Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
Mol Cell ; 81(19): 4041-4058.e15, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34624217

ABSTRACT

Deregulation of oncogenic signals in cancer triggers replication stress. Immediate early genes (IEGs) are rapidly and transiently expressed following stressful signals, contributing to an integrated response. Here, we find that the orphan nuclear receptor NR4A1 localizes across the gene body and 3' UTR of IEGs, where it inhibits transcriptional elongation by RNA Pol II, generating R-loops and accessible chromatin domains. Acute replication stress causes immediate dissociation of NR4A1 and a burst of transcriptionally poised IEG expression. Ectopic expression of NR4A1 enhances tumorigenesis by breast cancer cells, while its deletion leads to massive chromosomal instability and proliferative failure, driven by deregulated expression of its IEG target, FOS. Approximately half of breast and other primary cancers exhibit accessible chromatin domains at IEG gene bodies, consistent with this stress-regulatory pathway. Cancers that have retained this mechanism in adapting to oncogenic replication stress may be dependent on NR4A1 for their proliferation.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Immediate-Early Proteins/metabolism , Mitosis , Neoplastic Cells, Circulating/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , 3' Untranslated Regions , Animals , Antineoplastic Agents/pharmacology , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chromatin Assembly and Disassembly , Female , Gene Expression Regulation, Neoplastic , Genomic Instability , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Indoles/pharmacology , MCF-7 Cells , Mice, Inbred NOD , Mice, SCID , Mitosis/drug effects , Neoplastic Cells, Circulating/drug effects , Neoplastic Cells, Circulating/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phenylacetates/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , R-Loop Structures , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Signal Transduction , Transcription Elongation, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Proc Natl Acad Sci U S A ; 120(32): e2220849120, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37494420

ABSTRACT

The ovarian follicle reserve, formed pre- or perinatally, comprises all oocytes for lifetime reproduction. Depletion of this reserve results in infertility. Steroidogenic factor 1 (SF-1; Nr5a1) and liver receptor homolog 1 (LRH-1; Nr5a2) are two orphan nuclear receptors that regulate adult endocrine function, but their role in follicle formation is unknown. We developed models of conditional depletion of SF-1 or LRH-1 from prenatal ovaries. Depletion of SF-1, but not LRH-1, resulted in dramatically smaller ovaries and fewer primordial follicles. This was mediated by increased oocyte death, resulting from increased ovarian inflammation and increased Notch signaling. Major dysregulated genes were Iroquois homeobox 3 and 5 and their downstream targets involved in the establishment of the ovarian laminin matrix and oocyte-granulosa cell gap junctions. Disruptions of these pathways resulted in follicles with impaired basement membrane formation and compromised oocyte-granulosa communication networks, believed to render them more prone to atresia. This study identifies SF-1 as a key regulator of the formation of the ovarian reserve.


Subject(s)
Ovarian Reserve , Pregnancy , Female , Humans , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Ovarian Reserve/genetics , Ovarian Follicle/metabolism , Ovary/metabolism , Granulosa Cells/metabolism
3.
BMC Pulm Med ; 24(1): 130, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38491484

ABSTRACT

Bronchopulmonary dysplasia (BPD) is characterized by alveolar dysplasia, and evidence indicates that interferon regulatory factor 4 (IRF4) is involved in the pathogenesis of various inflammatory lung diseases. Nonetheless, the significance and mechanism of IRF4 in BPD remain unelucidated. Consequently, we established a mouse model of BPD through hyperoxia exposure, and ELISA was employed to measure interleukin-17 A (IL-17 A) and interleukin-6 (IL-6) expression levels in lung tissues. Western blotting was adopted to determine the expression of IRF4, surfactant protein C (SP-C), and podoplanin (T1α) in lung tissues. Flow cytometry was utilized for analyzing the percentages of FOXP3+ regulatory T cells (Tregs) and FOXP3+RORγt+ Tregs in CD4+ T cells in lung tissues to clarify the underlying mechanism. Our findings revealed that BPD mice exhibited disordered lung tissue structure, elevated IRF4 expression, decreased SP-C and T1α expression, increased IL-17 A and IL-6 levels, reduced proportion of FOXP3+ Tregs, and increased proportion of FOXP3+RORγt+ Tregs. For the purpose of further elucidating the effect of IRF4 on Treg phenotype switching induced by hyperoxia in lung tissues, we exposed neonatal mice with IRF4 knockout to hyperoxia. These mice exhibited regular lung tissue structure, increased proportion of FOXP3+ Tregs, reduced proportion of FOXP3+RORγt+ Tregs, elevated SP-C and T1α expression, and decreased IL-17 A and IL-6 levels. In conclusion, our findings demonstrate that IRF4-mediated Treg phenotype switching in lung tissues exacerbates alveolar epithelial cell injury under hyperoxia exposure.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Animals , Mice , Alveolar Epithelial Cells/pathology , T-Lymphocytes, Regulatory/metabolism , Interleukin-17/metabolism , Interleukin-6/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Hyperoxia/complications , Bronchopulmonary Dysplasia/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Phenotype , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism
4.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33853949

ABSTRACT

Fibroblast growth factor 23 (FGF23), a hormone generally derived from bone, is important in phosphate and vitamin D homeostasis. In acute kidney injury (AKI) patients, high-circulating FGF23 levels are associated with disease progression and mortality. However, the organ and cell type of FGF23 production in AKI and the molecular mechanism of its excessive production are still unidentified. For insight, we investigated folic acid (FA)-induced AKI in mice. Interestingly, simultaneous with FGF23, orphan nuclear receptor ERR-γ expression is increased in the liver of FA-treated mice, and ectopic overexpression of ERR-γ was sufficient to induce hepatic FGF23 production. In patients and in mice, AKI is accompanied by up-regulated systemic IL-6, which was previously identified as an upstream regulator of ERR-γ expression in the liver. Administration of IL-6 neutralizing antibody to FA-treated mice or of recombinant IL-6 to healthy mice confirms IL-6 as an upstream regulator of hepatic ERR-γ-mediated FGF23 production. A significant (P < 0.001) interconnection between high IL-6 and FGF23 levels as a predictor of AKI in patients that underwent cardiac surgery was also found, suggesting the clinical relevance of the finding. Finally, liver-specific depletion of ERR-γ or treatment with an inverse ERR-γ agonist decreased hepatic FGF23 expression and plasma FGF23 levels in mice with FA-induced AKI. Thus, inverse agonist of ERR-γ may represent a therapeutic strategy to reduce adverse plasma FGF23 levels in AKI.


Subject(s)
Acute Kidney Injury/physiopathology , Fibroblast Growth Factor-23/metabolism , Receptors, Estrogen/metabolism , Acute Kidney Injury/metabolism , Animals , Disease Models, Animal , Fibroblast Growth Factor-23/genetics , Folic Acid/adverse effects , Folic Acid/pharmacology , Interleukin-6/metabolism , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/metabolism , Receptors, Estrogen/genetics , Transcriptional Activation
5.
Int J Mol Sci ; 25(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38474099

ABSTRACT

Hypercapnia occurs when the partial pressure of carbon dioxide (CO2) in the blood exceeds 45 mmHg. Hypercapnia is associated with several lung pathologies and is transcriptionally linked to suppression of immune and inflammatory signalling through poorly understood mechanisms. Here we propose Orphan Nuclear Receptor Family 4A (NR4A) family members NR4A2 and NR4A3 as potential transcriptional regulators of the cellular response to hypercapnia in monocytes. Using a THP-1 monocyte model, we investigated the sensitivity of NR4A family members to CO2 and the impact of depleting NR4A2 and NR4A3 on the monocyte response to buffered hypercapnia (10% CO2) using RNA-sequencing. We observed that NR4A2 and NR4A3 are CO2-sensitive transcription factors and that depletion of NR4A2 and NR4A3 led to reduced CO2-sensitivity of mitochondrial and heat shock protein (Hsp)-related genes, respectively. Several CO2-sensitive genes were, however, refractory to depletion of NR4A2 and NR4A3, indicating that NR4As regulate certain elements of the cellular response to buffered hypercapnia but that other transcription factors also contribute. Bioinformatic analysis of conserved CO2-sensitive genes implicated several novel putative CO2-sensitive transcription factors, of which the ETS Proto-Oncogene 1 Transcription Factor (ETS-1) was validated to show increased nuclear expression in buffered hypercapnia. These data give significant insights into the understanding of immune responses in patients experiencing hypercapnia.


Subject(s)
Orphan Nuclear Receptors , Receptors, Steroid , Humans , Orphan Nuclear Receptors/genetics , Monocytes/metabolism , Hypercapnia , Carbon Dioxide , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Receptors, Steroid/metabolism , DNA-Binding Proteins , Receptors, Thyroid Hormone
6.
J Obstet Gynaecol Res ; 48(7): 1501-1512, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35445497

ABSTRACT

AIM: This review aims to summarize the research focused upon the functions of nuclear hormone receptor 4A (NR4A) in the human reproductive system. The research questions addressed are to decipher what role the NR4A subfamily plays in the regulation of the human reproductive system and effects upon fertility issues through regulation of the expression of the NR4A subfamily. METHODS: The electronic database PubMed was searched for studies published before November 2021. Keywords included "NR4A," "trophoblast," "decidualization," "folliculogenesis," "estrogen," "pregnancy," "Leydig cells," "fertility," and "reproductive." Relevant references from retrieved manuscripts and review articles were also searched manually. RESULTS: NR4A subfamily are involved in trophoblast differentiation, endometrial decidualization, embryo adhesion, secretion of related hormones, and regulation of spontaneous term labor. Besides, many studies have provided strong evidence that they play critical roles in spermatogenesis. Furthermore, Multiple mechanisms can affect the expression of NR4As. Broadly, NR4A family receptors affect the human reproductive system in multiple ways. CONCLUSIONS: Further research is needed to specifically dissect the functions and regulatory mechanisms of these receptors and their pharmaceutical antagonists and agonists. The connection between the NR4A subfamily and a variety of reproductive disorders needs to be proven experimentally such that further examination of human tissue is required to assess the role of these receptors in human reproductive diseases.


Subject(s)
Gene Expression Regulation , Nuclear Receptor Subfamily 4, Group A, Member 1 , Endometrium/metabolism , Female , Humans , Male , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Signal Transduction , Trophoblasts/metabolism
7.
Genes Chromosomes Cancer ; 60(2): 61-72, 2021 02.
Article in English | MEDLINE | ID: mdl-33094510

ABSTRACT

Colorectal cancer (CRC) is a high incidence cancer and major cause of cancer mortality. Though disease-causing tumor suppressors for major syndromes are well characterized, about 10% of CRC is familial but without mutations in known tumor suppressors. We exhaustively screened 100 polyposis families for APC germline mutations and identified 13, which are APC mutation-negative, microsatellite-stable (MSS), and with undetectable mutation in known tumor suppressors. Whole exome sequencing in three probands uncovered two with germline frameshift NR0B2 mutations, c.293_301delTTGGGTTGGinsAC and c.227delT. Sanger Sequencing identified a third proband with NR0B2 c.157_166delCATCGCACCT frameshift mutation. All three mutations deleted the C-terminus activation/repression domain of NR0B2, thus are loss-of-function mutations. Real-time RT-PCR performed on tumor and matched mucosa of one patient revealed that NR0B2 downstream targets, SMAD3 was derepressed while GLI1 was downregulated in the colonic mucosa compared to healthy controls. Truncated NR0B2 molecule was predicted to have weakened binding with interacting partners SMAD3, GLI1, BCL2, and RXRα, implying perturbation of TGF-ß, Hedgehog, anti-apoptotic and nuclear hormone receptor signaling pathways. Immunostaining also revealed nuclear retention of the most severely truncated NR0B2 molecule compared to the wildtype. Microsatellite and sequencing analysis did not detect loss of wildtype allele in probands' tumors. The patient who acquired somatic KRAS mutation progressed rapidly whist the other two patients manifested with late-onset obesity and diabetes. We propose that haploinsufficiency of NR0B2 is associated with a novel CRC syndrome with metabolic phenotypes.


Subject(s)
Carcinoma/genetics , Colorectal Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Adenomatous Polyposis Coli Protein/genetics , Adult , Age of Onset , Carcinoma/pathology , Colorectal Neoplasms/pathology , Female , Haploinsufficiency , Humans , Male , Microsatellite Repeats/genetics , Middle Aged , Mutation , Pedigree , Protein Binding , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoid X Receptor alpha/metabolism , Smad3 Protein/metabolism , Zinc Finger Protein GLI1/metabolism
8.
Int J Mol Sci ; 22(19)2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34639006

ABSTRACT

Breast cancer development is associated with macrophage infiltration and differentiation in the tumor microenvironment. Our previous study highlights the crucial function of reactive oxygen species (ROS) in enhancing macrophage infiltration during the disruption of mammary tissue polarity. However, the regulation of ROS and ROS-associated macrophage infiltration in breast cancer has not been fully determined. Previous studies identified retinoid orphan nuclear receptor alpha (RORα) as a potential tumor suppressor in human breast cancer. In the present study, we showed that retinoid orphan nuclear receptor alpha (RORα) significantly decreased ROS levels and inhibited ROS-mediated cytokine expression in breast cancer cells. RORα expression in mammary epithelial cells inhibited macrophage infiltration by repressing ROS generation in the co-culture assay. Using gene co-expression and chromatin immunoprecipitation (ChIP) analyses, we identified complex I subunits NDUFS6 and NDUFA11 as RORα targets that mediated its function in suppressing superoxide generation in mitochondria. Notably, the expression of RORα in 4T1 cells significantly inhibited cancer metastasis, reduced macrophage accumulation, and enhanced M1-like macrophage differentiation in tumor tissue. In addition, reduced RORα expression in breast cancer tissue was associated with an increased incidence of cancer metastasis. These results provide additional insights into cancer-associated inflammation, and identify RORα as a potential target to suppress ROS-induced mammary tumor progression.


Subject(s)
Electron Transport Complex I/metabolism , Neoplasms/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Reactive Oxygen Species/metabolism , Animals , Biomarkers , Cell Respiration , Cytokines/metabolism , Energy Metabolism , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
9.
Int J Mol Sci ; 22(11)2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34199599

ABSTRACT

Orphan nuclear receptor estrogen-related receptor γ (ERRγ) is an important transcription factor modulating gene transcription involved in endocrine control of liver metabolism. Transferrin receptor 2 (TFR2), a carrier protein for transferrin, is involved in hepatic iron overload in alcoholic liver disease (ALD). However, TFR2 gene transcriptional regulation in hepatocytes remains largely unknown. In this study, we described a detailed molecular mechanism of hepatic TFR2 gene expression involving ERRγ in response to an endocannabinoid 2-arachidonoylglycerol (2-AG). Treatment with 2-AG and arachidonyl-2'-chloroethylamide, a selective cannabinoid receptor type 1 (CB1) receptor agonist, increased ERRγ and TFR2 expression in hepatocytes. Overexpression of ERRγ was sufficient to induce TFR2 expression in both human and mouse hepatocytes. In addition, ERRγ knockdown significantly decreased 2-AG or alcohol-mediated TFR2 gene expression in cultured hepatocytes and mouse livers. Finally, deletion and mutation analysis of the TFR2 gene promoter demonstrated that ERRγ directly modulated TFR2 gene transcription via binding to an ERR-response element. This was further confirmed by chromatin immunoprecipitation assay. Taken together, these results reveal a previously unrecognized role of ERRγ in the transcriptional regulation of TFR2 gene expression in response to alcohol.


Subject(s)
Liver Diseases, Alcoholic/genetics , Liver/drug effects , Receptor, Cannabinoid, CB1/genetics , Receptors, Estrogen/genetics , Receptors, Transferrin/genetics , Alcohols/pharmacology , Animals , Arachidonic Acids/pharmacology , Endocannabinoids/pharmacology , Gene Expression Regulation/drug effects , Glycerides/pharmacology , Hep G2 Cells , Hepatocytes/drug effects , Humans , Iron/metabolism , Liver/metabolism , Liver/pathology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Mice , Promoter Regions, Genetic , Receptor, Cannabinoid, CB1/agonists , Sequence Deletion/genetics , Transferrin/genetics , Transferrin/metabolism
10.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114319

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) is a ligand-sensing transcription factor and presents as a potential drug target in metabolic diseases and cancer. In humans, mutations in the HNF4α gene cause maturity-onset diabetes of the young (MODY), and the elevated activity of this protein has been associated with gastrointestinal cancers. Despite the high therapeutic potential, available ligands and structure-activity relationship knowledge for this nuclear receptor are scarce. Here, we disclose a chemically diverse collection of orthogonally validated fragment-like activators as well as inverse agonists, which modulate HNF4α activity in a low micromolar range. These compounds demonstrate the druggability of HNF4α and thus provide a starting point for medicinal chemistry as well as an early tool for chemogenomics.


Subject(s)
Hepatocyte Nuclear Factor 4/chemistry , Hepatocyte Nuclear Factor 4/metabolism , Small Molecule Libraries/pharmacology , Calorimetry , Drug Discovery , Drug Evaluation, Preclinical , Fructose-Bisphosphatase/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Humans , Ligands , Molecular Structure , Small Molecule Libraries/chemistry , Structure-Activity Relationship
11.
Am J Physiol Lung Cell Mol Physiol ; 317(5): L615-L624, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31461311

ABSTRACT

Nur77 is an orphan nuclear receptor implicated in the regulation of a wide range of biological processes, including the maintenance of systemic blood vessel homeostasis. Although Nur77 is known to be expressed in the lung, its role in regulating pulmonary vascular functions remains entirely unknown. In this study, we found that Nur77 is expressed at high levels in the lung, and its expression is markedly upregulated in response to LPS administration. While the pulmonary vasculature of mice that lacked Nur77 appeared to function normally under homeostatic conditions, we observed a dramatic decrease in its barrier functions after exposure to LPS, as demonstrated by an increase in serum proteins in the bronchoalveolar lavage fluid and a reduction in the expression of endothelial junctional proteins, such as vascular endothelial cadherin (VE-cadherin) and ß-catenin. Similarly, we found that siRNA knockdown of Nur77 in lung microvascular endothelial cells also reduced VE-cadherin and ß-catenin expression and increased the quantity of fluorescein isothiocyanate-labeled dextran transporting across LPS-injured endothelial monolayers. Consistent with Nur77 playing a vascular protective role, we found that adenoviral-mediated overexpression of Nur77 both enhanced expression of VE-cadherin and ß-catenin and augmented endothelial barrier protection to LPS in cultured cells. Mechanistically, Nur77 appeared to mediate its protective effects, at least in part, by binding to ß-catenin and preventing its degradation. Our findings demonstrate a key role for Nur77 in the maintenance of lung endothelial barrier protection to LPS and suggest that therapeutic strategies aimed at augmenting Nur77 levels might be effective in treating a wide variety of inflammatory vascular diseases of the lung.


Subject(s)
Acute Lung Injury/complications , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Lipopolysaccharides/adverse effects , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Pneumonia/prevention & control , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Mice, Knockout , Pneumonia/etiology , Pneumonia/pathology
12.
J Pineal Res ; 67(2): e12579, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30958896

ABSTRACT

Exercise-induced physiological hypertrophy provides protection against cardiovascular disease, whereas disease-induced pathological hypertrophy leads to heart failure. Emerging evidence suggests pleiotropic roles of melatonin in cardiac disease; however, the effects of melatonin on physiological vs pathological cardiac hypertrophy remain unknown. Using swimming-induced physiological hypertrophy and pressure overload-induced pathological hypertrophy models, we found that melatonin treatment significantly improved pathological hypertrophic responses accompanied by alleviated oxidative stress in myocardium but did not affect physiological cardiac hypertrophy and oxidative stress levels. As an important mediator of melatonin, the retinoid-related orphan nuclear receptor-α (RORα) was significantly decreased in human and murine pathological hypertrophic cardiomyocytes, but not in swimming-induced physiological hypertrophic murine hearts. In vivo and in vitro loss-of-function experiments indicated that RORα deficiency significantly aggravated pathological cardiac hypertrophy, and notably weakened the anti-hypertrophic effects of melatonin. Mechanistically, RORα mediated the cardioprotection of melatonin in pathological hypertrophy mainly by transactivation of manganese-dependent superoxide dismutase (MnSOD) via binding to the RORα response element located in the promoter region of the MnSOD gene. Furthermore, MnSOD overexpression reversed the pro-hypertrophic effects of RORα deficiency, while MnSOD silencing abolished the anti-hypertrophic effects of RORα overexpression in pathological cardiac hypertrophy. Collectively, our findings provide the first evidence that melatonin exerts an anti-hypertrophic effect on pathological but not physiological cardiac hypertrophy via alleviating oxidative stress through transactivation of the antioxidant enzyme MnSOD in a RORα-dependent manner.


Subject(s)
Cardiomegaly/metabolism , Melatonin/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Signal Transduction , Superoxide Dismutase/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Disease Models, Animal , Mice , Mice, Mutant Strains , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Superoxide Dismutase/genetics
13.
Bioorg Med Chem Lett ; 29(12): 1463-1470, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31010722

ABSTRACT

We have previously reported the syntheses of a series of 3,6-disubstituted quinolines as modulators of the retinoic acid receptor-related orphan receptor gamma t (RORγt). These molecules are potent binders but are high molecular weight and they exhibited poor solubility at pH 2 and pH 7. This manuscript details our efforts at improving physical chemical properties for this series of compounds by increasing the diversity at the 3-position (i.e. introducing heteroatoms and lowering the molecular weight). These efforts have led to molecules which are potent binders with improved solubility.


Subject(s)
Drug Inverse Agonism , Quinolines/agonists , Animals , Humans , Molecular Structure , Structure-Activity Relationship
14.
J Biochem Mol Toxicol ; 33(6): e22309, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30801912

ABSTRACT

Glucocorticoids impair testosterone synthesis by an unknown mechanism. Stallions treated with the synthetic glucocorticoid dexamethasone had testes collected at 6 or 12 hours postinjection. The testicular expression of selected genes encoding nuclear receptors and steroidogenic enzymes was measured. At 6 hours, dexamethasone treatment decreased levels of NR0B2, NR4A1, NR5A1, and NR5A2 messenger RNAs (mRNAs) and NR5A2 mRNA levels remained depressed at 12 hours. In contrast, dexamethasone increased levels of NFKBIA mRNA at both time points. At 6 hours, dexamethasone did not alter levels of NR0B1, NR2F1, NR2F2, NR3C1, CYP11A1, CYP17A1, CYP19A1, DHCR24, GSTA3, HSD3B2, HSD17B3, LHCGR, or STAR mRNAs. In primary cultures of Leydig cells, 10 -9 and 10 -7 M dexamethasone decreased levels of NR4A1 and NR5A1 mRNAs and increased those of NFKBIA mRNA. Our discovery that dexamethasone downregulates NR4A1, NR5A1, and NR5A2 genes, known to be important for testicular functions, may be part of the mechanism by which glucocorticoids acutely decreases testosterone.


Subject(s)
Dexamethasone/adverse effects , Down-Regulation/drug effects , Leydig Cells/metabolism , Orphan Nuclear Receptors/biosynthesis , Testosterone/biosynthesis , Animals , Cytochrome P-450 Enzyme System/biosynthesis , Dexamethasone/pharmacology , Horses , Male
15.
Molecules ; 23(3)2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29498706

ABSTRACT

Medullary thyroid cancer (MTC) is a relatively rare thyroid cancer responsible for a substantial fraction of thyroid cancer mortality. More effective therapeutic drugs with low toxicity for MTC are urgently needed. Orphan nuclear receptor 4A1 (NR4A1) plays a pivotal role in regulating the proliferation and apoptosis of a variety of tumor cells. Based on the NR4A1 protein structure, 2-imino-6-methoxy-2H-chromene-3-carbothioamide (IMCA) was identified from the Specs compounds database using the protein structure-guided virtual screening approach. Computationally-based molecular modeling studies suggested that IMCA has a high affinity for the ligand binding pocket of NR4A1. MTT [3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide] and apoptosis assays demonstrated that IMCA resulted in significant thyroid cancer cell death. Immunofluorescence assays showed that IMCA induced NR4A1 translocation from the nucleus to the cytoplasm in thyroid cancer cell lines, which may be involved in the cell apoptotic process. In this study, the quantitative polymerase chain reaction results showed that the IMCA-induced upregulation of sestrin1 and sestrin2 was dose-dependent in thyroid cancer cell lines. Western blot showed that IMCA increased phosphorylation of adenosine 5'-monophosphate-activated protein kinase (AMPK) and decreased phosphorylation of ribosomal protein S6 kinase (p70S6K), which is the key enzyme in the mammalian target of rapamycin (mTOR) pathway. The experimental results suggest that IMCA is a drug candidate for MTC therapy and may work by increasing the nuclear export of NR4A1 to the cytoplasm and the tumor protein 53 (p53)-sestrins-AMPK-mTOR signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Thyroid Gland/drug effects , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzopyrans/chemistry , Binding Sites , Cell Line, Tumor , Humans , Molecular Docking Simulation , Molecular Targeted Therapy/methods , Nuclear Receptor Subfamily 4, Group A, Member 1/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thyroid Gland/metabolism , Thyroid Gland/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Bioorg Med Chem Lett ; 27(23): 5277-5283, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29079472

ABSTRACT

We identified 6-substituted quinolines as modulators of the retinoic acid receptor-related orphan receptor gamma t (RORγt). The synthesis of this class of RORγt modulators is reported, and optimization of the substituents at the quinoline 6-position that produced compounds with high affinity for the receptor is detailed. This effort identified molecules that act as potent, full inverse agonists in a RORγt-driven cell-based reporter assay. The X-ray crystal structures of two full inverse agonists from this chemical series bound to the RORγt ligand binding domain are disclosed, and we highlight the interaction of a hydrogen-bond acceptor on the 6-position substituent of the inverse agonist with Glu379:NH as a conserved binding contact.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Quinolines/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Ligands , Models, Molecular , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 27(9): 2047-2057, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28318945

ABSTRACT

A high-throughput screen of the ligand binding domain of the nuclear receptor retinoic acid-related orphan receptor gamma t (RORγt) employing a thermal shift assay yielded a quinoline tertiary alcohol hit. Optimization of the 2-, 3- and 4-positions of the quinoline core using structure-activity relationships and structure-based drug design methods led to the discovery of a series of modulators with improved RORγt inhibitory potency and inverse agonism properties.


Subject(s)
Drug Design , Drug Inverse Agonism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Quinolines/chemistry , Quinolines/pharmacology , Humans , Molecular Docking Simulation , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Structure-Activity Relationship , Th17 Cells/drug effects
18.
Cell Mol Life Sci ; 73(1): 95-117, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26511867

ABSTRACT

The aryl hydrocarbon receptor (AHR) is a cytoplasmic transcription factor, which plays an essential role in the xenobiotic metabolism in a wide variety of cells. The AHR gene is evolutionarily conserved and it has a central role not only in the differentiation and maturation of many tissues, but also in the toxicological metabolism of the cell by the activation of metabolizing enzymes. Several lines of evidence support that both AHR agonists and antagonists have profound immunological effects; and recently, the AHR has been implicated in antibacterial host defense. According to recent studies, the AHR is essential for the differentiation and activation of T helper 17 (Th17) cells. It is well known that Th17 cells have a central role in the development of inflammation, which is crucial in the defense against pathogens. In addition, Th17 cells play a major role in the pathogenesis of several autoimmune diseases such as rheumatoid arthritis. Therefore, the AHR may provide connection between the environmental chemicals, the immune regulation, and autoimmunity. In the present review, we summarize the role of the AHR in the Th17 cell functions.


Subject(s)
Cell Differentiation , Lymphocyte Activation , Receptors, Aryl Hydrocarbon/immunology , Th17 Cells/immunology , Animals , Autoimmune Diseases/immunology , Cytokines/immunology , Drug Discovery , Humans , Immunity, Cellular , Inflammation/immunology , Receptors, Aryl Hydrocarbon/analysis , Th17 Cells/cytology
19.
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
20.
Biochem J ; 470(2): 181-93, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26348907

ABSTRACT

Bile acids are primarily synthesized from cholesterol in the liver and have important roles in dietary lipid absorption and cholesterol homoeostasis. Detailed roles of the orphan nuclear receptors regulating cholesterol 7α-hydroxylase (CYP7A1), the rate-limiting enzyme in bile acid synthesis, have not yet been fully elucidated. In the present study, we report that oestrogen-related receptor γ (ERRγ) is a novel transcriptional regulator of CYP7A1 expression. Activation of cannabinoid receptor type 1 (CB1 receptor) signalling induced ERRγ-mediated transcription of the CYP7A1 gene. Overexpression of ERRγ increased CYP7A1 expression in vitro and in vivo, whereas knockdown of ERRγ attenuated CYP7A1 expression. Deletion analysis of the CYP7A1 gene promoter and a ChIP assay revealed an ERRγ-binding site on the CYP7A1 gene promoter. Small heterodimer partner (SHP) inhibited the transcriptional activity of ERRγ and thus regulated CYP7A1 expression. Overexpression of ERRγ led to increased bile acid levels, whereas an inverse agonist of ERRγ, GSK5182, reduced CYP7A1 expression and bile acid synthesis. Finally, GSK5182 significantly reduced hepatic CB1 receptor-mediated induction of CYP7A1 expression and bile acid synthesis in alcohol-treated mice. These results provide the molecular mechanism linking ERRγ and bile acid metabolism.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/biosynthesis , Liver/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Estrogen/metabolism , Animals , Bile Acids and Salts/metabolism , Cells, Cultured , Cholesterol 7-alpha-Hydroxylase/genetics , Drug Inverse Agonism , Ethanol/pharmacology , Gene Expression , Glycerides/pharmacology , HEK293 Cells , Hepatocytes/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL