Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 371
Filtrer
1.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892044

RÉSUMÉ

Anxiety is a common comorbidity of obesity, resulting from prescribing long-term caloric restriction diets (CRDs); patients with a reduced food intake lose weight but present anxious behaviors, poor treatment adherence, and weight regain in the subsequent 5 years. Intermittent fasting (IF) restricts feeding time to 8 h during the activity phase, reducing patients' weight even with no caloric restriction; it is unknown whether an IF regime with ad libitum feeding avoids stress and anxiety development. We compared the corticosterone blood concentration between male Wistar rats fed ad libitum or calorie-restricted with all-day or IF food access after 4 weeks, along with their anxiety parameters when performing the elevated plus maze (EPM). As the amygdalar thyrotropin-releasing hormone (TRH) is believed to have anxiolytic properties, we evaluated its expression changes in association with anxiety levels. The groups formed were the following: a control which was offered food ad libitum (C-adlib) or 30% of C-adlib's energy requirements (C-CRD) all day, and IF groups provided food ad libitum (IF-adlib) or 30% of C-adlib's requirements (IF-CRD) with access from 9:00 to 17:00 h. On day 28, the rats performed the EPM and, after 30 min, were decapitated to analyze their amygdalar TRH mRNA expression by in situ hybridization and corticosterone serum levels. Interestingly, circadian feeding synchronization reduced the body weight, food intake, and animal anxiety levels in both IF groups, with ad libitum (IF-adlib) or restricted (IF-CRD) food access. The anxiety levels of the experimental groups resulted to be negatively associated with TRH expression, which supported its anxiolytic role. Therefore, the low anxiety levels induced by synchronizing feeding with the activity phase would help patients who are dieting to improve their diet therapy adherence.


Sujet(s)
Amygdale (système limbique) , Anxiété , Restriction calorique , Rythme circadien , Corticostérone , Rat Wistar , Hormone de libération de la thyréostimuline , Animaux , Anxiété/métabolisme , Rats , Mâle , Amygdale (système limbique)/métabolisme , Hormone de libération de la thyréostimuline/métabolisme , Hormone de libération de la thyréostimuline/génétique , Restriction calorique/méthodes , Corticostérone/sang , Régulation négative , Comportement alimentaire , Jeûne , Consommation alimentaire , Poids
2.
Endocr J ; 70(8): 805-814, 2023 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-37211401

RÉSUMÉ

The secretion of several hypothalamic peptide hormones is activated during the preovulatory period. Hypothalamic thyrotropin-releasing hormone (TRH) is one such hormone with reproductive and/or metabolic significance. However, it remains unclear whether thyroid-stimulating hormone (TSH)-producing thyrotrophs are produced during the preovulatory period. We previously found a transient increase in the expression of the nuclear receptor NR4A3, a well-known immediate early gene, in the proestrus afternoon in the anterior pituitary glands of rats. To investigate the relationship between TRH secretion and pituitary NR4A3 expression during proestrus, we used proestrus and thyroidectomized rats to identify NR4A3-expressing cells and examined the regulation of Nr4a3 gene expression via the hypothalamus-pituitary-thyroid (HPT) axis. The percentage of NR4A3-expressing cells increased in thyrotrophs at 14:00 h of proestrus. Incubation of rat primary pituitary cells with TRH transiently stimulated Nr4a3 expression. Thyroidectomy to attenuate the negative feedback effects led to increased serum TSH levels and Nr4a3 gene expression in the anterior pituitary, whereas thyroxine (T4) administration conversely suppressed Nr4a3 expression. Additionally, the administration of T4 or TRH antibodies significantly suppressed the increase in Nr4a3 expression at 14:00 h of proestrus. These results demonstrate that pituitary NR4A3 expression is regulated by the HPT axis, and that TRH stimulates thyrotrophs and induces NR4A3 expression during the proestrus afternoon. This suggests the potential involvement of NR4A3 in the regulation of the HPT axis during pre- and post-ovulatory periods.


Sujet(s)
Cellules thyréotropes , Hormone de libération de la thyréostimuline , Femelle , Rats , Animaux , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme , Cellules thyréotropes/métabolisme , Prooestrus , Thyréostimuline , Hypophyse/métabolisme , Thyroxine/métabolisme
3.
Endocrinology ; 164(3)2023 01 09.
Article de Anglais | MEDLINE | ID: mdl-36423209

RÉSUMÉ

In contrast to mammals, birds have a higher basal metabolic rate and undertake wide range of energy-demanding activities. As a consequence, food deprivation for birds, even for a short period, poses major energy challenge. The energy-regulating hypothalamic homeostatic mechanisms, although extensively studied in mammals, are far from clear in the case of birds. We focus on the interplay between neuropeptide Y (NPY) and thyrotropin-releasing hormone (TRH), 2 of the most important hypothalamic signaling agents, in modulating the energy balance in a bird model, the zebra finch, Taeniopygia guttata. TRH neurons were confined to a few nuclei in the preoptic area and hypothalamus, and fibers widely distributed. The majority of TRH neurons in the hypothalamic paraventricular nucleus (PVN) whose axons terminate in median eminence were contacted by NPY-containing axons. Compared to fed animals, fasting significantly reduced body weight, PVN pro-TRH messenger RNA (mRNA) and TRH immunoreactivity, but increased NPY mRNA and NPY immunoreactivity in the infundibular nucleus (IN, avian homologue of mammalian arcuate nucleus) and PVN. Refeeding for a short duration restored PVN pro-TRH and IN NPY mRNA, and PVN NPY innervation to fed levels. Compared to control tissues, treatment of the hypothalamic superfused slices with NPY or an NPY-Y1 receptor agonist significantly reduced TRH immunoreactivity, a response blocked by treatment with a Y1-receptor antagonist. We describe a detailed neuroanatomical map of TRH-equipped elements, identify new TRH-producing neuronal groups in the avian brain, and demonstrate rapid restoration of the fasting-induced suppression of PVN TRH following refeeding. We further show that NPY via Y1 receptors may regulate PVN TRH neurons to control energy balance in T. guttata.


Sujet(s)
Fringillidae , Hormone de libération de la thyréostimuline , Animaux , Mâle , Hormone de libération de la thyréostimuline/génétique , Neuropeptide Y/métabolisme , Hypothalamus/métabolisme , Noyau paraventriculaire de l'hypothalamus/métabolisme , ARN messager/métabolisme , Mammifères/génétique
4.
Int J Mol Sci ; 23(24)2022 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-36555554

RÉSUMÉ

Prolactin (PRL) is an important hormone that is secreted by the pituitary gland and plays an important role in the growth, development and reproduction of organisms. Thyrotropin-releasing hormone (TRH) is a common prolactin-releasing factor that regulates the synthesis and secretion of prolactin. In recent studies, microRNAs (miRNAs) have been found to play a key role in the regulation of pituitary hormones. However, there is a lack of systematic studies on the regulatory role that TRH plays on the pituitary transcriptome, and the role of miRNAs in the regulation of PRL synthesis and secretion by TRH lacks experimental evidence. In this study, we first investigated the changes in PRL synthesis and secretion in the rat pituitary gland after TRH administration. The results of transcriptomic analysis after TRH treatment showed that 102 genes, including those that encode Nppc, Fgf1, PRL, Cd63, Npw, and Il23a, were upregulated, and 488 genes, including those that encode Lats1, Cacna2d1, Top2a, and Tfap2a, were downregulated. These genes are all involved in the regulation of prolactin expression. The gene expression of miR-126a-5p, which regulates the level of PRL in the pituitary gland, was screened by analysis prediction software and by a dual luciferase reporter system. The data presented in this study demonstrate that TRH can regulate prolactin synthesis and secretion through miR-126a-5p, thereby improving our understanding of the molecular mechanism of TRH-mediated PRL secretion and providing a theoretical basis for the role of miRNAs in regulating the secretion of pituitary hormones.


Sujet(s)
microARN , Adénohypophyse , Animaux , Rats , microARN/génétique , microARN/métabolisme , Adénohypophyse/métabolisme , Hormones hypophysaires/métabolisme , Prolactine/génétique , Prolactine/métabolisme , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme
5.
Biomolecules ; 12(10)2022 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-36291567

RÉSUMÉ

Acute myeloid leukemia (AML) is a biologically and genetically heterogeneous hematological malignance with an unsatisfactory risk stratification system. Recently, through the novel single-cell RNA sequencing technology, we revealed heterogeneous leukemia myeloblasts in RUNX1-RUNX1T1 AML. Thyrotropin-releasing hormone (TRH), as biomarkers of CD34+CD117bri myeloblasts, were found to be prognostic in RUNX1-RUNX1T1 AML. However, the clinical and genetic features of TRH in AML patients are poorly understood. Here, with data from TCGA AML, TRH was found to be downregulated in patients older than 60 years old, with DNMT3A and NPM1 mutations, while overexpressed in patients with KIT mutations. This was further validated in three other cohorts of primary AML including Beat AML (n = 223), GSE6891 (n = 461), and GSE17855 (n = 237). Furthermore, we demonstrated that the expression of TRH in AML could be used to improve the ELN 2017 risk stratification system. In conclusion, our preliminary analysis revealed that TRH, a novel biomarker for AML patients, could be used to evaluate the survival of AML.


Sujet(s)
Sous-unité alpha 2 du facteur CBF , Leucémie aigüe myéloïde , Humains , Adulte d'âge moyen , Sous-unité alpha 2 du facteur CBF/génétique , Hormone de libération de la thyréostimuline/génétique , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Mutation , Marqueurs biologiques , Protéines nucléaires/génétique
6.
Brain Res ; 1796: 148083, 2022 12 01.
Article de Anglais | MEDLINE | ID: mdl-36108782

RÉSUMÉ

The dorsomedial nucleus of the hypothalamus (DMH) is part of the brain circuits that modulate organism responses to the circadian cycle, energy balance, and psychological stress. A large group of thyrotropin-releasing hormone (Trh) neurons is localized in the DMH; they comprise about one third of the DMH neurons that project to the lateral hypothalamus area (LH). We tested their response to various paradigms. In male Wistar rats, food restriction during adulthood, or chronic variable stress (CVS) during adolescence down-regulated adult DMH Trh mRNA levels compared to those in sedentary animals fed ad libitum; two weeks of voluntary wheel running during adulthood enhanced DMH Trh mRNA levels compared to pair-fed rats. Except for their magnitude, female responses to exercise were like those in male rats; in contrast, in female rats CVS did not change DMH Trh mRNA levels. A very strong negative correlation between DMH Trh mRNA levels and serum corticosterone concentration in rats of either sex was lost in CVS rats. CVS canceled the response to food restriction, but not that to exercise in either sex. TRH receptor 1 (Trhr) cells were numerous along the rostro-caudal extent of the medial LH. In either sex, fasting during adulthood reduced DMH Trh mRNA levels, and increased LH Trhr mRNA levels, suggesting fasting may inhibit the activity of TRHDMH->LH neurons. Thus, in Wistar rats DMH Trh mRNA levels are regulated by negative energy balance, exercise and chronic variable stress through sex-dependent and -independent pathways.


Sujet(s)
Hypothalamus , Hormone de libération de la thyréostimuline , Animaux , Femelle , Mâle , Rats , Corticostérone , Hypothalamus/métabolisme , Noyau dorsomédial du thalamus , Activité motrice , Rat Wistar , Récepteur TRH/génétique , Récepteur TRH/métabolisme , ARN messager/métabolisme , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme
7.
Environ Sci Pollut Res Int ; 29(49): 74851-74860, 2022 Oct.
Article de Anglais | MEDLINE | ID: mdl-35641749

RÉSUMÉ

We have previously reported that filtered air (FA) intervention reduces inflammation and hypothalamus-pituitary-adrenal axis activation after fine particulate matter (PM2.5 exposure). Whether FA also modulates the hypothalamic-pituitary-thyroid (HPT) and hypothalamic-pituitary-gonadal (HPG) axes in rats after PM2.5 exposure is still unknown. Adult Sprague-Dawley rats were exposed to PM2.5 by using a "real-world" PM2.5 exposure system, and the FA intervention was conducted by renewing for 15 days. PM2.5 inhalation decreased thyrotropin-releasing hormone (TRH) and thyroxine (T4) levels in both male and female rats, and thyroid-stimulating hormone (TSH) level in male rats. FA intervention attenuated the reduction in TRH and TSH levels in male rats and reduction in T4 level in female rats. PM2.5 inhalation also reduced testosterone (T) level in male rats, and estradiol (E2) and progesterone (PROG) levels in female rats, and these changes were attenuated after FA intervention. The FA intervention attenuated the decreases in CD8 T cells and T cells induced by PM2.5 inhalation in female rats only by flow cytometry analysis. In blood, FA interventions ameliorated IL-6 and IL-1ß mRNA levels in both male and female rats after PM2.5 exposure. FA intervention restored the IL-4 and IL-10 levels in female rats after PM2.5 exposure. Moreover, FA intervention ameliorated the inflammatory responses induced by PM2.5 inhalation in the thyroid and gonads in both male and female rats. These data indicate that FA intervention exerted an effect on modulating the hormonal balance of the HPT and HPG axes, and this may be related to a reduction in the inflammatory responses in the thyroid and gonads of PM2.5-treated rats, respectively.


Sujet(s)
Glande thyroide , Hormone de libération de la thyréostimuline , Animaux , Oestradiol/pharmacologie , Femelle , Gonades/composition chimique , Interleukine-10 , Interleukine-4/pharmacologie , Interleukine-6 , Mâle , Matière particulaire/pharmacologie , Progestérone , ARN messager/génétique , Rats , Rat Sprague-Dawley , Testostérone , Thyréostimuline , Hormone de libération de la thyréostimuline/analyse , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/pharmacologie , Thyroxine
8.
Gene ; 834: 146580, 2022 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-35598680

RÉSUMÉ

Thyrotropin releasing hormone degrading enzyme (TRHDE) gene is implicated in Thyrotropin releasing hormone (TRH) mediated prolactin secretion. It has been shown that the prolactin secretion alters the Gonadotropin-releasinghormone(GnRH) mediated estrous cycle. Therefore, TRHDE may also regulate postpartum anestrus. Earlier studies reported the role of non-synonymous single nucleotide polymorphism (SNPs) in various pathophysiological conditions by altering the structure and function of the proteins. Hence, in the present study, we identified SNPs in the putative promoter, first exon, middle exon and 3'-UTR containing the last exon of TRHDE gene and determined their association with postpartum anestrus (PPA) in Murrah buffaloes. We found one non synonymous SNP (G > C at 118095875 bp on chromosome 4) in the first exon of TRHDE and performed its association analysis in a population sample of 50 extreme PPA (residual PPAI: 123.06 ± 12.98 days) and 50 normal (residual PPAI: -80.46 ± 3.19 days) buffaloes. The residual PPAI value was the observed PPAI adjusted for the effect of 38 non-genetic factors. The analysis showed a significant (P < 0.004167) association of this SNP with PPA in buffaloes. Molecular dynamics simulations (MDS) also supported that the C allele altering Glutamine to Histidine at the amino acid 148 of TRHDE could enhance the stability and rigidity of TRHDE protein, which may lower its activity, increase TRH and prolactin, and reduce GnRH in PPA buffaloes. The MDS analysis further strengthens the association of the SNP (G > C) in the TRHDE gene with PPA condition in Murrah buffaloes. However, further investigation is needed to prove the MDS observations.


Sujet(s)
Anoestrus , Buffles , Animaux , Buffles/génétique , Femelle , Hormone de libération des gonadotrophines/génétique , Polymorphisme de nucléotide simple , Période du postpartum/génétique , Prolactine/génétique , Hormone de libération de la thyréostimuline/génétique
9.
Int J Neurosci ; 132(8): 787-801, 2022 Aug.
Article de Anglais | MEDLINE | ID: mdl-33080155

RÉSUMÉ

INTRODUCTION: The role of extra-hypothalamic thyrotropin-releasing hormone (TRH) has been investigated by pharmacological studies using TRH or its analogues and found to produce a wide array of effects in the central nervous system. METHODS: Immunofluorescence, In situ labeling of DNA (TUNEL), in situ hybridization chain reaction and quantitative real-time polymerase chain reaction were used in this study. RESULTS: We found that the granular cells of the dentate gyrus expressed transiently a significant amount of TRH-like immunoreactivity and TRH mRNA during the 6-24 h period following global cerebral ischemia/reperfusion injury. TUNEL showed that apoptosis of neurons in the CA1 region occurred from 48 h and almost disappeared at 7 days. TRH administration 30 min before or 24 h after the injury could partially inhibit neuronal loss, and improve the survival of neurons in the CA1 region. CONCLUSION: These data suggest that endogenous TRH expressed transiently in the dentate gyrus of the hippocampus may play an important role in the survival of neurons during the early stage of ischemia/reperfusion injury and that delayed application of TRH still produced neuroprotection. This delayed application of TRH has a promising therapeutic significance for clinical situations.


Sujet(s)
Encéphalopathie ischémique , Lésion d'ischémie-reperfusion , Animaux , Encéphalopathie ischémique/métabolisme , Hippocampe/métabolisme , Peptides/métabolisme , ARN messager/métabolisme , Rats , Lésion d'ischémie-reperfusion/métabolisme , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme
10.
Genes (Basel) ; 12(11)2021 11 18.
Article de Anglais | MEDLINE | ID: mdl-34828419

RÉSUMÉ

Midbrain raphe nuclei (MRNs) contain a large number of serotonergic neurons associated with the regulation of numerous types of psychoemotional states and physiological processes. The aim of this work was to study alterations of the MRN transcriptome in mice with prolonged positive or negative fighting experience and to identify key gene networks associated with the regulation of serotonergic system functioning. Numerous genes underwent alterations of transcription in the MRNs of male mice that either manifested aggression or experienced social defeat in daily agonistic interactions. The expression of the Tph2 gene encoding the rate-limiting enzyme of the serotonin synthesis pathway correlated with the expression of many genes, 31 of which were common between aggressive and defeated mice and were downregulated in the MRNs of mice of both experimental groups. Among these common differentially expressed genes (DEGs), there were genes associated with behavior, learning, memory, and synaptic signaling. These results suggested that, in the MRNs of the mice, the transcriptome changes associated with serotonergic regulation of various processes are similar between the two groups (aggressive and defeated). In the MRNs, more DEGs correlating with Tph2 expression were found in defeated mice than in the winners, which is probably a consequence of deeper Tph2 downregulation in the losers. It was shown for the first time that, in both groups of experimental mice, the changes in the transcription of genes controlling the synthesis and transport of serotonin directly correlate with the expression of genes Crh and Trh, which control the synthesis of corticotrophin- and thyrotropin-releasing hormones. Our findings indicate that CRH and TRH locally produced in MRNs are related to serotonergic regulation of brain processes during a chronic social conflict.


Sujet(s)
Agressivité , Corticolibérine/métabolisme , Noyaux du raphé/métabolisme , Sérotonine/biosynthèse , Défaite sociale , Hormone de libération de la thyréostimuline/métabolisme , Animaux , Corticolibérine/génétique , Mâle , Souris , Souris de lignée C57BL , Sérotonine/génétique , Hormone de libération de la thyréostimuline/génétique , Tryptophane 5-monooxygenase/génétique , Tryptophane 5-monooxygenase/métabolisme
11.
Mol Pharmacol ; 100(3): 203-216, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34158361

RÉSUMÉ

Norepinephrine (NE) controls many vital body functions by activating adrenergic receptors (ARs). Average core body temperature (CBT) in mice is 37°C. Of note, CBT fluctuates between 36 and 38°C within 24 hours, but little is known about the effects of CBT changes on the pharmacodynamics of NE. Here, we used Peltier element-controlled incubators and challenged murine hypothalamic mHypoA -2/10 cells with temperature changes of ±1°C. We observed enhanced NE-induced activation of a cAMP-dependent luciferase reporter at 36 compared with 38°C. mRNA analysis and subtype specific antagonists revealed that NE activates ß 2- and ß 3-AR in mHypoA-2/10 cells. Agonist binding to the ß 2-AR was temperature insensitive, but measurements of cytosolic cAMP accumulation revealed an increase in efficacy of 45% ± 27% for NE and of 62% ± 33% for the ß 2-AR-selective agonist salmeterol at 36°C. When monitoring NE-promoted cAMP efflux, we observed an increase in the absolute efflux at 36°C. However, the ratio of exported to cytosolic accumulated cAMP is higher at 38°C. We also stimulated cells with NE at 37°C and measured cAMP degradation at 36 and 38°C afterward. We observed increased cAMP degradation at 38°C, indicating enhanced phosphodiesterase activity at higher temperatures. In line with these data, NE-induced activation of the thyreoliberin promoter was found to be enhanced at 36°C. Overall, we show that physiologic temperature changes fine-tune NE-induced cAMP signaling in hypothalamic cells via ß 2-AR by modulating cAMP degradation and the ratio of intra- and extracellular cAMP. SIGNIFICANCE STATEMENT: Increasing cytosolic cAMP levels by activation of G protein-coupled receptors (GPCR) such as the ß 2-adrenergic receptor (AR) is essential for many body functions. Changes in core body temperature are fundamental and universal factors of mammalian life. This study provides the first data linking physiologically relevant temperature fluctuations to ß 2-AR-induced cAMP signaling, highlighting a so far unappreciated role of body temperature as a modulator of the prototypic class A GPCR.


Sujet(s)
AMP cyclique/métabolisme , Cytosol/métabolisme , Récepteurs bêta-2 adrénergiques/physiologie , Xanthine(isobutyl-3 methyl-1)/pharmacologie , Facteurs de transcription ARNTL/métabolisme , Aminopyridines/pharmacologie , Animaux , Lignée cellulaire , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Facteurs de transcription Forkhead/métabolisme , Sous-unités alpha Gq-G11 des protéines G/physiologie , Sous-unités alpha Gs des protéines G/physiologie , Hypothalamus/physiologie , Souris , Neurones/physiologie , Norépinéphrine/pharmacologie , Récepteurs bêta-2 adrénergiques/biosynthèse , Récepteurs bêta-3 adrénergiques/biosynthèse , Récepteurs bêta-3 adrénergiques/physiologie , Facteurs de transcription STAT/métabolisme , Xinafoate de salmétérol/pharmacologie , Transduction du signal/physiologie , Température , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme
12.
Endocrinology ; 162(8)2021 08 01.
Article de Anglais | MEDLINE | ID: mdl-34043769

RÉSUMÉ

The hypothalamus-pituitary-thyroid (HPT) axis regulates energy balance through the pleiotropic action of thyroid hormones. HPT basal activity and stimulation by cold or voluntary exercise are repressed by previous chronic stress in adults. Maternal separation (MS) modifies HPT basal activity; we thus studied the response of the axis to energy demands and analyzed possible epigenetic changes on Trh promoter. Nonhandled (NH) or MS male Wistar rats were cold exposed 1 h at adulthood; Trh expression in the hypothalamic paraventricular nucleus (PVN) and serum thyrotropin (TSH) concentration were increased only in NH rats. Two weeks of voluntary exercise decreased fat mass and increased Trh expression, and thyroid hormones concentration changed proportionally to running distance in NH male rats and MS male rats. Although NH females ran more than MS and much more than males, exercise decreased body weight and fat mass only in NH rats with no change on any parameter of the HPT axis but increased Pomc expression in arcuate-nucleus of NH and Npy in MS females. Overall, the methylation pattern of PVN Trh gene promoter was similar in NH males and females; MS modified methylation of specific CpG sites, a thyroid hormone receptor (THR)-binding site present after the initiation site was hypomethylated in MS males; in MS females, the THR binding site of the proximal promoter (site 4) and 2 sites in the first intron were hypermethylated. Our studies showed that, in a sex-dimorphic manner, MS blunted the responses of HPT axis to energy demands in adult animals and caused methylation changes on Trh promoter that could alter T3 feedback.


Sujet(s)
Méthylation de l'ADN , Séparation d'avec la mère , Caractères sexuels , Hormones thyroïdiennes/métabolisme , Hormone de libération de la thyréostimuline/génétique , Animaux , Métabolisme énergétique , Femelle , Axe hypothalamohypophysaire/métabolisme , Mâle , Pro-opiomélanocortine/génétique , Rat Wistar , Stress psychologique/métabolisme , Glande thyroide/métabolisme
13.
Brain Res Bull ; 172: 14-21, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-33862124

RÉSUMÉ

The adenosine A1 receptor is important for body temperature regulation in mammals; however, little is known about its function in avian species. In this study, we investigated the effects of the adenosine A1 receptor agonist and antagonist (adenosine 5'-monophosphate [5'-AMP] and 8 p-sulfophenyl theophylline [8-SPT], respectively) on thermoregulation in chickens. Male chicks were used in this study. After administration of 5'-AMP and 8-SPT, the rectal temperature, plasma metabolites, and gene expressions in the hypothalamus and liver were measured. The rectal temperature was reduced by peripheral administration of 5'-AMP, and the hypothermic effect of 5'-AMP was attenuated by central injection of 8-SPT in chicks. In the hypothalamus, the mRNA level of the agouti-related protein (AgRP) was increased by 5'-AMP administration, whereas it was suppressed by 8-SPT. The plasma levels of free fatty acid were elevated in 5'-AMP-treated chicks and that elevation was suppressed by the 8-SPT treatment. The gene expression of proopiomelanocortin in the hypothalamus was affected by 8-SPT. Nevertheless, the gene expressions of the thermoregulation-related genes, such as the thyrotropin-releasing hormone, were not affected by 5'-AMP and 8-SPT. Hepatic gene expressions related to lipid intake and metabolism were suppressed by 5'-AMP. However, the gene expression of the uncoupling protein was upregulated by 5'-AMP. Based on these results, birds, like mammals, will undergo adenosine A1 receptor-induced hypothermia. In conclusion, it is suggested that 5'-AMP-mediated hypothermia via the adenosine A1 receptor may affect the central melanocortin system and suppress hepatic lipid metabolism in chickens.


Sujet(s)
AMP/pharmacologie , Régulation de la température corporelle/effets des médicaments et des substances chimiques , Hypothalamus/effets des médicaments et des substances chimiques , Hypothermie provoquée , Foie/effets des médicaments et des substances chimiques , Protéine apparentée à Agouti/génétique , Protéine apparentée à Agouti/métabolisme , Animaux , Glycémie , Poulets , Acide gras libre/sang , Expression des gènes/effets des médicaments et des substances chimiques , Hypothalamus/métabolisme , Foie/métabolisme , Mâle , Pro-opiomélanocortine/génétique , Pro-opiomélanocortine/métabolisme , Théophylline/analogues et dérivés , Théophylline/pharmacologie , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme
14.
Mol Cell Endocrinol ; 529: 111269, 2021 06 01.
Article de Anglais | MEDLINE | ID: mdl-33819522

RÉSUMÉ

Prolactin (PRL), mainly synthesized and secreted by the lactotrophs and somatolactotrophs of the anterior pituitary, is a pleiotropic hormone that regulates lactation. In the last decade, nesfatin-1 (NESF) and NESF-like peptide (NLP), encoded in nucleobindin 1 and 2 (NUCB1 and NUCB2), respectively, were characterized as metabolic factors with a potential role in the control of pituitary hormones. We hypothesized that NUCBs and their encoded peptides (NESF and NLP) suppress PRL transcription in the pituitary. The main objective of this research was to determine whether exogenous NESF and NLP, and/or endogenous NUCB1 and NUCB2 regulate the expression of prl and preb mRNAs. Using immortalized rat somatolactotrophs (GH3 cells), dose-response studies were performed to test whether NESF and NLP affect prl and preb. Moreover, the ability of these peptides to modulate the effects of the PRL stimulator thyrotropin releasing hormone (TRH) was studied. Besides, the effects of siRNA-mediated knockdown of endogenous NUCBs on prl and preb mRNAs were determined. NESF and NLP reduced the transcription of prl and preb in GH3 cells. Both NESF and NLP also prevented the stimulatory effects of TRH prl and preb expression. The knockdown of endogenous NUCB1 attenuates both basal prl and TRH-induced expression of prl and preb, while the silencing of NUCBs did not affect the actions of exogenous NESF or NLP. Overall, this work reveals that NUCBs and encoded-peptides are novel regulators of PRL. Future research should test whether the effects observed here in GH3 cells are preserved both in vivo and at the post-transcriptional level.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Facteurs d'échange de nucléotides guanyliques/génétique , Cellules lactotropes/effets des médicaments et des substances chimiques , Nucléobindines/pharmacologie , Peptides/pharmacologie , Prolactine/génétique , Cellules somatotropes/effets des médicaments et des substances chimiques , Facteurs de transcription/génétique , Animaux , Lignée de cellules transformées , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Protéines de liaison à l'ADN/métabolisme , Régulation de l'expression des gènes , Facteurs d'échange de nucléotides guanyliques/antagonistes et inhibiteurs , Facteurs d'échange de nucléotides guanyliques/métabolisme , Cellules lactotropes/cytologie , Cellules lactotropes/métabolisme , Nucléobindines/antagonistes et inhibiteurs , Nucléobindines/génétique , Nucléobindines/métabolisme , Prolactine/antagonistes et inhibiteurs , Prolactine/métabolisme , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Rats , Transduction du signal , Cellules somatotropes/cytologie , Cellules somatotropes/métabolisme , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme
15.
PLoS One ; 15(11): e0242380, 2020.
Article de Anglais | MEDLINE | ID: mdl-33201916

RÉSUMÉ

Thyroid hormone (T3) inhibits thyrotropin-releasing hormone (TRH) synthesis in the hypothalamic paraventricular nucleus (PVN). Although the T3 receptor (TR) ß2 is known to mediate the negative regulation of the prepro-TRH gene, its molecular mechanism remains unknown. Our previous studies on the T3-dependent negative regulation of the thyrotropin ß subunit (TSHß) gene suggest that there is a tethering mechanism, whereby liganded TRß2 interferes with the function of the transcription factor, GATA2, a critical activator of the TSHß gene. Interestingly, the transcription factors Sim1 and Arnt2, the determinants of PVN differentiation in the hypothalamus, are reported to induce expression of TRß2 and GATA2 in cultured neuronal cells. Here, we confirmed the expression of the GATA2 protein in the TRH neuron of the rat PVN using immunohistochemistry with an anti-GATA2 antibody. According to an experimental study from transgenic mice, a region of the rat prepro-TRH promoter from nt. -547 to nt. +84 was able to mediate its expression in the PVN. We constructed a chloramphenicol acetyltransferase (CAT) reporter gene containing this promoter sequence (rTRH(547)-CAT) and showed that GATA2 activated the promoter in monkey kidney-derived CV1 cells. Deletion and mutation analyses identified a functional GATA-responsive element (GATA-RE) between nt. -357 and nt. -352. When TRß2 was co-expressed, T3 reduced GATA2-dependent promoter activity to approximately 30%. Unexpectedly, T3-dependent negative regulation was maintained after mutation of the reported negative T3-responsive element, site 4. T3 also inhibited the GATA2-dependent transcription enhanced by cAMP agonist, 8-bromo-cAMP. A rat thyroid medullary carcinoma cell line, CA77, is known to express the preproTRH mRNA. Using a chromatin immunoprecipitation assay with this cell line where GATA2 expression plasmid was transfected, we observed the recognition of the GATA-RE by GATA2. We also confirmed GATA2 binding using gel shift assay with the probe for the GATA-RE. In CA77 cells, the activity of rTRH(547)-CAT was potentiated by overexpression of GATA2, and it was inhibited in a T3-dependent manner. These results suggest that GATA2 transactivates the rat prepro-TRH gene and that liganded TRß2 interferes with this activation via a tethering mechanism as in the case of the TSHß gene.


Sujet(s)
Facteur de transcription GATA-2/métabolisme , Récepteurs bêta des hormones thyroïdiennes/métabolisme , Hormone de libération de la thyréostimuline/métabolisme , Animaux , Lignée cellulaire , Facteur de transcription GATA-2/physiologie , Régulation de l'expression des gènes/génétique , Gènes rapporteurs/génétique , Ligands , Mâle , Noyau paraventriculaire de l'hypothalamus/métabolisme , Noyau paraventriculaire de l'hypothalamus/anatomopathologie , Régions promotrices (génétique)/génétique , Précurseurs de protéines , Rats , Rat Wistar , Récepteurs des hormones thyroïdiennes/métabolisme , Récepteurs bêta des hormones thyroïdiennes/génétique , Hormones thyroïdiennes , Sous-unité bêta de la thyrotropine/métabolisme , Hormone de libération de la thyréostimuline/génétique , Facteurs de transcription , Activation de la transcription , Tri-iodothyronine/métabolisme
16.
J Physiol Biochem ; 76(4): 599-608, 2020 Nov.
Article de Anglais | MEDLINE | ID: mdl-32914279

RÉSUMÉ

Thyrotropin-releasing hormone (TRH) plays several roles as a hormone/neuropeptide. Diencephalic TRH (dTRH) participates in the regulation of blood pressure in diverse animal models, independently of the thyroid status. The present study aimed to evaluate whether chronic overexpression of TRH in mice affects cardiovascular and metabolic variables. We developed a transgenic (TG) mouse model that overexpresses dTrh. Despite having higher food consumption and water intake, TG mice showed significantly lower body weight respect to controls. Also, TG mice presented higher blood pressure, heart rate, and locomotor activity independently of thyroid hormone levels. These results and the higher urine noradrenaline excretion observed in TG mice suggest a higher metabolic rate mediated by sympathetic overflow. Cardiovascular changes were impeded by siRNA inhibition of the diencephalic Trh overexpression. Also, the silencing of dTRH in the TG mice normalized urine noradrenaline excretion, supporting the view that the cardiovascular effects of TRH involve the sympathetic system. Overall, we show that congenital dTrh overexpression leads to an increase in blood pressure accompanied by changes in body weight and food consumption mediated by a higher sympathetic overflow. These results provide new evidence confirming the participation of TRH in cardiovascular and body weight regulation.


Sujet(s)
Métabolisme basal , Pression sanguine , Poids , Hormone de libération de la thyréostimuline , Animaux , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/physiologie
17.
Life Sci ; 261: 118346, 2020 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-32853656

RÉSUMÉ

Doxorubicin is an antineoplastic in the anthracycline class widely used for the treatment of several solid tumors and blood cancers. Cardiotoxicity is the major dose-limiting adverse effect of the drug. Chronic and accumulated doxorubicin administration cause myocyte damage and myocardial fibrosis. Doxorubicin-associated cardiotoxicity can be also observed after a short-course drug treatment even without clinical evidence of cardiac disease. Nevertheless, acute underlying mechanisms involved in the initiation of drug-induced cardiotoxicity remain poorly explored despite their similarities with pathophysiological conditions where cardiac TRH (cTRH) plays a central role. We showed that cTRH mediates myocardial injury induced by hypertension, and angiotensin II. Further, cTRH overexpression induces cardiac apoptosis, hypertrophy and fibrosis. AIM: To demonstrate that cTRH could mediate acute doxorubicin cardiotoxicity. MAIN METHOD: A single injection of doxorubicin (10 mg kg/day i.p.) was used to evaluate acute cardiac damage in a short-term experimental model of doxorubicin-induced cardiotoxicity. While inhibiting cTRH by small interfering RNA (siRNA), we evaluated the progression of cardiotoxicity. KEY FINDINGS: We found a doxorubicin-induced TRH overexpression in the LV, which was associated with apoptosis, hypertrophy and fibrosis. siRNA-mediated cTRH suppression prevented the doxorubicin-associated cardiac histological lesions. SIGNIFICANCES: doxorubicin requires an active cardiac TRH system to promote heart injury.


Sujet(s)
Antibiotiques antinéoplasiques/toxicité , Cardiotoxicité/étiologie , Doxorubicine/toxicité , Hormone de libération de la thyréostimuline/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Cardiomégalie/induit chimiquement , Cardiotoxicité/physiopathologie , Évolution de la maladie , Fibrose/induit chimiquement , Mâle , Souris , Souris de lignée C57BL , Petit ARN interférent , Hormone de libération de la thyréostimuline/génétique
18.
Insect Biochem Mol Biol ; 122: 103376, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32339620

RÉSUMÉ

EFLamide (EFLa) is a neuropeptide known for a long time from crustaceans, chelicerates and myriapods. Recently, EFLa-encoding genes were identified in the genomes of apterygote hexapods including basal insect species. In pterygote insects, however, evidence of EFLa was limited to partial sequences in the bed bug (Cimex), migratory locust and a few phasmid species. Here we present identification of a full length EFLa-encoding transcript in the linden bug, Pyrrhocoris apterus (Heteroptera). We created complete null mutants allowing unambiguous anatomical location of this peptide in the central nervous system. Only 2-3 EFLa-expressing cells are located very close to each other near to the surface of the lateral protocerebrum with dense neuronal arborization. Homozygous null EFLa mutants are fully viable and do not have any visible defect in development, reproduction, lifespan, diapause induction or circadian rhythmicity. Phylogenetic analysis revealed that EFLa-encoding transcripts are produced by alternative splicing of a gene that also produces Prohormone-4. However, this Proh-4/EFLa connection is found only in Hemiptera and Locusta, whereas EFLa-encoding transcripts in apterygote hexapods, chelicerates and crustaceans are clearly distinct from Proh-4 genes. The exact mechanism leading to the fused Proh-4/EFLa transcript is not yet determined, and might be a result of canonical cis-splicing, cis-splicing of adjacent genes (cis-SAG), or trans-splicing.


Sujet(s)
Heteroptera/génétique , Protéines d'insecte/génétique , Neuropeptides/génétique , Séquence d'acides aminés , Animaux , Femelle , Heteroptera/métabolisme , Protéines d'insecte/composition chimique , Protéines d'insecte/métabolisme , Mâle , Neuropeptides/composition chimique , Neuropeptides/métabolisme , Phylogenèse , Alignement de séquences , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme
19.
Front Biosci (Landmark Ed) ; 25(7): 1305-1323, 2020 03 01.
Article de Anglais | MEDLINE | ID: mdl-32114434

RÉSUMÉ

Starvation induces tertiary hypothyroidism in adult rodents. Response of the hypothalamus-pituitary-thyroid (HPT) axis to starvation is stronger in adult males than in females. To improve the description of this sexual dimorphism, we analyzed the dynamics of HPT axis response to fasting at multiple levels. In adult rats of the same cohort, 24 and 48 h of starvation inhibited paraventricular nucleus Trh expression and serum concentrations of TSH and T4 earlier in males than in females, with lower intensity in females than in males. In adult females fasted for 36-72 h, serum TSH concentration decreased after 36 h, when the activity of thyrotropin-releasing hormone (TRH)-degrading ectoenzyme was increased in the median eminence. The kinetics of these events were distinct from those previously observed in male rats. We suggest that the sex difference in TSH secretion kinetics is driven not only at the level of paraventricular nucleus TRH neurons, but also by differences in post-secretory catabolism of TRH, with enhancement of TRH-degrading activity more sustained in male than female animals.


Sujet(s)
Jeûne/métabolisme , Régulation de l'expression des gènes , Noyau paraventriculaire de l'hypothalamus/métabolisme , Glande thyroide/métabolisme , Animaux , Femelle , Mâle , Rat Wistar , Récepteur CRH/génétique , Récepteur CRH/métabolisme , Récepteurs aux glucocorticoïdes/génétique , Récepteurs aux glucocorticoïdes/métabolisme , Récepteur TRH/génétique , Récepteur TRH/métabolisme , Facteurs sexuels , Thyréostimuline/sang , Thyréostimuline/métabolisme , Hormone de libération de la thyréostimuline/génétique , Hormone de libération de la thyréostimuline/métabolisme , Facteurs temps
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE