Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Cell ; 178(4): 949-963.e18, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353221

ABSTRACT

Estrogen receptor-positive (ER+) breast cancers frequently remain dependent on ER signaling even after acquiring resistance to endocrine agents, prompting the development of optimized ER antagonists. Fulvestrant is unique among approved ER therapeutics due to its capacity for full ER antagonism, thought to be achieved through ER degradation. The clinical potential of fulvestrant is limited by poor physicochemical features, spurring attempts to generate ER degraders with improved drug-like properties. We show that optimization of ER degradation does not guarantee full ER antagonism in breast cancer cells; ER "degraders" exhibit a spectrum of transcriptional activities and anti-proliferative potential. Mechanistically, we find that fulvestrant-like antagonists suppress ER transcriptional activity not by ER elimination, but by markedly slowing the intra-nuclear mobility of ER. Increased ER turnover occurs as a consequence of ER immobilization. These findings provide proof-of-concept that small molecule perturbation of transcription factor mobility may enable therapeutic targeting of this challenging target class.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/pharmacology , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/therapeutic use , Female , Fulvestrant/therapeutic use , HEK293 Cells , Heterografts , Humans , Indazoles/pharmacology , Ligands , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Polymorphism, Single Nucleotide , Proteolysis/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects
2.
Immunity ; 49(4): 615-626.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332629

ABSTRACT

Macrophages polarize into distinct phenotypes in response to complex environmental cues. We found that the nuclear receptor PPARγ drove robust phenotypic changes in macrophages upon repeated stimulation with interleukin (IL)-4. The functions of PPARγ on macrophage polarization in this setting were independent of ligand binding. Ligand-insensitive PPARγ bound DNA and recruited the coactivator P300 and the architectural protein RAD21. This established a permissive chromatin environment that conferred transcriptional memory by facilitating the binding of the transcriptional regulator STAT6 and RNA polymerase II, leading to robust production of enhancer and mRNAs upon IL-4 re-stimulation. Ligand-insensitive PPARγ binding controlled the expression of an extracellular matrix remodeling-related gene network in macrophages. Expression of these genes increased during muscle regeneration in a mouse model of injury, and this increase coincided with the detection of IL-4 and PPARγ in the affected tissue. Thus, a predominantly ligand-insensitive PPARγ:RXR cistrome regulates progressive and/or reinforcing macrophage polarization.


Subject(s)
Epigenesis, Genetic/immunology , Epigenomics/methods , Gene Expression Regulation/immunology , Macrophage Activation/immunology , Macrophages/immunology , PPAR gamma/immunology , Animals , Cell Line , Cells, Cultured , Interleukin-4/immunology , Interleukin-4/pharmacology , Ligands , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism
3.
Mol Cell Proteomics ; 23(3): 100741, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38387774

ABSTRACT

Exogenous glucocorticoids are frequently used to treat inflammatory disorders and as adjuncts for the treatment of solid cancers. However, their use is associated with severe side effects and therapy resistance. Novel glucocorticoid receptor (GR) ligands with a patient-validated reduced side effect profile have not yet reached the clinic. GR is a member of the nuclear receptor family of transcription factors and heavily relies on interactions with coregulator proteins for its transcriptional activity. To elucidate the role of the GR interactome in the differential transcriptional activity of GR following treatment with the selective GR agonist and modulator dagrocorat compared to classic (ant)agonists, we generated comprehensive interactome maps by high-confidence proximity proteomics in lung epithelial carcinoma cells. We found that dagrocorat and the antagonist RU486 both reduced GR interaction with CREB-binding protein/p300 and the mediator complex compared to the full GR agonist dexamethasone. Chromatin immunoprecipitation assays revealed that these changes in GR interactome were accompanied by reduced GR chromatin occupancy with dagrocorat and RU486. Our data offer new insights into the role of differential coregulator recruitment in shaping ligand-specific GR-mediated transcriptional responses.


