Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Int J Mol Sci ; 23(19)2022 Oct 08.
Article in English | MEDLINE | ID: mdl-36233235

ABSTRACT

Adult-onset hypothyroidism is associated with learning and cognitive dysfunctions, which may be related to alterations in synaptic plasticity. Local reduced levels of thyroid hormones (THs) may impair glia morphology and activity, and promote the increase of pro-inflammatory cytokine levels mainly in the hippocampus. Given that neuroinflammation induces memory impairments, hypothyroidism-related glia dysfunction may participate in brain disorders. Thus, we investigated the mechanisms linking hypothyroidism and neuroinflammation, from a protective perspective. We induced hypothyroidism in adult C57BL/6J and wild-derived WSB/EiJ male mice by a seven-week propylthiouracil (PTU) treatment. We previously showed that WSB/EiJ mice were resistant to high-fat diet (HFD)-induced obesity, showing no neuroinflammatory response through adaptive abilities, unlike C57BL/6J. As PTU and HFD treatments are known to induce comparable inflammatory responses, we hypothesized that WSB/EiJ mice might also be protected against hypothyroidism-induced neuroinflammation. We showed that hypothyroid WSB/EiJ mice depicted no hippocampal neuroinflammatory response and were able to maintain their hippocampal thyroid signalling despite low circulatisng TH levels. In contrast, C57BL/6J mice exhibited disturbed hippocampal TH signalling, accompanied by neuroinflammation and memory impairment. Our results reinforce the preponderance of the hippocampal TH regulatory system over TH circulating levels in the hippocampal glial reactivity.


Subject(s)
Hypothyroidism , Propylthiouracil , Animals , Cytokines , Hippocampus , Hypothyroidism/complications , Male , Mice , Mice, Inbred C57BL , Neuroglia , Propylthiouracil/pharmacology , Thyroid Hormones
2.
Opt Lett ; 47(19): 4865-4868, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36181136

ABSTRACT

We demonstrate a method to image an object using a self-probing approach based on semiconductor high-harmonic generation. On the one hand, ptychography enables high-resolution imaging from the coherent light diffracted by an object. On the other hand, high-harmonic generation from crystals is emerging as a new source of extreme-ultraviolet ultrafast coherent light. We combine these two techniques by performing ptychography measurements with nanopatterned crystals serving as the object as well as the generation medium of the harmonics. We demonstrate that this strong field in situ approach can provide structural information about an object. With the future developments of crystal high harmonics as a compact short-wavelength light source, our demonstration can be an innovative approach for nanoscale imaging of photonic and electronic devices in research and industry.

3.
RMD Open ; 8(2)2022 06.
Article in English | MEDLINE | ID: mdl-35701010

ABSTRACT

BACKGROUND: Considering non-classical environmental risk factors for osteoarthritis (OA), a systematic literature review (SLR) was performed to summarise existing knowledge on associations between OA and pollutants. METHODS: PubMed was used to identify studies reporting data on OA and pollutants in humans (examples of MeSH terms: "Pesticides" or "Polychlorinated Biphenyls" or 'Lead'). Reports included epidemiological clinical studies, pollutant assessments in ex vivo OA joint, and in vitro effects of pollutants on chondrocytes. RESULTS: Among the 193 potentially relevant articles, 14 were selected and combined with 9 articles obtained by manual search. Among these 23 articles there were: (1) 11 epidemiological studies on the relationship between OA and pollutants exposure, (2) 8 on pollutant concentrations in ex vivo OA joint, (3) 4 on the in vitro effects of pollutants on human chondrocytes. Epidemiological studies investigating mainly chlorinated and fluorinated pollutants suggested a possible link with OA. In cross-sectional studies, radiographic knee OA prevalence increased with higher serum lead levels. There was also a relationship between serum lead levels and serum/urine joint biomarkers. A high concentration of heavy metals in the cartilage tidemark was found in ex vivo joints. In vitro, the viability of chondrocytes was reduced in presence of some pollutants. However, the level of knowledge currently remains low, justifying the need for new methodologically sound studies. CONCLUSIONS: This SLR supports the hypothesis of a possible involvement of pollutants in OA disease risk. Large-scale epidemiological and biological studies and ideally big-data analysis are needed to confirm that pollutants could be risk factors for OA.


