Abstract Former studies indicate that nuclear receptor subfamily 4 group A member 2 (Nurr1, NR4A2), a transcription factor, is regarded as a potential therapeutic target for central nervous system diseases, and many studies have focused on the development and optimization of agonists of Nurr1. Recent studies have shown that Nurr1 is upregulated in many other diseases. However, there is still a lack of effective inverse Nurr1 agonists as a therapeutic strategy or as pharmacological tools to counteract the receptor’s inherent activity. In this study, we screened Nurr1 ligands through a high-throughput screening system and identified a novel Nurr1 inverse agonist (K-strophanthoside). We further validated the binding site of K-strophanthoside on Nurr1 and investigated its effect on regulating Nurr1 function. K-strophanthoside directly binds to the ligand-binding domain of Nurr1 (Glu445, Glu514, Arg515, and His516) and mimics the function of Nurr1 knockdown by suppressing the intrinsic Nurr1 transcriptional activity. Our study contributes a valuable chemical tool for Nurr1 modulators and provides a potential treatment target for Nurr1-related disorders. __________________________________________________________________ graphic file with name ao5c06698_0008.jpg __________________________________________________________________ graphic file with name ao5c06698_0006.jpg Introduction Nuclear receptor subfamily 4 group A member 2 (Nurr1, also known as NR4a2) is a transcription factor that plays a significant role in various diseases, and its dysregulation is linked to neurodegenerative diseases, − autoimmune diseases, , and cancers. − Our previous study documented a synthetic Nurr1 activator that has a strong antiparkinsonian effect in animal models of PD. Downregulation of Nurr1 aggravates the pathology of AD involving neurodegeneration, Aβ accumulation, and neuroinflammation. Moreover, Nurr1 is increased in the peripheral blood of ALS patients and in the spinal cord of superoxide dismutase 1 (SOD1)-G93A transgenic mice in the early phase, which is regarded as a protective anti-inflammatory mechanism. However, in some diseases, increased Nurr1 is regarded as a pathogenic factor. ,, Previous studies have found abnormal expression of Nurr1 in multiple sclerosis (MS). ,, However, the relation between Nurr1 and MS is conflicting. Nurr1 was considered as a protective factor in MS; , however, recent studies have further found that continuous upregulation of Nurr1 due to chronic inflammation in MS leads to neuronal death, indicating the critical importance of maintaining balanced Nurr1 expression for the onset and progression of the disease. Furthermore, Nurr1 expression is increased in tumors, , associated with tumor growth and resistance. ,,,, Nurr1 mediated the ERK and AKT signaling pathways to facilitate tumor development in cervical cancer, and their inhibitors could halt tumor progression. In glioblastoma, the expression of Nurr1 is significantly upregulated in microglia, and both genetic and pharmacological inhibition of Nurr1 enhance the therapeutic efficacy against the tumor while suppressing its growth. , Therefore, Nurr1 represents a potential therapeutic target for a range of diseases. Nurr1 belongs to the nuclear receptor family of nonligand-dependent transcription factors. Its protein structure includes a nonligand-dependent transcription activation region 1 (AF1), a highly conserved zinc finger DNA binding domain (DBD), and a C-terminal ligand-binding domain (LBD) containing AF2. Previous study showed that unlike other nuclear receptors, the Nurr1-LBD lacks a conventional ligand-binding cavity and binding sites for coregulatory factors, leading to a lack of identifiable Nurr1 ligands. However, a series of small-molecule compounds were identified to modulate Nurr1 transcriptional activities by physically binding to Nurr1-LBD, including synthetic (amodiaquine, AQ; chloroquine, CQ) and natural small-molecule compounds (docosahexaenoic acid, DHA; 5,6-dihydroxyindole, DHI; prostaglandin E1, PGE1; and prostaglandin A1, PGA1). In addition to the above agonists, a number of inverse agonists have also been identified as chemical tools, including oxaprozin, the indole-based derivative, fatty acid mimetics, and nonsteroidal anti-inflammatory drugs. All of these results indicate the presence of Nurr1 ligands that bind to Nurr1-LBD and regulate its function. The important role of Nurr1 in related diseases indicates that the discovery of Nurr1 modulators remains an urgent necessity. However, most current research has focused on the development and validation of Nurr1 agonists, and there is still a lack of Nurr1 inverse agonists as both a potential therapeutic strategy for diseases and a valuable tool in elucidating Nurr1’s role in disease pathology. In this study, we screened a new inverse Nurr1 agonist (K-strophanthoside) from the Chemdiv compound library by structure-based virtual screening. Intriguingly, K-strophanthoside appears to modulate Nurr1 monomer activity more effectively than the Nurr1 homodimer and heterodimer, and it mimics the effects of Nurr1 silencing on various biological pathways. These findings warrant further exploration of the biological functions and potential applications of Nurr1 modulators. Results Compound 7 as a Potential Inverse Agonist for Nurr1 The ChemDiv compound library, containing about 1.5 million compounds, was selected as the small-molecule library for virtual screening. The three-dimensional structure of Nurr1-LBD in the Protein Data Bank (PDB code: [37]6DDA) was used to dock with compounds in the ChemDiv compound library. The absolute values of docking scores were ranked from high to low, and the top 10 compounds are shown in [38]Figure S1. To explore the transcriptional activity of Nurr1 regulated by the top 10 compounds, we preliminarily evaluated their toxicity using CCK-8, and the results showed that compounds 1 and 2 exhibited cytotoxicity, while others did not show cytotoxicity even at a higher concentration of 100 μM ([39]Figure S2). Then, we constructed HEK293T cells for the Nurr1-mediated transcriptional activity detection system by transfecting a luciferase plasmid containing the Nurr1 binding site ([40]Figure S3). The cells cultured with 8 other compounds (0, 6.25, 12.5, 25, 50, and 100 μM) were subjected to a luciferase reporter gene assay to determine the transcriptional activity of Nurr1. As shown in [41]Table , compounds 7 and 9 significantly inhibited the Nurr1 transcriptional activity by up to 51% (P < 0.01) and 18% (P < 0.05), compounds 3 and 10 increased the activities by 1.38 and 1.36 folds compared with the control group at a concentration of 100 μM (P < 0.01), respectively, and other compounds did not show significant changes in the Nurr1 transcriptional activity regulation ([42]Table ). According to the change in folds of Nurr1 transcriptional activities, compound 7 was selected as the primary target compound for further verification. 1. Summarized Transcriptional Regulation of Nurr1 by Compounds^ . concentration(μM) __________________________________________________________________ compound 0 6.25 12.5 25 50 100 3 1.00 ± 0.042 1.04 ± 0.074 0.91 ± 0.066 1.06 ± 0.046 1.10 ± 0.027 1.38 ± 0.039** 4 1.00 ± 0.043 0.97 ± 0.067 1.16 ± 0.065 1.23 ± 0.073 1.29 ± 0.088* 1.23 ± 0.089 5 1.00 ± 0.073 0.92 ± 0.036 0.87 ± 0.046 0.91 ± 0.010 0.88 ± 0.026 0.90 ± 0.011 6 1.00 ± 0.042 0.90 ± 0.019 1.06 ± 0.021 0.99 ± 0.023 1.10 ± 0.14 1.01 ± 0.066 7 1.00 ± 0.035 1.13 ± 0.065 0.89 ± 0.086 0.74 ± 0.13 0.76 ± 0.024 0.49 ± 0.017** 8 1.00 ± 0.017 0.93 ± 0.037 0.86 ± 0.029 1.19 ± 0.049 1.07 ± 0.086 1.10 ± 0.076 9 1.00 ± 0.048 0.97 ± 0.011 0.95 ± 0.07 0.88 ± 0.035 0.85 ± 0.045 0.82 ± 0.024* 10 1.00 ± 0.036 0.97 ± 0.040 1.15 ± 0.040 1.06 ± 0.070 1.23 ± 0.051* 1.36 ± 0.036** [43]Open in a new tab ^a The transcriptional activity of Nurr1 regulated by compounds 3–10 in HEK293T cells was determined in a cellular reporter gene assay. *P < 0.05, **P < 0.01. Data are mean ± S.E.M., n ≥ 3. Direct Binding of K-Strophanthoside to Nurr1-LBD The name of compound 7 was K-strophanthoside, which was found by blasting the structure of compound 7 in the SciFinder database. To further verify the interaction between K-strophanthoside and the Nurr1-LBD, the computer molecular docking predicted that K-strophanthoside exhibited suitable spatial complementarity with the Nurr1-LBD by forming hydrogen bonds with Glu445, Leu509, Glu514, Arg515, and His516 ([44]Figure A–C). However, there was no structural complementarity between K-strophanthoside and other members of the NR4A family with known 3D structures ([45]Figure S4A,B). The results suggested that K-strophanthoside was potentially bound to Nurr1-LBD. Then, a surface plasmon resonance (SPR) experiment was performed to verify the direct binding between K-strophanthoside and Nurr1, with AQ as a positive control. The results showed that K-strophanthoside bound to Nurr1 in a concentration-dependent manner, similar to AQ ([46]Figure ). The SPR fitting results showed that the association rate constant (k [a]) of the complexes between K-strophanthoside and the Nurr1 protein was 3.352 × 10^2 M^–1 s^–1, and the dissociation rate constant (k [d]) was 7.744 × 10^–3 s^–1. In particular, the dissociation constant (K [D]) of the complex for K-strophanthoside with the Nurr1 protein was 2.310 × 10^–5 M, showing stronger affinity than AQ (K [D] = 3.695 × 10^–5 M, [47]Table ). Furthermore, we constructed site-directed mutagenesis of the Nurr1 plasmids, and the mutations did not affect Nurr1 activity compared to the wild-type Nurr1 ([48]Figure S4C). Mutations of Glu445, Glu514, Arg515, and His516 significantly improved the transcriptional activity of Nurr1 induced by K-strophanthoside, while mutation of Leu509 did not show significant changes ([49]Figure D). Therefore, the amino acid residues (Glu445, Glu514, Arg515, His516) of Nurr1 that formed hydrogen bonds with K-strophanthoside are essential for K-strophanthoside-induced Nurr1 transcriptional activity. However, these mutations did not affect the AQ-induced Nurr1 activity ([50]Figure S10). Collectively, these findings suggest that K-strophanthoside is a ligand for Nurr1. 1. [51]1 [52]Open in a new tab Binding model of K-strophanthoside to the Nurr1 protein. (A) Two-dimensional binding pattern of K-strophanthoside to the Nurr1 protein (PDB code: 6DDA). (B) Three-dimensional binding mode of K-strophanthoside to Nurr1-LBD. K-strophanthoside is shown as a yellow rod. The surrounding residues in the binding bag are green. Hydrogen bonds are represented by the dotted red line. (C) Electrostatic surface rendering of K-strophanthoside with Nurr-LBD. Blue and red surfaces indicate positively and negatively charged surfaces, respectively. K-strophanthoside is shown as a yellow rod. (D) Effects of Nurr1 mutations in the potential K-strophanthoside binding sites on the transcriptional activity of Nurr1. The transcriptional activity of wild-type (WT) and mutant constructs (E445A, L509A, E514A, R515A, and H516A) was examined by luciferase reporter assay with or without K-strophanthoside (100 μM). *P < 0.05, **P < 0.01. Data are mean ± S.E.M., n ≥ 3, one-way ANOVA in comparison expression with control (WT-Nurr1). 2. [53]2 [54]Open in a new tab Binding between Nurr1 and small-molecule compounds was determined by SPR. The Nurr1 protein was coupled to a CM5 chip and exposed to samples at various doses. (A) Concentration-dependent binding of AQ to Nurr1. AQ was injected at increasing doses (1.953, 3.906, 7.813, 15.63, 31.25, 62.5, and 125 μM). (B) Steady-state affinity fitting diagram of AQ binding with Nurr1. (C) Concentration-dependent binding of K-strophanthoside to Nurr1. K-strophanthoside was injected at increasing doses (3.906, 7.813, 15.63, 31.25, 62.5, 125, and 250 μM). The black curves represent the fitting lines. 2. Dissociation Constant (K [D]) Values for the Binding of AQ and K-strophanthoside with the Nurr1 Protein. protein compounds K [D](M) R [max] (RU) Nurr1 protein amodiaquine (AQ) 3.695 × 10^–5 4.441 K-strophanthoside 2.310 × 10^–5 10 [55]Open in a new tab Regulation of Nurr1-Associated Gene Expression by K-Strophanthoside Nurr1 is an important transcriptional factor of genes (tyrosine hydroxylase, TH; L-aromatic amino acid decarboxylase, AADC, etc.) involved in the process of DA synthesis. To explore the regulation of K-strophanthoside on Nurr1 function, we cultured Neuro-2a cells with different concentrations of K-strophanthoside and screened for concentrations without cytotoxicity ([56]Figure S5). The results of Western blot showed that K-strophanthoside did not change the expression of Nurr1 ([57]Figure A,D), while the expression levels of TH and AADC were significantly downregulated to 47% and 35% by K-strophanthoside at a concentration of 30 μM (P < 0.05, [58]Figure A–C). Real-time PCR analysis showed that K-strophanthoside reduced the expression of TH and AADC to 75% and 62% compared with the control groups, respectively (P < 0.05, [59]Figure E). Notably, the expression levels of Nurr77, Nor1, and Nurr1 remained unchanged under the same conditions ([60]Figure S9). Immunofluorescence results showed that the levels of TH and AADC were decreased by K-strophanthoside with a concentration dependence ([61]Figure F,G). Moreover, we constructed Neuro-2a cells with Nurr1 downregulation ([62]Figure S6). Then, transcriptome analysis was conducted on Nurr1 knockdown cells and Neuro-2a cells treated with 30 μM K-strophanthoside by RNA sequencing to verify the role of K-strophanthoside in Nurr1-associated gene expression. A total of 1785 differential expression genes (DEGs), including 950 downregulated and 835 upregulated genes (fold change ≥ 1.5), were identified in Nurr1 knockdown cells ([63]Figure A), while 535 downregulated and 403 upregulated genes were detected in cells treated with K-strophanthoside ([64]Figure B). Analysis showed that there were 93 genes with common expression changes between cells treated with K-strophanthoside and Nurr1 knockdown cells, including 40 upregulated and 53 downregulated genes ([65]Figure C). Using the bioinformatics approach, we found that 73 of these genes contain NBRE-like binding sites within their promoter regions. These results are detailed in [66]Supplementary Table 2, supporting the notion that a substantial portion of the DEGs may be directly regulated by Nurr1 through its canonical response elements. The clustered heatmap of commonly changed DEGs in Nurr1 knockdown cells and Neuro-2a cells treated with K-strophanthoside is shown in [67]Figure S7. 3. [68]3 [69]Open in a new tab Biological effects of K-strophanthoside in Neuro-2a cells. (A–D) Impact of K-strophanthoside on Nurr1 and Nurr1-associated gene (TH and AADC) protein expression in Neuro-2a cells for 24 h. (E) Effect of K-strophanthoside on Nurr1-associated gene mRNA expression of TH and AADC at a concentration of 30 μM. (F, G) Fluorescence intensity of TH and AADC was detected by immunofluorescence after administration of K-strophanthoside. *P < 0.05, **P < 0.01. Data are mean ± S.E.M., n ≥ 3, one-way ANOVA in comparison expression with 0 μM compound (DMSO only) in Western blot analysis, t-test in comparison expression with 0 μM compound (DMSO only) in real-time PCR analysis. 4. [70]4 [71]Open in a new tab Integrated identification and enrichment analysis of DEGs obtained from Nurr1 knockdown cells and K-strophanthoside-treated cells at 30 μM. (A, B) Volcano plots for DEGs from Nurr1 knockdown cells (A) and K-strophanthoside-treated cells (B). Red and blue dots represent upregulated genes (q-value < 0.05 and log2FC ≥ 0.58) and downregulated genes (q-value < 0.05 and log2FC ≤ −0.58), respectively. (C) Venn map of DEGs indicates common upregulated and downregulated DEGs between Nurr1 knockdown cells and K-strophanthoside-treated cells. (D–F) Top 3 GO enrichment pathways of commonly changed DEGs in three functional groups: Biological process (D), cellular component (E), and molecular function (F). (G–H) Bubble diagram of the top 3 KEGG enrichment pathways for common upregulated DEGs (G) and common downregulated DEGs (H). DEGs, differentially expressed genes. To further analyze the function of these DEGs, we conducted Gene Ontology (GO) and KEGG enrichment analyses. The GO biological process terms ([72]Figure D) enriched by common downregulated DEGs were the phosphatidylethanolamine biosynthetic process, the polyamine biosynthetic process, and the MAPK cascade, while those enriched by common upregulated DEGs were RNA polymerase II transcription, DNA-templated transcription, and muscle structure development. The commonly changed DEGs were significantly enriched in cellular compounds of the extracellular matrix, photoreceptor inner segment, MPP7-DLG1-LIN7 complex, nucleus, nucleoplasm, and protein–DNA complex ([73]Figure E). The molecular functions enriched by commonly changed DEGs contain RNA polymerase II-specific DNA-binding transcription factor activity, RNA polymerase II cis-regulatory region sequence-specific DNA binding, DNA binding, oxidoreductase, hydrolyase, and lyase activity ([74]Figure F). Moreover, the enrichment and analysis of KEGG pathways by common upregulated DEGs were significantly enriched in lysine degradation, renal cell carcinoma, and HIF-1 signaling pathway ([75]Figure G), while arginine and proline metabolism, beta-alanine metabolism, and valine, leucine, and isoleucine degradation were enriched by common downregulated DEGs ([76]Figure H). Altogether, these results indicated that K-strophanthoside acts as a Nurr1 inverse agonist, mimicking the function of Nurr1 knockdown. Modulation of Nurr1 by K-Strophanthoside Depends on the DNA Response Element To fully understand the modulation of Nurr1 by K-strophanthoside in a physiological environment, it is essential to understand the effects of K-strophanthoside on the reporter activity via modulating the Nurr1 protein in its monomer, homodimer, or RXR heterodimer forms. We constructed luciferase reporter plasmids containing NGFI-B response element (NBRE) for the Nurr1 monomer, Nur-response element (NurRE) for the Nurr1 homodimer, and direct repeat 5 response element (DR5) for the Nurr1-RXR heterodimer ([77]Figure A). The luciferase reporter gene assay results showed that AQ robustly enhanced the activities of all three response elements in a concentration-dependent manner ([78]Figure B–E): AQ (100 μM) activated a 2.72-fold increase in NBRE, a 1.56-fold increase in NurRE, and a 1.69-fold increase in DR5. However, K-strophanthoside exhibited marked inverse agonism on the NBRE response element (49%) and a comparatively lower inverse agonism on NurRE (22%) and DR5 (10%) response elements at a concentration of 100 μM ([79]Figure B–E). These findings suggest that K-strophanthoside has a selective regulatory effect on Nurr1. 5. [80]5 [81]Open in a new tab Construction of luciferase reporter gene plasmids and luciferase reporter gene assays for Nurr1 modulation by small-molecule compounds. (A) DNA response elements for the human Nurr1 monomer (NBRE, AAGGTCACAAGGTCACAAGGTCACAAGGTCAC), the Nurr1 homodimer (NurRE, TGATATTTACCTCCAAATGCCA), or the Nurr1-RXR heterodimer (DR5, GGTTCACCGAAAGGTCA) were fused into pGL3-Basic vector to generate transgenic luciferase reporter gene plasmids. (B–D) Full-length Nurr1 reporter gene assays with human Nurr1 response elements in HEK293T cells for 24 h: (B) NBRE for the Nurr1 monomer, (C) NurRE for the Nurr1 homodimer, (D) DR5 for the Nurr1/RXRα heterodimer. (E) Transcriptional activity profiles of small-molecule compounds (100 μM) on Nurr1 modulation compared with the control group. *P < 0.05. Data are mean ± S.E.M., n ≥ 3, one-way ANOVA in comparison expression with 0 μM compound. Discussion Nurr1 is a transcription factor of the nuclear receptor family, whose expression disturbance is closely related to diseases. , The absence of Nurr1 exacerbates the pathology of neurodegenerative diseases, ,, such as neuroinflammation and the aggregation of pathological proteins, while Nurr1 overexpression or Nurr1 agonists could improve these changes. ,,− However, in some cancers − and autoimmune diseases, , Nurr1 upregulation is associated with disease progression. Understanding the function of Nurr1 in related diseases provides a potential target for the effective treatment of these diseases. Therefore, recent studies have increasingly focused on developing Nurr1 modulators. However, the absence of an effective intrinsic Nurr1 activity modulator has promoted an urgent need for exploring an effective Nurr1 inverse agonist. In this study, we selected the Chemdiv compound library, containing about 1.5 million compounds, as the VS small-molecule library to screen Nurr1 modulators. K-strophanthoside was identified as a Nurr1 modulator, exhibiting a significant inhibitory effect on the Nurr1 transcriptional activity and regulating the expression of Nurr1 target genes. Our finding provides a novel inverse agonist for Nurr1, further indicating the presence of ligands for Nurr1. Although Nurr1 is traditionally considered to lack a classical ligand-binding pocket, recent years have seen the exploration of compounds that bind to the Nurr1-LBD and modulate Nurr1 transcriptional activity. ,,, Our study reveals that K-strophanthoside binds directly to Nurr1-LBD at a micromolar concentration, exhibiting a stronger affinity compared to AQ. Amino acid residues (Glu445, Glu514, Arg515, and His516) participate in the regulation of Nurr1 transcriptional function mediated by K-strophanthoside. Interestingly, the sugar moieties of K-strophanthoside appear to contribute to its binding with Nurr1. Investigating the specific interactions between these glycosyl groups and the Nurr1 protein will be of significant interest in future studies, which may provide valuable insights into the structural basis of ligand recognition and receptor modulation. In addition, previous study identified that residues Ile403, Leu409, Tyr575, and Asp580 are essential for the interaction of AQ and Nurr1-LBD, which differ from the key residues involved in K-strophanthoside binding. This suggests that AQ and K-strophanthoside do not directly compete for the same binding site within Nurr1-LBD. Moreover, further studies are needed to determine whether K-strophanthoside competes with AQ for binding to Nurr1 in order to fully elucidate the mechanism of K-strophanthoside action. Previous research has proved that both synthetic and endogenous ligands, such as nonsteroidal anti-inflammatory drugs and unsaturated fatty acids, regulate Nurr1 activity. ,, However, our understanding of the cellular effects of ligands on Nurr1 activity within physiological environments and their biological correlations is still limited. To address this, we further explored the regulation of the cellular effects of K-strophanthoside on the Nurr1 activity in the physiological environment. A previous study demonstrated that K-strophanthoside is capable of penetrating the cell membrane and localizing to the nucleus. Our findings further indicate that K-strophanthoside inhibits the expression of TH and AADC in a dose-dependent manner, two rate-limiting enzymes of dopamine synthesis regulated by Nurr1, , while K-strophanthoside does not affect the expression of the Nurr1 protein. These results demonstrate that K-strophanthoside functions by inhibiting Nurr1 activity rather than altering its expression. Furthermore, K-strophanthoside affected the expression of Nurr1-regulated genes involved in the polyamine biosynthetic process, release of cytochrome c from mitochondria, reactive oxygen species metabolic process, apoptotic process, MAPK cascade, transcription process, and amino acid metabolism ([82]Supplement Table 1). Similarly, an inverse Nurr1 agonist study based on indole found that its inverse agonist changed Nurr1 target gene expression in T98G cells and mimicked neuroinflammation caused by Nurr1 deficiency. The transcription of all protein-coding genes in eukaryotic genomes requires the involvement of RNA polymerase II. As a transcription factor, Nurr1 inhibition impacts RNA polymerase II and the transcription of the DNA template. In the pathological environment, mitochondrial dysfunction can lead to the increase in ROS production, a key initiator of cytochrome c release into the cytosol. The cytochrome c-initiated pathway, involving its release from mitochondria and resulting in caspase activation and subsequent cell apoptosis, is a significant caspase activation pathway. Moreover, ROS mediates mitogen-activated protein kinase (MAPK) cascades, including ERK1/2, JNK, p38, and the BMK1/ERK5, which are the major intracellular signaling pathways that regulate a variety of cellular processes. Previous research indicated that AQ, a Nurr1 agonist, increased the level of phosphorylated P38 MAPK. Our study provides evidence that Nurr1 regulated cell apoptosis. In summary, our findings further demonstrate that K-strophanthoside inhibits Nurr1 transcriptional activity in the physiological environment and provide insights into other potential biological effects mediated by Nurr1. Upon ligand interaction with Nurr1, the conformation of the Nurr1 protein changes, and then, the activity of Nurr1 is regulated. As a transcription factor, Nurr1 can function as monomers, homodimers, and heterodimers to regulate the transcription of Nurr1 target genes via the specific DNA response elements. The Nurr1 monomer binds to the NBRE, the homodimer recognizes the NurRE, and the heterodimer formed by Nurr1 and RXR binds to DR5. , To comprehensively evaluate the regulation of Nurr1 by ligands, we investigate the impact of K-strophanthoside on the regulation of NBRE, NurRE, and DR5 by human full-length Nurr1 reporter gene experiments. However, unlike other inverse Nurr1 agonists, , K-strophanthoside appears to tend to modulate the transcriptional activity of the Nurr1 monomer in a dose-dependent manner rather than the homodimer or the heterodimer. Similarly, some nonsteroidal anti-inflammatory drugs (MFA, parecoxib, oxaprozin) have also been shown to act as inverse Nurr1 agonists. However, parecoxib and oxaprozin displayed inverse agonism on NBRE, NurRE, and DR5, while MFA only inhibited the activity of NurRE and DR5 but not NBRE. These results suggest that Nurr1 could potentially be selectively regulated by chemical tools. However, given the moderate potency of K-strophanthoside on Nurr1 inverse activation, it is necessary to identify the functional groups of K-strophanthoside and optimize the compounds to screen for the most effective small-molecule compounds in future studies. As members of the NR4A family, Nurr1, Nur77, and NOR1 share a significant structural homology. It is therefore essential to establish whether K-strophanthoside exhibits a specific binding affinity for Nurr1, rather than Nur77 or NOR1. The current unavailability of NOR1’s three-dimensional structure precludes a comprehensive investigation of its potential interaction with K-strophanthoside. Moreover, our study was limited to computational predictions, suggesting the absence of direct binding between K-strophanthoside and Nur77. However, this finding has not been experimentally validated. Consequently, future studies should be designed to address these limitations. In summary, we identified a novel inverse Nurr1 agonist, K-strophanthoside, which effectively inhibits the receptor’s intrinsic transcriptional activity, demonstrating that Nurr1 activity can be selectively regulated by its ligands. These results contribute to a better understanding of the activity of Nurr1 and promote the development of modulators aimed at different structural forms of Nurr1 receptors to verify the function of Nurr1 and assess the therapeutic effects of drugs targeting Nurr1. Materials and Methods Virtual Screening (VS) The dock module in the molecular operating environment (MOE) was used for structure-based VS (SBVS). Nurr1 in the PDB (PDB code: [83]6DDA) was defined as the receptor, and the binding site was set around the native ligand. The procedure of virtual screening is shown in [84]Figure S8. We initially conducted high-throughput rigid docking between Nurr1-LBD and 1.5 million compounds from the ChemDiv compound library, obtaining London dG scores. Subsequently, the top 15,000 compounds, ranked according to their London dG scores, were subjected to flexible redocking with Nurr1 using the “induced fit” protocol. The top 100 poses, ranked according to the London dG scores, performed the force field refinement and re-evaluated using the GBVI/WSA dG scoring function. Following the flexible docking of the 15,000 compounds, we employed fingerprint-based clustering to categorize them into structural clusters, applying a distance threshold of 0.5 between clusters. From each cluster, we identified the top 100 cluster centers as potential hits based on the highest-ranked compound within each cluster. Finally, the top 10 compounds were selected for further functional validation. The SBVS and molecular docking were performed by Wecomput Technology Co., Ltd. To further retrospectively validate our docking procedure, we chose a set of known Nurr1 modulators that have been assessed for their ability to bind to Nurr1-LBD. The Nurr1 protein structure (PDB code: [85]6DDA) was prepared by the QuickPrep module in MOE. All compounds were energy-minimized, docked with the same protocol, and ranked according to their docking scores. To evaluate the predictive performance of the docking approach, we generated an ROC curve. The AUC was 0.778 ([86]Figure S11), indicating good predictive accuracy of the docking method. Compounds and Antibodies The top 10 compounds were purchased from commercial sources (ChemDiv Inc.), with a purity of at least 95%. AQ was purchased from TargetMol. The antibodies were shown as follows: anti-Nurr1 antibody (Proteintech), anti-GAPDH antibody (Cell Signaling Technology), anti-TH antibody (Millipore), anti-AADC antibody (Santa Cruz Biotechnology), goat anti-mouse IgG H&L (Proteintech), goat anti-rabbit IgG H&L (Proteintech), goat anti-rabbit Alexa 594 (Cell Signaling Technology), and goat anti-mouse Alexa 488 (Cell Signaling Technology). SPR A Biacore T200 instrument was used for the SPR analysis to investigate the binding affinity between the human Nurr1 protein (Abcam) and compounds. The amino coupling method was adopted by the CM5 chip. The protein system contains Tris and was replaced with a pH 5.0 sodium acetate solution by an ultrafiltration tube. The protein concentration after ultrafiltration was about 84 μg/mL. The coupling conditions were as follows: a concentration of approximately 21 μg/mL, a system of pH 5.0 sodium acetate solution, a chip activation time of 420 s, and a sealing time of 420 s. The protein coupling amount of Nurr1 was 5950 RU. SPR binding experiments were performed in the same running buffer, PBS (pH 7.4) supplemented with 5% dimethyl sulfoxide (DMSO). Based on the solubility of the sample to be measured and the core surface test, a series of concentrations were prepared (AQ: 125, 62.5, 31.25, 15.63, 7.813, 3.906, 1.953 μM, compound 7: 250, 125, 62.5, 31.25, 15.63, 7.813, 3.906 μM). The test conditions for the compounds to be measured were an injection time of 60 s, a dissociation time of 60 s, and no regeneration. Binding curves were analyzed by using Biacore-provided software. Luciferase Reporter Assays To conduct transactivation assays based on full-length Nurr1, we constructed related reporter gene plasmids containing the human Nurr1 response elements DR5 (pGL3-Basic-DR5; GGTTCACCGAAAGGTCA), NBRE (pGL3-Basic-NBRE; AAGGTCACAAGGTCACAAGGTCACAAGGTCAC), or NurRE (pGL3-Basic-NurRE; TGATATTTACCTCCAAATGCCA). Nurr1 and RXRα were overexpressed for DR5. The Renilla luciferase expression vector pGL4.74 was obtained from Miaoling Biotechnology (Wuhan, China). The full-length human Nurr1 expression plasmid (pCMV-hNurr1) contains the full-length cDNA encoding human Nurr1 (amino acids 1–598). Site-directed mutagenesis was performed to generate specific Nurr1 point mutants. The following nucleotide substitutions were introduced to produce alanine substitutions at the indicated positions: Glu445 (GAA → GCA), Leu509 (CTG → GCG), Glu514 (GAG → GCG), Arg515 (AGA → GCA), and His516 (CAC → GCC). HEK293T cells were cultured and transfected as previously described. For assays on full-length human Nurr1, the plasmid mixtures were pCMV-hNurr1/pGL3-Basic-NBRE/pGL4.74 (NBRE), pCMV-hNurr1/pGL3-Basic-NurRE/pGL4.74 (NurRE), and pCMV-hNurr1/pCMV-RXRα/pGL3-Basic-DR5/pGL4.74 (DR5). The cells were cultured with respective concentrations of compounds and incubated for 24 h to detect the luciferase activity by a Dual Luciferase Reporter Gene Assay Kit (Beyotime). The firefly luciferase data were normalized by dividing them with the renilla luciferase data, followed by calculating the fold activation of compounds at various concentrations. In the Nurr1 mutagenesis experiments, fold changes were calculated by comparing normalized values in the presence versus absence of K-strophanthoside for each mutant. These values were subsequently compared to those from wild-type Nurr1 under the same conditions to determine the impact of each mutation. Cell Viability Assay Neuro-2a cells and HEK293T cells were seeded into 96-well plates (10^4 cells/well), and compounds at various concentrations were added to at least three wells. After incubation for 24 h, CCK-8 reagent (Meilunbio) was added, and the mixture was incubated at 37 °C and 5% CO[2] for 1–2 h. Absorbance at 450 nm was measured, and cell survival rates were calculated. Western Blot Cells after culture with K-strophanthoside for 24 h were resuspended in RIPA buffer (Beyotime) with protease inhibitor cocktails (Sigma-Aldrich) and centrifuged at 12,000 rpm for 15 min. BCA assay kit (Takara) was used to determine the protein concentration, and sodium dodecyl sulfate (SDS) loading buffer (Beyotime) was added to denature the proteins. The protein samples were separated by SDS-polyacrylamide gel electrophoresis (PAGE) and transferred to polyvinylidene difluoride (PVDF) membranes. The membranes were subsequently blocked and incubated with antibodies. Chemiluminescence detection kit (Abbkine) was used to visualize protein bands, and ImageJ was used to analyze the results. The intensities of proteins were normalized to those of GAPDH. Quantitative PCR Cells cultured with K-strophanthoside at 30 μM for 24 h were used for total RNA extraction using TRIzol reagent (Takara). After cDNA synthesis (TransGene Biotech), mRNA levels were analyzed by an SYBR Green RT-PCR kit (TransGene Biotech) and an ABI Prism 7500 assay system (Applied Biosystems). The results were analyzed by the 2-ΔΔCt formula. The primers for the real-time PCR assay were shown as follows: GAPDH (F: GCATTGTGGAAGGGCTCATG, R: AGGGATGATGTTCTGGGCAG); TH (F: CAATACAAGCAGGGTGAGCC, R: TAGCATAGAGGCCCTTCAGC); AADC (F: GAGCTGGAGACCGTGATGAT, R: TTAGTCCGAGCAGCCAGTAG); Nurr1 (F: ATTCCAGGTTCCAGGCAAAC, R: AGCAAAGCCAGGGATCTTCT). Immunofluorescence Staining After administration of K-strophanthoside for 24 h, cells were processed as previously described: they were blocked with 2% BSA, incubated with antibodies, and subsequently imaged by a confocal microscope (Nikon). RNA Isolation and Library Preparation Neuro-2a cells were cultured with K-strophanthoside at a concentration of 30 μM for 24 h, and the total RNA was extracted using the TRIzol reagent (Invitrogen). RNA purity, quantification, and integrity were evaluated. Then, the libraries were constructed using the VAHTS Universal V6 RNA-seq Library Prep Kit. RNA Sequencing and Differentially Expressed Gene Analysis The transcriptome sequencing and analysis were performed by OE Biotech Co., Ltd. Differential expression analysis was performed using DESeq2, with significance thresholds set at a q-value <0.05 and |log2foldchange| ≥ 0.58. Hierarchical cluster analysis, along with GO and KEGG pathway enrichment analysis of DEGs, was carried out using R (v 3.2.0). Statistical Analysis The experimental data obtained in this study were analyzed by the GraphPad Prism 7 system, and t-test or one-way ANOVA was used for statistical analysis. P < 0.05 was considered statistically significant. All data are presented as the mean ± standard error of the mean (S.E.M.). Supplementary Material [87]ao5c06698_si_001.pdf^ (1.3MB, pdf) Acknowledgments