cyclic GMP/metabolism , Erectile Dysfunction/drug therapy , Glucose Intolerance/blood , Homeostasis , Hypertension/drug therapy , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Obese , Obesity/drug therapy , Phosphodiesterase 5 Inhibitors/adverse effects , Phosphodiesterase 5 Inhibitors/therapeutic use , Signal Transduction , Sildenafil Citrate/therapeutic use
11.
J Endocrinol ; 235(2): 111-122, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28851749

ABSTRACT

Nesfatin-1 is a bioactive polypeptide expressed both in the brain and peripheral tissues and involved in the control of energy balance by reducing food intake. Central administration of nesfatin-1 significantly increases energy expenditure, as demonstrated by a higher dry heat loss; yet, the mechanisms underlying the thermogenic effect of central nesfatin-1 remain unknown. Therefore, in this study, we sought to investigate whether the increase in energy expenditure induced by nesfatin-1 is mediated by the central melanocortin pathway, which was previously reported to mediate central nesfatin-1´s effects on feeding and numerous other physiological functions. With the application of direct calorimetry, we found that intracerebroventricular nesfatin-1 (25 pmol) treatment increased dry heat loss and that this effect was fully blocked by simultaneous administration of an equimolar dose of the melanocortin 3/4 receptor antagonist, SHU9119. Interestingly, the nesfatin-1-induced increase in dry heat loss was positively correlated with body weight loss. In addition, as assessed with thermal imaging, intracerebroventricular nesfatin-1 (100 pmol) increased interscapular brown adipose tissue (iBAT) as well as tail temperature, suggesting increased heat production in the iBAT and heat dissipation over the tail surface. Finally, nesfatin-1 upregulated pro-opiomelanocortin and melanocortin 3 receptor mRNA expression in the hypothalamus, accompanied by a significant increase in iodothyronine deiodinase 2 and by a nonsignificant increase in uncoupling protein 1 and peroxisome proliferator-activated receptor gamma coactivator-1 alpha mRNA in the iBAT. Overall, we clearly demonstrate that nesfatin-1 requires the activation of the central melanocortin system to increase iBAT thermogenesis and, in turn, overall energy expenditure.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Melanocortins/metabolism , Nerve Tissue Proteins/metabolism , Thermogenesis/physiology , Animals , Biomarkers , Calcium-Binding Proteins/genetics , DNA-Binding Proteins/genetics , Ear , Hypothalamus/metabolism , Male , Melanocyte-Stimulating Hormones/pharmacology , Nerve Tissue Proteins/genetics , Nucleobindins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Melanocortin/antagonists & inhibitors , Receptors, Melanocortin/genetics , Receptors, Melanocortin/metabolism , Tail , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
12.
Endocrinology ; 158(6): 1977-1984, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28368510

ABSTRACT

3-Iodothyronamine (3-T1AM) is an endogenous thyroid hormone (TH)-derived metabolite that induces severe hypothermia in mice after systemic administration; however, the underlying mechanisms have remained enigmatic. We show here that the rapid 3-T1AM-induced loss in body temperature is a consequence of peripheral vasodilation and subsequent heat loss (e.g., over the tail surface). The condition is subsequently intensified by hypomotility and a lack of brown adipose tissue activation. Although the possible 3-T1AM targets trace amine-associated receptor 1 or α2a-adrenergic receptor were detected in tail artery and aorta respectively, myograph studies did not show any direct effect of 3-T1AM on vasodilation, suggesting that its actions are likely indirect. Intracerebroventricular application of 3-T1AM, however, replicated the phenotype of tail vasodilation and body temperature decline and led to neuronal activation in the hypothalamus, suggesting that the metabolite causes tail vasodilation through a hypothalamic signaling pathway. Consequently, the 3-T1AM response constitutes anapyrexia rather than hypothermia and closely resembles the heat-stress response mediated by hypothalamic temperature-sensitive neurons. Our results thus underline the well-known role of the hypothalamus as the body's thermostat and suggest an additional molecular link between TH signaling and the central control of body temperature.


Subject(s)
Brain/physiology , Tail/blood supply , Thyronines/pharmacology , Vasodilation/drug effects , Animals , Body Temperature Regulation/drug effects , Brain/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Infusions, Intraventricular , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Tail/drug effects , Thyronines/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...