Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Mol Oncol ; 16(22): 3975-3993, 2022 12.
Article in English | MEDLINE | ID: mdl-36217307

ABSTRACT

The THRA gene, encoding the thyroid hormone nuclear receptor TRα1, is expressed in an increasing gradient at the bottom of intestinal crypts, overlapping with high Wnt and Notch activities. Importantly, THRA is upregulated in colorectal cancers, particularly in the high-Wnt molecular subtype. The basis of this specific and/or altered expression pattern has remained unknown. To define the mechanisms controlling THRA transcription and TRα1 expression, we used multiple in vitro and ex vivo approaches. Promoter analysis demonstrated that transcription factors important for crypt homeostasis and altered in colorectal cancers, such as transcription factor 7-like 2 (TCF7L2; Wnt pathway), recombining binding protein suppressor of hairless (RBPJ; Notch pathway), and homeobox protein CDX2 (epithelial cell identity), modulate THRA activity. Specifically, although TCF7L2 and CDX2 stimulated THRA, RBPJ induced its repression. In-depth analysis of the Wnt-dependent increase showed direct regulation of the THRA promoter in cells and of TRα1 expression in murine enteroids. Given our previous results on the control of the Wnt pathway by TRα1, our new results unveil a complex regulatory loop and synergy between these endocrine and epithelial-cell-intrinsic signals. Our work describes, for the first time, the regulation of the THRA gene in specific cell and tumor contexts.


Subject(s)
Colorectal Neoplasms , Genes, erbA , Humans , Mice , Animals , Receptors, Thyroid Hormone/genetics , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormones/metabolism , Colorectal Neoplasms/genetics
2.
Development ; 148(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33757992

ABSTRACT

The thyroid hormone T3 and its nuclear receptor TRα1 control gut development and homeostasis through the modulation of intestinal crypt cell proliferation. Despite increasing data, in-depth analysis on their specific action on intestinal stem cells is lacking. By using ex vivo 3D organoid cultures and molecular approaches, we observed early responses to T3 involving the T3-metabolizing enzyme Dio1 and the transporter Mct10, accompanied by a complex response of stem cell- and progenitor-enriched genes. Interestingly, specific TRα1 loss-of-function (inducible or constitutive) was responsible for low ex vivo organoid development and impaired stem cell activity. T3 treatment of animals in vivo not only confirmed the positive action of this hormone on crypt cell proliferation but also demonstrated its key action in modulating the number of stem cells, the expression of their specific markers and the commitment of progenitors into lineage-specific differentiation. In conclusion, T3 treatment or TRα1 modulation has a rapid and strong effect on intestinal stem cells, broadening our perspectives in the study of T3/TRα1-dependent signaling in these cells.


Subject(s)
Cell Proliferation , Intestines , Signal Transduction , Stem Cells/metabolism , Thyroid Hormone Receptors alpha/metabolism , Triiodothyronine/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Female , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Male , Mice , Mice, Transgenic , Stem Cells/cytology , Thyroid Hormone Receptors alpha/genetics , Triiodothyronine/genetics
3.
Oncotarget ; 9(57): 30979-30996, 2018 Jul 24.
Article in English | MEDLINE | ID: mdl-30123421

ABSTRACT

Our previous work demonstrated a key function of the thyroid hormone nuclear receptor TRα1, a T3-modulated transcription factor, in controlling intestinal development and homeostasis via the Wnt and Notch pathways. Importantly, increased expression of TRα1 in the intestinal epithelium in a mutated Apc genetic background (vil-TRα1/Apc+/1638N mice) accelerated tumorigenesis and contributed to a more aggressive tumor phenotype compared to that of the Apc mutants alone. Therefore, the aim of this study was to determine the relevance of this synergistic effect in human colorectal cancers and to gain insights into the mechanisms involved. We analyzed cohorts of patients by in silico and experimental approaches and observed increased TRα1 expression and a significant correlation between TRα1 levels and Wnt activity. TRα1 loss-of-function and gain-of-function in Caco2 cell lines not only confirmed that TRα1 levels control Wnt activity but also demonstrated the role of TRα1 in regulating cell proliferation and migration. Finally, upon investigation of the molecular mechanisms responsible for the Wnt-TRα1 association, we described the repression by TRα1 of several Wnt inhibitors, including Frzb, Sox17 and Wif1. In conclusion, our results underline an important functional interplay between the thyroid hormone nuclear receptor TRα1 and the canonical Wnt pathway in intestinal cancer initiation and progression. More importantly, we show for the first time that the expression of TRα1 is induced in human colorectal cancers.

4.
Dev Biol ; 422(2): 71-82, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28069375

ABSTRACT

The thyroid hormones, T3 and T4, control several developmental and homeostatic processes. From a molecular point of view, most of their actions depend on the activity of the thyroid hormone nuclear receptors (TRs), which are T3-modulated transcription factors. Recent studies have not only highlighted that the physiological response induced by T3 within a cell depends on the expression of specific TRs, but also that the functions of TRs are coordinated by and integrated in other signalling pathways. This is particularly the case for the multilevel interactions between TRs and the Wnt signalling pathway. Interestingly both signals are involved in development and homeostasis, and their alterations are responsible for the development of pathologies, such as cancer. Here, we present findings on the complex crosstalk between TRs and Wnt in several organisms and in different tissue contexts, and speculate on the biological relevance of modulating TR-Wnt functionality in therapeutic approaches aimed to target cancer cells or applications for regenerative medicine.


Subject(s)
Receptors, Thyroid Hormone/metabolism , Thyroxine/metabolism , Triiodothyronine/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Humans , Wnt Signaling Pathway
5.
Development ; 142(16): 2764-74, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26286942

ABSTRACT

Thyroid hormones control various aspects of gut development and homeostasis. The best-known example is in gastrointestinal tract remodeling during amphibian metamorphosis. It is well documented that these hormones act via the TR nuclear receptors, which are hormone-modulated transcription factors. Several studies have shown that thyroid hormones regulate the expression of several genes in the Notch signaling pathway, indicating a possible means by which they participate in the control of gut physiology. However, the mechanisms and biological significance of this control have remained unexplored. Using multiple in vivo and in vitro approaches, we show that thyroid hormones positively regulate Notch activity through the TRα1 receptor. From a molecular point of view, TRα1 indirectly controls Notch1, Dll1, Dll4 and Hes1 expression but acts as a direct transcriptional regulator of the Jag1 gene by binding to a responsive element in the Jag1 promoter. Our findings show that the TRα1 nuclear receptor plays a key role in intestinal crypt progenitor/stem cell biology by controlling the Notch pathway and hence the balance between cell proliferation and cell differentiation.


Subject(s)
Cell Lineage/physiology , Hyperthyroidism/metabolism , Intestines/cytology , Receptor, Notch1/metabolism , Signal Transduction/physiology , Thyroid Hormone Receptors alpha/genetics , Animals , Blotting, Western , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Epithelial Cells/physiology , Immunohistochemistry , Intestines/physiology , Mice , Microscopy, Confocal
6.
Expert Rev Endocrinol Metab ; 2(1): 47-57, 2007 Jan.
Article in English | MEDLINE | ID: mdl-30743748

ABSTRACT

Thyroid hormone nuclear receptors (TRs) mediate thyroid hormone's activities in growth, differentiation, and development. Two TR genes (α and ß ) encode four thyroid hormone-binding receptors that regulate target gene expression. Mutations of the TRß gene cause the genetic syndrome of resistance to thyroid hormone. Studies indicate a close association between TRß mutations and several human cancers, suggesting their oncogenic role. A TRß gene knock-in mutant mouse (TRßPV/PV mouse) that spontaneously develops thyroid cancer allows elucidation of the oncogenic functions in vivo. TRßPV is a potent dominant negative mutant identified in a resistance to thyroid hormone patient. Molecular studies indicate that the PV mutant mediates its oncogenic activities via nucleus-initiated transcription and novel extranuclear actions. Thus, the deleterious effects of the gene mutations go beyond resistance to thyroid hormone and are more severe and extensive than previously envisioned. This newly identified oncogene exerts its tumorigenic effects via multiple signaling mechanisms.

SELECTION OF CITATIONS
SEARCH DETAIL