Subject(s)
Environmental Pollutants , Osteoarthritis, Knee , Biomarkers , Cross-Sectional Studies , Environmental Pollutants/toxicity , Humans , Lead , Risk Factors
4.
Sci Rep ; 9(1): 19696, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31873127

ABSTRACT

Energy imbalance due to excess of calories is considered to be a major player in the current worldwide obesity pandemic and could be accompanied by systemic and central inflammation and mitochondrial dysfunctions. This hypothesis was tested by comparing the wild-derived diet-induced obesity- (DIO-) resistant mouse strain WSB/EiJ to the obesity-prone C57BL/6J strain. We analysed circulating and hypothalamic markers of inflammatory status and hypothalamic mitochondrial activity in both strains exposed to high-fat diet (HFD). We further analysed the regulations of hypothalamic genes involved in inflammation and mitochondrial pathways by high throughput microfluidic qPCR on RNA extracted from laser micro-dissected arcuate (ARC) and paraventricular (PVN) hypothalamic nuclei. HFD induced increased body weight gain, circulating levels of leptin, cholesterol, HDL and LDL in C57BL/6J whereas WSB/EiJ mice displayed a lower inflammatory status, both peripherally (lower levels of circulating cytokines) and centrally (less activated microglia in the hypothalamus) as well as more reactive mitochondria in the hypothalamus. The gene expression data analysis allowed identifying strain-specific hypothalamic metabolic pathways involved in the respective responses to HFD. Our results point to the involvement of hypothalamic inflammatory and mitochondrial pathways as key factors in the control of energy homeostasis and the resistance to DIO.


Subject(s)
Inflammation/metabolism , Mitochondria/metabolism , Obesity/etiology , Obesity/metabolism , Animals , Cytokines/blood , Diet, High-Fat/adverse effects , Disease Models, Animal , Energy Metabolism , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation/genetics , Inflammation Mediators/metabolism , Leptin/blood , Lipid Metabolism , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Mitochondrial Dynamics , Obesity/genetics , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Species Specificity , Transcriptome
5.
Int J Mol Sci ; 19(7)2018 Jul 11.
Article in English | MEDLINE | ID: mdl-29997323

ABSTRACT

The obesity epidemic is well recognized as a significant global health issue. A better understanding of the energy homeostasis mechanisms could help to identify promising anti-obesity therapeutic strategies. It is well established that the hypothalamus plays a pivotal role governing energy balance. The hypothalamus consists of tightly interconnected and specialized neurons that permit the sensing and integration of several peripheral inputs, including metabolic and hormonal signals for an appropriate physiological response. Current evidence shows that thyroid hormones (THs) constitute one of the key endocrine factors governing the regulation and the integration of metabolic homeostasis at the hypothalamic level. THs modulate numerous genes involved in the central control of metabolism, as TRH (Thyrotropin-Releasing Hormone) and MC4R (Melanocortin 4 Receptor). THs act through their interaction with thyroid hormone receptors (TRs). Interestingly, TH signaling, especially regarding metabolic regulations, involves TRs crosstalk with other metabolically linked nuclear receptors (NRs) including PPAR (Peroxisome proliferator-activated receptor) and LXR (Liver X receptor). In this review, we will summarize current knowledge on the important role of THs integration of metabolic pathways in the central regulation of metabolism. Particularly, we will shed light on the crosstalk between TRs and other NRs in controlling energy homeostasis. This could be an important track for the development of attractive therapeutic compounds.


