Next Article in Journal
The Correlation Between Plasma Vitamin D and Blood Parameters in Prenatal Women
Previous Article in Journal
Type 2 Diabetes and the Multifaceted Gut-X Axes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

NR4A1 Acts as a Nutrient Sensor That Inhibits the Effects of Aging

Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
Nutrients 2025, 17(16), 2709; https://doi.org/10.3390/nu17162709
Submission received: 15 July 2025 / Revised: 11 August 2025 / Accepted: 18 August 2025 / Published: 21 August 2025
(This article belongs to the Section Geriatric Nutrition)

Abstract

Orphan nuclear receptor 4A1 (NR4A1) is a member of the NR4A subfamily that was initially discovered as an intermediate early gene expressed in response to stressors, including inflammatory agents. This review addresses the hypothesis that NR4A1 is a key nutrient sensor that contributes to the anti-aging and health-protective effects of receptor ligands, dietary phenolics, and other diet-derived compounds. There is evidence in animal models including humans that NR4A1 serves as an important gene that decreases the rate of aging and its associated diseases. For example, in humans and mice, NR4A1 expression decreases with age and loss of NR4A1 enhances disease susceptibility, and survival curves show that NR4A1-deficient mice live 4 months less than wild-type animals. An extensive comparison of inflammatory diseases, immune dysfunction, and fibrosis in multiple tissues shows that in NR4A1−/− mice and rats these diseases and injuries are enhanced compared to wild-type NR4A1−/− animals. There is evidence showing that structurally diverse NR4A1 ligands reverse the induced adverse effects in NR4A1 wild-type mice. This raises an important question regarding the mechanisms of NR4A1-dependent inhibition of the aging process and the potential for this receptor as a nutrient sensor. It has been well established that polyphenolics, including flavonoids, resveratrol, and other compounds in the diet, are health-protective and decrease the aging process. Recent studies show that resveratrol and flavonoids such as quercetin and kaempferol bind NR4A1 and exhibit protective NR4A1-dependent inhibition of endometriosis and cancer. These limited studies support a role for NR4A1 as a potential dietary sensor of nutrients that are known to be health-protective and a potential nutrient target for improving health.

Graphical Abstract

1. Introduction

The rate of aging in individuals is complex and is influenced by multiple factors, including genetics, environmental factors, diet, and lifestyle [1,2,3,4]. The goal of decreasing mortality by addressing genetic deficiencies is difficult and requires heightened awareness and early treatment of specific genetic-induced diseases. Increased health benefits are more attainable by modifying diet/lifestyle and exposure to adverse environmental factors. Diet is an important consideration, since population studies show decreased rates of mortality in vegetarians and individuals consuming the Mediterranean diet and other diets enriched in fruits, nuts, and vegetables and also coffee [5,6,7,8,9,10]. These health benefits are primarily due to modulation of key pathways/genes by dietary components, which include flavonoids and other polyphenolics, caloric restriction, vitamins, and microbiome-derived compounds.
Some of the genes/pathways involved in aging include sirtuin-1 (SIRT-1), SIRT-3, SIRT-7, telomerase components, AMPK/mTOR signaling, FOX03, NRF2, NFkB, and polyamine production. The functions of these genes are variable, and they are involved in redox regulation, anti-inflammatory pathways, mTOR pathway inhibition, maintenance of DNA integrity (e.g., DNA repair mechanisms), enhanced mitochondrial functions, and extracellular matrix remodeling [1,2,3,4]. The aging process is accompanied by the decline in many of the important pathways/genes required for maintaining cellular homeostasis, and dietary interventions that decrease mortality modulate the aging process by targeting these specific pathways and related genes. The identities of some genes/pathways involved in aging and many of the dietary compounds that modulate and enhance health benefits such as decreased mortality are known; however, the mechanisms of individual and compound-induced protective responses are not completely understood. Identification of specific pathways/genes targeted by health-promoting dietary compounds would facilitate the development of a more precision nutrition approach for enhancing diet-induced health and the development of related dietary interventions and therapeutics.
We hypothesize that orphan nuclear receptor 4A1 (NR4A1) plays a protective role against stressor/inflammation-induced cell damage that accompanies the aging process. In addition, synthetic NR4A1 ligands further enhance NR4A1-mediated protection, and recent discoveries show that some health-promoting polyphenolics also bind NR4A1, suggesting that this receptor may also play a role as a nutrient sensor.

