Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Commun Biol ; 6(1): 1253, 2023 12 11.
Article in English | MEDLINE | ID: mdl-38081939

ABSTRACT

Thyroid hormone (3,5,3'-triiodothyronine, T3) is a key regulator of pituitary gland function. The response to T3 is thought to hinge crucially on interactions of nuclear T3 receptors with enhancers but these sites in pituitary chromatin remain surprisingly obscure. Here, we investigate genome-wide receptor binding in mice using tagged endogenous thyroid hormone receptor ß (TRß) and analyze T3-regulated open chromatin using an anterior pituitary-specific Cre driver (Thrbb2Cre). Strikingly, T3 regulates histone modifications and chromatin opening primarily at sites that maintain TRß binding regardless of T3 levels rather than at sites where T3 abolishes or induces de novo binding. These sites associate more frequently with T3-activated than T3-suppressed genes. TRß-deficiency blunts T3-regulated gene expression, indicating that TRß confers transcriptional sensitivity. We propose a model of gene activation in which poised receptor-enhancer complexes facilitate adjustable responses to T3 fluctuations, suggesting a genomic basis for T3-dependent pituitary function or pituitary dysfunction in thyroid disorders.


Subject(s)
Chromatin , Thyroid Hormones , Mice , Animals , Chromatin/genetics , Chromatin/metabolism , Thyroid Hormones/metabolism , Triiodothyronine/pharmacology , Triiodothyronine/metabolism , Pituitary Gland/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism
2.
Thyroid ; 33(7): 867-876, 2023 07.
Article in English | MEDLINE | ID: mdl-37166378

ABSTRACT

Background: Thyrotropin-releasing hormone (TRH) neurons in the paraventricular nucleus of the hypothalamus (PVN) have been identified as direct regulators of thyrotropin (TSH) and thyroid hormone (TH) levels. They play a significant role in context of negative feedback by TH at the level of TRH gene expression and during fasting when TH levels fall due, in part, to suppression of TRH gene expression. Methods: To test these functions directly for the first time, we used a chemogenetic approach and activated PVN TRH neurons in both fed and fasted mice. Next, to demonstrate the signals that regulate the fasting response in TRH neurons, we activated or inhibited agouti-related protein (AgRP)/neuropeptide Y (NPY) neurons in the arcuate nucleus of the hypothalamus of fed or fasted mice, respectively. To determine if the same TRH neurons responsive to melanocortin signaling mediate negative feedback by TH, we disrupted the thyroid hormone receptor beta (TRß) in all melanocortin 4 receptor (MC4R) neurons in the PVN. Results: Activation of TRH neurons led to increased TSH and TH levels within 2 hours demonstrating the specific role of PVN TRH neurons in the regulation of the hypothalamic-pituitary-thyroid (HPT) axis. Moreover, activation of PVN TRH neurons prevented the fall in TH levels in fasting mice. Stimulation of AgRP/NPY neurons led to a fall in TH levels despite increasing feeding. Inhibition of these same neurons prevented the fall in TH levels during a fast presumably via their ability to directly regulate PVN TRH neurons via, in part, the MC4R. Surprisingly, TH-mediated feedback was not impaired in mice lacking TRß in MC4R neurons. Conclusions: TRH neurons are major regulators of the HPT axis and the fasting-induced suppression of TH levels. The latter relies, at least in part, on the activation of AgRP/NPY neurons in the arcuate nucleus. Interestingly, present data do not support an important role for TRß signaling in regulating MC4R neurons in the PVN. Thus, it remains possible that different subsets of TRH neurons in the PVN mediate responses to energy balance and to TH feedback.


Subject(s)
Thyrotropin-Releasing Hormone , Thyrotropin , Mice , Animals , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Thyroid Gland/metabolism , Pituitary Hormone-Releasing Hormones/metabolism , Hypothalamus , Thyroid Hormones/metabolism , Paraventricular Hypothalamic Nucleus , Neurons/metabolism
4.
Cell Biosci ; 12(1): 118, 2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35902915

ABSTRACT

BACKGROUND: The activation of the hypothalamic-pituitary-adrenal (HPA) axis is essential for metabolic adaptation in response to fasting. However, the neurocircuitry connecting changes in the peripheral energy stores to the activity of hypothalamic paraventricular corticotrophin-releasing factor (CRFPVN) neurons, the master controller of the HPA axis activity, is not completely understood. Our main goal was to determine if hypothalamic arcuate nucleus (ARC) POMC and AgRP neurons can communicate fasting-induced changes in peripheral energy stores, associated to a fall in plasma leptin levels, to CRFPVN neurons to modulate the HPA axis activity in mice. RESULTS: We observed increased plasma corticosterone levels associate with increased CRFPVN mRNA expression and increased CRFPVN neuronal activity in 36 h fasted mice. These responses were associated with a fall in plasma leptin levels and changes in the mRNA expression of Agrp and Pomc in the ARC. Fasting-induced decrease in plasma leptin partially modulated these responses through a change in the activity of ARC neurons. The chemogenetic activation of POMCARC by DREADDs did not affect fasting-induced activation of the HPA axis. DREADDs inhibition of AgRPARC neurons reduced the content of CRFPVN and increased its accumulation in the median eminence but had no effect on corticosterone secretion induced by fasting. CONCLUSION: Our data indicate that AgRPARC neurons are part of the neurocircuitry involved in the coupling of PVNCRF activity to changes in peripheral energy stores induced by prolonged fasting.

5.
Mol Metab ; 53: 101315, 2021 11.
Article in English | MEDLINE | ID: mdl-34390859

ABSTRACT

OBJECTIVE: The nuclear receptor corepressor 1 (NCOR1) and the silencing mediator of retinoic acid and thyroid hormone (SMRT, also known as NCOR2) play critical and specific roles in nuclear receptor action. NCOR1, both in vitro and in vivo specifically regulates thyroid hormone (TH) action in the context of individual organs such as the liver, and systemically in the context of the hypothalamic-pituitary-thyroid (HPT) axis. In contrast, selective deletion of SMRT in the liver or globally has shown that it plays very little role in TH signaling. However, both NCOR1 and SMRT have some overlapping roles in hepatic metabolism and lipogenesis. Here, we determine the roles of NCOR1 and SMRT in global physiologic function and find if SMRT could play a compensatory role in the regulation of TH action, globally. METHODS: We used a postnatal deletion strategy to disrupt both NCOR1 and SMRT together in all tissues at 8-9 weeks of age in male and female mice. This was performed using a tamoxifen-inducible Cre recombinase (UBC-Cre-ERT2) to KO (knockout) NCOR1, SMRT, or NCOR1 and SMRT together. We used the same strategy to KO HDAC3 in male and female mice of the same age. Metabolic parameters, gene expression, and thyroid function tests were analyzed. RESULTS: Surprisingly, adult mice that acquired NCOR1 and SMRT deletion rapidly became hypoglycemic and hypothermic and perished within ten days of deletion of both corepressors. Postnatal deletion of either NCOR1 or SMRT had no impact on mortality. NCOR1/SMRT KO mice rapidly developed hepatosteatosis and mild elevations in liver function tests. Additionally, alterations in lipogenesis, beta oxidation, along with hepatic triglyceride and glycogen levels suggested defects in hepatic metabolism. The intestinal function was intact in the NCOR1/SMRT knockout (KO) mice. The KO of HDAC3 resulted in a distinct phenotype from the NCOR1/SMRT KO mice, whereas none of the HDAC3 KO mice succumbed after tamoxifen injection. CONCLUSIONS: The KO of NCOR1 and SMRT rapidly leads to significant metabolic abnormalities that do not survive - including hypoglycemia, hypothermia, and weight loss. Hepatosteatosis rapidly developed along with alterations in hepatic metabolism suggesting a contribution to the dramatic phenotype from liver injury. Glucose production and absorption were intact in NCOR1/SMRT KO mice, demonstrating a multifactorial process leading to their demise. HDAC3 KO mice have a distinct phenotype from the NCOR1/SMRT KO mice-which implies that NCOR1/SMRT together regulate a critical pathway that is required for survival in adulthood and is separate from HDAC3.


Subject(s)
Homeostasis , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Co-Repressor 1/deficiency , Nuclear Receptor Co-Repressor 2/deficiency
7.
PLoS One ; 14(8): e0220717, 2019.
Article in English | MEDLINE | ID: mdl-31404087

ABSTRACT

Silencing Mediator of Retinoid and Thyroid Hormone Receptors (SMRT) and the nuclear receptor co-repressor1 (NCoR1) are paralogs and regulate nuclear receptor (NR) function through the recruitment of a multiprotein complex that includes histone deacetylase activity. Previous genetic strategies which deleted SMRT in a specific tissue or which altered the interaction between SMRT and NRs have suggested that it may regulate adiposity and insulin sensitivity. However, the full role of SMRT in adult mice has been difficult to establish because its complete deletion during embryogenesis is lethal. To elucidate the specific roles of SMRT in mouse target tissues especially in the context of thyroid hormone (TH) signaling, we used a tamoxifen-inducible post-natal disruption strategy. We found that global SMRT deletion causes dramatic obesity even though mice were fed a standard chow diet and exhibited normal food intake. This weight gain was associated with a decrease in energy expenditure. Interestingly, the deletion of SMRT had no effect on TH action in any tissue but did regulate retinoic acid receptor (RAR) function in the liver. We also demonstrate that the deletion of SMRT leads to profound hepatic steatosis in the setting of obesity. This is unlike NCoR1 deletion, which results in hepatic steatosis due to the upregulation of lipogenic gene expression. Taken together, our data demonstrate that SMRT plays a unique and CoR specific role in the regulation of body weight and has no role in TH action. This raises the possibility that additional role of CoRs besides NCoR1 and SMRT may exist to regulate TH action.


Subject(s)
Body Weight/physiology , Nuclear Receptor Co-Repressor 2/physiology , Thyroid Hormones/physiology , Animals , Blotting, Western , Cholesterol/analysis , Echocardiography , Energy Metabolism , Glucose Tolerance Test , Lipids/blood , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Thyroid Hormones/blood , Thyrotropin/blood , Thyrotropin/physiology , Thyroxine/blood , Thyroxine/physiology , Triglycerides/analysis , Weight Gain/physiology
8.
Mol Cell Endocrinol ; 458: 127-135, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28286327

ABSTRACT

Thyroid hormones are a critical regulator of mammalian physiology. Much of their action is due to effects in the nucleus where T3 engages thyroid hormone receptor isoforms to mediate its effects. In order to function properly the TR isoforms must be recruited to regulatory sequences within genes that they up-regulate. On these positive regulated target genes the TR can activate or repress depending upon whether the receptor is bound to T3 or not and the type of co-regulatory proteins present in that cell type. In contrast to T3 mediated activation, the mechanism by which the TR represses transcription in the presence of T3 remains unclear. Herein we will review the components of the transcriptional response to T3 within the nucleus and attempt to highlight the outstanding questions in the field.


Subject(s)
Gene Regulatory Networks , Receptors, Thyroid Hormone/metabolism , Thyroid Hormones/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Humans , Regulatory Elements, Transcriptional , Signal Transduction , Up-Regulation
9.
J Clin Endocrinol Metab ; 101(12): 4512-4520, 2016 12.
Article in English | MEDLINE | ID: mdl-27691051

ABSTRACT

CONTEXT: Challenges and opportunities face the next generation (Next-Gen) of endocrine researchers and clinicians, the lifeblood of the field of endocrinology for the future. A symposium jointly sponsored by The Endocrine Society and the Endocrine Society of Australia was convened to discuss approaches to addressing the present and future Next-Gen needs. EVIDENCE ACQUISITION: Data collection by literature review, assessment of previously completed questionnaires, commissioning of a new questionnaire, and summarization of symposium discussions were studied. EVIDENCE SYNTHESIS: Next-Gen endocrine researchers face diminishing grant funding in inflation-adjusted terms. The average age of individuals being awarded their first independent investigator funding has increased to age 45 years. For clinicians, a workforce gap exists between endocrinologists needed and those currently trained. Clinicians in practice are increasingly becoming employees of integrated hospital systems, resulting in greater time spent on nonclinical issues. Workforce data and published reviews identify challenges specifically related to early career women in endocrinology. Strategies to Address Issues: Recommendations encompassed the areas of grant support for research, mentoring, education, templates for career development, specific programs for Next-Gen members by senior colleagues as outlined in the text, networking, team science, and life/work integration. Endocrine societies focusing on Next-Gen members provide a powerful mechanism to support these critical areas. CONCLUSIONS: A concerted effort to empower, train, and support the next generation of clinical endocrinologists and endocrine researchers is necessary to ensure the viability and vibrancy of our discipline and to optimize our contributions to improving health outcomes. Collaborative engagement of endocrine societies globally will be necessary to support our next generation moving forward.


Subject(s)
Endocrinologists , Endocrinology , Societies, Medical , Endocrinologists/economics , Endocrinologists/education , Endocrinologists/standards , Endocrinology/economics , Endocrinology/education , Endocrinology/standards , Humans , Societies, Medical/standards
10.
Endocrinology ; 156(10): 3853-65, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26241124

ABSTRACT

A key function of the thyroid hormone receptor ß (Thrb) gene is in the development of auditory function. However, the roles of the 2 receptor isoforms, TRß1 and TRß2, expressed by the Thrb gene are unclear, and it is unknown whether these isoforms promote the maintenance as well as development of hearing. We investigated the function of TRß1 in mice with a Thrb(b1) reporter allele that expresses ß-galactosidase instead of TRß1. In the immature cochlea, ß-galactosidase was detected in the greater epithelial ridge, sensory hair cells, spiral ligament, and spiral ganglion and in adulthood, at low levels in the hair cells, support cells and root cells of the outer sulcus. Although deletion of all TRß isoforms causes severe, early-onset deafness, deletion of TRß1 or TRß2 individually caused no obvious hearing loss in juvenile mice. However, over subsequent months, TRß1 deficiency resulted in progressive loss of hearing and loss of hair cells. TRß1-deficient mice had minimal changes in serum thyroid hormone and thyrotropin levels, indicating that hormonal imbalances were unlikely to cause hearing loss. The results suggest mutually shared roles for TRß1 and TRß2 in cochlear development and an unexpected requirement for TRß1 in the maintenance of hearing in adulthood.


Subject(s)
Aging , Hair Cells, Auditory/metabolism , Hearing Loss/genetics , Thyroid Hormone Receptors beta/genetics , Animals , Cochlea/embryology , Cochlea/growth & development , Cochlea/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/pathology , Hearing Loss/blood , Hearing Loss/metabolism , Immunohistochemistry , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ of Corti/embryology , Organ of Corti/growth & development , Organ of Corti/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormones/blood , Thyrotropin/blood , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
11.
Endocrinology ; 155(10): 4061-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25004090

ABSTRACT

Type 3 deiodinase (D3), the physiologic inactivator of thyroid hormones, is induced during tissue injury and regeneration. This has led to the hypotheses that D3 impacts injury tolerance by reducing local T3 signaling and contributes to the fall in serum triiodothyronine (T3) observed in up to 75% of sick patients (termed the low T3 syndrome). Here we show that a novel mutant mouse with hepatocyte-specific D3 deficiency has normal local responses to toxin-induced hepatonecrosis, including normal degrees of tissue necrosis and intact regeneration, but accelerated systemic recovery from illness-induced hypothyroxinemia and hypotriiodothyroninemia, demonstrating that peripheral D3 expression is a key modulator of the low T3 syndrome.


Subject(s)
Chemical and Drug Induced Liver Injury/rehabilitation , Hepatocytes/metabolism , Iodide Peroxidase/genetics , Liver Regeneration/genetics , Liver/pathology , Animals , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/physiopathology , Female , Iodide Peroxidase/deficiency , Liver/drug effects , Male , Mice , Mice, Knockout , Necrosis/chemically induced , Necrosis/genetics , Organ Specificity/genetics , Recovery of Function/genetics , Toxins, Biological
12.
Mol Cell Biol ; 34(9): 1564-75, 2014 May.
Article in English | MEDLINE | ID: mdl-24550004

ABSTRACT

Resistance to thyroid hormone (RTH), a human syndrome, is characterized by high thyroid hormone (TH) and thyroid-stimulating hormone (TSH) levels. Mice with mutations in the thyroid hormone receptor beta (TRß) gene that cannot bind steroid receptor coactivator 1 (SRC-1) and Src-1(-/-) mice both have phenotypes similar to that of RTH. Conversely, mice expressing a mutant nuclear corepressor 1 (Ncor1) allele that cannot interact with TRß, termed NCoRΔID, have low TH levels and normal TSH. We hypothesized that Src-1(-/-) mice have RTH due to unopposed corepressor action. To test this, we crossed NCoRΔID and Src-1(-/-) mice to create mice deficient for coregulator action in all cell types. Remarkably, NCoR(ΔID/ΔID) Src-1(-/-) mice have normal TH and TSH levels and are triiodothryonine (T(3)) sensitive at the level of the pituitary. Although absence of SRC-1 prevented T(3) activation of key hepatic gene targets, NCoR(ΔID/ΔID) Src-1(-/-) mice reacquired hepatic T(3) sensitivity. Using in vivo chromatin immunoprecipitation assays (ChIP) for the related coactivator SRC-2, we found enhanced SRC-2 recruitment to TR-binding regions of genes in NCoR(ΔID/ΔID) Src-1(-/-) mice, suggesting that SRC-2 is responsible for T(3) sensitivity in the absence of NCoR1 and SRC-1. Thus, T(3) targets require a critical balance between NCoR1 and SRC-1. Furthermore, replacement of NCoR1 with NCoRΔID corrects RTH in Src-1(-/-) mice through increased SRC-2 recruitment to T(3) target genes.


Subject(s)
Co-Repressor Proteins/metabolism , Nuclear Receptor Coactivator 1/metabolism , Nuclear Receptor Coactivator 2/metabolism , Signal Transduction , Thyroid Hormone Resistance Syndrome/metabolism , Thyroid Hormones/metabolism , Animals , Female , Gene Deletion , Humans , Male , Mice , Mutation , Nuclear Receptor Coactivator 1/genetics , Pituitary Gland/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormone Resistance Syndrome/blood , Thyroid Hormone Resistance Syndrome/genetics , Thyroid Hormones/blood , Thyrotropin/blood , Thyrotropin/metabolism , Triiodothyronine/metabolism
13.
Mol Cell Endocrinol ; 365(1): 84-94, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-23000398

ABSTRACT

Thyrotropin-releasing hormone (TRH) in the paraventricular nucleus (PVN) of the hypothalamus is regulated by thyroid hormone (TH). cAMP response element binding protein (CREB) has also been postulated to regulate TRH expression but its interaction with TH signaling in vivo is not known. To evaluate the role of CREB in TRH regulation in vivo, we deleted CREB from PVN neurons to generate the CREB1(ΔSIM1) mouse. As previously shown, loss of CREB was compensated for by an up-regulation of CREM in euthyroid CREB1(ΔSIM1) mice but TSH, T4 and T3 levels were normal, even though TRH mRNA levels were elevated. Interestingly, TRH mRNA expression was also increased in the PVN of CREB1(ΔSIM1) mice in the hypothyroid state but became normal when made hyperthyroid. Importantly, CREM levels were similar in CREB1(ΔSIM1) mice regardless of thyroid status, demonstrating that the regulation of TRH by T3 in vivo likely occurs independently of the CREB/CREM family.


Subject(s)
Cyclic AMP Response Element Modulator/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Female , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/cytology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/physiology , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Thyrotropin-Releasing Hormone/genetics , Triiodothyronine/metabolism
14.
Cell Metab ; 14(6): 780-90, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22100407

ABSTRACT

Fasting-induced suppression of the hypothalamic-pituitary-thyroid (HPT) axis is an adaptive response to decrease energy expenditure during food deprivation. Previous studies demonstrate that leptin communicates nutritional status to the HPT axis through thyrotropin-releasing hormone (TRH) in the paraventricular nucleus (PVN) of the hypothalamus. Leptin targets TRH neurons either directly or indirectly via the arcuate nucleus through pro-opiomelanocortin (POMC) and agouti-related peptide/neuropeptide Y (AgRP/NPY) neurons. To evaluate the role of these pathways in vivo, we developed double knockout mice that lack both the melanocortin 4 receptor (MC4R) and NPY. We show that NPY is required for fasting-induced suppression of Trh expression in the PVN. However, both MC4R and NPY are required for activation of hepatic pathways that metabolize T(4) during the fasting response. Thus, these signaling pathways play a key role in the communication of fasting signals to reduce thyroid hormone levels both centrally and through a peripheral hepatic circuit.


Subject(s)
Fasting/physiology , Hypothalamo-Hypophyseal System/metabolism , Neuropeptide Y/metabolism , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction/physiology , Thyroid Hormones/metabolism , Absorptiometry, Photon , Analysis of Variance , Animals , Body Composition , Female , In Situ Hybridization , Leptin/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Knockout , Models, Biological , Neuropeptide Y/genetics , Paraventricular Hypothalamic Nucleus/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Melanocortin, Type 4/genetics , Thyrotropin-Releasing Hormone/metabolism
15.
Mol Endocrinol ; 25(2): 212-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21239618

ABSTRACT

The role of nuclear receptor corepressor (NCoR) in thyroid hormone (TH) action has been difficult to discern because global deletion of NCoR is embryonic lethal. To circumvent this, we developed mice that globally express a modified NCoR protein (NCoRΔID) that cannot be recruited to the thyroid hormone receptor (TR). These mice present with low serum T(4) and T(3) concentrations accompanied by normal TSH levels, suggesting central hypothyroidism. However, they grow normally and have increased energy expenditure and normal or elevated TR-target gene expression across multiple tissues, which is not consistent with hypothyroidism. Although these findings imply an increased peripheral sensitivity to TH, the hypothalamic-pituitary-thyroid axis is not more sensitive to acute changes in TH concentrations but appears to be reset to recognize the reduced TH levels as normal. Furthermore, the thyroid gland itself, although normal in size, has reduced levels of nonthyroglobulin-bound T(4) and T(3) and demonstrates decreased responsiveness to TSH. Thus, the TR-NCoR interaction controls systemic TH sensitivity as well as the set point at all levels of the hypothalamic-pituitary-thyroid axis. These findings suggest that NCoR levels could alter cell-specific TH action that would not be reflected by the serum TSH.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Receptors, Thyroid Hormone/metabolism , Thyroid Gland/metabolism , Thyroid Hormones/metabolism , Animals , Blotting, Western , Gene Expression , Hypothyroidism , In Situ Hybridization , Mice , Mutant Proteins/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Polymerase Chain Reaction , Receptors, Thyroid Hormone/genetics , Thyrotropin/blood , Thyroxine/blood , Triiodothyronine/blood
16.
Mol Endocrinol ; 24(12): 2366-81, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20943814

ABSTRACT

Fasting-induced suppression of thyroid hormone levels is an adaptive response to reduce energy expenditure in both humans and mice. This suppression is mediated by the hypothalamic-pituitary-thyroid axis through a reduction in TRH levels expressed in neurons of the paraventricular nucleus of the hypothalamus (PVN). TRH gene expression is positively regulated by leptin. Whereas decreased leptin levels during fasting lead to a reduction in TRH gene expression, the mechanisms underlying this process are still unclear. Indeed, evidence exists that TRH neurons in the PVN are targeted by leptin indirectly via the arcuate nucleus, whereas correlative evidence for a direct action exists as well. Here we provide both in vivo and in vitro evidence that the activity of hypothalamic-pituitary-thyroid axis is regulated by both direct and indirect leptin regulation. We show that both leptin and α-MSH induce significant neuronal activity mediated through a postsynaptic mechanism in TRH-expressing neurons of PVN. Furthermore, we provide in vivo evidence indicating the contribution of each pathway in maintaining serum levels of thyroid hormone.


Subject(s)
Adiposity/physiology , Neurons/physiology , Paraventricular Hypothalamic Nucleus/physiology , Thyrotropin-Releasing Hormone/biosynthesis , Adiposity/drug effects , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Fasting/blood , Fasting/metabolism , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Leptin/metabolism , Leptin/pharmacology , Melanocortins/agonists , Melanocortins/pharmacology , Mice , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Rats , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Hormones/blood , Thyroid Hormones/metabolism , Thyrotropin-Releasing Hormone/antagonists & inhibitors , Thyrotropin-Releasing Hormone/genetics , alpha-MSH/metabolism , alpha-MSH/pharmacology
17.
PLoS One ; 5(8): e12324, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20808804

ABSTRACT

BACKGROUND: Mice with a targeted deletion of the basic helix-loop-helix transcription factor, Nescient Helix-Loop-Helix 2 (Nhlh2), display adult-onset obesity with significant increases in their fat depots, abnormal responses to cold exposure, and reduced spontaneous physical activity levels. These phenotypes, accompanied by the hypothalamic expression of Nhlh2, make the Nhlh2 knockout (N2KO) mouse a useful model to study the role of central nervous system (CNS) control on peripheral tissue such as adipose tissue. METHODOLOGY: Differences in body temperature and serum analysis of leptin were performed in fasted and ad lib fed wild-type (WT) and N2KO mice. Histological analysis of white (WAT) and brown adipose tissue (BAT) was performed. Gene and protein level expression of inflammatory and metabolic markers were compared between the two genotypes. PRINCIPAL FINDINGS: We report significant differences in serum leptin levels and body temperature in N2KO mice compared with WT mice exposed to a 24-hour fast, suggestive of a defect in both white (WAT) and brown adipose tissue (BAT) function. As compared to WT mice, N2KO mice showed increased serum IL-6 protein and WAT IL-6 mRNA levels. This was accompanied by slight elevations of mRNA for several macrophage markers, including expression of macrophage specific protein F4/80 in adipose, suggestive of macrophage infiltration of WAT in the mutant animals. The mRNAs for beta3-adrenergic receptors (beta3-AR), beta2-AR and uncoupling proteins were significantly reduced in WAT and BAT from N2KO mice compared with WT mice. CONCLUSIONS: These studies implicate Nhlh2 in the central control of WAT and BAT function, with lack of Nhlh2 leading to adipose inflammation and altered gene expression, impaired leptin response to fasting, all suggestive of a deficient torpor response in mutant animals.


Subject(s)
Adipose Tissue/metabolism , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Energy Metabolism , Gene Deletion , Gene Expression Regulation , Receptors, Adrenergic, beta/metabolism , Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipose Tissue/cytology , Animals , Antigens, Differentiation/metabolism , Biomarkers/metabolism , Body Temperature , Fasting , Gene Expression Profiling , Gene Knockout Techniques , Inflammation/genetics , Leptin/metabolism , Macrophages/metabolism , Male , Mice , Receptors, Adrenergic, beta/genetics
18.
Endocrinology ; 151(2): 793-801, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20032051

ABSTRACT

The expression of the TRH gene in the paraventricular nucleus (PVH) of the hypothalamus is required for the normal production of thyroid hormone (TH) in rodents and humans. In addition, the regulation of TRH mRNA expression by TH, specifically in the PVH, ensures tight control of the set point of the hypothalamic-pituitary-thyroid axis. Although many studies have assumed that the regulation of TRH expression by TH is at the level of transcription, there is little data available to demonstrate this. We used two in vivo model systems to show this. In the first model system, we developed an in situ hybridization (ISH) assay directed against TRH heteronuclear RNA to measure TRH transcription directly in vivo. We show that in the euthyroid state, TRH transcription is present both in the PVH and anterior/lateral hypothalamus. In the hypothyroid state, transcription is activated in the PVH only and can be shut off within 5 h by TH. In the second model system, we employed transgenic mice that express the Cre recombinase under the control of the genomic region containing the TRH gene. Remarkably, TH regulates Cre expression in these mice in the PVH only. Taken together, these data affirm that TH regulates TRH at the level of transcription in the PVH only and that genomic elements surrounding the TRH gene mediate its regulation by T(3). Thus, it should be possible to identify the elements within the TRH locus that mediate its regulation by T(3) using in vivo approaches.


Subject(s)
Gene Expression Regulation/physiology , Thyrotropin-Releasing Hormone/genetics , Transcription, Genetic , Animals , Genes, Reporter , Green Fluorescent Proteins/genetics , Immunohistochemistry , Integrases/genetics , Mice , Mice, Inbred C57BL , Propylthiouracil/pharmacology , RNA, Heterogeneous Nuclear/genetics , RNA, Messenger/genetics , Thyrotropin-Releasing Hormone/metabolism , Transcription, Genetic/drug effects
20.
Front Biosci ; 12: 3983-93, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17485352

ABSTRACT

Multiple regulatory pathways exist to control the expression levels of neuropeptides in response to body weight and energy availability changes. Since many neuropeptides are first synthesized in a pro-neuropeptide form, the availability of processing enzymes in a neuron can control the amount of active mature neuropeptide produced at any given time. In this review, we will focus on the regulation of prohormone convertase 1 (PC1) and prohormone convertase 2 (PC2), as well as downstream neuropeptide genes. Evidence from our laboratory suggests that Nescient helix-loop-helix 2 (Nhlh2) regulates the transcription of PC1 and PC2, possibly in conjunction with the leptin-stimulated transcription factor, STAT3. Furthermore, Nhlh2 itself is a target of leptin and other energy availability signals, with high levels of expression during energy surplus, and low levels of expression in conditions of reduced energy availability such as food deprivation or cold exposure. Overall, coordinate regulation of Nhlh2, PC1, PC2 and downstream hypothalamic neuropeptides such as thyrotropin releasing hormone (TRH) and pro-opiomelanocortin (POMC) does lead to energy balance modulation and ensuing long-term changes in body weight.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Energy Metabolism , Gene Expression Regulation , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...