Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
2.
Subcell Biochem ; 95: 57-85, 2020.
Article in English | MEDLINE | ID: mdl-32297296

ABSTRACT

Retinoic acid (RA), a major natural active metabolite of vitamin A (VA) is well known to play critical roles in embryonic development. The effects of RA are mediated by nuclear receptors (RARs), which regulate the expression of gene batteries involved in cell growth and differentiation. Since the early 1990s several laboratories have focused on understanding how RA-regulated genes and RAR binding sites operate by studying the differentiation of embryonal carcinoma cells and embryonic stem cells. The development of hybridization-based microarray technology and high performance software analysis programs has allowed the characterization of thousands of RA-regulated genes. During the two last decades, publication of the genome sequence of various organisms has allowed advances in massive parallel sequencing and bioinformatics analysis of genome-wide data sets. These new generation sequencing (NGS) technologies have revolutionized the field by providing a global integrated picture of RA-regulated gene networks and the regulatory programs involved in cell fate decisions during embryonal carcinoma and embryonic stem cells differentiation. Now the challenge is to reconstruct the RA-regulated gene networks at the single cell level during the development of specialized embryonic tissues.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks , Genomics , Tretinoin/metabolism , Animals , Cell Differentiation/genetics , Humans , Receptors, Retinoic Acid/metabolism
3.
Oncogene ; 38(14): 2482-2500, 2019 04.
Article in English | MEDLINE | ID: mdl-30532072

ABSTRACT

All trans-retinoic acid (ATRA) is used in the treatment of acute promyelocytic leukemia (APL) and it is a promising agent also in solid tumors. The pharmacological activity of ATRA is mediated by the ligand-activated RAR and RXR transcription factors. In the present study, we define the basal and ATRA dependent RARα interactome in a RARα-overexpressing breast cancer cellular model, identifying 28 nuclear proteins. We focus our attention on the S100A3 calcium-binding protein, which interacts with RARα constitutively. In ATRA-sensitive breast cancer cells, S100A3 binds to RARα in basal conditions and binding is reduced by the retinoid. The interaction of S100A3 with RARα is direct and in lung cancer, APL and acute-myeloid-leukemia (AML) cells. In APL, S100A3 interacts not only with RARα, but also with PML-RARα. The interaction surface maps to the RARα ligand-binding domain, where the I396 residue plays a crucial role. Binding of S100A3 to RARα/PML-RARα controls the constitutive and ATRA-dependent degradation of these receptors. S100A3 knockdown decreases the amounts of RARα in breast- and lung cancer cells, inducing resistance to ATRA-dependent anti-proliferative/differentiating effects. Conversely, S100A3 knockdown in PML-RARα+ APL and PML-RARα- AML cells reduces the amounts of RARα/PML-RARα and increases basal and ATRA-induced differentiation. In this cellular context, opposite effects on RARα/PML-RARα levels and ATRA-induced differentiation are observed upon S100A3 overexpression. Our results provide new insights into the molecular mechanisms controlling RARα activity and have practical implications, as S100A3 represents a novel target for rational drug combinations aimed at potentiating the activity of ATRA.


Subject(s)
Breast Neoplasms/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Lung Neoplasms/metabolism , Promyelocytic Leukemia Protein/metabolism , Retinoic Acid Receptor alpha/metabolism , S100 Proteins/metabolism , A549 Cells , Animals , COS Cells , Cell Differentiation/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Chlorocebus aethiops , Female , Humans , Receptors, Retinoic Acid/metabolism
4.
FEBS Lett ; 592(8): 1426-1433, 2018 04.
Article in English | MEDLINE | ID: mdl-29543331

ABSTRACT

The nuclear retinoic acid (RA) receptors (RARα, ß and γ) are ligand-dependent regulators of transcription. Upon activation by RA, they are recruited at the promoters of target genes together with several coregulators. Then, they are degraded by the ubiquitin proteasome system. Here, we report that the degradation of the RARα subtype involves ubiquitination and the tripartite motif protein TRIM24, which was originally identified as a ligand-dependent corepressor of RARα. We show that in response to RA, TRIM24 serves as an adapter linking RARα to the proteasome for its degradation. In addition, TRIM24 and the proteasome are recruited with RARα to the promoters of target genes and thus are inherently linked to RARα transcriptional activity.


Subject(s)
Carrier Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Retinoic Acid Receptor alpha/metabolism , Ubiquitination/physiology , Carrier Proteins/genetics , HeLa Cells , Humans , MCF-7 Cells , Proteasome Endopeptidase Complex/genetics , Retinoic Acid Receptor alpha/genetics , Tretinoin/pharmacology , Ubiquitination/drug effects
5.
Biochem Biophys Res Commun ; 495(1): 846-853, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29158086

ABSTRACT

Retinoic acid receptors (RARs) are classically considered as nuclear ligand-dependent regulators of transcription. Here we highlighted a novel face of the RARα subtype: RARα is present in low amounts in the cytoplasm of mouse embryonic fibroblasts (MEFs) where it interacts with profilin2a (PFN2A), a small actin-binding protein involved in filaments polymerization. The interaction involves the N-terminal proline-rich motif (PRM) of RARα and the SH3-like domain of PFN2a. When increased in the cytoplasm, RARα competes with other PFN2a-binding proteins bearing PRMs and involved in actin filaments elongation. Consequently, the actin filament network is altered and MEFs adhesion is decreased. This novel role opens novel avenues for the understanding of pathologies characterized by increased levels of cytoplasmic RARα.


Subject(s)
Actin Cytoskeleton/metabolism , Cytoplasm/metabolism , Fibroblasts/metabolism , Profilins/metabolism , Retinoic Acid Receptor alpha/metabolism , Animals , Cells, Cultured , Mice , Protein Binding , Protein Interaction Mapping
6.
PLoS One ; 12(1): e0171043, 2017.
Article in English | MEDLINE | ID: mdl-28125680

ABSTRACT

Retinoic acid (RA) plays key roles in cell differentiation and growth arrest through nuclear retinoic acid receptors (RARs), which are ligand-dependent transcription factors. While the main trigger of RAR activation is the binding of RA, phosphorylation of the receptors has also emerged as an important regulatory signal. Phosphorylation of the RARγ N-terminal domain (NTD) is known to play a functional role in neuronal differentiation. In this work, we investigated the phosphorylation of RARγ ligand binding domain (LBD), and present evidence that the phosphorylation status of the LBD affects the phosphorylation of the NTD region. We solved the X-ray structure of a phospho-mimetic mutant of the LBD (RARγ S371E), which we used in molecular dynamics simulations to characterize the consequences of the S371E mutation on the RARγ structural dynamics. Combined with simulations of the wild-type LBD, we show that the conformational equilibria of LBD salt bridges (notably R387-D340) are affected by the S371E mutation, which likely affects the recruitment of the kinase complex that phosphorylates the NTD. The molecular dynamics simulations also showed that a conservative mutation in this salt bridge (R387K) affects the dynamics of the LBD without inducing large conformational changes. Finally, cellular assays showed that the phosphorylation of the NTD of RARγ is differentially regulated by retinoic acid in RARγWT and in the S371N, S371E and R387K mutants. This multidisciplinary work highlights an allosteric coupling between phosphorylations of the LBD and the NTD of RARγ and supports the importance of structural dynamics involving electrostatic interactions in the regulation of RARs activity.


Subject(s)
Allosteric Regulation/physiology , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Humans , Ligands , Molecular Dynamics Simulation , Phosphorylation , Protein Binding , Retinoic Acid Receptor gamma
7.
Oncotarget ; 8(23): 37041-37060, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-27419624

ABSTRACT

Treatment of acute promyelocytic leukemia (APL) with all-trans retinoic acid (ATRA) is the first example of targeted therapy. In fact, the oncogenic fusion-protein (PML-RAR) typical of this leukemia contains the retinoid-nuclear-receptor RARα. PML-RAR is responsible for the differentiation block of the leukemic blast. Besides PML-RAR, two endogenous RARα proteins are present in APL blasts, i.e. RARα1 and RARα2. We developed different cell populations characterized by PML-RAR, RARα2 and RARα1 knock-down in the APL-derived NB4 cell-line. Unexpectedly, silencing of PML-RAR and RARα2 results in similar increases in the constitutive expression of several granulocytic differentiation markers. This is accompanied by enhanced expression of the same granulocytic markers upon exposure of the NB4 blasts to ATRA. Silencing of PML-RAR and RARα2 causes also similar perturbations in the whole genome gene-expression profiles of vehicle and ATRA treated NB4 cells. Unlike PML-RAR and RARα2, RARα1 knock-down blocks ATRA-dependent induction of several granulocytic differentiation markers. Many of the effects on myeloid differentiation are confirmed by over-expression of RARα2 in NB4 cells. RARα2 action on myeloid differentiation does not require the presence of PML-RAR, as it is recapitulated also upon knock-down in PML-RAR-negative HL-60 cells. Thus, relative to RARα1, PML-RAR and RARα2 exert opposite effects on APL-cell differentiation. These contrasting actions may be related to the fact that both PML-RAR and RARα2 interact with and inhibit the transcriptional activity of RARα1. The interaction surface is located in the carboxy-terminal domain containing the D/E/F regions and it is influenced by phosphorylation of Ser-369 of RARα1.


Subject(s)
Cell Differentiation/drug effects , Oncogene Proteins, Fusion/genetics , Retinoic Acid Receptor alpha/genetics , Tretinoin/pharmacology , Acute Disease , Animals , Antineoplastic Agents/pharmacology , COS Cells , Cell Differentiation/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Chlorocebus aethiops , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Oncogene Proteins, Fusion/metabolism , RNA Interference , Retinoic Acid Receptor alpha/metabolism
8.
PLoS One ; 11(6): e0157290, 2016.
Article in English | MEDLINE | ID: mdl-27362937

ABSTRACT

Retinoic acid (RA), the main active vitamin A metabolite, controls multiple biological processes such as cell proliferation and differentiation through genomic programs and kinase cascades activation. Due to these properties, RA has proven anti-cancer capacity. Several breast cancer cells respond to the antiproliferative effects of RA, while others are RA-resistant. However, the overall signaling and transcriptional pathways that are altered in such cells have not been elucidated. Here, in a large-scale analysis of the phosphoproteins and in a genome-wide analysis of the RA-regulated genes, we compared two human breast cancer cell lines, a RA-responsive one, the MCF7 cell line, and a RA-resistant one, the BT474 cell line, which depicts several alterations of the "kinome". Using high-resolution nano-LC-LTQ-Orbitrap mass spectrometry associated to phosphopeptide enrichment, we found that several proteins involved in signaling and in transcription, are differentially phosphorylated before and after RA addition. The paradigm of these proteins is the RA receptor α (RARα), which was phosphorylated in MCF7 cells but not in BT474 cells after RA addition. The panel of the RA-regulated genes was also different. Overall our results indicate that RA resistance might correlate with the deregulation of the phosphoproteome with consequences on gene expression.


Subject(s)
Breast Neoplasms/metabolism , Phosphoproteins/metabolism , Proteome/drug effects , Transcriptome/drug effects , Tretinoin/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphorylation/drug effects , Signal Transduction/drug effects
9.
Am J Physiol Heart Circ Physiol ; 310(11): H1773-89, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27084391

ABSTRACT

To determine whether hepatic depletion of vitamin A (VA) stores has an effect on the postnatal heart, studies were carried out with mice lacking liver retinyl ester stores fed either a VA-sufficient (LRVAS) or VA-deficient (LRVAD) diet (to deplete circulating retinol and extrahepatic stores of retinyl esters). There were no observable differences in the weights or gross morphology of hearts from LRVAS or LRVAD mice relative to sex-matched, age-matched, and genetically matched wild-type (WT) controls fed the VAS diet (WTVAS), but changes in the transcription of functionally relevant genes were consistent with a state of VAD in LRVAS and LRVAD ventricles. In silico analysis revealed that 58/67 differentially expressed transcripts identified in a microarray screen are products of genes that have DNA retinoic acid response elements. Flow cytometric analysis revealed a significant and cell-specific increase in the number of proliferating Sca-1 cardiac progenitor cells in LRVAS animals relative to WTVAS controls. Before myocardial infarction, LRVAS and WTVAS mice had similar cardiac systolic function and structure, as measured by echocardiography, but, unexpectedly, repeat echocardiography demonstrated that LRVAS mice had less adverse remodeling by 1 wk after myocardial infarction. Overall, the results demonstrate that the adult heart is responsive to retinoids, and, most notably, reducing hepatic VA stores (while maintaining circulating levels of VA) impacts ventricular gene expression profiles, progenitor cell numbers, and response to injury.


Subject(s)
Liver/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Receptors, Retinoic Acid/metabolism , Retinoids/metabolism , Vitamin A Deficiency/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Echocardiography , Heart/physiopathology , Mice , Mice, Knockout , Myocardial Infarction/physiopathology , Vitamin A Deficiency/genetics , Vitamin A Deficiency/physiopathology , Retinoic Acid Receptor gamma
10.
Can J Physiol Pharmacol ; 93(12): 1065-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26459513

ABSTRACT

Vitamin A or retinol is a multifunctional vitamin that is essential at all stages of life from embryogenesis to adulthood. Up to now, it has been accepted that the effects of vitamin A are exerted by active metabolites, the major ones being 11-cis retinal for vision, and all trans-retinoic acid (RA) for cell growth and differentiation. Basically RA binds nuclear receptors, RARs, which regulate the expression of a battery of target genes in a ligand dependent manner. During the last decade, new scenarios have been discovered, providing a rationale for the understanding of other long-noted but not explained functions of retinol. These novel scenarios involve: (i) other nuclear receptors such as PPAR ß/δ, which regulate the expression of other target genes with other functions; (ii) extranuclear and nontranscriptional effects, such as the activation of kinases, which phosphorylate RARs and other transcription factors, thus expanding the list of the RA-activated genes; (iii) finally, vitamin A is active per se and can work as a cytokine that regulates gene transcription by activating STRA6. New effects of vitamin A and RA are continuously being discovered in new fields, revealing new targets and new mechanisms thus improving the understanding the pleiotropicity of their effects.


Subject(s)
Cell Nucleus/drug effects , Vitamin A/pharmacology , Vitamin A/therapeutic use , Animals , Cell Differentiation/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Humans , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism
11.
Mol Aspects Med ; 41: 1-115, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25543955

ABSTRACT

Retinoic acid (RA), the major bioactive metabolite of retinol or vitamin A, induces a spectrum of pleiotropic effects in cell growth and differentiation that are relevant for embryonic development and adult physiology. The RA activity is mediated primarily by members of the retinoic acid receptor (RAR) subfamily, namely RARα, RARß and RARγ, which belong to the nuclear receptor (NR) superfamily of transcription factors. RARs form heterodimers with members of the retinoid X receptor (RXR) subfamily and act as ligand-regulated transcription factors through binding specific RA response elements (RAREs) located in target genes promoters. RARs also have non-genomic effects and activate kinase signaling pathways, which fine-tune the transcription of the RA target genes. The disruption of RA signaling pathways is thought to underlie the etiology of a number of hematological and non-hematological malignancies, including leukemias, skin cancer, head/neck cancer, lung cancer, breast cancer, ovarian cancer, prostate cancer, renal cell carcinoma, pancreatic cancer, liver cancer, glioblastoma and neuroblastoma. Of note, RA and its derivatives (retinoids) are employed as potential chemotherapeutic or chemopreventive agents because of their differentiation, anti-proliferative, pro-apoptotic, and anti-oxidant effects. In humans, retinoids reverse premalignant epithelial lesions, induce the differentiation of myeloid normal and leukemic cells, and prevent lung, liver, and breast cancer. Here, we provide an overview of the biochemical and molecular mechanisms that regulate the RA and retinoid signaling pathways. Moreover, mechanisms through which deregulation of RA signaling pathways ultimately impact on cancer are examined. Finally, the therapeutic effects of retinoids are reported.


Subject(s)
Neoplasms/metabolism , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Animals , Antineoplastic Agents/therapeutic use , Female , Humans , Male , Neoplasms/drug therapy , Receptors, Retinoic Acid/genetics , Response Elements/physiology , Retinoic Acid Receptor alpha , Retinoic Acid Receptor gamma
12.
Biochim Biophys Acta ; 1851(1): 66-75, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24768681

ABSTRACT

Retinoic acid (RA), the active derivative of vitamin A, a fat-soluble vitamin, plays key roles in cell growth and differentiation by activating nuclear receptors, RARs (α, ß and γ), which are ligand dependent regulators of transcription. The past years highlighted several novelties in the field that increased the complexity of RA effects. Indeed, in addition to its classical genomic effects, RA also has extranuclear and non-transcriptional effects. RA induces the rapid and transient activation of kinase cascades, which are integrated in the nucleus via the phosphorylation of RARs at a conserved serine residue located in the N-terminal domain and their coregulators. In order to investigate the relevance of RARs' phosphorylation in cell differentiation, mouse embryonic stem (mES) cells were used as a model. When treated with RA, these pluripotent cells give rise to neuronal cells. Cells invalidated for each RAR were generated as well as stable rescue lines expressing RARs mutated in phosphor acceptor sites. Such a strategy revealed that RA-induced neuronal differentiation involves the RARγ2 subtype and requires RARγ2 phosphorylation. Moreover, in gene expression profiling experiments, the phosphorylated form of RARγ2 was found to regulate a small subset of genes through binding a novel RA response element consisting of two direct repeats with a 7 base pair spacer. These new findings suggest an important role for RAR phosphorylation during cell differentiation, and pave the way for further investigations with other cell types and during embryonic development. This article is part of a Special Issue entitled Linking transcription to physiology in lipodomics.


Subject(s)
Embryonic Stem Cells/physiology , Tretinoin/metabolism , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Phosphorylation , Receptors, Retinoic Acid/metabolism , Signal Transduction
14.
Subcell Biochem ; 70: 1-20, 2014.
Article in English | MEDLINE | ID: mdl-24962878

ABSTRACT

The discovery of retinoic acid receptors arose from research into how vitamins are essential for life. Early studies indicated that Vitamin A was metabolized into an active factor, retinoic acid (RA), which regulates RNA and protein expression in cells. Each step forward in our understanding of retinoic acid in human health was accomplished by the development and application of new technologies. Development cDNA cloning techniques and discovery of nuclear receptors for steroid hormones provided the basis for identification of two classes of retinoic acid receptors, RARs and RXRs, each of which has three isoforms, α, ß and É£. DNA manipulation and crystallographic studies revealed that the receptors contain discrete functional domains responsible for binding to DNA, ligands and cofactors. Ligand binding was shown to induce conformational changes in the receptors that cause release of corepressors and recruitment of coactivators to create functional complexes that are bound to consensus promoter DNA sequences called retinoic acid response elements (RAREs) and that cause opening of chromatin and transcription of adjacent genes. Homologous recombination technology allowed the development of mice lacking expression of retinoic acid receptors, individually or in various combinations, which demonstrated that the receptors exhibit vital, but redundant, functions in fetal development and in vision, reproduction, and other functions required for maintenance of adult life. More recent advancements in sequencing and proteomic technologies reveal the complexity of retinoic acid receptor involvement in cellular function through regulation of gene expression and kinase activity. Future directions will require systems biology approaches to decipher how these integrated networks affect human stem cells, health, and disease.


Subject(s)
Receptors, Retinoic Acid/history , Retinoid X Receptors/history , Tretinoin/metabolism , Vitamin A/metabolism , Animals , Cloning, Molecular , Gene Expression Regulation , History, 20th Century , History, 21st Century , Humans , Mice , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/history , Protein Isoforms/metabolism , Protein Structure, Tertiary , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Response Elements , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Signal Transduction , Tretinoin/chemistry , Vitamin A/chemistry
15.
Subcell Biochem ; 70: 103-27, 2014.
Article in English | MEDLINE | ID: mdl-24962883

ABSTRACT

The nuclear retinoic acid receptors (RAR α, ß and γ) and their isoforms are ligand-dependent regulators of transcription Transcription , which mediate the effects of all-trans retinoic acid (RA), the active endogenous metabolite of Vitamin A. They heterodimerize with Retinoid X Receptors (RXRs α, ß and γ), and regulate the expression of a battery of target genes Target genes involved in cell growth and differentiation Differentiation . During the two last decades, the description of the crystallographic structures of RARs, the characterization of the polymorphic response elements of their target genes Target genes , and the identification of the multiprotein complexes involved in their transcriptional activity have provided a wealth of information on their pleiotropic effects. However, the regulatory scenario became even more complicated once it was discovered that RARs are phosphoproteins and that RA can activate kinase signaling cascades via a pool of RARs present in membrane lipid rafts. Now it is known that these RA-activated kinases Kinases translocate to the nucleus where they phosphorylate RARs and other retinoid signaling factors. The phosphorylation Phosphorylation state of the RARs dictates whether the transcriptional programs which are known to be induced by RA are facilitated and/or switched on. Thus, kinase signaling pathways appear to be crucial for fine-tuning the appropriate physiological activity of RARs.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Tretinoin/metabolism , Gene Expression Regulation , Humans , Ligands , Mitogen-Activated Protein Kinases/genetics , Models, Molecular , Phosphorylation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Response Elements , Retinoid X Receptors/chemistry , Retinoid X Receptors/genetics , Signal Transduction , Tretinoin/chemistry
16.
J Cell Sci ; 127(Pt 9): 2095-105, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24569880

ABSTRACT

Retinoic acid (RA) plays key roles in cell differentiation and growth arrest by activating nuclear RA receptors (RARs) (α, ß and γ), which are ligand-dependent transcription factors. RARs are also phosphorylated in response to RA. Here, we investigated the in vivo relevance of the phosphorylation of RARs during RA-induced neuronal differentiation of mouse embryonic stem cells (mESCs). Using ESCs where the genes encoding each RAR subtype had been inactivated, and stable rescue lines expressing RARs mutated in phospho-acceptor sites, we show that RA-induced neuronal differentiation involves RARγ2 and requires RARγ2 phosphorylation. By gene expression profiling, we found that the phosphorylated form of RARγ2 regulates a small subset of genes through binding an unusual RA response element consisting of two direct repeats with a seven-base-pair spacer. These new findings suggest an important role for RARγ phosphorylation during cell differentiation and pave the way for further investigations during embryonic development.


Subject(s)
Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Animals , Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , Phosphorylation , Retinoic Acid Receptor gamma
17.
Mol Endocrinol ; 28(2): 260-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24422634

ABSTRACT

Retinoic acid (RA) controls many aspects of embryonic development by binding to specific receptors (retinoic acid receptors [RARs]) that regulate complex transcriptional networks. Three different RAR subtypes are present in vertebrates and play both common and specific roles in transducing RA signaling. Specific activities of each receptor subtype can be correlated with its exclusive expression pattern, whereas shared activities between different subtypes are generally assimilated to functional redundancy. However, the question remains whether some subtype-specific activity still exists in regions or organs coexpressing multiple RAR subtypes. We tackled this issue at the transcriptional level using early zebrafish embryo as a model. Using morpholino knockdown, we specifically invalidated the zebrafish endogenous RAR subtypes in an in vivo context. After building up a list of RA-responsive genes in the zebrafish gastrula through a whole-transcriptome analysis, we compared this panel of genes with those that still respond to RA in embryos lacking one or another RAR subtype. Our work reveals that RAR subtypes do not have fully redundant functions at the transcriptional level but can transduce RA signal in a subtype-specific fashion. As a result, we define RAR subtype-specific transcriptotypes that correspond to repertoires of genes activated by different RAR subtypes. Finally, we found genes of the RA pathway (cyp26a1, raraa) the regulation of which by RA is highly robust and can even resist the knockdown of all RARs. This suggests that RA-responsive genes are differentially sensitive to alterations in the RA pathway and, in particular, cyp26a1 and raraa are under a high pressure to maintain signaling integrity.


Subject(s)
Gastrula/metabolism , Receptors, Retinoic Acid/metabolism , Zebrafish/genetics , Animals , Base Sequence , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Retinoic Acid 4-Hydroxylase , Signal Transduction , Transcription, Genetic , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins
18.
J Cell Sci ; 127(Pt 3): 521-33, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24357724

ABSTRACT

Nuclear retinoic acid (RA) receptors (RARα, ß and γ) are ligand-dependent transcription factors that regulate the expression of a battery of genes involved in cell differentiation and proliferation. They are also phosphoproteins and we previously showed the importance of their phosphorylation in their transcriptional activity. In the study reported here, we conducted a genome-wide analysis of the genes that are regulated by RARs in mouse embryonic fibroblasts (MEFs) by comparing wild-type MEFs to MEFs lacking the three RARs. We found that in the absence of RA, RARs control the expression of several gene transcripts associated with cell adhesion. Consequently the knockout MEFs are unable to adhere and to spread on substrates and they display a disrupted network of actin filaments, compared with the WT cells. In contrast, in the presence of the ligand, RARs control the expression of other genes involved in signaling and in RA metabolism. Taking advantage of rescue cell lines expressing the RARα or RARγ subtypes (either wild-type or mutated at the N-terminal phosphorylation sites) in the null background, we found that the expression of RA-target genes can be controlled either by a specific single RAR or by a combination of RAR isotypes, depending on the gene. We also selected genes that require the phosphorylation of the receptors for their regulation by RA. Our results increase the repertoire of genes that are regulated by RARs and highlight the complexity and diversity of the transcriptional programs regulated by RARs, depending on the gene.


Subject(s)
Cell Adhesion/genetics , Receptors, Retinoic Acid/biosynthesis , Animals , Cell Differentiation/genetics , Cell Proliferation , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Mice , Phosphorylation , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Signal Transduction , Retinoic Acid Receptor gamma
19.
Protein Expr Purif ; 95: 113-20, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333369

ABSTRACT

Gene activation by retinoic acid nuclear receptors (RAR) is regulated by a number of molecular events such as ligand binding, interaction with cognate DNA sequences and co-regulatory proteins, and phosphorylation. Among the several phosphorylation sites that are involved in the non-genomic regulatory pathways of the RAR, two are located in a proline rich domain (PRD) within the N-terminal domain (NTD) of the receptor. This region is predicted to be intrinsically disordered, complicating its production and purification. We present here an approach enabling the high yield production of RAR fragments encompassing the PRD and the DNA binding domain (DBD). We found that expression levels were dependent on where the position of the N-terminal boundary of the fragment was placed within the RAR sequence. The purification protocol involves the use of maltose binding protein as a solubilising tag and extensive centrifugation steps at critical points of the purification process. This protocol is suitable to express (15)N, (13)C labeled proteins enabling nuclear magnetic resonance studies. The resulting proteins were characterized by biophysical methods including Small Angle X-ray Scattering and NMR. These studies showed that PRD extension of RARγ is disordered in solution, a state that is compatible with modifications such as phosphorylation.


Subject(s)
Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , DNA/chemistry , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Mice , Molecular Sequence Data , Proline , Protein Structure, Tertiary , Receptors, Retinoic Acid/genetics , Recombinant Proteins/genetics , Sequence Alignment , Retinoic Acid Receptor gamma
20.
Virchows Arch ; 462(6): 619-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23639973

ABSTRACT

Retinoid receptors (RRs) play a key role in cell proliferation and differentiation. We characterized the expression of RA receptors and retinoid X receptors (RARs and RXRs) in a series of 111 thyroid tumors and investigated the mechanisms responsible for their deregulation: hypermethylation of the RARB2 promoter, loss of heterozygosity (LOH) in the regions of RARB and RXRA, and altered expression of CRBP1 and enzymes involved in RA biosynthesis (RDH10 and RALDH2). Expression of RALDH2 and RDH10 was conserved in 100 % of adenomas and in 90 and 98 %, respectively, of carcinomas, whereas staining for CRBP1 was decreased in 9 % of FAs and 28 % of carcinomas, mainly anaplastic carcinomas (55 %). We found an abnormal expression of RARA, RARB, RXRA, and RXRB in 67, 69, 66, and 73 %, respectively, of thyroid carcinomas (n = 78) and in 9, 9, 9, and 33 % of follicular adenomas (n = 33) (p < 0.001). An abnormal staining pattern of at least two of these markers had 90 % sensitivity and 91 % specificity for a diagnosis of malignancy. Promoter hypermethylation of RARB2 was observed in some anaplastic carcinomas (14 %). LOH was found to be common at the RARB locus (3p24-3p25) and the RXRA locus (9q34), respectively, in 44 and 55 % of carcinomas and in 27 and 43 % of adenomas. In conclusion, immunohistochemical staining for RARs and RXRs may help in the differential diagnosis between well-differentiated carcinoma and follicular adenoma. Further investigation should be carried out to determine whether the characterization of RR expression might identify patients who could benefit from therapy with RA derivatives.


Subject(s)
Adenocarcinoma/diagnosis , Adenoma/diagnosis , Biomarkers, Tumor/metabolism , Retinoid X Receptors/metabolism , Thyroid Neoplasms/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenoma/genetics , Adenoma/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Aldehyde Dehydrogenase 1 Family , Biomarkers, Tumor/genetics , Child , Diagnosis, Differential , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Loss of Heterozygosity , Male , Methylation , Middle Aged , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinal Dehydrogenase/genetics , Retinal Dehydrogenase/metabolism , Retinoid X Receptors/genetics , Retinol-Binding Proteins, Cellular/genetics , Retinol-Binding Proteins, Cellular/metabolism , Sensitivity and Specificity , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...