Subject(s)
Benzamides , Chromatin , Phenanthrenes , Receptors, Glucocorticoid , Humans , Receptors, Glucocorticoid/genetics , Mifepristone/pharmacology , Mediator Complex/metabolism , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Dexamethasone/pharmacology
4.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34452998

ABSTRACT

Efforts to improve estrogen receptor-α (ER)-targeted therapies in breast cancer have relied upon a single mechanism, with ligands having a single side chain on the ligand core that extends outward to determine antagonism of breast cancer growth. Here, we describe inhibitors with two ER-targeting moieties, one of which uses an alternate structural mechanism to generate full antagonism, freeing the side chain to independently determine other critical properties of the ligands. By combining two molecular targeting approaches into a single ER ligand, we have generated antiestrogens that function through new mechanisms and structural paradigms to achieve antagonism. These dual-mechanism ER inhibitors (DMERIs) cause alternate, noncanonical structural perturbations of the receptor ligand-binding domain (LBD) to antagonize proliferation in ER-positive breast cancer cells and in allele-specific resistance models. Our structural analyses with DMERIs highlight marked differences from current standard-of-care, single-mechanism antiestrogens. These findings uncover an enhanced flexibility of the ER LBD through which it can access nonconsensus conformational modes in response to DMERI binding, broadly and effectively suppressing ER activity.


Subject(s)
Breast Neoplasms/drug therapy , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Crystallography, X-Ray , Female , Humans , Protein Binding , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Nat Chem Biol ; 17(3): 307-316, 2021 03.
Article in English | MEDLINE | ID: mdl-33510451

ABSTRACT

Glucocorticoids display remarkable anti-inflammatory activity, but their use is limited by on-target adverse effects including insulin resistance and skeletal muscle atrophy. We used a chemical systems biology approach, ligand class analysis, to examine ligands designed to modulate glucocorticoid receptor activity through distinct structural mechanisms. These ligands displayed diverse activity profiles, providing the variance required to identify target genes and coregulator interactions that were highly predictive of their effects on myocyte glucose disposal and protein balance. Their anti-inflammatory effects were linked to glucose disposal but not muscle atrophy. This approach also predicted selective modulation in vivo, identifying compounds that were muscle-sparing or anabolic for protein balance and mitochondrial potential. Ligand class analysis defined the mechanistic links between the ligand-receptor interface and ligand-driven physiological outcomes, a general approach that can be applied to any ligand-regulated allosteric signaling system.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Glucose Transporter Type 4/genetics , Muscular Atrophy/drug therapy , Receptors, Glucocorticoid/chemistry , Signal Transduction/drug effects , A549 Cells , Allosteric Regulation , Animals , Anti-Inflammatory Agents/chemical synthesis , Cell Line, Transformed , Gene Expression Regulation , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Lipopolysaccharides/administration & dosage , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Structure-Activity Relationship
6.
Int J Mol Sci ; 24(1)2022 Dec 24.
Article in English | MEDLINE | ID: mdl-36613751

ABSTRACT

Angelman Syndrome (AS) is a severe neurodevelopmental disorder, caused by the neuronal absence of the ubiquitin protein ligase E3A (UBE3A). UBE3A promotes ubiquitin-mediated protein degradation and functions as a transcriptional coregulator of nuclear hormone receptors, including the glucocorticoid receptor (GR). Previous studies showed anxiety-like behavior and hippocampal-dependent memory disturbances in AS mouse models. Hippocampal GR is an important regulator of the stress response and memory formation, and we therefore investigated whether the absence of UBE3A in AS mice disrupted GR signaling in the hippocampus. We first established a strong cortisol-dependent interaction between the GR ligand binding domain and a UBE3A nuclear receptor box in a high-throughput interaction screen. In vivo, we found that UBE3A-deficient AS mice displayed significantly more variation in circulating corticosterone levels throughout the day compared to wildtypes (WT), with low to undetectable levels of corticosterone at the trough of the circadian cycle. Additionally, we observed an enhanced transcriptomic response in the AS hippocampus following acute corticosterone treatment. Surprisingly, chronic corticosterone treatment showed less contrast between AS and WT mice in the hippocampus and liver transcriptomic responses. This suggests that UBE3A limits the acute stimulation of GR signaling, likely as a member of the GR transcriptional complex. Altogether, these data indicate that AS mice are more sensitive to acute glucocorticoid exposure in the brain compared to WT mice. This suggests that stress responsiveness is altered in AS which could lead to anxiety symptoms.


