Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 55
1.
Bioorg Chem ; 141: 106887, 2023 12.
Article En | MEDLINE | ID: mdl-37801784

Docosahexaenoic acid (DHA) has a strong anti-inflammatory effect and is reported to bind to the ligand-binding domain (LBD) of the anti-inflammatory modulator Nur77. Recently, we have found that DHA ethanolamine (DHA-EA) exerts anti-inflammatory activity as a Nur77 modulator. Herein, using a fragment splicing-based drug design strategy, nineteen new DHA-EA derivatives were synthesized starting from DHA algae oil and then evaluated for their anti-inflammatory activity. The cell-based cytotoxicity assays showed that compounds J2, J9, and J18 had no noticeable effect on the cell morphology and viability of RAW 264.7, LO2, and MCR-5 cells. Meanwhile, J9 was identified as the most potent anti-inflammatory molecule in LPS-stimulated RAW 264.7 cells. Also, the molecular docking study and SPR assay demonstrated that J9 exhibited in vitro Nur77-binding affinity (KD = 8.58 × 10-6 M). Moreover, the mechanism studies revealed that the anti-inflammatory activity of J9 was associated with its inhibition of NF-κB activation in a Nur77-dependent manner. Notably, J9 could attenuate LPS-induced inflammation in the mouse acute lung injury (ALI) model. Overall, the DHA-EA derivative J9 targeting Nur77 is a potential candidate for developing anti-inflammatory and ALI-treating agents.


Docosahexaenoic Acids , Nuclear Receptor Subfamily 4, Group A, Member 1 , Animals , Mice , Anti-Inflammatory Agents/adverse effects , Docosahexaenoic Acids/pharmacology , Inflammation/drug therapy , Lipopolysaccharides , Molecular Docking Simulation , Ethanolamines/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors
2.
Eur J Med Chem ; 244: 114849, 2022 Dec 15.
Article En | MEDLINE | ID: mdl-36274272

Encouraged by our previous findings and in continuation of our ongoing study project in designing and synthesis of novel Nur77-targeting anti-cancer agents, a series of 5-((4-(pyridin-3-yl)pyrimidin-2-yl)amino)-1H-indole-2-carboxamide derivatives were designed, synthesized and biologically evaluated as potent Nur77 modulators. Among synthesized compounds, 8b maintained good potency against different liver cancer cell lines and other types of cancer cell lines while exhibiting lower toxicity than the positive compound celastrol. Moreover, 8b displayed excellent Nur77-binding activity, superior to the lead compound 10g and comparable to the reference compound celastrol. The cytotoxic action of 8b towards cancer cells was associated with its induction of Nur77-mitochondrial targeting and Nur77-dependent apoptosis. Notably, 8b has good in vivo safety and anti-hepatocellular carcinoma (HCC) activity. Altogether, this study reveals that 8b is a novel Nur77 modulator with great promise for further research.


Antineoplastic Agents , Carcinoma, Hepatocellular , Indoles , Liver Neoplasms , Nuclear Receptor Subfamily 4, Group A, Member 1 , Pentacyclic Triterpenes , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Indoles/chemistry , Indoles/pharmacology , Indoles/therapeutic use , Liver Neoplasms/drug therapy , Structure-Activity Relationship , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/therapeutic use , Apoptosis/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Molecular Targeted Therapy
3.
Mol Carcinog ; 61(1): 73-84, 2022 01.
Article En | MEDLINE | ID: mdl-34699643

Paraspeckles compound 1 (PSPC1) is a multifunctional protein that plays an important role in cancer cells, where PSPC1 is a master regulator of pro-oncogenic responses that includes activation of TGFß (TGFß1), TGFß-dependent EMT, and metastasis. The pro-oncogenic activities of PSPC1 closely resembled those observed for the orphan nuclear receptor 4A1 (NR4A1, Nur77) and knockdown of NR4A1 decreased expression of PSPC1 in MDA-MB-231 breast, H1299 lung, and SNU449 liver cancer cells. Similar results were observed in these same cell lines after treatment with bisindole-derived (CDIMs) NR4A1 antagonists. Moreover, PSPC1-dependent regulation of TGFß, genes associated with cancer stem cells and epithelial to mesenchymal transition (EMT) were also downregulated after NR4A1 silencing or treatment of breast, lung, and liver cancer cells with CDIM/NR4A1 antagonists. Results of chromatin immunoprecipitation (ChIP) assays suggest that NR4A1 regulates PSPC1 through interaction with an NBRE sequence in the PSPC1 gene promoter. These results coupled with in vivo studies showing that NR4A1 antagonists inhibit breast tumor growth and downregulate PSPC1 in tumors indicate that the pro-oncogenic nuclear PSPC1 factor can be targeted by CDIM/NR4A1 antagonists.


Breast Neoplasms/drug therapy , Methane/administration & dosage , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Promoter Regions, Genetic , RNA-Binding Proteins/genetics , A549 Cells , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , HCT116 Cells , Hep G2 Cells , Humans , Methane/pharmacology , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , PC-3 Cells , Promoter Regions, Genetic/drug effects , RNA-Binding Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Xenograft Model Antitumor Assays
4.
Mol Cell ; 81(19): 4041-4058.e15, 2021 10 07.
Article En | MEDLINE | ID: mdl-34624217

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.


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
5.
J Cell Mol Med ; 25(11): 5099-5112, 2021 06.
Article En | MEDLINE | ID: mdl-33942481

Nuclear receptor subfamily 4, group A, member 1 (NR4A1) can aggravate ischaemia-reperfusion (I/R) injury in the heart, kidney and brain. Thus, the present study aimed to unravel the role of NR4A1 on hepatic I/R injury. For this purpose, the mouse hepatic I/R model and H/R-exposed mouse hepatocytes model were established to stimulate the hepatic and hepatocellular damage. Then, the levels of ALT and AST as well as TNF-α and IL-1ß expression were measured in the mouse serum and supernatant of hepatocyte s, respectively. Thereafter, we quantified the levels of NR4A1, CYR61, NF-kB p65 and TGFß1 under pathological conditions, and their interactions were analysed using ChIP and dual-luciferase reporter gene assays. The in vivo and in vitro effects of NR4A1, CYR61, NF-kB p65 and TGFß1 on I/R-induced hepatic and H/R-induced hepatocellular damage were evaluated using gain- and loss-of-function approaches. NR4A1 was up-regulated in the hepatic tissues of I/R-operated mice and in H/R-treated hepatocytes. Silencing NR4A1 relieved the I/R-induced hepatic injury, as supported by suppression of ALT and AST as well as TNF-α and IL-1ß. Meanwhile, NR4A1 knockdown attenuated the H/R-induced hepatocellular damage by inhibiting the apoptosis of hepatocyte s. Moreover, we also found that NR4A1 up-regulated the expression of CYR61 which resulted in the activation of the NF-κB signalling pathway, thereby enhancing the transcription of TGFß1, which was validated to be the mechanism underlying the contributory role of NR4A1 in hepatic I/R injury. Taken together, NR4A1 silencing reduced the expression of CYR61/NF-κB/TGFß1, thereby relieving the hepatic I/R injury.


Cysteine-Rich Protein 61/antagonists & inhibitors , Inflammation/prevention & control , Liver Diseases/prevention & control , NF-kappa B/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Reperfusion Injury/complications , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Protective Agents , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
6.
Bioorg Chem ; 112: 104912, 2021 07.
Article En | MEDLINE | ID: mdl-33933804

Orphan nuclear receptor Nur77 is a unique member of the NR4A nuclear receptor subfamily, which is critical for cellular processes especially the inflammatory responses. Many efforts have been made to discover novel scaffold small molecules targeting Nur77. Herein, we evaluated the previously reported binding sites in crystal structures of Nur77 with small molecules, and then discovered compound 13 as a hit of Nur77 via virtual screening targeting the best-scored binding site. Based on the results of fluorescence titration assay, structure-activity relationship (SAR) analysis was summarized for compound 13 and its analogs. Among these analogs, compound 13e displayed the most potent binding affinity (0.54 ± 0.02 µM). The binding mode of compound 13e was predicted via molecule docking. Moreover, 13e exhibited significant anti-inflammation activity in TNF-α induced HepG2 cell model. Taken together, these results provided a new insight into the understanding the functions of specific binding sites on Nur77 for small molecular compounds, and the development of new scaffold Nur77 modulators.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Binding Sites/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship
7.
Molecules ; 26(8)2021 Apr 16.
Article En | MEDLINE | ID: mdl-33923503

The orphan nuclear receptor 4A1 (NR4A1) is overexpressed in pancreatic cancer and exhibits pro-oncogenic activity, and NR4A1 silencing and treatment with its inactivators has been shown to inhibit pancreatic cancer cells and tumor growth. In this study, we identified broussochalcone A (BCA) as a new NR4A1 inhibitor and demonstrated that BCA inhibits cell growth partly by inducing NR4A1-mediated apoptotic pathways in human pancreatic cancer cells. BCA downregulated specificity protein 1 (Sp1)-mediated expression of an anti-apoptotic protein, survivin, and activated the endoplasmic reticulum (ER) stress-mediated apoptotic pathway. These results suggest that NR4A1 inactivation contributes to the anticancer effects of BCA, and that BCA represents a potential anticancer agent targeting NR4A1 that is overexpressed in many types of human cancers.


Chalcones/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Resorcinols/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Reactive Oxygen Species/metabolism , Pancreatic Neoplasms
8.
Theranostics ; 11(7): 3376-3391, 2021.
Article En | MEDLINE | ID: mdl-33537093

Background: Colorectal cancer (CRC) and the associated metastatic lesions are reported to be hypoxic. Hypoxia is a common feature in the tumor microenvironment and a potent stimulant of CRC. We have identified a regulatory role of Nur77 on Akt activation to enhance ß-catenin signaling essential for CRC progression under hypoxic conditions. Methods: The functional role of Nur77 in hypoxia-induced EMT was examined by scattering assays to monitor the morphologies of CRC cell lines under 1% O2. Sphere formation assays were performed to investigate whether Nur77 induced cancer stem cell-like properties in hypoxic CRC cells. The expression of various epithelial-to-mesenchymal transition (EMT) and stemness markers was analyzed by qPCR and Western blotting. Finally, Nur77 function and signaling in vivo was ascertained in subcutaneous tumor xenograft or liver metastasis model in nude mice using CRC cells stably transfected with appropriate constructs. Results: Herein, we show, for the first time, that Nur77 is a novel regulator of microRNA biogenesis that may underlie its significant tumor-promoting activities in CRC cells under hypoxia. Mechanistically, Nur77 interacted with the tumor suppressor protein p63, leading to the inhibition of p63-dependent transcription of Dicer, an important miRNA processor and subsequent decrease in the biogenesis of let-7i-5p which targeted the 3'UTR of p110α mRNA and regulated its stability. Knockdown of Nur77 or overexpression of let-7i-5p inhibited the tumor metastasis in vivo. Conclusion: Our data uncovered a novel mechanistic link connecting Nur77, Akt, and invasive properties of CRC in the hypoxic microenvironment.


Adenocarcinoma/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/genetics , DEAD-box RNA Helicases/genetics , Hypoxia/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Proto-Oncogene Proteins c-akt/genetics , Ribonuclease III/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DEAD-box RNA Helicases/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/metabolism , Hypoxia/mortality , Hypoxia/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Mice , Mice, Nude , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribonuclease III/metabolism , Signal Transduction , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Burden , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
9.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article En | MEDLINE | ID: mdl-33562500

Fibrosis is a hallmark of adverse cardiac remodeling, which promotes heart failure, but it is also an essential repair mechanism to prevent cardiac rupture, signifying the importance of appropriate regulation of this process. In the remodeling heart, cardiac fibroblasts (CFs) differentiate into myofibroblasts (MyoFB), which are the key mediators of the fibrotic response. Additionally, cardiomyocytes are involved by providing pro-fibrotic cues. Nuclear receptor Nur77 is known to reduce cardiac hypertrophy and associated fibrosis; however, the exact function of Nur77 in the fibrotic response is yet unknown. Here, we show that Nur77-deficient mice exhibit severe myocardial wall thinning, rupture and reduced collagen fiber density after myocardial infarction and chronic isoproterenol (ISO) infusion. Upon Nur77 knockdown in cultured rat CFs, expression of MyoFB markers and extracellular matrix proteins is reduced after stimulation with ISO or transforming growth factor-ß (TGF-ß). Accordingly, Nur77-depleted CFs produce less collagen and exhibit diminished proliferation and wound closure capacity. Interestingly, Nur77 knockdown in neonatal rat cardiomyocytes results in increased paracrine induction of MyoFB differentiation, which was blocked by TGF-ß receptor antagonism. Taken together, Nur77-mediated regulation involves CF-intrinsic promotion of CF-to-MyoFB transition and inhibition of cardiomyocyte-driven paracrine TGF-ß-mediated MyoFB differentiation. As such, Nur77 provides distinct, cell-specific regulation of cardiac fibrosis.


Cardiomyopathies/metabolism , Myocytes, Cardiac/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Gene Knockdown Techniques , Heart Rupture/genetics , Heart Rupture/metabolism , Heart Rupture/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Models, Cardiovascular , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Rats , Transforming Growth Factor beta/metabolism , Ventricular Remodeling/genetics , Ventricular Remodeling/physiology
10.
Acta Biochim Pol ; 67(4): 521-529, 2020 Dec 17.
Article En | MEDLINE | ID: mdl-33332077

This study aims to investigate the protection of dexmedetomidine (Dex) against pulmonary ischemia-reperfusion injury (PIRI) in the mouse model and reveal the mechanism in hypoxia reoxygenation (H/R)-induced mouse pulmonary vascular endothelial cells (MPVECs). The lung wet-to-dry weight ratio, histopathological features, and malondialdehyde (MDA) concentrations were measured. The H/R-induced MPVECs were exposed to Dex, and the cell viability, cell apoptosis and protein expressions were assessed by the Cell Counting Kit-8 (CCK8) assay, flow cytometry and western blot, respectively. In addition, the regulatory relationship between miR-21-5p and orphan nuclear receptor 4A1 (Nr4a1) was revealed by several assays, including the dual-luciferase reporter assay, real-time quantitative polymerase chain reaction (RT-qPCR) and western blot. We found that the Dex treatment significantly alleviated pulmonary injury and decreased the level of MDA and wet/dry weight ratio in PIRI mice. Dex treatment also increased cell viability, reduced apoptotic ratio and downregulated expression levels of Cleaved Caspase-3 and Cleaved Caspase-9 in H/R induced MPVECs. Furthermore, the expression of miR-21-5p was upregulated, while Nr4a1 was downregulated by Dex in a concentration-dependent manner in H/R induced MPVECs. Moreover, Nr4a1 was verified as a target of miR-497-5p. Overexpression of Nr4a1 could reverse the protective effects of Dex on alleviating H/R-induced injury in MPVECs. Taken together, Dex treatment attenuated ischemia-reperfusion induced pulmonary injury through modulating the miR-21-5p/Nr4a1 signaling pathway.


Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , Endothelial Cells/drug effects , MicroRNAs/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Reperfusion Injury/drug therapy , Animals , Apoptosis/drug effects , Base Pairing , Base Sequence , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Survival/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation , HEK293 Cells , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Malondialdehyde/antagonists & inhibitors , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction
11.
Endocrinology ; 161(4)2020 04 01.
Article En | MEDLINE | ID: mdl-32099996

Endometriosis is an inflammatory disease that primarily affects women during their reproductive years, and since current hormonal therapies are of concern, new hormone-independent treatment regimens are needed. The orphan nuclear receptor 4A1 (NR4A1, Nur77) is expressed in patient-derived (stromal) endometriotic cells and also epithelial cell lines, and we observed that knockdown of NR4A1 in patient-derived ectopic endometrium-isolated ovarian endometrioma (ESECT)-7 and ESECT-40 cells decreased cell proliferation and induced apoptosis. Moreover, the treatment of these cells with bis-indole derived NR4A1 ligands 1,1-bis(3'-indolyl)-1-(p-hydroxyphenyl)methane (DIM-C-pPhOH) and its buttressed 3-chloro-5-methoxy analog (DIM-C-pPhOH-3-Cl-5-OCH3) inhibited cell growth and induced apoptosis and related genes. The compounds exhibit NR4A1 antagonist activities in both functional and transactivation assays whereas these effects were not observed in normal endometrial cells. We also observed that NR4A1 knockdown and treatment with NR4A1 antagonists decreased fibrosis, α-smooth muscle actin, and related pro-fibrotic genes in ESECT-7 and ESECT-40 cells, and similar results were observed in epithelial-derived endometriotic cell lines. Moreover, in an endometriosis mouse model with auto-transplantation and also in severe combined immune deficiency mice transplanted with human endometriotic cells treatment with 25 mg/kg/day DIM-C-pPhOH-3-Cl-5-OCH3 significantly inhibited growth and expansion of endometriotic lesions. Thus, bis-indole-derived NR4A1 ligands represent a novel class of drugs as nonhormonal therapy for endometriosis.


Apoptosis/drug effects , Cell Proliferation/drug effects , Endometriosis/drug therapy , Endometrium/drug effects , Indoles/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Phenols/pharmacology , Animals , Disease Models, Animal , Endometriosis/metabolism , Endometrium/metabolism , Female , Gene Knockdown Techniques , Indoles/therapeutic use , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phenols/therapeutic use
12.
Apoptosis ; 25(5-6): 321-340, 2020 06.
Article En | MEDLINE | ID: mdl-31993850

Arterial media calcification is related to mitochondrial dysfunction. Protective mitophagy delays the progression of vascular calcification. We previously reported that lactate accelerates osteoblastic phenotype transition of VSMC through BNIP3-mediated mitophagy suppression. In this study, we investigated the specific links between lactate, mitochondrial homeostasis, and vascular calcification. Ex vivo, alizarin S red and von Kossa staining in addition to measurement of calcium content, RUNX2, and BMP-2 protein levels revealed that lactate accelerated arterial media calcification. We demonstrated that lactate induced mitochondrial fission and apoptosis in aortas, whereas mitophagy was suppressed. In VSMCs, lactate increased NR4A1 expression, leading to activation of DNA-PKcs and p53. Lactate induced Drp1 migration to the mitochondria and enhanced mitochondrial fission through NR4A1. Western blot analysis of LC3-II and p62 and mRFP-GFP-LC3 adenovirus detection showed that NR4A1 knockdown was involved in enhanced autophagy flux. Furthermore, NR4A1 inhibited BNIP3-related mitophagy, which was confirmed by TOMM20 and BNIP3 protein levels, and LC3-II co-localization with TOMM20. The excessive fission and deficient mitophagy damaged mitochondrial structure and impaired respiratory function, determined by mPTP opening rate, mitochondrial membrane potential, mitochondrial morphology under TEM, ATP production, and OCR, which was reversed by NR4A1 silencing. Mechanistically, lactate enhanced fission but halted mitophagy via activation of the NR4A1/DNA-PKcs/p53 pathway, evoking apoptosis, finally accelerating osteoblastic phenotype transition of VSMC and calcium deposition. This study suggests that the NR4A1/DNA-PKcs/p53 pathway is involved in the mechanism by which lactate accelerates vascular calcification, partly through excessive Drp-mediated mitochondrial fission and BNIP3-related mitophagy deficiency.


Diabetes Mellitus, Experimental/genetics , Lactic Acid/pharmacology , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Vascular Calcification/genetics , Animals , Aorta/drug effects , Aorta/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cholecalciferol/pharmacology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat/adverse effects , Dynamins/genetics , Dynamins/metabolism , Gene Expression Regulation , Male , Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proteins/metabolism , Mitophagy/drug effects , Mitophagy/genetics , Nicotine/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Organ Culture Techniques , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Streptozocin/administration & dosage , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Calcification/metabolism , Vascular Calcification/pathology
13.
Cancer Res ; 80(5): 1011-1023, 2020 03 01.
Article En | MEDLINE | ID: mdl-31911554

PD-L1 is expressed in tumor cells and its interaction with PD-1 plays an important role in evading immune surveillance; this can be overcome using PD-L1 or PD-1 immunotherapy antibodies. This study reports a novel approach for targeting PD-L1. In human breast cancer cell lines and 4T1 mouse mammary tumor cells, PD-L1 expression was regulated by the nuclear receptor NR4A1/Sp1 complex bound to the proximal germinal center (GC)-rich region of the PD-L1 gene promoter. Treatment of breast cancer cells with bis-indole-derived NR4A1 antagonists including 1,1-bis(3'-indolyl)-1-(3-chloro-4-hydroxy-5-methoxyphenyl)methane (Cl-OCH3) decreased expression of PD-L1 mRNA, promoter-dependent luciferase activity, and protein. In in vivo studies using a syngeneic mouse model bearing orthotopically injected 4T1 cells, Cl-OCH3 decreased tumor growth and weight and inhibited lung metastasis. Cl-OCH3 also decreased expression of CD3+/CD4+/CD25+/FoxP3+ regulatory T cells and increased the Teff/Treg ratio. Therefore, the potent anticancer activities of NR4A1 antagonists are also accompanied by enhanced antitumor immunity in PD-L1-expressing triple-negative breast cancer and thus represent a novel class of drugs that mimic immunotherapy. SIGNIFICANCE: These findings show that the orphan nuclear receptor NR4A1 controls PD-L1 expression and identify a chemical probe capable of disrupting this regulatory axis.


Antineoplastic Agents/pharmacology , B7-H1 Antigen/metabolism , Lung Neoplasms/prevention & control , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy/methods , Indoles/pharmacology , Indoles/therapeutic use , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Proteolysis/drug effects , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
14.
J Immunol ; 203(8): 2163-2170, 2019 10 15.
Article En | MEDLINE | ID: mdl-31527196

Glucocorticoid (GC) signaling in thymocytes shapes the TCR repertoire by antagonizing thymocyte negative selection. The transcription factors Nur77 and Helios, which are upregulated in TCR-signaled thymocytes, have been implicated in negative selection. In this study, we found that GCs inhibited Helios and, to a lesser extent, Nur77 upregulation in TCR-stimulated mouse thymocytes. Inhibition was increased by GC preincubation, and reductions in mRNA were prevented by a protein synthesis inhibitor, suggesting that GCs suppress indirectly via an intermediary factor. Upregulation of Helios in TCR-stimulated thymocytes was unaffected by deletion of Nur77, indicating Nur77 and Helios are regulated independently. Whereas CD4+ thymocytes are positively selected in wild-type AND TCR-transgenic B6 mice, loss of GC receptor expression resulted in increased negative selection. Correspondingly, Helios and Nur77 levels were elevated in TCRhiCD4+CD8+ (TCR-signaled) thymocytes. Notably, deletion of Helios fully reversed this negative selection, whereas deletion of Nur77 had no effect on CD4+CD8+ cell numbers but reversed the loss of mature CD4+ thymocytes. Thus, Nur77 and Helios are GC targets that play nonredundant roles in setting the signaling threshold for thymocyte negative selection.


DNA-Binding Proteins/antagonists & inhibitors , Glucocorticoids/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Thymocytes/drug effects , Transcription Factors/antagonists & inhibitors , Animals , DNA-Binding Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Thymocytes/metabolism , Transcription Factors/metabolism
15.
Mol Cancer Res ; 17(11): 2221-2232, 2019 11.
Article En | MEDLINE | ID: mdl-31462501

Nuclear receptor 4A1 (NR4A1, Nur77) is overexpressed in rhabdomyosarcoma (RMS), and inactivation of NR4A1 (siNR4A1) or treatment with the NR4A1 antagonist 1,1-bis(3'-indoly)-1-(p-hydroxy-phenyl)methane (DIM-C-pPhOH) has antiproliferative and proapoptotic effects on RMS cells. However, the mechanism by which NR4A1 inhibition exerts these effects is poorly defined. Here, we report that NR4A1 silencing or inhibition resulted in accumulation of reactive oxygen species (ROS) and ROS-dependent induction of the tumor suppressor-like cytokine IL24 in RMS cells. Mechanistically, NR4A1 was found to regulate the expression of the proreductant genes thioredoxin domain-containing 5 (TXNDC5) and isocitrate dehydrogenase 1 (IDH1), which are downregulated in RMS cells following NR4A1 knockdown or inhibition. Silencing TXNDC5 and IDH1 also induced ROS accumulation and IL24 expression in RMS cells, suggesting that NR4A1 antagonists mediate their antiproliferative and apoptotic effects through modulation of proreductant gene expression. Finally, cotreatment with the antioxidant glutathione or IL24-blocking antibody reversed the effects of NR4A1 inhibition, demonstrating the importance of both ROS and IL24 in mediating the cellular responses. IMPLICATIONS: Overall, these data elucidate the mechanism by which NR4A1 inhibition functions to inhibit the proliferation, survival, and migration of RMS cells.


Gene Expression Regulation, Neoplastic , Interleukins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Reactive Oxygen Species/metabolism , Rhabdomyosarcoma/therapy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Down-Regulation , Glutathione/therapeutic use , Humans , Indoles/therapeutic use , Interleukins/antagonists & inhibitors , Interleukins/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phenols/therapeutic use , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology
16.
Exp Mol Pathol ; 111: 104303, 2019 12.
Article En | MEDLINE | ID: mdl-31465766

Renal ischemia-reperfusion injury (IRI), a major cause of acute kidney injury as well as a contributor to a rapid kidney dysfunction and high mortality rates, is a complex yet not fully understood process. Investigation on the underlying molecular mechanism including the inflammation initiation and progression can help to have a better understanding of the disease, and thereby lead to a potential therapeutic approach. We established renal IRI mouse model groups differing in their ages. These renal IRI mice were treated either only with si-nuclear receptor subfamily 4, group A, member 1 (NR4A1) or together with si-ß-catenin by tail vein injection to analyze the role of NR4A1 and ß-catenin in the development of renal IRI. Serum creatinine (SCr) and blood urea nitrogen (BUN) levels were examined for renal function analysis. Levels of the apoptosis markers B-cell lymphoma-2 (Bcl-2), Bcl-2 associated protein X (Bax), and cleaved caspase-3 were determined. NR4A1 gene was up-regulated in the renal tissues of all mice with IRI, which showed a much higher level in the old mice with IRI. si-NR4A1 treatment resulted in reduced SCr and BUN levels and a decrease of cell apoptosis, indicated by lower expression of Bax and cleaved Caspase-3, while in contrast increased levels of Bcl-2 were detected. Interestingly, also the ß-catenin level was increased by knockdown of NR4A1. Furthermore, si-ß-catenin reversed the effect of knockdown of NR4A1, leading to aggravated renal function damage, severe pathological injury and increased apoptosis. Thus, silencing NR4A1 ameliorates renal IRI via ß-catenin signaling pathway activation. Down-regulated NR4A1 confirms renoprotective properties against renal IRI via the activation of ß-catenin signaling pathway in old mice.


Apoptosis , Kidney Diseases/prevention & control , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Protective Agents/metabolism , Reperfusion Injury/prevention & control , beta Catenin/metabolism , Animals , Disease Models, Animal , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , beta Catenin/genetics
17.
Gynecol Oncol ; 154(1): 218-227, 2019 07.
Article En | MEDLINE | ID: mdl-31053403

OBJECTIVES: NR4A1 is overexpressed in many solid tumors, and the objectives of this study were to investigate the expression and functional role of this receptor in endometrial cancer cells and demonstrate that NR4A1 antagonist inhibit mTOR. METHODS: Ishikawa and Hec-1B endometrial cells were used as models to investigate the parallel effects of NR4A1 knockdown by RNA interference (siNR4A1) and treatment with bis-indole-derived NR4A1 ligands (antagonists) on cell growth and survival by determining cell numbers and effects on Annexin V staining. Western blot analysis of whole cell lysates was used to determine effects of these treatments on expression of growth promoting, survival and apoptotic genes and mTOR signaling. Effects of NR4A1 antagonists on tumor growth were determined in athymic nude mice bearing Hec-1B cells as xenografts. RESULTS: siNR4A1 or treatment with bis-indole-derived NR4A1 antagonists inhibited growth of endometrial cancer cells in vitro and endometrial tumors in vivo and this was accompanied by decreased expression of growth promoting and survival genes and mTOR inhibition. CONCLUSIONS: NR4A1 exhibited pro-oncogenic activity in endometrial cells due, in part, to regulation of cell growth, survival and mTOR signaling, and all of these pathways and their associated gene products were inhibited after treatment with bis-indole-derived NR4A1 antagonists. Moreover, these compounds also blocked endometrial tumor growth in vivo demonstrating that NR4A1 is a potential novel drug target for treatment of endometrial cancer.


Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Indoles/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Aged , Animals , Apoptosis/drug effects , Cell Line, Tumor , Endometrial Neoplasms/pathology , Female , Gene Knockdown Techniques , Humans , Ligands , Mice , Mice, Nude , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Random Allocation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
18.
Breast Cancer Res Treat ; 177(1): 29-40, 2019 Aug.
Article En | MEDLINE | ID: mdl-31119568

BACKGROUND: Nuclear receptor 4A1 (NR4A1) is overexpressed in mammary tumors, and the methylene-substituted bis-indole derivative 1,1-bis(3'-indolyl)-1-(p-hydroxyphenyl)methane (DIM-C-pPhOH) acts as an NR4A1 antagonist (inverse agonist) and inhibits NR4A1-regulated pro-oncogenic pathways/genes in breast and other cancer cells. METHODS: Buttressed analogs of DIM-C-pPhOH were synthesized by condensation of the substituted p-hydroxybenzaldehydes with indole. Breast cancer cell growth, survival, and migration assays were carried out by cell counting, Annexin V staining, and Boyden chamber assays, respectively. Changes in RNA and protein expression were determined by RT-PCR and western blots, respectively. Analysis of RNAseq results was carried out using Ingenuity Pathway Analysis, and in vivo potencies of NR4A1 antagonists were determined in athymic nude mice bearing MDA-MB-231 cells in an orthotopic model. RESULTS: Ingenuity Pathway analysis of common genes modulated by NR4A1 knockdown or treatment with DIM-C-pPhOH showed that changes in gene expression were consistent with the observed decreased functional responses, namely inhibition of growth and migration and increased apoptosis. DIM-C-pPhOH is rapidly metabolized and the effects and potencies of buttressed analogs of DIM-C-pPhOH which contain one or two substituents ortho to the hydroxyl groups were investigated using NR4A1-regulated gene/gene products as endpoints. The buttressed analogs were more potent than DIM-C-pPhOH in both in vitro assays and as inhibitors of mammary tumor growth. Moreover, using 1,1-bis(3'-indolyl)-1-(3-chloro-4-hydroxy-5-methoxyphenyl)methane (DIM-C-pPhOh-3-Cl-5-OCH3) significant tumor growth inhibition was observed at doses as low as 2 mg/kg/d which was at least an order of magnitude more potent than DIM-C-pPhOH. CONCLUSIONS: These buttressed analogs represent a more potent set of second generation NR4A1 antagonists as inhibitors of breast cancer.


Antineoplastic Agents/pharmacology , Indoles/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Phenols/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Indoles/chemistry , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phenols/chemistry , Xenograft Model Antitumor Assays
19.
Eur J Med Chem ; 177: 171-187, 2019 Sep 01.
Article En | MEDLINE | ID: mdl-31132532

Nur77, an orphan member of the nuclear receptor superfamily, plays an important role in the regulation of inflammatory processes. Our previous work found that celastrol, a pentacyclic triterpene, bound to Nur77 to inhibit inflammation in a Nur77-dependent manner. Celastrol binding to Nur77 promotes Nur77 translocation from nucleus to cytoplasm, resulting in clearance of inflamed mitochondria and then alleviation of inflammation. Here, we report the design, synthesis, SAR study and biological evaluation of a series of celastrol analogs. A total of 24 celastrol derivatives were made. Compound 3a with a Kd of 0.87 µM was found to be less toxic than celastrol and could be a hit molecule for further optimization.


Anti-Inflammatory Agents/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Triterpenes/pharmacology , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/toxicity , Binding Sites , Drug Design , Hep G2 Cells , Humans , Molecular Docking Simulation , Molecular Structure , Nuclear Receptor Subfamily 4, Group A, Member 1/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Pentacyclic Triterpenes , Protein Binding/drug effects , Sequestosome-1 Protein/metabolism , Structure-Activity Relationship , TNF Receptor-Associated Factor 2/metabolism , Triterpenes/chemical synthesis , Triterpenes/metabolism , Triterpenes/toxicity , Zebrafish
20.
Thorac Cancer ; 10(4): 715-727, 2019 04.
Article En | MEDLINE | ID: mdl-30806032

BACKGROUND: Although cigarette smoking is considered one of the key risk factors for lung cancer, 15% of male patients and 53% of female patients with lung cancer are non-smokers. Metabolic changes are critical features of cancer. Therapeutic target identification from a metabolic perspective in non-small cell lung cancer (NSCLC) tissue of female non-smokers has long been ignored. RESULTS: Based on microarray data retrieved from Affymetrix expression arrays E-GEOD-19804, we found that the downregulated genes in non-smoking female NSCLC patients tended to participate in protein/amino acid and lipid metabolism, while upregulated genes were more involved in protein/amino acid and carbohydrate metabolism. Combining nutrient metabolic co-expression, protein-protein interaction network construction and overall survival assessment, we identified NR4A1 and TIE1 as potential therapeutic targets for NSCLC in female non-smokers. To accelerate the drug development for non-smoking female NSCLC patients, we identified nilotinib as a potential agonist targeting NR4A1 encoded protein by molecular docking and molecular dynamic stimulation. We also show that nilotinib inhibited proliferation and induced senescence of cells in non-smoking female NSCLC patients in vitro. CONCLUSIONS: These results not only uncover nutrient metabolic characteristics in non-smoking female NSCLC patients, but also provide a new paradigm for identifying new targets and drugs for novel therapy for such patients.


Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Pyrimidines/pharmacology , Receptor, TIE-1/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Non-Smokers/statistics & numerical data , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/chemistry , Protein Interaction Maps , Pyrimidines/therapeutic use , Receptor, TIE-1/metabolism , Survival Analysis
...