Subject(s)
Hypothalamus/metabolism , Obesity/metabolism , Receptors, Thyroid Hormone/metabolism , Thyroid Hormones/metabolism , Animals , Energy Metabolism , Homeostasis , Humans , Receptor, Melanocortin, Type 4/metabolism , Thyrotropin-Releasing Hormone/metabolism
7.
PLoS One ; 9(9): e106983, 2014.
Article in English | MEDLINE | ID: mdl-25229406

ABSTRACT

Reversing the escalating rate of obesity requires increased knowledge of the molecular mechanisms controlling energy balance. Liver X receptors (LXRs) and thyroid hormone receptors (TRs) are key physiological regulators of energetic metabolism. Analysing interactions between these receptors in the periphery has led to a better understanding of the mechanisms involved in metabolic diseases. However, no data is available on such interactions in the brain. We tested the hypothesis that hypothalamic LXR/TR interactions could co-regulate signalling pathways involved in the central regulation of metabolism. Using in vivo gene transfer we show that LXR activation by its synthetic agonist GW3965 represses the transcriptional activity of two key metabolic genes, Thyrotropin-releasing hormone (Trh) and Melanocortin receptor type 4 (Mc4r) in the hypothalamus of euthyroid mice. Interestingly, this repression did not occur in hypothyroid mice but was restored in the case of Trh by thyroid hormone (TH) treatment, highlighting the role of the triiodothyronine (T3) and TRs in this dialogue. Using shLXR to knock-down LXRs in vivo in euthyroid newborn mice, not only abrogated Trh repression but actually increased Trh transcription, revealing a potential inhibitory effect of LXR on the Hypothalamic-Pituitary-Thyroid axis. In vivo chromatin immunoprecipitation (ChIP) revealed LXR to be present on the Trh promoter region in the presence of T3 and that Retinoid X Receptor (RXR), a heterodimerization partner for both TR and LXR, was never recruited simultaneously with LXR. Interactions between the TR and LXR pathways were confirmed by qPCR experiments. T3 treatment of newborn mice induced hypothalamic expression of certain key LXR target genes implicated in metabolism and inflammation. Taken together the results indicate that the crosstalk between LXR and TR signalling in the hypothalamus centres on metabolic and inflammatory pathways.


Subject(s)
Hypothalamus/metabolism , Orphan Nuclear Receptors/metabolism , Thyroid Gland/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Chromatin Immunoprecipitation , Female , Liver X Receptors , Mice , Orphan Nuclear Receptors/genetics , Pregnancy , Protein Binding , Thyrotropin-Releasing Hormone/genetics
8.
PLoS One ; 9(4): e95064, 2014.
Article in English | MEDLINE | ID: mdl-24747825

ABSTRACT

Mammalian thyroid hormone receptors (TRs) have multiple isoforms, including the bona fide receptors that bind T3 (TRα1, TRß1 and TRß2) and a non-hormone-binding variant, TRα2. Intriguingly, TRα2 is strongly expressed in the brain, where its mRNA levels exceed those of functional TRs. Ablation of TRα2 in mice results in over-expression of TRα1, and a complex phenotype with low levels of free T3 and T4, without elevated TSH levels, suggesting an alteration in the negative feedback at the hypothalamic-pituitary level. As the hypothesis of a potential TRH response defect has never been tested, we explored the functional role of TRα2 in negative feedback on transcription of hypothalamic thyrotropin, Trh. The in vivo transcriptional effects of TRα2 on hypothalamic Trh were analysed using an in vivo reporter gene approach. Effects on Trh-luc expression were examined to that of two, T3 positively regulated genes used as controls. Applying in vivo gene transfer showed that TRα2 over-expression in the mouse hypothαlamus abrogates T3-dependent repression of Trh and T3 activation of positively regulated promoters, blocking their physiological regulation. Surprisingly, loss of function studies carried out by introducing a shTRα2 construct in the hypothalamus also blocked physiological T3 dependent regulation. Thus, modulating hypothalamic TRα2 expression by either gain or loss of function abrogated T3 dependent regulation of Trh transcription, producing constant transcriptional levels insensitive to feedback. This loss of physiological regulation was reflected at the level of the endogenous Trh gene, were gain or loss of function held mRNA levels constant. These results reveal the as yet undescribed dominant negative role of TRα2 over TRα1 effect on hypothalamic Trh transcription.


Subject(s)
Hypothalamus/metabolism , Thyroid Hormone Receptors alpha/physiology , Transcription, Genetic , Animals , Mice , Polymerase Chain Reaction , Thyroid Hormone Receptors alpha/genetics
9.
Mol Cell Endocrinol ; 381(1-2): 115-23, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-23896434

ABSTRACT

How Retinoid X receptors (RXR) and thyroid hormone receptors (TR) interact on negative TREs and whether RXR subtype specificity is determinant in such regulations is unknown. In a set of functional studies, we analyzed RXR subtype effects in T3-dependent repression of hypothalamic thyrotropin-releasing hormone (Trh). Two-hybrid screening of a hypothalamic paraventricular nucleus cDNA bank revealed specific, T3-dependent interaction of TRs with RXRß. In vivo chromatin immuno-precipitation showed recruitment of RXRs to the TRE-site 4 region of the Trh promoter in the absence of T3. In vivo overexpression of RXRα in the mouse hypothalamus heightened T3-independent Trh transcription, whereas RXRß overexpression abrogated this activity. Loss of function of RXRα and ß by shRNAs induced inverse regulations. Thus, RXRα and RXRß display specific roles in modulating T3-dependent regulation of Trh. These results provide insight into the actions of these different TR heterodimerization partners within the context of a negatively regulated gene.


Subject(s)
Retinoid X Receptor alpha/metabolism , Retinoid X Receptor beta/metabolism , Thyrotropin-Releasing Hormone/genetics , Transcription, Genetic , Animals , Gene Expression Regulation , Male , Mice , Mice, Nude , Paraventricular Hypothalamic Nucleus/metabolism , Promoter Regions, Genetic , Retinoid X Receptor alpha/genetics , Retinoid X Receptor beta/genetics , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Thyrotropin-Releasing Hormone/metabolism , Triiodothyronine/physiology
10.
Mol Ther Nucleic Acids ; 2: e89, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23612115

ABSTRACT

RNA interference (RNAi) is a major tool for basic and applied investigations. However, obtaining RNAi data that have physiological significance requires investigation of regulations and therapeutic strategies in appropriate in vivo settings. To examine in vivo gene regulation and protein function in the adult neural stem cell (NSC) niche, we optimized a new non-viral vector for delivery of siRNA into the subventricular zone (SVZ). This brain region contains the neural stem and progenitor cells populations that express the stem cell marker, SOX2. Temporally and spatially controlled Sox2 knockdown was achieved using the monocationic lipid vector, IC10. siRNA/IC10 complexes were stable over time and smaller (<40 nm) than jetSi complexes (≈400 nm). Immunocytochemistry showed that siRNA/IC10 complexes efficiently target both the progenitor and stem cell populations in the adult SVZ. Injection of the complexes into the lateral brain ventricle resulted in specific knockdown of Sox2 in the SVZ. Furthermore, IC10-mediated transient in vivo knockdown of Sox2-modulated expression of several genes implicated in NSC maintenance. Taken together, these data show that IC10 cationic lipid formulation can efficiently vectorize siRNA in a specific area of the adult mouse brain, achieving spatially and temporally defined loss of function.Molecular Therapy-Nucleic Acids (2013) 2, e89; doi:10.1038/mtna.2013.8; published online 23 April 2013.

11.
Endocr Res ; 38(1): 15-23, 2013.
Article in English | MEDLINE | ID: mdl-22746188

ABSTRACT

OBJECTIVES: Although a relationship between obesity and metabolic consequences with thyroid function has been reported, the underlying pathogenesis is not completely known. In the current study, we evaluated the thyroid function in obese and/or diabetic patients compared to healthy normal weight peers, exploring the possible association between components of metabolic syndrome and thyroid function parameters. METHODS: We recruited 108 subjects (56 male and 52 female). In all subjects, thyroid stimulating hormone (TSH), free thyroxine (FT4), fasting plasma levels of insulin and glucose, homeostasis model assessment for insulin resistance, and obesity parameters were assessed. RESULTS: We found that circulating levels of TSH and FT4 were significantly increased in overweight and obese subjects. However, the data do not reveal any change of these hormones in diabetics. Multivariate linear regression analysis showed that TSH was directly associated with both obesity and insulin resistance parameters (p < 0.05). FT4 was negatively associated only with obesity parameters (p < 0.05). CONCLUSIONS: Our data strongly support that the changes of thyroid hormones may be influenced by adiposity and its metabolic consequences, such as insulin resistance. This relationship can be explained by a cross talk between adipose tissue release and thyroid function. Nevertheless, metformin treatment seems to affect thyroid function in diabetic patients by maintaining plasma thyrotropin levels to subnormal levels.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Obesity/physiopathology , Thyroid Gland/physiopathology , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Female , Humans , Insulin/blood , Insulin Resistance , Male , Metabolic Syndrome , Middle Aged , Obesity/blood , Thyroid Function Tests , Thyrotropin/blood , Thyroxine/blood , Tunisia
12.
Acta Diabetol ; 50(2): 227-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-21604201

ABSTRACT

Cellular resistance to insulin caused by reduced glucose transport and metabolism is a primary defect leading to the development of metabolic disease. While the etiology of insulin resistance is multifactorial, reduced insulin action is associated with impaired activity of the glucose transporter GLUT4 in insulin-sensitive tissues. Yet, the role of adipose tissue GLUT4 deregulation in the pathogenesis of insulin resistance, obesity, and diabetes is still unclear. In this study, we assessed the relative GLUT4 level in human subcutaneous adipose tissue from obese, diabetic, and diabetic obese versus control subjects, using a real-time PCR method. GLUT4 mRNA levels were considerably decreased among type 2 diabetic patients compared with those of the controls (P < 0.01), whereas no such difference was found between obese and normal-weight controls. Multiple linear regressions analysis in both diabetic non-obese and diabetic obese groups showed a negative correlation between GLUT4 mRNA expression and both markers of obesity or insulin resistance (P < 0.01). However, in obese group, GLUT4 was inversely associated only with HOMA-IR (P < 0.01). Our findings showed that adipose GLUT4 gene expression changes were more related to insulin resistance and type 2 diabetes rather than to obesity.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Gene Expression , Glucose Transporter Type 4/genetics , Obesity/metabolism , Subcutaneous Fat/metabolism , Body Mass Index , Female , Humans , Insulin Resistance , Linear Models , Male , Middle Aged , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Subcutaneous Fat/chemistry
13.
Tunis Med ; 88(8): 569-72, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20711963

ABSTRACT

BACKGROUND: The incidence of obesity has dramatically increased in overall the world. It is a consequence of imbalance between energy intake and energy expenditure. Leptin is a fat derived adipokine that has emerged over the past decade as a key hormone in the regulation of food intake and energy expenditure. Elevated leptin levels are found in obese humans, suggesting a role of leptin in regulating body weight and adiposity. AIM: The aim of this study was to investigate the change of leptin mRNA expression level and its correlation with obesity and several metabolic variables in Tunisian patients. METHODS: Real time quantitative polymerase chain reaction (QPCR) analysis was carried out among two groups who underwent an abdominal surgery: controls (n = 9) and obese patients (n = 7). RESULTS: Leptin mRNA expression in subcutaneous adipose tissue was markedly increased in obese patients (p < 0.01). It was positively correlated with measures of obesity waist circumference (WC) (r = 0, 71, p < 0.01) and body mass index (BMI) (r = 0, 68, p < 0.01). Interestingly, leptin gene expression was also correlated to insulin resistance index (r = 0, 72, p < 0.01). CONCLUSION: The present study is the first investigation of leptin regulation in subcutaneous adipose tissue of Tunisian population. Our data showed that leptin levels are higher in obese subjects than in control subjects. This indicates that the subcutaneous adipose plays an important role in impaired adipokine regulation, and consequently in developing metabolic disorder.


Subject(s)
Leptin , Obesity/genetics , Obesity/metabolism , RNA, Messenger , Subcutaneous Fat/metabolism , Body Mass Index , Humans , Leptin/blood , Leptin/genetics , Leptin/physiology , Middle Aged , Obesity/blood , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tunisia , Waist Circumference
14.
Mol Cell Endocrinol ; 323(2): 172-82, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20399831

ABSTRACT

The hypothalamus integrates metabolic and endocrine signals. As such it represents a potential target for a wide spectrum of endocrine disrupting chemicals (EDCs). We investigated hypothalamic effects of two environmentally abundant xenobiotics, the flame-retardant tetrabromo bisphenol A (TBBPA) and the anti-fouling agent tributyltin (TBT). These EDCs affect endocrine signalling through different nuclear receptors including the thyroid hormone receptor (TR) or its partner, the retinoid X receptor (RXR). Promoter sequences of two hypothalamic genes implicated in metabolic control and regulated by thyroid hormone, thyrotropin-releasing hormone (Trh) and type 4 melanocortin receptor (Mc4r), were studied in vivo using reporter assays. Chronic exposure of gestating dams or acute exposure of their newborns to TBBPA abrogated activation of both Trh and Mc4r transcription. Exposure of lactating dams to TBT amplified activation of Trh without affecting Mc4r transcription. Thus, perinatal exposure to EDCs affecting nuclear receptor signalling modulates hypothalamic set-points controlling metabolic responses.


Subject(s)
Environmental Pollutants/pharmacology , Estrogens, Non-Steroidal/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Phenols/pharmacology , Thyroid Hormones/metabolism , Trialkyltin Compounds/pharmacology , Animals , Animals, Newborn , Benzhydryl Compounds , Female , Genes, Reporter , Homeostasis , Mice , Pregnancy , Promoter Regions, Genetic , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Signal Transduction/drug effects , Thyroid Hormones/genetics , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism , Transcription, Genetic
15.
Proc Natl Acad Sci U S A ; 107(9): 4471-6, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20160073

ABSTRACT

The type 4 melanocortin receptor MC4R, a key relay in leptin signaling, links central energy control to peripheral reserve status. MC4R activation in different brain areas reduces food intake and increases energy expenditure. Mice lacking Mc4r are obese. Mc4r is expressed by hypothalamic paraventricular Thyrotropin-releasing hormone (TRH) neurons and increases energy usage through activation of Trh and production of the thyroid hormone tri-iodothyronine (T(3)). These facts led us to test the hypothesis that energy homeostasis should require negative feedback by T(3) on Mc4r expression. Quantitative PCR and in situ hybridization showed hyperthyroidism reduces Mc4r mRNA levels in the paraventricular nucleus. Comparative in silico analysis of Mc4r regulatory regions revealed two evolutionarily conserved potential negative thyroid hormone-response elements (nTREs). In vivo ChIP assays on mouse hypothalamus demonstrated association of thyroid hormone receptors (TRs) with a region spanning one nTRE. Further, in vivo gene reporter assays revealed dose-dependent T(3) repression of transcription from the Mc4r promoter in mouse hypothalamus, in parallel with T(3)-dependent Trh repression. Mutagenesis of the nTREs in the Mc4r promoter demonstrated direct regulation by T(3), consolidating the ChIP results. In vivo shRNA knockdown, TR over-expression approaches and use of mutant mice lacking specific TRs showed that both TRalpha and TRbeta contribute to Mc4r regulation. T(3) repression of Mc4r transcription ensures that the energy-saving effects of T(3) feedback on Trh are not overridden by MC4R activation of Trh. Thus parallel repression by T(3) on hypothalamic Mc4r and Trh contributes to energy homeostasis.


Subject(s)
Feedback , Hypothalamus/metabolism , Receptor, Melanocortin, Type 4/genetics , Triiodothyronine/physiology , Animals , Chromatin Immunoprecipitation , In Situ Hybridization , Mice , Mice, Knockout , Polymerase Chain Reaction , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/physiology
16.
Mol Cell Endocrinol ; 317(1-2): 44-52, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-19900503

ABSTRACT

Thyroid hormone receptor (TR) and peroxisome proliferator-activated receptor gamma (PPARgamma) co-regulate numerous peripheral metabolic responses. To examine potential crosstalk between PPARgamma and TRbeta in the hypothalamus, thyrotropin-releasing hormone (Trh) regulation in the newborn mouse hypothalamus was followed. QPCR showed PPARgamma to be expressed in the hypothalamus at this developmental stage. Intracerebral injection of PPARgamma agonists modified transcription from a TRH-luc construct introduced into the hypothalamus and increased serum thyroxine levels. Furthermore, shRNA-based in vivo PPARgamma knockdown amplified T(3)-independent transcription and PPARgamma overexpression dose-dependently abrogated T(3)-dependent Trh repression. Overexpression of retinoid X receptor-alpha (RXRalpha), the common heterodimeric partner of PPARgamma and TRbeta, rescued PPARgamma abrogation of T(3)-dependent repression. Thus, competition for RXR could represent one mechanism underlying this hypothalamic crosstalk between PPARgamma and TRbeta. These demonstrations of PPARgamma effects on hypothalamic Trh transcription in vivo consolidate the role of the TRH neuron as a central integrator of energy homeostasis.


Subject(s)
Gene Expression Regulation , Hypothalamus/metabolism , PPAR gamma/metabolism , Thyrotropin-Releasing Hormone/genetics , Anilides/pharmacology , Animals , Animals, Newborn , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Hypothalamus/drug effects , Injections, Intraventricular , Mice , PPAR gamma/genetics , Pioglitazone , Promoter Regions, Genetic/genetics , Retinoid X Receptor alpha/metabolism , Rosiglitazone , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology , Thyroid Hormone Receptors beta/metabolism , Thyrotropin-Releasing Hormone/metabolism , Thyroxine/metabolism , Transfection , Triiodothyronine/pharmacology
17.
Neurosci Lett ; 406(3): 240-3, 2006 Oct 09.
Article in English | MEDLINE | ID: mdl-16930836

ABSTRACT

RNA interference mediated by small interfering RNAs (siRNAs) is a powerful tool for evaluating gene function in vivo. In particular it should be able to provide tissue-specific and developmental stage-specific knock-down of target genes in physiological contexts. However, demonstrations of its use on neuronal specific genes in vivo are lacking. We examined whether a recently developed cationic lipid based approach was applicable to study the differential effects of the two beta thyroid hormone receptor (TR) isoforms, TRbeta1 and TRbeta2, on T3-transcriptional repression of the hypothalamic gene, TRH. The cationic lipid based technique used, JetSI/DOPE, was previously shown to efficiently knock-down reporter gene mRNA in vivo. Here we now show that its use to vectorise siRNA against TRbeta1 and TRbeta2 mRNA abrogates T3-mediated repression of hypothalamic TRH transcription. In particular, when using siRNA against either TRbeta1 or TRbeta2 differential effects are revealed. siRNA directed against TRbeta1 blocks both T3 independent activation and T3 dependent modulation of TRH transcription. In contrast, siRNA directed against TRbeta2 abrogates only T3 repression of transcription. These results corroborate our previous findings obtained in mutant TRbeta(-/-) mice, showing that the TRbeta1 and TRbeta2 isoforms have differential effects on T3-TRH transcription. The data thus show that the cationic lipid-based siRNA strategy can effectively be used to reveal fine, tissue specific and isoform specific effects on neuronal gene transcription in vivo.


Subject(s)
Gene Expression Regulation/drug effects , Hypothalamus/drug effects , RNA, Small Interfering/pharmacology , Thyroid Hormone Receptors beta/physiology , Thyrotropin-Releasing Hormone/metabolism , Animals , Animals, Newborn , Drug Delivery Systems , Green Fluorescent Proteins/metabolism , Mice , Phosphatidylethanolamines/pharmacology , Thyrotropin-Releasing Hormone/genetics
18.
EMBO Rep ; 7(10): 1035-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16936638

ABSTRACT

Transcriptional control of hypothalamic thyrotropin-releasing hormone (TRH) integrates central regulation of the hypothalamo-hypophyseal-thyroid axis and hence thyroid hormone (triiodothyronine (T(3))) homeostasis. The two beta thyroid hormone receptors, TRbeta1 and TRbeta2, contribute to T(3) feedback on TRH, with TRbeta1 having a more important role in the activation of TRH transcription. How TRbeta1 fulfils its role in activating TRH gene transcription is unknown. By using a yeast two-hybrid screening of a mouse hypothalamic complementary DNA library, we identified a novel partner for TRbeta1, hepatitis virus B X-associated protein 2 (XAP2), a protein first identified as a co-chaperone protein. TR-XAP2 interactions were TR isoform specific, being observed only with TRbeta1, and were enhanced by T(3) both in yeast and mammalian cells. Furthermore, small inhibitory RNA-mediated knockdown of XAP2 in vitro affected the stability of TRbeta1. In vivo, siXAP2 abrogated specifically TRbeta1-mediated (but not TRbeta2) activation of hypothalamic TRH transcription. This study provides the first in vivo demonstration of a regulatory, physiological role for XAP2.


Subject(s)
Hypothalamus/metabolism , Proteins/metabolism , Proteins/physiology , Thyrotropin-Releasing Hormone/metabolism , Transcriptional Activation , Animals , Gene Expression Regulation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Paraventricular Hypothalamic Nucleus/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Thyroid Hormone Receptors beta/metabolism , Tissue Distribution , Transfection
19.
Exp Neurol ; 183(2): 338-45, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14552875

ABSTRACT

An endogenous antipyresis has been observed around parturition in several species, including rats. It has been proposed that the neuropeptide vasopressin is responsible for this antipyresis via an action on the V(1a) receptor subtype, but this concept is controversial. We therefore addressed the question of the regulation of V(1a) receptor expression within the rat hypothalamus around parturition, to assess its possible involvement in the antipyresis phenomenon observed at term. We analyzed V(1a) receptor mRNA and protein levels in the hypothalamus/preoptic area of female rats at Days 15 and 22 (parturition) of gestation, and at Day 5 of lactation. We used quantitative RT-PCR to assess the mRNA levels and designed a semiquantitative Western blot assay to analyze changes in protein levels between the three stages studied. No significant changes either in V(1a) receptor mRNA or protein levels were observed between the three stages, suggesting that variations in the hypothalamic V(1a) receptor expression levels alone cannot account for the endogenous antipyresis observed at term.


Subject(s)
Body Temperature/physiology , Hypothalamus/metabolism , Parturition/metabolism , Receptors, Vasopressin/metabolism , Animals , Blotting, Western , Female , Gestational Age , Pregnancy , Preoptic Area/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Vasopressin/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...