Subject(s)
Angelman Syndrome , Mice , Animals , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Corticosterone/metabolism , Hippocampus/metabolism , Brain/metabolism , Neurons/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Ubiquitin-Protein Ligases/metabolism , Disease Models, Animal
7.
Chem Res Toxicol ; 34(2): 313-329, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33405908

ABSTRACT

Identification of chemicals that affect hormone-regulated systems will help to predict endocrine disruption. In our previous study, a 46 gene biomarker was found to be an accurate predictor of estrogen receptor (ER) α modulation in chemically treated MCF-7 cells. Here, potential ERα modulators were identified using the biomarker by screening a microarray compendium consisting of ∼1600 gene expression comparisons representing exposure to ∼1200 chemicals. A total of ∼170 chemicals were identified as potential ERα modulators. In the Connectivity Map 2.0 collection, 75 and 39 chemicals were predicted to activate or suppress ERα, and they included 12 and six known ERα agonists and antagonists/selective ERα modulators, respectively. Nineteen and eight of the total number were also identified as active in an ERα transactivation assay carried out in an MCF-7-derived cell line used to screen the Tox21 10K chemical library in agonist or antagonist modes, respectively. Chemicals predicted to modulate ERα in MCF-7 cells were examined further using global and targeted gene expression in wild-type and ERα-null cells, transactivation assays, and cell-free ERα coregulator interaction assays. Environmental chemicals classified as weak and very weak agonists were confirmed to activate ERα including apigenin, kaempferol, and oxybenzone. Novel activators included digoxin, nabumetone, ivermectin, and six progestins. Novel suppressors included emetine, mifepristone, niclosamide, and proscillaridin. Our strategy will be useful to identify environmentally relevant ERα modulators in future high-throughput transcriptomic screens.


Subject(s)
Biomarkers, Tumor/genetics , Estrogen Receptor Modulators/analysis , Estrogen Receptor alpha/genetics , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Female , Gene Expression Profiling , Humans , MCF-7 Cells , Tumor Cells, Cultured
8.
Bioorg Med Chem Lett ; 30(16): 127293, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32631515

ABSTRACT

LRH-1 is a nuclear receptor that regulates lipid metabolism and homeostasis, making it an attractive target for the treatment of diabetes and non-alcoholic fatty liver disease. Building on recent structural information about ligand binding from our labs, we have designed a series of new LRH-1 agonists that further engage LRH-1 through added polar interactions. While the current synthetic approach to this scaffold has, in large part, allowed for decoration of the agonist core, significant variation of the bridgehead substituent is mechanistically precluded. We have developed a new synthetic approach to overcome this limitation, identified that bridgehead substitution is necessary for LRH-1 activation, and described an alternative class of bridgehead substituents for effective LRH-1 agonist development. We determined the crystal structure of LRH-1 bound to a bridgehead-modified compound, revealing a promising opportunity to target novel regions of the ligand binding pocket to alter LRH-1 target gene expression.


Subject(s)
Aniline Compounds/pharmacology , Drug Development , Receptors, Cytoplasmic and Nuclear/agonists , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Oxidation-Reduction , Photochemical Processes , Receptors, Cytoplasmic and Nuclear/genetics , Structure-Activity Relationship
9.
Neuroendocrinology ; 109(3): 266-276, 2019.
Article in English | MEDLINE | ID: mdl-30884490

ABSTRACT

Glucocorticoid hormones have important effects on brain function in the context of acute and chronic stress. Many of these are mediated by the glucocorticoid receptor (GR). GR has transcriptional activity which is highly context-specific and differs between tissues and even between cell types. The outcome of GR-mediated transcription depends on the interactome of associated coregulators. Selective GR modulators (SGRMs) are a class of GR ligands that can be used to activate only a subset of GR-coregulator interactions, thereby giving the possibility to induce a unique combination of agonistic and antagonistic GR properties. We describe SGRM action in animal models of brain function and pathology, and argue for their utility as molecular filters, to characterize context-specific GR interactome and transcriptional activity that are responsible for particular glucocorticoid-driven effects in cognitive processes such as memory consolidation. The ultimate objective of this approach is to identify molecular processes that are responsible for adaptive and maladaptive effects of glucocorticoids in the brain.


Subject(s)
Brain/metabolism , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Humans
10.
J Recept Signal Transduct Res ; 38(2): 112-121, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29447503

ABSTRACT

Estrogen receptor alpha (ERα) and retinoic acid receptors (RARs) play important and opposite roles in breast cancer growth. While exposure to ERα agonists such as 17ß-estradiol (E2) is related to proliferation, RAR agonists such as all-trans retinoic acid (AtRA) induce anti-proliferative effects. Although crosstalk between these pathways has been proposed, the molecular mechanisms underlying this interplay are still not completely unraveled. The aim of this study was to evaluate the effects of AtRA on ERα-mediated signaling in the ERα positive cell lines MCF7/BUS and U2OS-ERα-Luc to investigate some of the possible underlying modes of action. To do so, this study assessed the effects of AtRA on different ERα-related events such as ERα-mediated cell proliferation and gene expression, ERα-coregulator binding and ERα subcellular localization. AtRA-mediated antagonism of E2-induced signaling was observed in the proliferation and gene expression studies. However, AtRA showed no remarkable effects on the E2-driven coregulator binding and subcellular distribution of ERα. Interestingly, in the absence of E2, ERα-mediated gene expression, ERα-coregulator binding and ERα subcellular mobilization were increased upon exposure to micromolar concentrations of AtRA found to inhibit cell proliferation after long-term exposure. Nevertheless, experiments using purified ERα showed that direct binding of AtRA to ERα does not occur. Altogether, our results using MCF7/BUS and U2OS-ERα-Luc cells suggest that AtRA, without being a direct ligand of ERα, can indirectly interfere on basal ERα-coregulator binding and basal ERα subcellular localization in addition to the previously described crosstalk mechanisms such as competition of ERs and RARs for DNA binding sites.


Subject(s)
Estrogens/pharmacology , Receptors, Estrogen/metabolism , Signal Transduction , Tretinoin/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Luciferases/metabolism , MCF-7 Cells , Protein Binding/drug effects , Signal Transduction/drug effects
11.
Mol Syst Biol ; 12(4): 864, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27107013

ABSTRACT

Some estrogen receptor-α (ERα)-targeted breast cancer therapies such as tamoxifen have tissue-selective or cell-specific activities, while others have similar activities in different cell types. To identify biophysical determinants of cell-specific signaling and breast cancer cell proliferation, we synthesized 241 ERα ligands based on 19 chemical scaffolds, and compared ligand response using quantitative bioassays for canonical ERα activities and X-ray crystallography. Ligands that regulate the dynamics and stability of the coactivator-binding site in the C-terminal ligand-binding domain, called activation function-2 (AF-2), showed similar activity profiles in different cell types. Such ligands induced breast cancer cell proliferation in a manner that was predicted by the canonical recruitment of the coactivators NCOA1/2/3 and induction of the GREB1 proliferative gene. For some ligand series, a single inter-atomic distance in the ligand-binding domain predicted their proliferative effects. In contrast, the N-terminal coactivator-binding site, activation function-1 (AF-1), determined cell-specific signaling induced by ligands that used alternate mechanisms to control cell proliferation. Thus, incorporating systems structural analyses with quantitative chemical biology reveals how ligands can achieve distinct allosteric signaling outcomes through ERα.


Subject(s)
Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Signal Transduction/drug effects , Binding Sites , Cell Proliferation/drug effects , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , MCF-7 Cells , Models, Molecular , Molecular Structure , Peptide Library , Protein Binding
12.
Biochim Biophys Acta Proteins Proteom ; 1865(9): 1195-1206, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28642153

ABSTRACT

Retinoic Acid Receptor alpha (RARα/NR1B1), Retinoic Acid Receptor beta (RARß/NR1B2) and Retinoic Acid Receptor gamma (RARγ/NR1B3) are transcription factors regulating gene expression in response to retinoids. Within the RAR genomic pathways, binding of RARs to coregulators is a key intermediate regulatory phase. However, ligand-dependent interactions between the wide variety of coregulators that may be present in a cell and the different RAR subtypes are largely unknown. The aim of this study is to characterize the coregulator binding profiles of RARs in the presence of the pan-agonist all-trans-Retinoic Acid (AtRA); the subtype-selective agonists Am80 (RARα), CD2314 (RARß) and BMS961 (RARγ); and the antagonist Ro415253. To this end, we used a microarray assay for coregulator-nuclear receptor interactions to assess RAR binding to 154 motifs belonging to >60 coregulators. The results revealed a high number of ligand-dependent RAR-coregulator interactions among all RAR variants, including many binding events not yet described in literature. Next, this work confirmed a greater ligand-independent activity of RARß compared to the other RAR subtypes based on both higher basal and lower ligand-driven coregulator binding. Further, several coregulator motifs showed selective binding to a specific RAR subtype. Next, this work showed that subtype-selective agonists can be successfully discriminated by using coregulator binding assays. Finally this study demonstrated the possible applications of a coregulator binding assay as a tool to discriminate between agonistic/antagonistic actions of ligands. The RAR-coregulator interactions found will be of use to direct further studies to better understand the mechanisms driving the eventual actions of retinoids.


Subject(s)
Receptors, Retinoic Acid/chemistry , Retinoic Acid Receptor alpha/chemistry , Amino Acid Motifs , Anthracenes/pharmacology , Benzoates/pharmacology , Binding Sites , Chromans , Protein Array Analysis , Protein Binding , Protein Domains , Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/antagonists & inhibitors , Recombinant Proteins/metabolism , Response Elements , Retinoic Acid Receptor alpha/agonists , Retinoic Acid Receptor alpha/antagonists & inhibitors , Retinoids/pharmacology , Structure-Activity Relationship , Tetrahydronaphthalenes/pharmacology , Thiophenes/pharmacology , Tretinoin/pharmacology , Retinoic Acid Receptor gamma
13.
Biochim Biophys Acta ; 1851(12): 1577-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26434697

ABSTRACT

BACKGROUND: Liver X receptors (LXRs) are transcription factors activated by cholesterol metabolites containing an oxidized side chain. Due to their ability to regulate lipid metabolism and cholesterol transport, they have become attractive pharmacological targets. LXRs are closely related to DAF-12, a nuclear receptor involved in nematode lifespan and regulated by the binding of C-27 steroidal acids. Based on our recent finding that the lack of the C-25 methyl group does not abolish their DAF-12 activity, we evaluated the effect of removing it from the (25R)-cholestenoic acid, a LXR agonist. METHODS: The binding mode and the molecular basis of action of 27-nor-5-cholestenoic acid were evaluated using molecular dynamics simulations. The biological activity was investigated using reporter gene expression assays and determining the expression levels of endogenous target genes. The in vitro MARCoNI assay was used to analyze the interaction with cofactors. RESULTS: 27-Nor-5-cholestenoic acid behaves as an inverse agonist. This correlates with the capacity of the complex to better bind corepressors rather than coactivators. The C-25 methyl moiety would be necessary for the maintenance of a torsioned conformation of the steroid side chain that stabilizes an active LXRß state. CONCLUSION: We found that a 27-nor analog is able to act as a LXR ligand. Interestingly, this minimal structural change on the steroid triggered a drastic change in the LXR response. GENERAL SIGNIFICANCE: Results contribute to improve our understanding on the molecular basis of LXRß mechanisms of action and provide a new scaffold in the quest for selective LXR modulators.


Subject(s)
Cholestenes/pharmacology , Gene Expression Regulation/drug effects , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/metabolism , Binding Sites , HEK293 Cells , Hep G2 Cells , Humans , Ligands , Liver X Receptors , Orphan Nuclear Receptors/genetics
14.
BMC Genomics ; 17(1): 832, 2016 10 26.
Article in English | MEDLINE | ID: mdl-27782803

ABSTRACT

BACKGROUND: NR2E1 (Tlx) is an orphan nuclear receptor that regulates the maintenance and self-renewal of neural stem cells, and promotes tumourigenesis. Nr2e1-null mice exhibit reduced cortical and limbic structures and pronounced retinal dystrophy. NR2E1 functions mainly as a repressor of gene transcription in association with the co-repressors atrophin-1, LSD1, HDAC and BCL11A. Recent evidence suggests that NR2E1 also acts as an activator of gene transcription. However, co-activator complexes that interact with NR2E1 have not yet been identified. In order to find potential novel co-regulators for NR2E1, we used a microarray assay for real-time analysis of co-regulator-nuclear receptor interaction (MARCoNI) that contains peptides representing interaction motifs from potential co-regulatory proteins, including known co-activator nuclear receptor box sequences (LxxLL motif). RESULTS: We found that NR2E1 binds strongly to an atrophin-1 peptide (Atro box) used as positive control and to 19 other peptides that constitute candidate NR2E1 partners. Two of these proteins, p300 and androgen receptor (AR), were further validated by reciprocal pull-down assays. The specificity of NR2E1 binding to peptides in the array was evaluated using two single amino acid variants, R274G and R276Q, which disrupted the majority of the binding interactions observed with wild-type NR2E1. The decreased binding affinity of these variants to co-regulators was further validated by pull-down assays using atrophin1 as bait. Despite the high conservation of arginine 274 in vertebrates, its reduced interactions with co-regulators were not significant in vivo as determined by retinal phenotype analysis in single-copy Nr2e1-null mice carrying the variant R274G. CONCLUSIONS: We showed that MARCoNI is a specific assay to test interactions of NR2E1 with candidate co-regulators. In this way, we unveiled 19 potential co-regulator partners for NR2E1, including eight co-activators. All the candidates here identified need to be further validated using in vitro and in vivo models. This assay was sensitive to point mutations in NR2E1 ligand binding domain making it useful to identify mutations and/or small molecules that alter binding of NR2E1 to protein partners.


Subject(s)
Ligands , Orphan Nuclear Receptors/agonists , Receptors, Cytoplasmic and Nuclear/agonists , Amino Acid Sequence , Animals , Carrier Proteins , Drug Discovery , Genetic Variation , Humans , Mice , Mice, Knockout , Orphan Nuclear Receptors/chemistry , Orphan Nuclear Receptors/metabolism , Phenotype , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism
15.
Breast Cancer Res ; 18(1): 123, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27927249

ABSTRACT

BACKGROUND: To date, all studies conducted on breast cancer diagnosis have focused on the expression of the full-length 66-kDa estrogen receptor alpha (ERα66). However, much less attention has been paid to a shorter 46-kDa isoform (ERα46), devoid of the N-terminal region containing the transactivation function AF-1. Here, we investigated the expression levels of ERα46 in breast tumors in relation to tumor grade and size, and examined the mechanism of its generation and its specificities of coregulatory binding and its functional activities. METHODS: Using approaches combining immunohistochemistry, Western blotting, and proteomics, antibodies allowing ERα46 detection were identified and the expression levels of ERα46 were quantified in 116 ERα-positive human breast tumors. ERα46 expression upon cellular stress was studied, and coregulator bindings, transcriptional, and proliferative response were determined to both ERα isoforms. RESULTS: ERα46 was expressed in over 70% of breast tumors at variable levels which sometimes were more abundant than ERα66, especially in differentiated, lower-grade, and smaller-sized tumors. We also found that ERα46 can be generated via internal ribosome entry site-mediated translation in the context of endoplasmic reticulum stress. The binding affinities of both unliganded and fully-activated receptors towards co-regulator peptides revealed that the respective potencies of ERα46 and ERα66 differ significantly, contributing to the differential transcriptional activity of target genes to 17ß estradiol (E2). Finally, increasing amounts of ERα46 decrease the proliferation rate of MCF7 tumor cells in response to E2. CONCLUSIONS: We found that, besides the full-length ERα66, the overlooked ERα46 isoform is also expressed in a majority of breast tumors. This finding highlights the importance of the choice of antibodies used for the diagnosis of breast cancer, which are able or not to detect the ERα46 isoform. In addition, since the function of both ERα isoforms differs, this work underlines the need to develop new technologies in order to discriminate ERα66 and ERα46 expression in breast cancer diagnosis which could have potential clinical relevance.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Alternative Splicing , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cluster Analysis , Endoplasmic Reticulum Stress , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Protein Binding , Protein Biosynthesis , Protein Isoforms , Proteome , Proteomics/methods , Retrospective Studies
16.
Toxicol Appl Pharmacol ; 313: 138-148, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27773686

ABSTRACT

Chemicals that alter normal function of farnesoid X receptor (FXR) have been shown to affect the homeostasis of bile acids, glucose, and lipids. Several structural classes of environmental chemicals and drugs that modulated FXR transactivation were previously identified by quantitative high-throughput screening (qHTS) of the Tox21 10K chemical collection. In the present study, we validated the FXR antagonist activity of selected structural classes, including avermectin anthelmintics, dihydropyridine calcium channel blockers, 1,3-indandione rodenticides, and pyrethroid pesticides, using in vitro assay and quantitative structural-activity relationship (QSAR) analysis approaches. (Z)-Guggulsterone, chlorophacinone, ivermectin, and their analogs were profiled for their ability to alter CDCA-mediated FXR binding using a panel of 154 coregulator motifs and to induce or inhibit transactivation and coactivator recruitment activities of constitutive androstane receptor (CAR), liver X receptor alpha (LXRα), or pregnane X receptor (PXR). Our results showed that chlorophacinone and ivermectin had distinct modes of action (MOA) in modulating FXR-coregulator interactions and compound selectivity against the four aforementioned functionally-relevant nuclear receptors. These findings collectively provide mechanistic insights regarding compound activities against FXR and possible explanations for in vivo toxicological observations of chlorophacinone, ivermectin, and their analogs.


Subject(s)
Indans/pharmacology , Ivermectin/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , HEK293 Cells , Humans , Ivermectin/analogs & derivatives , Structure-Activity Relationship
17.
Proc Natl Acad Sci U S A ; 110(19): 7910-5, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23613579

ABSTRACT

Glucocorticoid receptor (GR) antagonism may be of considerable therapeutic value in stress-related psychopathology such as depression. However, blockade of all GR-dependent processes in the brain will lead to unnecessary and even counteractive effects, such as elevated endogenous cortisol levels. Selective GR modulators are ligands that can act both as agonist and as antagonist and may be used to separate beneficial from harmful treatment effects. We have discovered that the high-affinity GR ligand C108297 is a selective modulator in the rat brain. We first demonstrate that C108297 induces a unique interaction profile between GR and its downstream effector molecules, the nuclear receptor coregulators, compared with the full agonist dexamethasone and the antagonist RU486 (mifepristone). C108297 displays partial agonistic activity for the suppression of hypothalamic corticotropin-releasing hormone (CRH) gene expression and potently enhances GR-dependent memory consolidation of training on an inhibitory avoidance task. In contrast, it lacks agonistic effects on the expression of CRH in the central amygdala and antagonizes GR-mediated reduction in hippocampal neurogenesis after chronic corticosterone exposure. Importantly, the compound does not lead to disinhibition of the hypothalamus-pituitary-adrenal axis. Thus, C108297 represents a class of ligands that has the potential to more selectively abrogate pathogenic GR-dependent processes in the brain, while retaining beneficial aspects of GR signaling.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Animals , Brain/embryology , Brain/physiology , Corticotropin-Releasing Hormone/antagonists & inhibitors , Dexamethasone/pharmacology , Hippocampus/metabolism , Ligands , Male , Mifepristone/pharmacology , Nuclear Receptor Coactivator 1/metabolism , Peptides/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/metabolism , Steroids/metabolism , Time Factors , Transcription, Genetic , Two-Hybrid System Techniques
18.
Chem Res Toxicol ; 27(2): 304-308, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24460184

ABSTRACT

Heterotropic cooperativity of human cytochrome P450 (P450) 3A4/3A5 by the teratogen thalidomide was recently demonstrated by H. Yamazaki et al. ( ( 2013 ) Chem. Res. Toxicol. 26 , 486 - 489 ) using the model substrate midazolam in various in vitro and in vivo models. Chimeric mice with humanized liver also displayed enhanced midazolam clearance upon pretreatment with orally administered thalidomide, presumably because of human P450 3A induction. In the current study, we further investigated the regulation of human hepatic drug metabolizing enzymes. Thalidomide enhanced levels of P450 3A4 and 2B6 mRNA, protein expression, and/or oxidation activity in human hepatocytes, indirectly suggesting the activation of upstream transcription factors involved in detoxication, e.g., the nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR). A key event after ligand binding is an alteration of nuclear receptor conformation and recruitment of coregulator proteins that alter chromatin accessibility of target genes. To investigate direct engagement and functional alteration of PXR and CAR by thalidomide, we utilized a peptide microarray with 154 coregulator-derived nuclear receptor-interaction motifs and coregulator and nuclear receptor boxes, which serves as a sensor for nuclear receptor conformation and activity status as a function of ligand. Thalidomide and its human proximate metabolite 5-hydroxythalidomide displayed significant modulation of coregulator interaction with PXR and CAR ligand-binding domains, similar to established agonists for these receptors. These results collectively suggest that thalidomide acts as a ligand for PXR and CAR and causes enzyme induction leading to increased P450 enzyme activity. The possibilities of drug interactions during thalidomide therapy in humans require further evaluation.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Hepatocytes/metabolism , Receptors, Steroid/metabolism , Thalidomide/pharmacology , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Biotransformation , Cell Line , Cells, Cultured , Constitutive Androstane Receptor , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/genetics , Humans , Pregnane X Receptor , Protein Array Analysis , Receptors, Cytoplasmic and Nuclear/metabolism
19.
Bone Res ; 12(1): 44, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164247

ABSTRACT

The vitamin D receptor (VDR) plays a critical role in the regulation of mineral and bone homeostasis. Upon binding of 1α,25-dihydroxyvitamin D3 to the VDR, the activation function 2 (AF2) domain repositions and recruits coactivators for the assembly of the transcriptional machinery required for gene transcription. In contrast to coactivator-induced transcriptional activation, the functional effects of coactivator-independent VDR signaling remain unclear. In humans, mutations in the AF2 domain are associated with hereditary vitamin D-resistant rickets, a genetic disorder characterized by impaired bone mineralization and growth. In the present study, we used mice with a systemic or conditional deletion of the VDR-AF2 domain (VdrΔAF2) to study coactivator-independent VDR signaling. We confirm that ligand-induced transcriptional activation was disabled because the mutant VDRΔAF2 protein was unable to interact with coactivators. Systemic VdrΔAF2 mice developed short, undermineralized bones with dysmorphic growth plates, a bone phenotype that was more pronounced than that of systemic Vdr knockout (Vdr-/-) mice. Interestingly, a rescue diet that is high in calcium, phosphate, and lactose, normalized this phenotype in Vdr-/-, but not in VdrΔAF2 mice. However, osteoblast- and osteoclast-specific VdrΔAF2 mice did not recapitulate this bone phenotype indicating coactivator-independent VDR effects are more important in other organs. In addition, RNA-sequencing analysis of duodenum and kidney revealed a decreased expression of VDR target genes in systemic VdrΔAF2 mice, which was not observed in Vdr-/- mice. These genes could provide new insights in the compensatory (re)absorption of minerals that are crucial for bone homeostasis. In summary, coactivator-independent VDR effects contribute to mineral and bone homeostasis.


Subject(s)
Calcium , Lactose , Phosphates , Receptors, Calcitriol , Rickets , Signal Transduction , Animals , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/genetics , Mice , Rickets/metabolism , Rickets/genetics , Rickets/pathology , Rickets/prevention & control , Phosphates/metabolism , Calcium/metabolism , Lactose/metabolism , Mice, Knockout , Diet , Mice, Inbred C57BL
20.
Chem Res Toxicol ; 26(3): 336-46, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23383871

ABSTRACT

Testing chemicals for their endocrine-disrupting potential, including interference with estrogen receptor (ER) signaling, is an important aspect of chemical safety testing. Because of the practical drawbacks of animal testing, the development of in vitro alternatives for the uterotrophic assay and other in vivo (anti)estrogenicity tests has high priority. It was previously demonstrated that an in vitro assay that profiles ligand-induced binding of ERα to a microarray of coregulator-derived peptides might be a valuable candidate for a panel of in vitro assays aiming at an ultimate replacement of the uterotrophic assay. In the present study, the reproducibility and robustness of this coregulator binding assay was determined by measuring the binding profiles of 14 model compounds that are recommended by the Office of Prevention, Pesticides and Toxic Substances for testing laboratory proficiency in estrogen receptor transactivation assays. With a median coefficient of variation of 5.0% and excellent correlation (R(2) = 0.993) between duplicate measurements, the reproducibility of the ERα-coregulator binding assay was better than the reproducibility of other commonly used in vitro ER functional assays. In addition, the coregulator binding assay is correctly predicting the estrogenicity for 13 out of 14 compounds tested. When the potency of the ER-agonists to induce ERα-coregulator binding was compared to their ER binding affinity, their ranking was similar, and the correlation between the EC50 values was excellent (R(2) = 0.96), as was the correlation with their potency in a transactivation assay (R(2) = 0.94). Moreover, when the ERα-coregulator binding profiles were hierarchically clustered using Euclidian cluster distance, the structurally related compounds were found to cluster together, whereas the steroid test compounds having an aromatic A-ring were separated from those with a cyclohexene A-ring. We concluded that this assay is capable of distinguishing ERα agonists and antagonists and that it even reflects the structural similarity of ERα agonists, indicating a potential to achieve identification and classification of ERα endocrine disruptors with high fidelity.


Subject(s)
Drug Evaluation, Preclinical/methods , Endocrine Disruptors/chemistry , Endocrine Disruptors/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Protein Array Analysis/methods , Cell Line , Estrogen Antagonists/chemistry , Estrogen Antagonists/pharmacology , Humans , Ligands , Protein Binding , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL