scholarly journals Thyroid hormone receptor alpha sumoylation modulates white adipose tissue stores

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yan-Yun Liu ◽  
Jingjing Jiang ◽  
Sujie Ke ◽  
Anna Milanesi ◽  
Kiyomi Abe ◽  
...  

AbstractThyroid hormone (TH) and thyroid hormone receptor (THR) regulate stem cell proliferation and differentiation during development, as well as during tissue renewal and repair in the adult. THR undergoes posttranslational modification by small ubiquitin-like modifier (SUMO). We generated the THRA (K283Q/K288R)−/− mouse model for in vivo studies and used human primary preadipocytes expressing the THRA sumoylation mutant (K283R/K288R) and isolated preadipocytes from mutant mice for in vitro studies. THRA mutant mice had reduced white adipose stores and reduced adipocyte cell diameter on a chow diet, compared to wild-type, and these differences were further enhanced after a high fat diet. Reduced preadipocyte proliferation in mutant mice, compared to wt, was shown after in vivo labeling of preadipocytes with EdU and in preadipocytes isolated from mice fat stores and studied in vitro. Mice with the desumoylated THRA had disruptions in cell cycle G1/S transition and this was associated with a reduction in the availability of cyclin D2 and cyclin-dependent kinase 2. The genes coding for cyclin D1, cyclin D2, cyclin-dependent kinase 2 and Culin3 are stimulated by cAMP Response Element Binding Protein (CREB) and contain CREB Response Elements (CREs) in their regulatory regions. We demonstrate, by Chromatin Immunoprecipitation (ChIP) assay, that in mice with the THRA K283Q/K288R mutant there was reduced CREB binding to the CRE. Mice with a THRA sumoylation mutant had reduced fat stores on chow and high fat diets and reduced adipocyte diameter.

PLoS ONE ◽  
2020 ◽  
Vol 15 (12) ◽  
pp. e0240338
Author(s):  
Xuan G. Luong ◽  
Sarah K. Stevens ◽  
Andreas Jekle ◽  
Tse-I Lin ◽  
Kusum Gupta ◽  
...  

Thyroid hormones are important modulators of metabolic activity in mammals and alter cholesterol and fatty acid levels through activation of the nuclear thyroid hormone receptor (THR). Currently, there are several THRβ agonists in clinical trials for the treatment of non-alcoholic steatohepatitis (NASH) that have demonstrated the potential to reduce liver fat and restore liver function. In this study, we tested three THRβ-agonism-based NASH treatment candidates, GC-1 (sobetirome), MGL-3196 (resmetirom), and VK2809, and compared their selectivity for THRβ and their ability to modulate the expression of genes specific to cholesterol and fatty acid biosynthesis and metabolism in vitro using human hepatic cells and in vivo using a rat model. Treatment with GC-1 upregulated the transcription of CPT1A in the human hepatocyte-derived Huh-7 cell line with a dose-response comparable to that of the native THR ligand, triiodothyronine (T3). VK2809A (active parent of VK2809), MGL-3196, and VK2809 were approximately 30-fold, 1,000-fold, and 2,000-fold less potent than T3, respectively. Additionally, these relative potencies were confirmed by quantification of other direct gene targets of THR, namely, ANGPTL4 and DIO1. In primary human hepatocytes, potencies were conserved for every compound except for VK2809, which showed significantly increased potency that was comparable to that of its active counterpart, VK2809A. In high-fat diet fed rats, a single dose of T3 significantly reduced total cholesterol levels and concurrently increased liver Dio1 and Me1 RNA expression. MGL-3196 treatment resulted in concentration-dependent decreases in total and low-density lipoprotein cholesterol with corresponding increases in liver gene expression, but the compound was significantly less potent than T3. In conclusion, we have implemented a strategy to rank the efficacy of THRβ agonists by quantifying changes in the transcription of genes that lead to metabolic alterations, an effect that is directly downstream of THR binding and activation.


Endocrinology ◽  
2003 ◽  
Vol 144 (2) ◽  
pp. 552-558 ◽  
Author(s):  
H. C. van Beeren ◽  
W. M. C. Jong ◽  
E. Kaptein ◽  
T. J. Visser ◽  
O. Bakker ◽  
...  

Dronedarone (Dron), without iodine, was developed as an alternative to the iodine-containing antiarrhythmic drug amiodarone (AM). AM acts, via its major metabolite desethylamiodarone, in vitro and in vivo as a thyroid hormone receptor α1 (TRα1) and TRβ1 antagonist. Here we investigate whether Dron and/or its metabolite debutyldronedarone inhibit T3 binding to TRα1 and TRβ1in vitro and whether dronedarone behaves similarly to amiodarone in vivo. In vitro , Dron had a inhibitory effect of 14% on the binding of T3 to TRα1, but not on TRβ1. Desethylamiodarone inhibited T3 binding to TRα1 and TRβ1 equally. Debutyldronedarone inhibited T3 binding to TRα1 by 77%, but to TRβ1 by only 25%. In vivo , AM increased plasma TSH and rT3, and decreased T3. Dron decreased T4 and T3, rT3 did not change, and TSH fell slightly. Plasma total cholesterol was increased by AM, but remained unchanged in Dron-treated animals. TRβ1-dependent liver low density lipoprotein receptor protein and type 1 deiodinase activities decreased in AM-treated, but not in Dron-treated, animals. TRα1-mediated lengthening of the QTc interval was present in both AM- and Dron-treated animals. The in vitro and in vivo findings suggest that dronedarone via its metabolite debutyldronedarone acts as a TRα1-selective inhibitor.


2020 ◽  
Author(s):  
Xuan G. Luong ◽  
Sarah K. Stevens ◽  
Andreas Jekle ◽  
Tse-I Lin ◽  
Kusum Gupta ◽  
...  

AbstractThyroid hormones are important modulators of metabolic activity in mammals and alter cholesterol and fatty acid levels through activation of the nuclear thyroid hormone receptor (THR). Currently, there are several THRβ agonists in clinical trials for the treatment of non-alcoholic steatohepatitis (NASH) that have demonstrated the potential to reduce liver fat and restore liver function. In this study, we tested three THRβ-agonism-based NASH treatment candidates, GC-1 (sobetirome), MGL-3196 (resmetirom), and VK2809, and compared their selectivity for THRβ and their ability to modulate the expression of genes specific to cholesterol and fatty acid biosynthesis and metabolism in vitro using human hepatic cells and in vivo using a rat model. Treatment with GC-1 upregulated the transcription of CPT1A in the human hepatocyte-derived Huh-7 cell line with a dose-response comparable to that of the native THR ligand, triiodothyronine (T3). VK2809A (active parent of VK2809), MGL-3196, and VK2809 were approximately 30-fold, 1,000-fold, and 2,000-fold less potent than T3, respectively. Additionally, these relative potencies were confirmed by quantification of other direct gene targets of THR, namely, ANGPTL4 and DIO1. In primary human hepatocytes, potencies were conserved for every compound except for VK2809, which showed significantly increased potency that was comparable to that of its active counterpart, VK2809A. In high-fat diet fed rats, a single dose of T3 significantly reduced total cholesterol levels and concurrently increased liver Dio1 and Me1 RNA expression. MGL-3196 treatment resulted in concentration-dependent decreases in total and low-density lipoprotein cholesterol with corresponding increases in liver gene expression, but the compound was significantly less potent than T3. In conclusion, we have implemented a strategy to rank the efficacy of THRβ agonists by quantifying changes in the transcription of genes that lead to metabolic alterations, an effect that is directly downstream of THR binding and activation.


Endocrinology ◽  
2014 ◽  
Vol 155 (9) ◽  
pp. 3713-3724 ◽  
Author(s):  
Negin P. Martin ◽  
Ezequiel Marron Fernandez de Velasco ◽  
Fengxia Mizuno ◽  
Erica L. Scappini ◽  
Bernd Gloss ◽  
...  

Abstract Several rapid physiological effects of thyroid hormone on mammalian cells in vitro have been shown to be mediated by the phosphatidylinositol 3-kinase (PI3K), but the molecular mechanism of PI3K regulation by nuclear zinc finger receptor proteins for thyroid hormone and its relevance to brain development in vivo have not been elucidated. Here we show that, in the absence of hormone, the thyroid hormone receptor TRβ forms a cytoplasmic complex with the p85 subunit of PI3K and the Src family tyrosine kinase, Lyn, which depends on two canonical phosphotyrosine motifs in the second zinc finger of TRβ that are not conserved in TRα. When hormone is added, TRβ dissociates and moves to the nucleus, and phosphatidylinositol (3, 4, 5)-trisphosphate production goes up rapidly. Mutating either tyrosine to a phenylalanine prevents rapid signaling through PI3K but does not prevent the hormone-dependent transcription of genes with a thyroid hormone response element. When the rapid signaling mechanism was blocked chronically throughout development in mice by a targeted point mutation in both alleles of Thrb, circulating hormone levels, TRβ expression, and direct gene regulation by TRβ in the pituitary and liver were all unaffected. However, the mutation significantly impaired maturation and plasticity of the Schaffer collateral synapses on CA1 pyramidal neurons in the postnatal hippocampus. Thus, phosphotyrosine-dependent association of TRβ with PI3K provides a potential mechanism for integrating regulation of development and metabolism by thyroid hormone and receptor tyrosine kinases.


2001 ◽  
Author(s):  
Γεωργία Μπράλιου

Η θυρεοειδής ορμόνη (Τ3) παίζει πολλαπλό και σημαντικό ρόλο στην ομοιόσταση, στην ανάπτυξη, στην μεταμόρφωση των αμφίβιων, στη διαφοροποίηση και στη νεοπλασία. Η δράση της Τ3 διενεργείται μέσω της σύνδεσής της με τον υποδοχέα θυρεοειδούς ορμόνης (TR), ο οποίος με τη σειρά του συνδέεται σε ειδικές αλληλουχίες DNA που ονομάζονται στοιχεία απόκρισης στη θυρεοειδή ορμόνη (TREs). Ο TR παρουσία Τ3 ενεργοποιεί τη μεταγραφή ενώ χωρίς ορμόνη ο TR την καταστέλλει χρησιμοποιώντας πληθώρα μηχανισμών. Η v-ErbΑ ογκοπρωτεϊνη είναι μια ιική μεταλλαγμένη παραλλαγή του TRα που κωδικοποιείται από τον ιό της ερυθροβλάστωσης των πτηνών (AEV). Η συνεργασία του v-erbA με το v-erbB, το δεύτερο ογκογονίδιο του AEV με δραστικότητα κινάσης τυροσίνης, οδηγεί σε ερυθρολευχαιμία ως αποτέλεσμα του ανεξέλεγκτου πολλαπλασιασμού και της παύσης της διαφοροποίησης. Λόγω μεταλλάξεων, το ν-erbΑ δεν μπορεί να δεσμεύσει Τ3 και έτσι δεν μπορεί να ενεργοποιήσει τη μεταγραφή, όπως το κυτταρικό ομόλογό του, ο TR. Η υπόθεση ότι το v-ErbA συμπεριφέρεται ως TR χωρίς προσδεμένη ορμόνη είναι ελκυστική, αλλά αντιβαίνει σε μια σειρά από πειραματικές παρατηρήσεις. Ο στόχος των μελετών της παρούσας διατριβής είναι να διαλευκάνουν τους μοριακούς μηχανισμούς που υπαγορεύουν την ογκογόνο δράση του v-erbA.Χρησιμοποιώντας χαρτογράφηση υπερευαισθησίας σε DNAάση Ι εντοπίσαμε δύο υποψήφιες περιοχές (HS1 και HS2) ρύθμισης της μεταγραφής του ερυθροειδικού γονιδίου της καρβονικής ανυδράσης ΙΙ (CA II) από το v-erbA. Η HS1 βρίσκεται 7 kb ανοδικά (3’) ενώ HS2 βρίσκεται 8 kb καθοδικά (5’) της θέσης έναρξης μεταγραφής, εντός του δεύτερου ιντρονίου. Πειράματα ανοσοκατακρήμνισης συμπλόκων v-ErbA/DNA καθώς και in vitro και in vivo footprinting πειράματα απέδειξαν ότι η ιντρονική περιοχή HS2 έχει μία θέση δέσμευσης για v-ErbΑ (VRE). Σημαντικό είναι ότι η in vivo κατάληψη του VRE από το v-ErbΑ συμπίπτει με καταστολή της μεταγραφής της CA ΙΙ, ενώ όταν τα κύτταρα ενεργοποιούνται για διαφοροποίηση και εκφράζεται η CA II, τότε το v-ErbA δεν προσδένεται στο VRE. Χρησιμοποιώντας ανάλυση μεταλλαξιγένεσης και πειράματα επιμόλυνσης κυττάρων αποδείξαμε ότι η περιοχή HS2 ενεργοποιεί τη μεταγραφή ανεξάρτητα από τη θέση και τον προσανατολισμό της σε σχέση με υποκινητές, και ότι η δραστικότητα ως ενισχυτή της HS2 εξαρτάται από ερυθροειδικούς παράγοντες που προσδένονται στις GATA αλληλουχίες που βρίσκονται γειτονικά στο VRE. Έτσι, το ν-ErbΑ φαίνεται να εξουδετερώνει την θετική μεταγραφική δραστικότητα των ερυθροειδικών GATA-παραγόντων και η HS2 συμπεριφέρεται ως ισχυρός ερυθροειδικός ενισχυτής που καταστέλλεται από το v-ErbΑ. Δείξαμε επίσης ότι η αρχιτεκτονική της χρωματίνης της HS2 ρυθμίζεται από το δεσμευμένο με ορμόνη TR σε ερυθροειδή πρωταρχικά κύτταρα και ότι το v-ErbΑ εμποδίζει την πρόσδεση του δεσμευμένου με ορμόνη TR στο VRE, με αποτέλεσμα την άρση της μεταγραφικής ενεργοποίησης που προκαλεί η Τ3. Αυτή η δράση του v-ErbA μπορεί να συσχετίζεται με την παρατηρούμενη ογκογονικότητά του. Με πειράματα ανοσοκαθίζησης δείξαμε επίσης ότι το v-ErbΑ μπορεί να αλληλεπιδράσει με τον ενδογενή συν-καταστολέα της μεταγραφής, τον NCoR. Ωστόσο, ο NCoR δεν φαίνεται να είναι ζωτικής σημασίας για την καταστολή μέσω ν-ErbΑ, ενώ άλλοι συν-καταστολείς μπορεί να είναι πιο δραστικοί σε σύμπλοκα με το ν-ErbA.Συμπερασματικά, οι μελέτες μας δίνουν νέα δεδομένα για τον μηχανισμό με τον οποίο το ν-erbΑ καταστέλλει τη μεταγραφή, δηλαδή μέσω της ρύθμισης του ιντρονικού ενισχυτή HS2 του ερυθροειδικού γονιδίου της CA II και διαμορφώνοντας την αρχιτεκτονική της χρωματίνης στην περιοχή της CA II μέσω αλληλεπιδράσεων με άλλες πρωτεΐνες, και παρουσιάζουν πιθανούς μηχανισμούς που εξηγούν το ρόλο του v-erbΑ στο λευχαιμικό μετασχηματισμό.


Endocrinology ◽  
2011 ◽  
Vol 152 (3) ◽  
pp. 1136-1142 ◽  
Author(s):  
Carmen Grijota-Martínez ◽  
Eric Samarut ◽  
Thomas S. Scanlan ◽  
Beatriz Morte ◽  
Juan Bernal

Thyroid hormone analogs with selective actions through specific thyroid hormone receptor (TR) subtypes are of great interest. They might offer the possibility of mimicking physiological actions of thyroid hormone with receptor subtype or tissue specificity with therapeutic aims. They are also pharmacological tools to dissect biochemical pathways mediated by specific receptor subtypes, in a complementary way to mouse genetic modifications. In this work, we studied the in vivo activity in developing rats of two thyroid hormone agonists, the TRβ-selective GC-24 and the TRα-selective CO23. Our principal goal was to check whether these compounds were active in the rat brain. Analog activity was assessed by measuring the expression of thyroid hormone target genes in liver, heart, and brain, after administration to hypothyroid rats. GC-24 was very selective for TRβ and lacked activity on the brain. On the other hand, CO23 was active in liver, heart, and brain on genes regulated by either TRα or TRβ. This compound, previously shown to be TRα-selective in tadpoles, displayed no selectivity in the rat in vivo.


2019 ◽  
Vol 116 (45) ◽  
pp. 22754-22763 ◽  
Author(s):  
Teresa G. Krieger ◽  
Carla M. Moran ◽  
Alberto Frangini ◽  
W. Edward Visser ◽  
Erik Schoenmakers ◽  
...  

Mutations in the thyroid hormone receptor α 1 gene (THRA) have recently been identified as a cause of intellectual deficit in humans. Patients present with structural abnormalities including microencephaly, reduced cerebellar volume and decreased axonal density. Here, we show that directed differentiation of THRA mutant patient-derived induced pluripotent stem cells to forebrain neural progenitors is markedly reduced, but mutant progenitor cells can generate deep and upper cortical layer neurons and form functional neuronal networks. Quantitative lineage tracing shows that THRA mutation-containing progenitor cells exit the cell cycle prematurely, resulting in reduced clonal output. Using a micropatterned chip assay, we find that spatial self-organization of mutation-containing progenitor cells in vitro is impaired, consistent with down-regulated expression of cell–cell adhesion genes. These results reveal that thyroid hormone receptor α1 is required for normal neural progenitor cell proliferation in human cerebral cortical development. They also exemplify quantitative approaches for studying neurodevelopmental disorders using patient-derived cells in vitro.


2004 ◽  
Vol 24 (20) ◽  
pp. 9026-9037 ◽  
Author(s):  
Daniel R. Buchholz ◽  
Akihiro Tomita ◽  
Liezhen Fu ◽  
Bindu D. Paul ◽  
Yun-Bo Shi

ABSTRACT Thyroid hormone (T3) has long been known to be important for vertebrate development and adult organ function. Whereas thyroid hormone receptor (TR) knockout and transgenic studies of mice have implicated TR involvement in mammalian development, the underlying molecular bases for the resulting phenotypes remain to be determined in vivo, especially considering that T3 is known to have both genomic, i.e., through TRs, and nongenomic effects on cells. Amphibian metamorphosis is an excellent model for studying the role of TR in vertebrate development because of its total dependence on T3. Here we investigated the role of TR in metamorphosis by developing a dominant positive mutant thyroid hormone receptor (dpTR). In the frog oocyte transcription system, dpTR bound a T3-responsive promoter and activated the promoter independently of T3. Transgenic expression of dpTR under the control of a heat shock-inducible promoter in premetamorphic tadpoles led to precocious metamorphic transformations. Molecular analyses showed that dpTR induced metamorphosis by specifically binding to known T3 target genes, leading to increased local histone acetylation and gene activation, similar to T3-bound TR during natural metamorphosis. Our experiments indicated that the metamorphic role of T3 is through genomic action of the hormone, at least on the developmental parameters tested. They further provide the first example where TR is shown to mediate directly and sufficiently these developmental effects of T3 in individual organs by regulating target gene expression in these organs.


Sign in / Sign up

Export Citation Format

Share Document