Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 167(3): 843-857.e14, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27720451

ABSTRACT

Glucagon and thyroid hormone (T3) exhibit therapeutic potential for metabolic disease but also exhibit undesired effects. We achieved synergistic effects of these two hormones and mitigation of their adverse effects by engineering chemical conjugates enabling delivery of both activities within one precisely targeted molecule. Coordinated glucagon and T3 actions synergize to correct hyperlipidemia, steatohepatitis, atherosclerosis, glucose intolerance, and obesity in metabolically compromised mice. We demonstrate that each hormonal constituent mutually enriches cellular processes in hepatocytes and adipocytes via enhanced hepatic cholesterol metabolism and white fat browning. Synchronized signaling driven by glucagon and T3 reciprocally minimizes the inherent harmful effects of each hormone. Liver-directed T3 action offsets the diabetogenic liability of glucagon, and glucagon-mediated delivery spares the cardiovascular system from adverse T3 action. Our findings support the therapeutic utility of integrating these hormones into a single molecular entity that offers unique potential for treatment of obesity, type 2 diabetes, and cardiovascular disease.


Subject(s)
Glucagon/therapeutic use , Metabolic Diseases/drug therapy , Triiodothyronine/drug effects , Animals , Atherosclerosis/drug therapy , Body Weight/drug effects , Bone and Bones/drug effects , Chemical Engineering/methods , Cholesterol/metabolism , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Drug Combinations , Drug Delivery Systems , Drug Synergism , Glucagon/adverse effects , Glucagon/chemistry , Glucagon/pharmacology , Hyperglycemia/drug therapy , Liver/drug effects , Liver/metabolism , Mice , Molecular Targeted Therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Triiodothyronine/adverse effects , Triiodothyronine/chemistry , Triiodothyronine/pharmacology
2.
EMBO Rep ; 24(9): e57020, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37424431

ABSTRACT

Cell identity is specified by a core transcriptional regulatory circuitry (CoRC), typically limited to a small set of interconnected cell-specific transcription factors (TFs). By mining global hepatic TF regulons, we reveal a more complex organization of the transcriptional regulatory network controlling hepatocyte identity. We show that tight functional interconnections controlling hepatocyte identity extend to non-cell-specific TFs beyond the CoRC, which we call hepatocyte identity (Hep-ID)CONNECT TFs. Besides controlling identity effector genes, Hep-IDCONNECT TFs also engage in reciprocal transcriptional regulation with TFs of the CoRC. In homeostatic basal conditions, this translates into Hep-IDCONNECT TFs being involved in fine tuning CoRC TF expression including their rhythmic expression patterns. Moreover, a role for Hep-IDCONNECT TFs in the control of hepatocyte identity is revealed in dedifferentiated hepatocytes where Hep-IDCONNECT TFs are able to reset CoRC TF expression. This is observed upon activation of NR1H3 or THRB in hepatocarcinoma or in hepatocytes subjected to inflammation-induced loss of identity. Our study establishes that hepatocyte identity is controlled by an extended array of TFs beyond the CoRC.


Subject(s)
Gene Expression Regulation , Transcription Factors , Transcription Factors/metabolism , Hepatocytes/metabolism , Liver/metabolism , Gene Regulatory Networks
3.
Liver Int ; 44(1): 125-138, 2024 01.
Article in English | MEDLINE | ID: mdl-37872645

ABSTRACT

OBJECTIVE: Progressive hepatic fibrosis can be considered the final stage of chronic liver disease. Hepatic stellate cells (HSC) play a central role in liver fibrogenesis. Thyroid hormones (TH, e.g. thyroxine; T4 and triiodothyronine; T3) significantly affect development, growth, cell differentiation and metabolism through activation of TH receptor α and/or ß (TRα/ß). Here, we evaluated the influence of TH in hepatic fibrogenesis. DESIGN: Human liver tissue was obtained from explanted livers following transplantation. TRα-deficient (TRα-KO) and wild-type (WT) mice were fed a control or a profibrogenic methionine-choline deficient (MCD) diet. Liver tissue was assessed by qRT-PCR for fibrogenic gene expression. In vitro, HSC were treated with TGFß in the presence or absence of T3. HSC with stable TRα knockdown and TRα deficient mouse embryonic fibroblasts (MEF) were used to determine receptor-specific function. Activation of HSC and MEF was assessed using the wound healing assay, Western blotting, and qRT-PCR. RESULTS: TRα and TRß expression is downregulated in the liver during hepatic fibrogenesis in humans and mice. TRα represents the dominant isoform in HSC. In vitro, T3 blunted TGFß-induced expression of fibrogenic genes in HSC and abrogated wound healing by modulating TGFß signalling, which depended on TRα presence. In vivo, TRα-KO enhanced MCD diet-induced liver fibrogenesis. CONCLUSION: These observations indicate that TH action in non-parenchymal cells is highly relevant. The interaction of TRα with TH regulates the phenotype of HSC via the TGFß signalling pathway. Thus, the TH-TR axis may be a valuable target for future therapy of liver fibrosis.


Subject(s)
Fibroblasts , Hepatic Stellate Cells , Animals , Mice , Humans , Hepatic Stellate Cells/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Transforming Growth Factor beta
4.
FASEB J ; 34(9): 12072-12082, 2020 09.
Article in English | MEDLINE | ID: mdl-32776612

ABSTRACT

Mammals adapt to seasons using a neuroendocrine calendar defined by the photoperiodic change in the nighttime melatonin production. Under short photoperiod, melatonin inhibits the pars tuberalis production of TSHß, which, in turn, acts on tanycytes to regulate the deiodinase 2/3 balance resulting in a finely tuned seasonal control of the intra-hypothalamic thyroid hormone T3. Despite the pivotal role of this T3 signaling for synchronizing reproduction with the seasons, T3 cellular targets remain unknown. One candidate is a population of hypothalamic neurons expressing Rfrp, the gene encoding the RFRP-3 peptide, thought to be integral for modulating rodent's seasonal reproduction. Here we show that nighttime melatonin supplementation in the drinking water of melatonin-deficient C57BL/6J mice mimics photoperiodic variations in the expression of the genes Tshb, Dio2, Dio3, and Rfrp, as observed in melatonin-proficient mammals. Notably, we report that this melatonin regulation of Rfrp expression is no longer observed in mice carrying a global mutation of the T3 receptor, TRα, but is conserved in mice with a selective neuronal mutation of TRα. In line with this observation, we find that TRα is widely expressed in the tanycytes. Altogether, our data demonstrate that the melatonin-driven T3 signal regulates RFRP-3 neurons through non-neuronal, possibly tanycytic, TRα.


Subject(s)
Gene Expression Regulation/drug effects , Melatonin/pharmacology , Neuropeptides/biosynthesis , Receptors, Thyroid Hormone/metabolism , Triiodothyronine/metabolism , Animals , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Mice , Mice, Knockout , Neuropeptides/genetics , Receptors, Thyroid Hormone/genetics , Triiodothyronine/genetics , Iodothyronine Deiodinase Type II
5.
FASEB J ; 34(11): 15480-15491, 2020 11.
Article in English | MEDLINE | ID: mdl-32969079

ABSTRACT

Thyroid hormones are important for homeostatic control of energy metabolism and body temperature. Although skeletal muscle is considered a key site for thyroid action, the contribution of thyroid hormone receptor signaling in muscle to whole-body energy metabolism and body temperature has not been resolved. Here, we show that T3-induced increase in energy expenditure requires thyroid hormone receptor alpha 1 (TRα1 ) in skeletal muscle, but that T3-mediated elevation in body temperature is achieved in the absence of muscle-TRα1 . In slow-twitch soleus muscle, loss-of-function of TRα1 (TRαHSACre ) alters the fiber-type composition toward a more oxidative phenotype. The change in fiber-type composition, however, does not influence the running capacity or motivation to run. RNA-sequencing of soleus muscle from WT mice and TRαHSACre mice revealed differentiated transcriptional regulation of genes associated with muscle thermogenesis, such as sarcolipin and UCP3, providing molecular clues pertaining to the mechanistic underpinnings of TRα1 -linked control of whole-body metabolic rate. Together, this work establishes a fundamental role for skeletal muscle in T3-stimulated increase in whole-body energy expenditure.


Subject(s)
Energy Metabolism/drug effects , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/physiology , Thyroid Hormone Receptors alpha/physiology , Thyroid Hormones/pharmacology , Animals , Male , Mice , Mice, Knockout , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/cytology , Muscle Fibers, Slow-Twitch/drug effects , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Physical Conditioning, Animal , Transcriptome
6.
Proc Natl Acad Sci U S A ; 114(31): 8229-8234, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716910

ABSTRACT

Thyroid hormone receptors (TRs) are members of the nuclear hormone receptor superfamily that act as ligand-dependent transcription factors. Here we identified the ten-eleven translocation protein 3 (TET3) as a TR interacting protein increasing cell sensitivity to T3. The interaction between TET3 and TRs is independent of TET3 catalytic activity and specifically allows the stabilization of TRs on chromatin. We provide evidence that TET3 is required for TR stability, efficient binding of target genes, and transcriptional activation. Interestingly, the differential ability of different TRα1 mutants to interact with TET3 might explain their differential dominant activity in patients carrying TR germline mutations. So this study evidences a mode of action for TET3 as a nonclassical coregulator of TRs, modulating its stability and access to chromatin, rather than its intrinsic transcriptional activity. This regulatory function might be more general toward nuclear receptors. Indeed, TET3 interacts with different members of the superfamily and also enhances their association to chromatin.


Subject(s)
Chromatin/metabolism , Dioxygenases/metabolism , Thyroid Hormone Receptors alpha/metabolism , Catalytic Domain , Chromatin/genetics , Dioxygenases/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Immunoprecipitation , Mutation , Nitriles/pharmacology , Protein Interaction Domains and Motifs , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Thiazoles/pharmacology , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Transcription, Genetic , Ubiquitination
7.
J Vasc Res ; 55(4): 224-234, 2018.
Article in English | MEDLINE | ID: mdl-30092589

ABSTRACT

Thyroid hormone (TH) regulates gene transcription by binding to TH receptors (TRs). TRs regulate the genes of lipid metabolism and the renin-angiotensin system (RAS). We examined the effect of TRα deletion in ApoE-/- mice (DKO mice) on the following: (i) the expression of genes controlling cholesterol metabolism and tissue (t)RAS in the liver and aorta and (ii) the expression of these genes and the regulation of cholesterol content in cultured vascular smooth muscle cells (VSMCs). TRα deletion in ApoE-/- mice led to the repression of genes involved in the synthesis and influx of cholesterol in the liver. However, TRα deletion in the arterial wall suppressed the expression of genes involved in the esterification and excretion of cholesterol and enhanced the expression of angiotensinogen (AGT). The VSMCs of the ApoE-/- and DKO mice increased their cholesterol content during cholesterol loading, but failed to increase the expression of ATP-binding cassette transporter A1 (ABCA1). T3 addition partially corrected these abnormalities in the cells of the ApoE-/- mice but not those of the DKO mice. In conclusion, TRα deletion in ApoE-/- mice slightly increases the expression of tRAS in the aorta and aggravates the dysregulation of cholesterol content in the VSMCs.


Subject(s)
Apolipoproteins E/deficiency , Cholesterol/metabolism , Muscle, Smooth, Vascular/metabolism , Renin-Angiotensin System/physiology , Thyroid Hormone Receptors alpha/deficiency , ATP Binding Cassette Transporter 1/genetics , Animals , Aorta/chemistry , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Atherosclerosis/diagnostic imaging , Cells, Cultured , Cholesterol/administration & dosage , Cholesterol/genetics , Gene Expression , Hybridization, Genetic , Liver/chemistry , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/cytology , RNA, Messenger , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/physiology , Triiodothyronine/pharmacology , Ultrasonography
9.
J Biol Chem ; 291(1): 198-214, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26453307

ABSTRACT

MTORC2-AKT is a key regulator of carbohydrate metabolism and insulin signaling due to its effects on FOXO1 phosphorylation. Interestingly, both FOXO1 and thyroid hormone (TH) have similar effects on carbohydrate and energy metabolism as well as overlapping transcriptional regulation of many target genes. Currently, little is known about the regulation of MTORC2-AKT or FOXO1 by TH. Accordingly, we performed hepatic transcriptome profiling in mice after FOXO1 knockdown in the absence or presence of TH, and we compared these results with hepatic FOXO1 and THRB1 (TRß1) ChIP-Seq data. We identified a subset of TH-stimulated FOXO1 target genes that required co-regulation by FOXO1 and TH. TH activation of FOXO1 was directly linked to an increase in SIRT1-MTORC2 interaction and RICTOR deacetylation. This, in turn, led to decreased AKT and FOXO1 phosphorylation. Moreover, TH increased FOXO1 nuclear localization, DNA binding, and target gene transcription by reducing AKT-dependent FOXO1 phosphorylation in a THRB1-dependent manner. These events were associated with TH-mediated oxidative phosphorylation and NAD(+) production and suggested that downstream metabolic effects by TH can post-translationally activate other transcription factors. Our results showed that RICTOR/MTORC2-AKT can integrate convergent hormonal and metabolic signals to provide coordinated and sensitive regulation of hepatic FOXO1-target gene expression.


Subject(s)
Carrier Proteins/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Liver/metabolism , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Thyroid Hormones/pharmacology , Acetylation/drug effects , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Enzyme Activation/drug effects , Forkhead Box Protein O1 , Hep G2 Cells , Humans , Liver/drug effects , Male , Mechanistic Target of Rapamycin Complex 2 , Mice, Inbred C57BL , NAD/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Receptors, Thyroid Hormone/metabolism , Sirtuin 1/metabolism , Up-Regulation/drug effects
10.
Proc Natl Acad Sci U S A ; 111(42): 15108-13, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25288732

ABSTRACT

Several physiopathological processes require orientated cellular migration. This phenomenon highly depends on members of the RHO family of GTPases. Both excessive and deficient RHO activity impair directional migration. A tight control is thus exerted on these proteins through the regulation of their activation and of their stability. Here we show that the estrogen-related receptor α (ERRα) directly activates the expression of TNFAIP1, the product of which [BTB/POZ domain-containing adapter for Cullin3-mediated RhoA degradation 2 (BACURD2)] regulates RHOA protein turnover. Inactivation of the receptor leads to enhanced RHOA stability and activation. This results in cell disorientation, increased actin network, and inability to form a lamellipodium at the migration edge. As a consequence, directional migration, but not cell motility per se, is impaired in the absence of the receptor, under pathological as well as physiological conditions. Altogether, our results show that the control exerted by ERRα on RHOA stability is required for directional migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Receptors, Estrogen/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Cell Line, Tumor , Cullin Proteins/metabolism , Extracellular Matrix/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , Protein Stability , Protein Structure, Tertiary , Proteins/metabolism , Wound Healing , ERRalpha Estrogen-Related Receptor
11.
Physiol Genomics ; 48(2): 73-81, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26672044

ABSTRACT

The deletion of thyroid hormone receptor-α (TRα) in atherosclerosis-prone apolipoprotein E-deficient (ApoE(-/-)) mice (ApoE(-/-)TRα(0/0)) accelerates the formation of atherosclerotic plaques without aggravation of hypercholesterolemia. To evaluate other predisposition risk factors to atherosclerosis in this model, we studied blood pressure (BP) and cardiac and vascular functions, as well as exercise tolerance in young adult ApoE(-/-)TRα(0/0) mice before the development of atherosclerotic plaques. Telemetric BP recorded for 4 consecutive days showed that the spontaneous systolic BP was slightly decreased in ApoE(-/-)TRα(0/0) compared with ApoE(-/-) mice associated with a reduced locomotor activity. The percentage of animals that completed endurance (57% vs. 89%) and maximal running (0% vs. 89% at 46 cm/s speed in ApoE(-/-)TRα(0/0) and ApoE(-/-) mice, respectively) tests was lower in ApoE(-/-)TRα(0/0) mice. Moreover, during the maximal running test, both maximal running speed and running distance were significantly reduced in ApoE(-/-)TRα(0/0) mice, associated with a blunted BP response to exercise. Transthoracic echocardiography revealed a decreased interventricular septum thickness and an increased end-systolic left ventricular volume in ApoE(-/-)TRα(0/0) mice. Accordingly, left ventricular fractional shortening, ejection fraction, and stroke volume were all significantly decreased in ApoE(-/-)TRα(0/0) mice with a concomitant blunted cardiac output. No interstrain difference was observed in vascular reactivity, except that ApoE(-/-)TRα(0/0) mice exhibited an enhanced acetylcholine-induced relaxation in mesenteric and distal femoral arteries. In conclusion, the deletion of TRα in ApoE(-/-) mice alters cardiac structure and contractility; both could contribute to blunted BP response to physical exercise and impaired exercise performance.


Subject(s)
Heart/physiopathology , Physical Conditioning, Animal , Thyroid Hormone Receptors alpha/genetics , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Blood Pressure , Body Composition , Circadian Rhythm , Echocardiography , Gene Deletion , Hypercholesterolemia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Running , Stroke Volume , Systole , Thyroid Hormone Receptors alpha/deficiency
13.
J Biol Chem ; 288(42): 30365-30372, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-23995837

ABSTRACT

Hepatic gluconeogenesis is a concerted process that integrates transcriptional regulation with hormonal signals. A major regulator is thyroid hormone (TH), which acts through its nuclear receptor (TR) to induce the expression of the hepatic gluconeogenic genes, phosphoenolpyruvate carboxykinase (PCK1) and glucose-6-phosphatase (G6PC). Forkhead transcription factor FoxO1 also is an important regulator of these genes; however, its functional interactions with TR are not known. Here, we report that TR-mediated transcriptional activation of PCK1 and G6PC in human hepatic cells and mouse liver was FoxO1-dependent and furthermore required FoxO1 deacetylation by the NAD(+)-dependent deacetylase, SirT1. siRNA knockdown of FoxO1 decreased, whereas overexpression of FoxO1 increased, TH-dependent transcriptional activation of PCK1 and G6PC in cultured hepatic cells. FoxO1 siRNA knockdown also decreased TH-mediated transcription in vivo. Additionally, TH was unable to induce FoxO1 deacetylation or hepatic PCK1 gene expression in TH receptor ß-null (TRß(-/-)) mice. Moreover, TH stimulated FoxO1 recruitment to the PCK1 and G6PC gene promoters in a SirT1-dependent manner. In summary, our results show that TH-dependent deacetylation of a second metabolically regulated transcription factor represents a novel mechanism for transcriptional integration of nuclear hormone action with cellular energy status.


Subject(s)
Forkhead Transcription Factors/metabolism , Gluconeogenesis/physiology , Liver/metabolism , Thyroid Hormones/metabolism , Transcription, Genetic/physiology , Transcriptional Activation/physiology , Acetylation , Animals , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Glucose-6-Phosphatase/biosynthesis , Glucose-6-Phosphatase/genetics , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Phosphoenolpyruvate Carboxykinase (GTP)/biosynthesis , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Promoter Regions, Genetic/physiology , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Thyroid Hormones/genetics
14.
Biochim Biophys Acta ; 1830(7): 3900-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22704954

ABSTRACT

BACKGROUND: Thyroid hormone receptors TRα1, TRß1 and TRß2 are broadly expressed and exert a pleiotropic influence on many developmental and homeostatic processes. Extensive genetic studies in mice precisely defined their respective function. SCOPE OF REVIEW: The purpose of the review is to discuss two puzzling issues: MAJOR CONCLUSIONS: Mouse genetics support a balanced contribution of expression pattern and receptor intrinsic properties in defining the receptor respective functions. The molecular mechanisms sustaining cell specific response remain hypothetical and based on studies performed with other nuclear receptors. GENERAL SIGNIFICANCE: The isoform-specificity and cell-specificity questions have many implications for clinical research, drug development, and endocrine disruptor studies. This article is part of a Special Issue entitled Thyroid hormone signalling.


Subject(s)
Receptors, Thyroid Hormone/physiology , Animals , Humans , Protein Isoforms , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Signal Transduction , Thyroid Hormones/genetics , Thyroid Hormones/metabolism
15.
J Vasc Res ; 51(5): 350-9, 2014.
Article in English | MEDLINE | ID: mdl-25500991

ABSTRACT

OBJECTIVE: This study evaluated the consequences of thyroid hormone receptor-α (TRα) disruption on vascular reactivity. METHODS: The activity of superior mesenteric arteries isolated from TRα knockout mice generated in the SV129 background (TRα(0/0)SV) or in a pure C57BL/6 background (TRα(0/0)C57) was compared to that of their corresponding wild-type strains (SV129 or C57BL/6 mice). RESULTS: The wild-type SV129 mice exhibited an impaired acetylcholine (Ach)-induced mesenteric artery relaxation compared to C57BL/6 mice, associated with greater responses to angiotensin II (AII) and phenylephrine (PE). The disruption of TRα decreased the vascular response to sodium nitroprusside and PE in both the SV129 and C57BL/6 genetic backgrounds. Responses to Ach and AII were also blunted, but only in TRα(0/0)C57 mice. The administration of 3,3'5-triiodo-L-thyronine sodium salt (T3) elicited a vasodilatation in C57BL/6 mice even at the lowest concentration (10(-9)M); a maximal relaxation of more than 50% was observed with the concentrations between 10(-9) and 10(-8)M. However, the response to T3 was nearly absent in TRα(0/0)C57 mice. CONCLUSION: TRα is essential for the control of vascular tone, particularly in thyroid hormone-mediated relaxation. The difference in response to Ach observed between the two wild-type mice should be taken into account for interpreting the vascular responses of genetically engineered mice.


Subject(s)
Mesenteric Artery, Superior/metabolism , Thyroid Hormone Receptors alpha/deficiency , Vasodilation , Animals , Dose-Response Relationship, Drug , Genotype , Male , Mesenteric Artery, Superior/drug effects , Mesenteric Artery, Superior/physiopathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Species Specificity , Thyroid Hormone Receptors alpha/agonists , Thyroid Hormone Receptors alpha/genetics , Triiodothyronine/pharmacology , Vasoconstriction , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
16.
Thyroid ; 34(2): 261-273, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38115594

ABSTRACT

Background: Tanycytes are specialized glial cells within the mediobasal hypothalamus that have multiple functions, including hormone sensing and regulation of hypophysiotropic hormone secretion. There are ongoing discussions about the role of tanycytes in regulating the supply of hypothalamic thyroid hormones (THs) through the expression of TH transporters (Slc16a2, Slco1c1) and deiodinases (Dio2, Dio3). In this study, we investigated the potential feedback effect of thyrotropin (TSH) on the transcription of these gatekeeper genes on tanycytes. Methods: We analyzed the changes in the expression of TH-gatekeeper genes, in TSH-stimulated primary tanycytes, using quantitative polymerase chain reaction (qPCR). We also used RNAScope® in brain slices to further reveal the local distribution of the transcripts. In addition, we blocked intracellular pathways and used small-interfering RNA (siRNA) to elucidate differences in the regulation of the gatekeeper genes. Results: TSH elevated messenger RNA (mRNA) levels of Slco1c1, Dio2, and Dio3 in tanycytes, while Slc16a2 was mostly unaffected. Blockade and knockdown of the TSH receptor (TSHR) and antagonization of cAMP response element-binding protein (CREB) clearly abolished the increased expression induced by TSH, indicating PKA-dependent regulation through the TSHR. The TSH-dependent expression of Dio3 and Slco1c1 was also regulated by protein kinase C (PKC), and in case of Dio3, also by extracellular signal-regulated kinase (ERK) activity. Importantly, these gene regulations were specifically found in different subpopulations of tanycytes. Conclusions: This study demonstrates that TSH induces transcriptional regulation of TH-gatekeeper genes in tanycytes through the Tshr/Gαq/PKC pathway, in parallel to the Tshr/Gαs/PKA/CREB pathway. These differential actions of TSH on tanycytic subpopulations appear to be important for coordinating the supply of TH to the hypothalamus and aid its functions.


Subject(s)
Ependymoglial Cells , Thyrotropin , Humans , Thyrotropin/pharmacology , Thyrotropin/metabolism , Ependymoglial Cells/metabolism , Thyroid Hormones/metabolism , Thyroid Gland/metabolism , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Pituitary Hormone-Releasing Hormones/metabolism , Protein Kinase C/metabolism
17.
bioRxiv ; 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38260502

ABSTRACT

Protein translation is an energy-intensive ribosome-driven process that is reduced during nutrient scarcity to conserve cellular resources. During prolonged starvation, cells selectively translate specific proteins to enhance their survival (adaptive translation); however, this process is poorly understood. Accordingly, we analyzed protein translation and mRNA transcription by multiple methods in vitro and in vivo to investigate adaptive hepatic translation during starvation. While acute starvation suppressed protein translation in general, proteomic analysis showed that prolonged starvation selectively induced translation of lysosome and autolysosome proteins. Significantly, the expression of the orphan nuclear receptor, estrogen-related receptor alpha (Esrra) increased during prolonged starvation and served as a master regulator of this adaptive translation by transcriptionally stimulating 60S acidic ribosomal protein P1 (Rplp1) gene expression. Overexpression or siRNA knockdown of Esrra expression in vitro or in vivo led to parallel changes in Rplp1 gene expression, lysosome/autophagy protein translation, and autophagy. Remarkably, we have found that Esrra had dual functions by not only regulating transcription but also controling adaptive translation via the Esrra/Rplp1/lysosome/autophagy pathway during prolonged starvation.

18.
Mol Metab ; : 101997, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39032642

ABSTRACT

OBJECTIVE: Currently, little is known about the mechanism(s) regulating global and specific protein translation during metabolic dysfunction-associated steatohepatitis (MASH; previously known as non-alcoholic steatohepatitis, NASH). METHODS: Unbiased label-free quantitative proteome, puromycin-labelling and polysome profiling were used to understand protein translation activity in vitro and in vivo. RESULTS: We observed a global decrease in protein translation during lipotoxicity in human primary hepatocytes, mouse hepatic AML12 cells, and livers from a dietary mouse model of MASH. Interestingly, proteomic analysis showed that Rplp1, which regulates ribosome and translation pathways, was one of the most downregulated proteins. Moreover, decreased Esrra expression and binding to the Rplp1 promoter, diminished Rplp1 gene expression during lipotoxicity. This, in turn, reduced global protein translation and Esrra/Rplp1-dependent translation of lysosome (Lamp2, Ctsd) and autophagy (sqstm1, Map1lc3b) proteins. Of note, Esrra did not increase its binding to these gene promoters or their gene transcription, confirming its regulation of their translation during lipotoxicity. Notably, hepatic Esrra-Rplp1-dependent translation of lysosomal and autophagy proteins also was impaired in MASH patients and liver-specific Esrra knockout mice. Remarkably, alternate day fasting induced Esrra-Rplp1-dependent expression of lysosomal proteins, restored autophagy, and reduced lipotoxicity, inflammation, and fibrosis in hepatic cell culture and in vivo models of MASH. CONCLUSIONS: Esrra regulation of Rplp1-mediated translation of lysosome/autolysosome proteins was downregulated during MASH. Alternate day fasting activated this novel pathway and improved MASH, suggesting that Esrra and Rplp1 may serve as therapeutic targets for MASH. Our findings also provided the first example of a nuclear hormone receptor, Esrra, to not only regulate transcription but also protein translation, via induction of Rplp1.

19.
Leukemia ; 38(3): 570-578, 2024 03.
Article in English | MEDLINE | ID: mdl-38321107

ABSTRACT

Myeloproliferative neoplasms (MPNs) are a group of chronic hematologic malignancies that lead to morbidity and early mortality due to thrombotic complications and progression to acute leukemia. Clinical and mutational risk factors have been demonstrated to predict outcomes in patients with MPNs and are used commonly to guide therapeutic decisions, including allogenic stem cell transplant, in myelofibrosis. Adolescents and young adults (AYA, age ≤45 years) comprise less than 10% of all MPN patients and have unique clinical and therapeutic considerations. The prevalence and clinical impact of somatic mutations implicated in myeloid disease has not been extensively examined in this population. We conducted a retrospective review of patients evaluated at eight Canadian centers for MPN patients diagnosed at ≤45 years of age. In total, 609 patients were included in the study, with median overall survival of 36.8 years. Diagnosis of prefibrotic or overt PMF is associated with the lowest OS and highest risk of AP/BP transformation. Thrombotic complications (24%), including splanchnic circulation thrombosis (9%), were frequent in the cohort. Mutations in addition to those in JAK2/MPL/CALR are uncommon in the initial disease phase in our AYA population (12%); but our data indicate they may be predictive of transformation to post-ET/PV myelofibrosis.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Primary Myelofibrosis , Thrombocythemia, Essential , Thrombosis , Humans , Young Adult , Adolescent , Middle Aged , Primary Myelofibrosis/genetics , Primary Myelofibrosis/therapy , Polycythemia Vera/genetics , Thrombocythemia, Essential/genetics , Canada/epidemiology , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Thrombosis/genetics , Janus Kinase 2/genetics , Mutation , Calreticulin/genetics
20.
Endocrinology ; 164(4)2023 02 11.
Article in English | MEDLINE | ID: mdl-36801988

ABSTRACT

Thyroid hormone increases energy expenditure. Its action is mediated by TR, nuclear receptors present in peripheral tissues and in the central nervous system, particularly in hypothalamic neurons. Here, we address the importance of thyroid hormone signaling in neurons, in general for the regulation of energy expenditure. We generated mice devoid of functional TR in neurons using the Cre/LoxP system. In hypothalamus, which is the center for metabolic regulation, mutations were present in 20% to 42% of the neurons. Phenotyping was performed under physiological conditions that trigger adaptive thermogenesis: cold and high-fat diet (HFD) feeding. Mutant mice displayed impaired thermogenic potential in brown and inguinal white adipose tissues and were more prone to diet-induced obesity. They showed a decreased energy expenditure on chow diet and gained more weight on HFD. This higher sensitivity to obesity disappeared at thermoneutrality. Concomitantly, the AMPK pathway was activated in the ventromedial hypothalamus of the mutants as compared with the controls. In agreement, sympathetic nervous system (SNS) output, visualized by tyrosine hydroxylase expression, was lower in the brown adipose tissue of the mutants. In contrast, absence of TR signaling in the mutants did not affect their ability to respond to cold exposure. This study provides the first genetic evidence that thyroid hormone signaling exerts a significant influence in neurons to stimulate energy expenditure in some physiological context of adaptive thermogenesis. TR function in neurons to limit weight gain in response to HFD and this effect is associated with a potentiation of SNS output.


Subject(s)
Obesity , Thyroid Hormones , Male , Mice , Animals , Obesity/genetics , Obesity/metabolism , Thyroid Hormones/metabolism , Diet, High-Fat/adverse effects , Adipose Tissue, Brown/metabolism , Neurons/metabolism , Thermogenesis/physiology , Energy Metabolism/genetics
SELECTION OF CITATIONS
SEARCH DETAIL