Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Mol Cell Biol ; 42(2): e0036321, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34871063

ABSTRACT

Mutations in thyroid hormone receptor α (TRα), a ligand-inducible transcription factor, cause resistance to thyroid hormone α (RTHα). This disorder is characterized by tissue-specific hormone refractoriness and hypothyroidism due to the inhibition of target gene expression by mutant TRα-corepressor complexes. Using biophysical approaches, we show that RTHα-associated TRα mutants devoid of ligand-dependent transcription activation function unexpectedly retain the ability to bind thyroid hormone. Visualization of the ligand T3 within the crystal structure of a prototypic TRα mutant validates this notion. This finding prompted the synthesis of different thyroid hormone analogues, identifying a lead compound, ES08, which dissociates corepressor from mutant human TRα more efficaciously than T3. ES08 rescues developmental anomalies in a zebrafish model of RTHα and induces target gene expression in TRα mutation-containing cells from an RTHα patient more effectively than T3. Our observations provide proof of principle for developing synthetic ligands that can relieve transcriptional repression by the mutant TRα-corepressor complex for treatment of RTHα.


Subject(s)
Co-Repressor Proteins/genetics , Gene Expression/physiology , Genetic Predisposition to Disease/genetics , Thyroid Hormones/metabolism , Animals , Humans , Mutation/genetics , Phenotype , Receptors, Thyroid Hormone/genetics , Thyroid Hormone Receptors alpha/metabolism , Triiodothyronine/genetics
2.
PLoS One ; 15(10): e0241238, 2020.
Article in English | MEDLINE | ID: mdl-33104749

ABSTRACT

The NCoR corepressor plays critical roles in mediating transcriptional repression by both nuclear receptors and non-receptor transcription factors. Alternative mRNA splicing of NCoR produces a series of variants with differing molecular and biological properties. The NCoRω splice-variant inhibits adipogenesis whereas the NCoRδ splice-variant promotes it, and mice bearing a splice-specific knockout of NCoRω display enhanced hepatic steatosis and overall weight gain on a high fat diet as well as a greatly increased resistance to diet-induced glucose intolerance. We report here that the reciprocal NCoRδ splice-specific knock-out mice display the contrary phenotypes of reduced hepatic steatosis and reduced weight gain relative to the NCoRω-/- mice. The NCoRδ-/- mice also fail to demonstrate the strong resistance to diet-induced glucose intolerance exhibited by the NCoRω-/- animals. The NCoR δ and ω variants possess both unique and shared transcriptional targets, with expression of certain hepatic genes affected in opposite directions in the two mutants, others altered in one but not the other genotype, and yet others changed in parallel in both NCoRδ-/- and NCoRω-/- animals versus WT. Gene set expression analysis (GSEA) identified a series of lipid, carbohydrate, and amino acid metabolic pathways that are likely to contribute to their distinct steatosis and glucose tolerance phenotypes. We conclude that alternative-splicing of the NCoR corepressor plays a key role in the regulation of hepatic energy storage and utilization, with the NCoRδ and NCoRω variants exerting both opposing and shared functions in many aspects of this phenomenon and in the organism as a whole.


Subject(s)
Alternative Splicing/genetics , Liver/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Animals , Diet , Fatty Liver/complications , Fatty Liver/metabolism , Fatty Liver/pathology , Feeding Behavior , Gene Expression Regulation , Glucose Intolerance/complications , Insulin Resistance , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Weight Gain
3.
BMC Res Notes ; 12(1): 343, 2019 Jun 17.
Article in English | MEDLINE | ID: mdl-31208445

ABSTRACT

OBJECTIVE: The NCoR-1 and NCoR-2 corepressors are products of an early gene duplication near the beginning of vertebrate evolution and play both overlapping and divergent roles in development and physiology. Alternative-splicing of NCoR-1 and NCoR-2 further customizes their functions. To better understand the evolutionary basis of this phenomenon we extended our prior study of NCoR-1 and NCoR-2 alternative-splicing to an expanded series of species. RESULTS: Alternative-splicing of NCoR-2 was observed in all vertebrates examined whereas alternative-splicing of NCoR-1 was largely limited to placental mammals. Notably the most prominent of the NCoR-1 alternative-splicing events specific to the placental lineage, in exon 37 that plays a key role in murine metabolism, mimics in many features an analogous alternative-splicing event that appeared in NCoR-2 much earlier at the beginning of the vertebrate radiation. Detection of additional alternative-splicing events, at exons 28 in NCoR-1 or NCoR-2, was limited to the Rodentia or Primates examined, indicating both corepressor paralogs continued to acquire additional splice variations more recently and independently of one another. Our results suggest that the NCoR-1/NCoR-2 paralogs have been subject to a mix of shared and distinct selective pressures, resulting in the pattern of divergent and convergent alternative-splicing observed in extant species.


Subject(s)
Alternative Splicing , Mammals/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 2/genetics , RNA, Messenger/genetics , Animals , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Evolution, Molecular , Exons/genetics , Female , Humans , Mammals/classification , Mammals/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Phylogeny , Placenta , Pregnancy , Species Specificity
4.
Thyroid ; 28(12): 1708-1722, 2018 12.
Article in English | MEDLINE | ID: mdl-30235988

ABSTRACT

BACKGROUND: Thyroid hormone receptors (TRs) are tightly regulated by the corepressors nuclear receptor corepressor (NCoR) and silencing mediator of retinoic acid and thyroid hormone receptors. Three conserved corepressor/NR signature box motifs (CoRNR1-3) forming the nuclear receptor interaction domain have been identified in these corepressors. Whereas TRs regulate multiple normal physiological and developmental pathways, mutations in TRs can result in endocrine diseases and be associated with cancers due to impairment of corepressor release. Three mutants that are located in helix H11 of TRs are of special interest: TRα-M388I, a mutant associated with the development of renal clear cell carcinomas (RCCCs), and TRß-Δ430 and TRß-Δ432, two deletion mutants causing resistance to thyroid hormone syndrome. METHODS: Several cell-based and biophysical methods were used to measure the affinity between wild-type and mutant TRα and TRß and all the CoRNR motifs from corepressors to quantify the effects of different thyroid hormone analogues on these interactions. This study was coupled with the measurement of interactions between wild-type and mutant TRs in the context of a heterodimer with RXR to a NCoR fragment in the presence of the same ligands. Structural insights into the binding mode of corepressors to TRs were assessed in parallel by nuclear magnetic resonance spectroscopy. RESULTS: The study shows that TRs interact more avidly with the silencing mediator of retinoic acid and thyroid hormone receptors than with NCoR peptides, and that TRα binds most avidly to S-CoRNR3, whereas TRß binds preferentially to S-CoRNR2. In the studied TR mutants, a transfer of the CoRNR-specificity toward CoRNR1 was observed, coupled with a significant increase in the binding strength. In contrast to 3,5,3'-triiodothyronine (T3), the agonist TRIAC and the antagonist NH-3 were very efficient at dissociating the abnormally strong interactions between mutant TRßs and corepressors. A strong impairment of T3-binding for TRß mutants was shown compared to TRIAC and NH-3 and could explain the different efficiencies of the different ligands in releasing corepressors from the studied TRß mutants. Consequently, TRIAC was found to be more effective than T3 in facilitating coactivator recruitment and decreasing the dominant activity of TRß-Δ430. CONCLUSION: This study helps to clarify the specific interaction surfaces involved in the pathologic phenotype of TR mutants and demonstrates that TRIAC is a potential therapeutic agent for patients suffering from resistance to thyroid hormone syndromes.


Subject(s)
Co-Repressor Proteins/metabolism , Mutation , Receptors, Thyroid Hormone/metabolism , Thyroid Hormone Resistance Syndrome/metabolism , Thyroid Hormones/chemistry , Anisotropy , Humans , Kinetics , Ligands , Magnetic Resonance Spectroscopy , Nuclear Proteins/metabolism , Peptides/chemistry , Protein Binding , Receptors, Thyroid Hormone/genetics , Repressor Proteins/genetics , Spectrometry, Fluorescence , Thyroid Gland/metabolism , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormone Resistance Syndrome/genetics
5.
Mol Cell Endocrinol ; 447: 87-97, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28257829

ABSTRACT

Thyroid hormone receptors (TRs) play crucial roles in vertebrates. Wild-type (WT) TRs function primarily as hormone-regulated transcription factors. A human endocrine disease, Resistance to Thyroid Hormone (RTH)-Syndrome, is caused by inheritance of mutant TRs impaired in the proper regulation of target gene expression. To better understand the molecular basis of RTH we compared the target genes regulated by an RTH-TRß1 mutant (R429Q) to those regulated by WT-TRß1. With only a few potential exceptions, the vast majority of genes we were able to identify as regulated by the WT-TRß1, positively or negatively, were also regulated by the RTH-TRß1 mutant. We conclude that the actions of R429Q-TRß1 in RTH-Syndrome most likely reflect the reduced hormone affinity observed for this mutant rather than an alteration in target gene repertoire. Our results highlight the importance of target gene specificity in defining the disease phenotype and improve our understanding of how clinical treatments impact RTH-Syndrome.


Subject(s)
Mutation/genetics , Receptors, Thyroid Hormone/metabolism , Thyroid Hormone Resistance Syndrome/genetics , Adenoviridae/metabolism , Gene Expression Regulation/drug effects , Gene Ontology , Genetic Vectors/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Mutant Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Triiodothyronine/pharmacology
6.
BMC Evol Biol ; 16(1): 221, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27756201

ABSTRACT

BACKGROUND: SMRT and NCoR are corepressor paralogs that help mediate transcriptional repression by a variety of transcription factors, including the nuclear hormone receptors. The functions of both corepressors are extensively diversified in mice by alternative mRNA splicing, generating a series of protein variants that differ in different tissues and that exert different, even diametrically opposite, biochemical and biological effects from one another. RESULTS: We report here that the alternative splicing previously reported for SMRT appears to be a relatively recent evolutionary phenomenon, with only one of these previously identified sites utilized in a teleost fish and a limited additional number of the additional known sites utilized in a bird, reptile, and marsupial. In contrast, extensive SMRT alternative splicing at these sites was detected among the placental mammals. The alternative splicing of NCoR previously identified in mice (and shown to regulate lipid and carbohydrate metabolism) is likely to have arisen separately and after that of SMRT, and includes an example of convergent evolution. CONCLUSIONS: We propose that the functions of both SMRT and NCoR have been diversified by alternative splicing during evolution to allow customization for different purposes in different tissues and different species.


Subject(s)
Alternative Splicing/genetics , Co-Repressor Proteins/genetics , Evolution, Molecular , Animals , Co-Repressor Proteins/metabolism , Humans , Liver/metabolism , Mice , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Opossums/genetics , RNA Splice Sites/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sheep/genetics , Species Specificity , Xenopus/genetics , Zebrafish/genetics
7.
Mol Cell Endocrinol ; 413: 228-35, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26166430

ABSTRACT

Alternative mRNA splicing diversifies the products encoded by the NCoR and SMRT corepressor loci. There is a programmed alteration in NCoR mRNA splicing during adipocyte differentiation from an NCoRω isoform, which contains three nuclear receptor interaction domains, to an NCoRδ isoform that contains two nuclear receptor interaction domains. This alternative mRNA splicing of NCoR has profound effects on adiposity and on diabetes in mouse models. We report here that dexamethasone, a powerful regulator of metabolism and of adipocyte differentiation, confers this change in NCoR mRNA splicing in cultured adipocytes. We also demonstrate that changes in dietary components can consistently, if moderately, modulate the total transcript levels and the mRNA splicing of NCoR and SMRT in both cultured cells and intact mice. This ability of alternative corepressor mRNA splicing to respond to nutritional changes confirms its importance in regulating glucose and lipid metabolism, and its promise as a therapeutic candidate for metabolic disorders such as type 2 diabetes.


Subject(s)
Adipocytes/metabolism , Alternative Splicing/drug effects , Dexamethasone/pharmacology , Glucose/pharmacology , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/cytology , Alternative Splicing/genetics , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Glucose/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Mice , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 2/genetics , RNA, Messenger/genetics , Signal Transduction/genetics
8.
Mol Cell Biol ; 34(22): 4104-14, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25182530

ABSTRACT

Alternative mRNA splicing is an important means of diversifying function in higher eukaryotes. Notably, both NCoR and SMRT corepressors are subject to alternative mRNA splicing, yielding a series of distinct corepressor variants with highly divergent functions. Normal adipogenesis is associated with a switch in corepressor splicing from NCoRω to NCoRδ, which appears to help regulate this differentiation process. We report here that mimicking this development switch in mice by a splice-specific whole-animal ablation of NCoRω is very different from a whole-animal or tissue-specific total NCoR knockout and produces significantly enhanced weight gain on a high-fat diet. Surprisingly, NCoRω(-/-) mice are protected against diet-induced glucose intolerance despite enhanced adiposity and the presence of multiple additional, prodiabetic phenotypic changes. Our results indicate that the change in NCoR splicing during normal development both helps drive normal adipocyte differentiation and plays a key role in determining a metabolically appropriate storage of excess calories. We also conclude that whole-gene "knockouts" fail to reveal how important gene products are customized, tailored, and adapted through alternative mRNA splicing and thus do not reveal all the functions of the protein products of that gene.


Subject(s)
Alternative Splicing , Fatty Liver/genetics , Glucose Intolerance/genetics , Liver/pathology , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 2/genetics , Weight Gain , Adipogenesis , Animals , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Deletion , Glucose Intolerance/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
9.
Article in English | MEDLINE | ID: mdl-24744751

ABSTRACT

Thyroid hormones (THs) are essential for fetal and post-natal nervous system development and also play an important role in the maintenance of adult brain function. Of the two major THs, T4 (3,5,3',5'-tetraiodo-l-thyronine) is classically viewed as an pro-hormone that must be converted to T3 (3,5,3'-tri-iodo-l-thyronine) via tissue-level deiodinases for biological activity. THs primarily mediate their effects by binding to thyroid hormone receptor (TR) isoforms, predominantly TRα1 and TRß1, which are expressed in different tissues and exhibit distinctive roles in endocrinology. Notably, the ability to respond to T4 and to T3 differs for the two TR isoforms, with TRα1 generally more responsive to T4 than TRß1. TRα1 is also the most abundantly expressed TR isoform in the brain, encompassing 70-80% of all TR expression in this tissue. Conversion of T4 into T3 via deiodinase 2 in astrocytes has been classically viewed as critical for generating local T3 for neurons. However, deiodinase-deficient mice do not exhibit obvious defectives in brain development or function. Considering that TRα1 is well-established as the predominant isoform in brain, and that TRα1 responds to both T3 and T4, we suggest T4 may play a more active role in brain physiology than has been previously accepted.

10.
Mol Endocrinol ; 28(5): 745-57, 2014 May.
Article in English | MEDLINE | ID: mdl-24673558

ABSTRACT

T4 (3,5,3',5'-tetraiodo-l-thyronine) is classically viewed as a prohormone that must be converted to the T3 (3,5,3'-triiodo-l-thyronine) form for biological activity. We first determined that the ability of reporter genes to respond to T4 and to T3 differed for the different thyroid hormone receptor (TR) isoforms, with TRα1 generally more responsive to T4 than was TRß1. The response to T4 vs T3 also differed dramatically in different cell types in a manner that could not be attributed to differences in deiodinase activity or in hormone affinity, leading us to examine the role of TR coregulators in this phenomenon. Unexpectedly, several coactivators, such as steroid receptor coactivator-1 (SRC1) and thyroid hormone receptor-associated protein 220 (TRAP220), were recruited to TRα1 nearly equally by T4 as by T3 in vitro, indicating that TRα1 possesses an innate potential to respond efficiently to T4 as an agonist. In contrast, release of corepressors, such as the nuclear receptor coreceptor NCoRω, from TRα1 by T4 was relatively inefficient, requiring considerably higher concentrations of this ligand than did coactivator recruitment. Our results suggest that cells, by altering the repertoire and abundance of corepressors and coactivators expressed, may regulate their ability to respond to T4, raising the possibility that T4 may function directly as a hormone in specific cellular or physiological contexts.


Subject(s)
Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism , Thyroxine/physiology , 3T3-L1 Cells , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Mice , Protein Isoforms/agonists , Protein Isoforms/metabolism , Signal Transduction , Thyroid Hormone Receptors alpha/agonists , Thyroid Hormone Receptors beta/agonists , Transcriptional Activation , Triiodothyronine/physiology
11.
Mol Cell Endocrinol ; 383(1-2): 80-95, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24325866

ABSTRACT

Thyroid hormone receptors (TRs) are expressed primarily as two major isoforms, TRα1 and TRß1, which are expressed at different times in development and at different tissue abundances in the adult. The transcription properties and biological properties of TRα1 and TRß1 can differ. We report here that although overlapping, TRα1 and TRß1 recruit distinct panels of partner proteins that may account for their divergent biological functions, and which appear to explain their distinct target gene regulatory properties.


Subject(s)
Gene Expression Regulation , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/genetics , Animals , Genes, Reporter , HEK293 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Protein Binding , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sf9 Cells , Signal Transduction , Spodoptera , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism
12.
Environ Sci Technol ; 47(20): 11776-84, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24066858

ABSTRACT

Prenatal exposures to polybrominated diphenyl ethers (PBDEs) can harm neurodevelopment in humans and animals. In 2003-2004, PentaBDE and OctaBDE were banned in California and phased-out of US production; resulting impacts on human exposures are unknown. We previously reported that median serum concentrations of PBDEs and their metabolites (OH-PBDEs) among second trimester pregnant women recruited from San Francisco General Hospital (2008-2009; n = 25) were the highest among pregnant women worldwide. We recruited another cohort from the same clinic in 2011-2012 (n = 36) and now compare serum concentrations of PBDEs, OH-PBDEs, polychlorinated biphenyl ethers (PCBs) (structurally similar compounds banned in 1979), and OH-PCBs between two demographically similar cohorts. Between 2008-2009 and 2011-2012, adjusted least-squares geometric mean (LSGM) concentrations of ∑PBDEs decreased 65% (95% CI: 18, 130) from 90.0 ng/g lipid (95% CI: 64.7, 125.2) to 54.6 ng/g lipid (95% CI: 39.2, 76.2) (p = 0.004); ∑OH-PBDEs decreased 6-fold (p < 0.0001); and BDE-47, -99, and -100 declined more than BDE-153. There was a modest, nonsignificant (p = 0.13) decline in LSGM concentrations of ∑PCBs and minimal differences in ∑OH-PCBs between 2008-2009 and 2011-2012. PBDE exposures are likely declining due to regulatory action, but the relative stability in PCB exposures suggests PBDE exposures may eventually plateau and persist for decades.


Subject(s)
Halogenated Diphenyl Ethers/blood , Hospitals, General , Polychlorinated Biphenyls/blood , Pregnancy Trimester, Second/blood , Adolescent , Adult , Cohort Studies , Demography , Female , Humans , Least-Squares Analysis , Maternal Age , Multivariate Analysis , Pregnancy , San Francisco , Time Factors , Young Adult
13.
Mol Endocrinol ; 27(5): 840-59, 2013 May.
Article in English | MEDLINE | ID: mdl-23558175

ABSTRACT

Thyroid hormone receptors (TRs) are expressed as a series of interrelated isoforms that perform distinct biological roles. The TRß2 isoform is found predominantly in the hypothalamus, pituitary, retina, and cochlea and displays unique transcriptional properties relative to the other TR isoforms. To more fully understand the isoform-specific biological and molecular properties of TRß2, we have identified a series of previously unrecognized proteins that selectively interact with TRß2 compared with the more widely expressed TRß1. Several of these proteins preferentially enhance the transcriptional activity of TRß2 when coexpressed in cells and are likely to represent novel, isoform-specific coactivators. Additional proteins were also identified in our screen that bind equally to TRß1 and TRß2 and may function as isoform-independent auxiliary proteins for these and/or other nuclear receptors. We propose that a combination of isoform-specific recruitment and tissue-specific expression of these newly identified coregulator candidates serves to customize TR function for different biological purposes in different cell types.


Subject(s)
Nuclear Proteins/metabolism , Thyroid Hormone Receptors beta/metabolism , Amino Acid Sequence , Animals , Cell Extracts , Cell Line , Chromatography, Liquid , DNA/metabolism , Genes, Reporter , Humans , Immunoprecipitation , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Organ Specificity/genetics , Protein Binding , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Response Elements/genetics , Thyroid Hormone Receptors beta/chemistry , Thyroid Hormone Receptors beta/genetics , Transcriptional Activation
14.
J Clin Invest ; 122(7): 2428-38, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22684107

ABSTRACT

For more than a century, thyroid hormones (THs) have been known to exert powerful catabolic effects, leading to weight loss. Although much has been learned about the molecular mechanisms used by TH receptors (TRs) to regulate gene expression, little is known about the mechanisms by which THs increase oxidative metabolism. Here, we report that TH stimulation of fatty acid ß-oxidation is coupled with induction of hepatic autophagy to deliver fatty acids to mitochondria in cell culture and in vivo. Furthermore, blockade of autophagy by autophagy-related 5 (ATG5) siRNA markedly decreased TH-mediated fatty acid ß-oxidation in cell culture and in vivo. Consistent with this model, autophagy was altered in livers of mice expressing a mutant TR that causes resistance to the actions of TH as well as in mice with mutant nuclear receptor corepressor (NCoR). These results demonstrate that THs can regulate lipid homeostasis via autophagy and help to explain how THs increase oxidative metabolism.


Subject(s)
Lipid Metabolism , Liver/metabolism , Triiodothyronine/physiology , Animals , Autophagy , Autophagy-Related Protein 5 , Carnitine/analogs & derivatives , Carnitine/metabolism , Cell Line, Tumor , Fatty Acids/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Hepatocytes/metabolism , Hepatocytes/physiology , Hepatocytes/ultrastructure , Histone Deacetylases/metabolism , Humans , Ketone Bodies/metabolism , Male , Metabolic Networks and Pathways/genetics , Metabolome , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Phagosomes/metabolism , Protein Binding , RNA Interference , Receptors, Thyroid Hormone/metabolism
15.
Mol Cell Endocrinol ; 351(2): 306-16, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22266197

ABSTRACT

The SMRT and NCoR corepressors bind to, and mediate transcriptional repression by, many nuclear receptors. Both SMRT and NCoR are expressed by alternative mRNA splicing, generating a series of structurally and functionally distinct corepressor "variants". We report that a splice variant of SMRT, SMRTε, recognizes a restricted subset of nuclear receptors. Unlike the other corepressor variants characterized, SMRTε possesses only a single receptor interaction domain (RID) and exhibits an unusual specificity for a subset of nuclear receptors that includes the retinoic acid receptors (RARs). The ability of the single RID in SMRTε to efficiently interact with RARs appears to be enhanced by a recently recognized ß-strand/ß-strand interaction between corepressor and receptor. We suggest that alternative mRNA splicing of corepressors can restrict their function to specific nuclear receptor partnerships, and we propose that this may serve to customize the transcriptional repression properties of different cell types for different biological purposes.


Subject(s)
Nuclear Receptor Co-Repressor 2/metabolism , Receptors, Retinoic Acid/metabolism , Alternative Splicing , Animals , Cell Line, Tumor , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Humans , Liver X Receptors , Mice , Nuclear Receptor Co-Repressor 1/metabolism , Orphan Nuclear Receptors/metabolism , Protein Structure, Secondary/physiology , Retinoic Acid Receptor alpha , Retinoic Acid Receptor gamma
16.
J Biol Chem ; 286(52): 44988-99, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22065574

ABSTRACT

The SMRT and NCoR corepressors partner with, and help mediate repression by, a wide variety of nuclear receptors and non-receptor transcription factors. Both SMRT and NCoR are expressed by alternative mRNA splicing, resulting in the production of a series of interrelated corepressor variants that differ in their tissue distribution and in their biochemical properties. We report here that different corepressor splice variants can exert opposing transcriptional and biological effects during adipocyte differentiation. Most notably, the NCoRω splice variant inhibits, whereas the NCoRδ splice variant promotes, adipogenesis. Furthermore, the ratio of NCoRω to NCoRδ decreases during adipogenic differentiation. We propose that this alteration in corepressor splicing helps convert the cellular transcriptional program from one that maintains the pre-adipocyte in an undifferentiated state to a new transcriptional context that promotes differentiation and helps establish the proper physiology of the mature adipocyte.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Alternative Splicing/physiology , Cell Differentiation/physiology , Nuclear Receptor Co-Repressor 2/biosynthesis , Transcription, Genetic/physiology , 3T3-L1 Cells , Adipocytes/cytology , Animals , Mice , Nuclear Receptor Co-Repressor 2/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics
17.
J Thyroid Res ; 2011: 361304, 2011.
Article in English | MEDLINE | ID: mdl-21760978

ABSTRACT

Thyroid hormone, operating through its receptors, plays crucial roles in the control of normal human physiology and development; deviations from the norm can give rise to disease. Clinical endocrinologists often must confront and correct the consequences of inappropriately high or low thyroid hormone synthesis. Although more rare, disruptions in thyroid hormone endocrinology due to aberrations in the receptor also have severe medical consequences. This review will focus on the afflictions that are caused by, or are closely associated with, mutated thyroid hormone receptors. These include Resistance to Thyroid Hormone Syndrome, erythroleukemia, hepatocellular carcinoma, renal clear cell carcinoma, and thyroid cancer. We will describe current views on the molecular bases of these diseases, and what distinguishes the neoplastic from the non-neoplastic. We will also touch on studies that implicate alterations in receptor expression, and thyroid hormone levels, in certain oncogenic processes.

18.
Mol Endocrinol ; 25(7): 1111-25, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21622532

ABSTRACT

Thyroid hormone receptors (TR) are hormone-modulated transcription factors that regulate overall metabolic rate, lipid utilization, heart rate, and development. TR are expressed as a mix of interrelated receptor isoforms. The TRß2 isoform is expressed in the hypothalamus and pituitary, where it plays an important role in the feedback regulation of thyroid hormone levels. TRß2 exhibits unique transcriptional properties that parallel the ability of this isoform to bind to certain coactivators cooperatively through multiple contact surfaces. The more peripherally expressed TRß1 isoform, in contrast, appears to recruit these coactivators through a single contact mechanism. We report here that clusters of charged amino acids in the TR hormone-binding domain are required for this enhanced mode of coactivator recruitment and that mutations in these charge clusters, by disrupting TRß2 coactivator binding, are a molecular basis for pituitary resistance to thyroid hormone, a disease characterized by inappropriate thyroid hormone feedback regulation. We propose that the charge clusters allow wild-type TRß2 to assume a conformation compatible with its mode of multiple contact coactivator recruitment, whereas disruption of these charge clusters disrupts normal T(3) homeostasis by reducing TRß2 to a TRß1-like, single contact mode of coactivator binding.


Subject(s)
Nuclear Receptor Coactivator 1/metabolism , Thyroid Hormone Receptors beta/chemistry , Thyroid Hormone Resistance Syndrome/metabolism , Amino Acid Motifs , Arginine/chemistry , Cell Line , Genes, Reporter , Humans , Immobilized Proteins/chemistry , Luciferases/biosynthesis , Luciferases/genetics , Mutation, Missense , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Stability , Protein Structure, Tertiary , Response Elements , Surface Properties , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormone Resistance Syndrome/genetics , Transcription, Genetic
19.
Mol Endocrinol ; 25(8): 1311-25, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21622534

ABSTRACT

Thyroid hormone receptors (TRs) are hormone-regulated transcription factors that regulate a diverse array of biological activities, including metabolism, homeostasis, and development. TRs also serve as tumor suppressors, and aberrant TR function (via mutation, deletion, or altered expression) is associated with a spectrum of both neoplastic and endocrine diseases. A particularly high frequency of TR mutations has been reported in renal clear cell carcinoma (RCCC) and in hepatocellular carcinoma (HCC). We have shown that HCC-TR mutants regulate only a fraction of the genes targeted by wild-type TRs but have gained the ability to regulate other, unique, targets. We have suggested that this altered gene recognition may contribute to the neoplastic phenotype. Here, to determine the generality of this phenomenon, we examined a distinct set of TR mutants associated with RCCC. We report that two different TR mutants, isolated from independent RCCC tumors, possess greatly expanded target gene specificities that extensively overlap one another, but only minimally overlap that of the wild-type TRs, or those of two HCC-TR mutants. Many of the genes targeted by either or both RCCC-TR mutants have been previously implicated in RCCC and include a series of metallothioneins, solute carriers, and genes involved in glycolysis and energy metabolism. We propose as a hypothesis that TR mutations from RCCC and HCC may play tissue-specific roles in carcinogenesis, and that the divergent target gene recognition patterns of TR mutants isolated from the two different types of tumors may arise from different selective pressures during development of RCCC vs. HCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Kidney Neoplasms/genetics , Mutant Proteins/isolation & purification , Mutant Proteins/metabolism , Receptors, Thyroid Hormone/metabolism , Carcinoma, Hepatocellular/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Repressor Proteins/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Triiodothyronine/pharmacology
20.
J Mol Endocrinol ; 46(3): 233-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21389087

ABSTRACT

We previously showed that two thyroid hormone receptor (TR) isoforms--TRα1 and TRß1--differentially regulate thyroid hormone (triiodothyroxine, T(3))-stimulated adipogenesis in vivo. This study aims to understand the role of the nuclear receptor corepressor, NCoR1, in TR isoform-dependent adipogenesis. We found that T(3)-stimulated adipogenesis of 3T3-L1 cells was accompanied by progressive loss of NCoR1 protein levels. In 3T3-L1 cells stably expressing a mutated TRα1, PV (L1-α1PV cells), the T(3)-stimulated adipogenesis was more strongly inhibited than that in 3T3-L1 cells stably expressing an identical mutation in TRß1 (L1-ß1PV cells). The stronger inhibition of adipogenesis in L1-α1PV cells was associated with a higher NCoR1 protein level. These results indicate that the degree of loss of NCoR1 correlates with the extent of adipogenesis. siRNA knockdown of NCoR1 promoted adipogenesis of control 3T3-L1 cells and reversed the inhibited adipogenesis of L1-α1PV and L1-ß1PV cells, indicating that NCoR1 plays an essential role in TR isoform-dependent adipogenesis. An ubiquitin ligase, mSiah2, that targets NCoR1 for proteasome degradation was upregulated on day 1 before the onset of progressive loss of NCoR1. NCoR1 was found to associate with mSiah2 and with TR, TRα1PV, or TRß1PV, but a stronger interaction of NCoR1 with TRα1PV than with TRß1PV was detected. Furthermore, TRα1PV-NCoR1 complex was more avidly recruited than TRß1PV-NCoR1 to the promoter of the C/ebpα gene, leading to more inhibition in its expression. These results indicate that differential interaction of NCoR1 with TR isoforms accounted for the TR isoform-dependent regulation of adipogenesis and that aberrant interaction of NCoR1 with TR could underlie the pathogenesis of lipid disorders in hypothyroidism.


Subject(s)
Adipocytes/physiology , Adipogenesis/physiology , Nuclear Receptor Co-Repressor 1/metabolism , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/physiology , Chromatin Immunoprecipitation , Down-Regulation , Humans , Mice , Mutation , Nuclear Receptor Co-Repressor 1/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , Promoter Regions, Genetic , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/genetics , Time Factors , Transcriptional Activation , Triiodothyronine/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...