2. Orphan Nuclear Receptor 4A1 (NR4A1) and Aging

2.1. Background

Orphan nuclear receptor 4A1 (NR4A1, Nur77) is a member of the NR4A subfamily of nuclear receptors that includes NR4A2 (Nurr1) and NR4A3 (Nor1), and these receptors are classified as orphans since their endogenous ligands have not yet been identified [11,12,13]. NR4A1 and other members of this subfamily have a modular structure that exhibits similar percentages of amino acid identities in the C-terminal ligand-binding (E) and DNA-binding (C) domains but differs substantially in its N-terminal (A/B) domain. NR4A1 targets multiple promoter DNA-binding elements and also modulates gene expression through activation of regulated genes via protein–protein interaction (Figure 1) [14,15,16,17]. The three NR4A members are immediate early genes in non-transformed cells and they are induced by multiple stressors in various cell types [18,19,20,21], whereas in cancer cells individual NR4A genes exhibit tumor type-specific tumor suppressor- or tumor promoter-like activities [14]. Although the endogenous ligands for NR4A1 have not been identified, a large number of biochemicals including prostaglandins and fatty acids bind and activate the receptor at pharmacological concentrations. In addition, dietary compounds, structurally diverse natural products and their analogs, and a large number of synthetic chemicals including drugs also bind NR4A1 and activate the receptor [22,23,24]. Studies in this laboratory have identified and characterized a series of synthetic 1,1-bis(3′-indolyl)-1-(substitutedphenyl)methane (CDIM) compounds derived from 1,1-bis(3′-indolyl)methane (DIM) that bind NR4A, and the 3,5-disubstitutedphenyl (DIM-3,5) analogs bind both NR4A1 and NR4A2 [25] (Figure 2).

2.2. Effects of Aging on NR4A1 Expression

A characteristic of many key genes involved in reducing the rate of aging is their decreased expression during the aging process, and this has also been observed for NR4A1. For example, NR4A1 mRNA expression is significantly decreased in peripheral blood mononuclear cells from ages 20–40 to >75 years of age, and similar differences were observed in mice [26]. Moreover, in a subset of humans with normal and decreased cognitive function, NR4A1 expression was lower in the latter group. Experimental studies in mice showed that the age-dependent decrease was in the hippocampal CAI pyramidal neurons and the loss of NR4A1 and impairment of cognition was due to loss of Trk, an NR4A1-regulated gene [26]. Chronic kidney disease is age-related and associated with enhanced tubulointerstitial fibrosis; in mice there was an inverse correlation between TGFβ expression vs. NR4A1 expression and increased kidney damage. In older mice, the loss of NR4A1 activated TGFβ/SMAD signaling and subsequent renal toxicity [27]. The age-dependent decrease in NR4A1 was also observed in the mouse liver, kidney, and peri-adipose tissue; this was paralleled by a decrease in SIRT1 expression and was also observed in oxidative stress-induced HEK-293T cells [28]. NR4A1 indirectly regulates SIRT1 by decreasing the E3 ligase MDM2 and thereby enhancing SIRT1 expression by decreasing its proteasome-mediated degradation. This paper also compared the survival of wild-type and NR4A1−/− mice and showed that the loss of NR4A1 decreased % survival curves of mice by approximately 4 months [28]. Thus the mouse models showed that NR4A1 is a pro-survival transcription factor due in part to stabilizing SIRT1.
The extent of cardiac fibrosis is also age-dependent and comparisons between 6- and 15-month-old mice showed increases in cardiac fibrosis and damage that correlated with decreased expression of NR4A1 mRNA levels [29]. Moreover, in NR4A1−/− mice the markers of cardiac dysfunction were enhanced compared to wild-type mice. Mechanistic studies showed that NR4A1 deficiency correlated with decreased GSK-3β and increased β-catenin expression, and the former gene was a direct target of NR4A1. These results also link NR4A1 activity to SIRT3, which inhibits age-related fibrosis in the heart and other tissues by SIRT3-dependent deacetylation and activation of GSK-3β [30]. The contributions of NR4A1 to muscle growth were also investigated in wild-type and NR4A−/− mice at various time points [31]. Although age-dependent changes in NR4A1 were not provided, the loss of NR4A1 (globally and muscle-specific) decreased myofiber size in mice at E 18.5 and in 3-month-old mice. Moreover, the loss of NR4A1 was accompanied by induction of two negative regulators of muscle mass, SMAD2 and FOX01. Ovarian function also decreases in reproductively aging mice, and this is accompanied by decreased NR4A1 expression. Moreover, in human umbilical cord mesenchymal stem cell transplantation models in mice, AMPK/NR4A1 signaling enhanced ovarian function and decreased fibrosis [32,33,34]. Results of these age-dependent effects on expression of NR4A1 and mechanistic studies with NR4A1−/− mice demonstrate that like other pro-survival genes NR4A1 decreases with age and NR4A1 contributes to several health benefits. These studies also show that NR4A1 interacts with genes that are associated with decreased rates of mortality, including SIRT1, GSK-3β, and SIRT3. Moreover, NR4A1 also plays a protective role in maintaining DNA integrity. DNA-dependent protein kinase (DNA-PK) plays an important role in double-stranded break (DSB) repair and NR4A1 is involved in repair of DNA damage [35,36]. NR4A1 and other NR4As are recruited to the DNA damage sites and target poly-ADP-ribosylated DNA-PKs to enhance its autophosphorylation.
Thus the role of NR4A1 in aging shows that this transcription factor interacts with several key genes involved aging (Figure 3), including SIRT1 in oxidative stress responses [28], SIRT3 in age-related fibrosis [30], and NFkB in multiple stress-induced responses [18], DNA damage, and repair responses [35,36]. In addition, NR4A1 regulates mTOR signaling in angiotensinαII-induced cardiac hypertrophy [37]; NR4A1 also enhances muscle mass in mice through IGF1-induced activation of mTOR [38], and in a mouse model of Parkinson’s disease the anti-inflammatory effect of NR4A1 is also associated with NRF2 [39]. FOX03 expression inhibits mouse hepatocyte proliferation, and this is paralleled by downregulation of NR4A1 in a liver regeneration model [40]. Thus NR4A1 regulates or exhibits some association with many of the genes that play a role in aging; however, their interactions and responses and effects of stressors may be variable and cell context-dependent. The next section of this review briefly outlines several tissue-specific health-promoting functions of NR4A1, particularly with respect to protecting against stressor/inflammation-induced damage, which plays a role in the aging process. We hypothesize that orphan nuclear receptor 4A1 (NR4A1) plays a protective role against stressor/inflammation-induced cell damage that accompanies the aging process. In addition, synthetic NR4A1 ligands further enhance NR4A1-mediated protection, and recent discoveries show that some health-promoting polyphenolics also bind NR4A1, suggesting that this receptor may play a role as a nutrient sensor.

3. Tissue-Specific Functions of NR4A1

Although there are only a limited number of studies on the role of NR4A1 in aging, it has been shown that NR4A1 decreases with age, and NR4A1 deficiencies are accompanied by enhanced adverse responses. There is an increasing number of publications on the tissue-specific constitutive role of NR4A1 and these are primarily determined by examining responses after knockdown of the receptor, and this will be outlined in this section.

3.1. Cardiovascular

NR4A1 is expressed in the heart and is induced by VEGF and other vascular permeabilizing agents [41,42]. In mice, the loss of NR4A1 is accompanied by lower levels of basal vascular permeability (BVP), and it was concluded that NR4A1 plays a key role in BVP, chronic vascular hyperpermeability (CVH), and acute vascular hyperpermeability (AVH) [42,43]. In NR4A1-KO mice adverse cardio remodeling was associated with increased diastolic and systolic Ca+2 and this was accompanied by larger cardiomyocytes [44]. Protection of cardiac remodeling by NR4A1 was also due to NR4A1-dependent suppression of the sympathetic co-transmitter neuropeptide Y (NPY) [45]. NR4A1 also plays an important role in immune system-mediated cardiac remodeling, and this involves Ly-6Chigh monocytes and macrophages that infiltrate the damaged myocardium [46,47,48]. The loss of NR4A1 in macrophages and monocytes results in enhanced pro-inflammatory M1 macrophages and NR4A1-KO mice maintained on a Western diet developed increased atherosclerosis [47]. These results clearly demonstrate a protective role for NR4A1 in cardiovascular disease and there are also extensive studies on interactions of NR4A1 during induced cardiovascular damage [49], which will also be noted in the next section of this review.

3.2. Neuronal

NR4A1, NR4A2, and NR4A3 play an important role in long-term memory and NR4A ligands reactivate age-dependent memory decline [50,51,52] and this is associated with their regulation of endoplasmic reticulum chaperones [53]. Another study in mice reported that NR4A1 was necessary for object location whereas NR4A2 was required for long-term memory, object location, and recognition [54] and loss of NR4A1 in inhibitory GABAergic interneurons affects associative learning [55]. NR4A1 is also involved in post-stroke recovery, and deletion of NR4A1 in microglia results in increased expression of TNF and this results in increased brain injury [56].

3.3. Muscle Mass and Myofiber Size

NR4A1 overexpression in the skeletal muscle of mice increased muscle mass and loss of NR4A1 reduced muscle mass and myofiber size via activating IGF-1 growth-promoting pathways [38], and estrogen induces NR4A1 in skeletal muscle [57]. NR4A1 overexpression also promoted cell adhesion and fusion in myoblasts by regulating ZEB1 transcription [58].

3.4. Retina and Eye

In a genomic screen for transcriptional targets of circadian melatonin and dopamine signaling in mice, NR4A1 was identified as a candidate gene that regulated neurohormone release and functional adaptation and healthiness of the retinal and photoreceptor cells [59].

3.5. Obesity and Metabolic Disease

β-Cells play a critical role in insulin secretion and decreases in β-cell function are associated with development of type 1 and type 2 diabetes. Overexpression of NR4A1 (and NR4A3) enhances β-cell expansion, mitochondrial respiration, and insulin secretion; these effects are reversed in NR4A1-KO mice [60,61] and this complements other studies demonstrating a role for NR4A1 in mitochondrial function [62,63,64].
NR4A1 also plays an important role in muscle cells, which are prime sites of glucose metabolism and overall insulin sensitivity and obesity. For example, results of receptor knockdown in muscle cells show that NR4A1 is linked to genes that regulate lipid homeostasis, mitochondrial function, energy expenditure, and glucose metabolism in muscle cells and in vivo [62,63,64,65,66,67,68,69]. In the liver, NR4A1 is involved in gluconeogenesis and increased blood glucose levels, and deletion of the receptor enhances hepatic steatosis and increases expression of lipogenic genes [63,67]. NR4A1 and other NR4A members also play a role in lipid metabolism and are induced by multiple stimuli; however, overall NR4A1 expression inhibits adipogenesis and this is associated with inhibition of mitotic clonal expansion of adipocytes [68]. The overall mechanisms of NR4A1 action in adipose tissue includes NR4A1-mediated repression of PPARƔ2 in white adipose tissue [69], interactions between NR4A1 and STAT3 acetylation, and leptin sensitivity [70]. Moreover, there is also a report showing that female, but not male, NR4A1-deficient mice exhibit increased susceptibility to obesity when maintained on a high-fat diet [71].

3.6. Intestinal Inflammation

NR4A1 also plays a role in intestinal inflammatory conditions, which include colitis and inflammatory bowel disease. In NR4A1-deficient mice, elevated expression of genes related to extracellular matrix (ECM) production, metabolism, and cell proliferation were observed in intestinal smooth muscle cells [72]. Another study showed that loss of NR4A1 also increased intestinal mucosal ECM content and markers of cell proliferation and increased α-smooth muscle actin and collagen levels [73]. Similar protective effects of NR4A1 were observed in another study, and it was also reported that NR4A1 SNPs with lower activity than wild-type NR4A1 were associated with increased risks for ulcerative colitis and Crohn’s disease [74]. NR4A1 indirectly inhibited NFkB signaling by negatively regulating TLR-IL1R signaling and preventing TRAF6 deubiquitination [74]. These studies clearly demonstrate that NR4A1 expression inhibits both basal and dextran sodium sulfate-induced intestinal inflammation and subsequent damage.

3.7. Wound Healing and Angiogenesis

Wound healing is a critical step in resolving both basal and induced lesions and both gene knockdown and overexpression studies show that NRA1 regulates VEGF-induced angiogenesis and also the induction of integrins [75,76,77]. Interestingly, NR4A1 mediated induction of β4-integrin in HUVEC cells [2] and a subsequent study showed that induction of other integrins was also NR4A1-dependent [77].

3.8. Bone

NR4A1 is an important regulator of macrophage function, and Hamers and coworkers investigated the role of NR4A1 in bone marrow-derived macrophages (BMMs) in both wild-type and NR4A1-KO mice [78]. NR4A1 downregulates CXCL12 (SDF-1α) expression, enhances CX3CR1 expression, and represses NFkB, suggesting an anti-inflammatory effect of NR4A1 in BMMs. A subsequent report by this group [79] compared results of studies in wild-type classical NR4A-KO and novel NR4A1-KO (Cre) mouse models. It was demonstrated that the classical NR4A1-KO mice expressed the N-terminal aa1-117 region of NR4A1 whereas the NR4A1-KO (Cre) mice did not. Moreover, the classical NR4A1-KO mice (aa1-117) exhibited several functional and structural abnormalities not observed in wild-type or NR4A1-KO (Cre) mice [79]. In contrast, in both the classical and full-length NR4A1-KO mice there were decreased circulating Ly6Clow monocytes and decreased EGFR and VCAM1, which are bone marrow retention genes. Overall the report suggests that results obtained in NR4A1-knockout mice (whole body) expressing aa1-117 may need to be reinterpreted, particularly since this truncated form of NR4A1 (namely TR3β) is expressed and may be functional in some tissue/cell types, including human bone marrow cells and acute myeloid leukemia.

3.9. Autoimmunity

Loss of NR4A1 in T cells in mice is characterized by a highly proliferative phenotype and enhanced susceptibility to T cell-mediated inflammatory disease such as contact dermatitis and CNS autoimmunity [80]. NR4A1-deficient myeloid cells exhibit increased production of norepinephrine, and this resulted in acceleration of experimental autoimmune encephalomyelitis (AEA) and increased lumbar spinal cord Th expression [81]. Thus NR4A1 is an endogenous inhibitor in this model of induced multiple sclerosis and this is due, in part, to repression of norepinephrine and Th expression; a more recent study showed similar functions for NR4A1 in CNS autoimmunity [82]. It was also reported that NR4A1 was protective in a mouse model of lupus by inhibiting synaptic stripping by microglia [83].

3.10. Immune Responses

NR4A family members play a key role in regulating immune cell function and their expression is induced by T cell receptor signaling in the nucleus and NR4As play both overlapping and specific roles in T cells. NR4As enhance differentiation of CD4+ T cells into Treg cells in the thymus and peripheral tissues and this involves induction of Foxp3 and suppression of Th2 and Th17 cytokine genes [84,85,86,87,88]. NR4A1 (and NR4A3) acts to restrain B cell responses to antigen and interactions with T cells, and this is mediated by decreased expression of MYC and the basic leucine zipper ATF-like transcription factor (BATF) [89,90]. NR4A1 is also expressed in dendritic cells but loss of the receptor in mice did not significantly impact the development of dendritic cells in the spleen and lymph nodes; knockdown of NR4A1 in human monocyte-derived dendritic cells and murine dendritic cells enhanced inflammatory responses and T cell populations were increased [91]. Most studies with macrophages demonstrate that NR4A1 and other NR4As attenuate inflammation-induced responses [92]. Other examples of NR4A1-mediated effects in immune cells and resident tissues are discussed throughout this section.

3.11. Lung

In patients with pulmonary arterial hypertension (PAH), the pulmonary artery smooth muscle cells (PASMCs) exhibited increased proliferation and survival and decreased expression of NR4A1, NR4A2, and NR4A3 (protein and mRNA) compared to normal lung PASMCs [93]. In contrast, expression of the three NR4As in lung tissue is increased in patients with PAH compared to donors. Results of both human and mouse studies demonstrate that loss of NR4A1 in PASMCs results in decreased wound healing, increased cell proliferation, decreased axin, and increased β-catenin mRNAs. Studies in mice with hypoxia-induced pulmonary hypertension also demonstrate a protective role for NR4A1 [93].

3.12. Kidney

Several studies show that loss of NR4A1in the kidney enhances basal, genetic, and induced kidney damage. There was a decrease in NR4A1 in 20-month-old compared to 5-month-old mice and this correlated with increased renal tubular injury fibrosis, pSMAD2/3/SMAD2/3 ratio, collagen1, and Acta2 [27]. In addition, most of these same parameters were also increased in NR4A1-deficient mice, demonstrating the protective role of NR4A1 against unilateral ureteral obstruction (UUO)-induced fibrosis. Inhibition of the P13K/AKT pathway [94] and VEGF was associated with NR4A1-mediated inhibition of basal and unilateral ureteral obstruction-induced effects in mouse models [95]. In rat models loss of NR4A1 enhanced macrophage-mediated rat renal injury [96]. An in vivo study in mice and ITK-2 human kidney cells showed that UUO-induced effects in mice resulted in increased NR4A1, and fibrosis and similar results were observed in TGFβ-induced fibrosis in ITK-2 cells. Moreover, CsnB enhanced fibrosis [97]. This study did not examine the effects in NR4A1-deficient mice or cells and the results are in contrast to most other reports on NR4A1 and tissue injury.

3.13. Bladder

Urinary tract infection (UTI) of the bladder by UroPathogenic Escherichia Coli (UPEC) is a serious and common infection that is traditionally treated with antibacterials. It was recently observed in wild-type and NR4A1-KO mice that loss of NR4A1 increased the persistence of UPEC in bladder tissue, and this was accompanied by enhanced bacterial communities in the bladder of NR4A1-KO mice [98]. Thus NR4A1 plays a protective role in terms of decreasing UTI by acting on bacterial communities, and this response coupled with the effectiveness of treatment with CsnB is a highly novel approach for treating UTI.

3.14. Liver

The basal activity of NR4A1 in the liver is primarily derived from comparing liver function in normal and NR4A1-KO cells and in vivo. In FOX03-deficient mice and liver cells, loss of NR4A1 increased expression markers of hepatotoxicity [40]. Most studies show that NR4A1 plays a protective role in various stress-induced models of hepatotoxicity. For example, NR4A1 protects against homocysteine-induced hepatic steatosis, drug-induced liver injury, ischemic reperfusion injury, hypoxia–reperfusion injury, TGFβ-induced fibrosis, and genetic-induced liver damage [99,100,101,102,103,104,105,106,107].

3.15. Interactions with ROS

Reports on the potential NR4A1-mediated antioxidant activities in non-transformed tissues/cells are limited; however, there is some supporting data. For example, in vascular endothelial dysfunction the induction of ROS production is enhanced; this can be alleviated by overexpression of NR4A1 and the subsequent activation of genes that protect against ROS and nitric oxide production [108]. There is also evidence that NR4A1 “may be a sensor of oxidative stress and an inhibitor of vascular remodeling” [109], and a recent study showed that metformin inhibited myocardial ischemia–reperfusion injury and this is due, in part, to enhanced NR4A1 production and receptor-dependent induction of isocitrate dehydrogenase1 [110]. In a model of Parkinson’s disease, 1-methyl-4-phenylpyridinium (MMP+)-induced apoptosis and ROS in SH-SY5Y cells was inhibited by overexpression of YY1, which in turn induced NR4A1 expression [111]. These results suggest that NR4A1-mediated inhibition of ROS may be due to the activation of gene products that downregulate ROS or induce genes associated with enhance cellular reductant formation.

4. NR4A1 Ligands and Their Modulation of NR4A1-Dependent Responses

4.1. Introduction

Most but not all studies [112,113] confirm that NR4A1 plays a health-protective role in maintaining cellular homeostasis, as indicated above. Among the most commonly observedNR4A1/NR4A1 ligand-mediated anti-inflammatory responses is the inhibition of NFkB signaling, and this has been extensively reviewed [20,21,22,23,114,115]. Initial studies on the structures of NR4A proteins indicated that the ligand-binding domains contained bulky amino acid side chains and that these receptors were ligand-independent transcription factors [116]. This conclusion was consistent with the fact that endogenous ligands for orphan nuclear receptors such as NR4A have not been identified. However, a rapidly increasing number of structurally diverse compounds that directly interact with the ligand-binding domain of NR4A1 have been characterized [22,23,24] and act as agonists or inverse agonists that activate or inhibit expression of NR4A1-regulated genes, respectively. The agonist or inverse agonist activities of these ligands may be gene-, response-, and cell context-dependent, and this is not uncommon for ligands for other nuclear receptors. For example, tamoxifen, a selective estrogen receptor modulator (SERM), acts as a functional ER antagonist in breast cancer but exhibits functional ER agonist activities in the bone and endometrium [117,118]. This latter response has been associated with an increased incidence of endometrial cancer in women with breast cancer who were treated with tamoxifen for extended periods [119].

4.2. Effects of NR4A1 Ligands on Liver, Intestinal, Bladder, and Kidney Damage

Among the many structurally diverse compounds that bind NR4A1, the natural product cytosporone B (CsnB) has been the most widely used for determining effects on NR4A1-regulated non-cancer endpoints [120,121]. Thus the effect of this compound and other NR4A1 ligands serve as a model for the potential effects of naturally occurring dietary compounds that may act through NR4A1 to induce health benefits, and this will be discussed in the next section of this review. Table 1 summarizes several studies that have investigated the role of NR4A1 in multiple tissues/organs, and comparisons between wild-type NR4A and NR4A−/− mice invariably show that the loss of NR4A1 results in enhanced tissue/organ damage. Moreover the use of CsnB or other NR4A1 ligands such as 6-mercaptorpurine (6-MP), bis-indole-derived compounds (CDIMs), celastrol, or the Gly-Pro-Ala (GPA) peptide further enhance the protective effects of NR4A1. Results of these studies in non-cancer tissues provide insights into cell or tissue damage that may also be observed as part of the aging process and which is protected by treatment with NR4A1 ligands.
In cultured human hepatic stellate cells expressing the PNPLA3 I148M variant, a risk factor for fibrogenic liver disease, there was increased TGFβ signaling and decreased NR4A1 expression compared to wild-type cells [107]. These cells exhibited multiple dysfunctional characteristics. A series of experiments in wild-type and NR4A1-KO mouse-derived cells showed that loss of NR4A1 enhanced induced fibrosis and steatosis and NR4A1 ligands such as CsnB protected against these responses in NR4A1-expressing liver cells/tissues [99,104,107]. The response patterns in mouse and in cell culture models of intestinal inflammation were similar to those observed in the liver, where induced inflammatory responses are enhanced in cells/tissues deficient in NR4A1 compared to wild-type, and NR4A1 and ligands such CsnB or 6-MP ameliorated cell damage phenotypes in wild-type NR4A1-expressing intestines [72,73,74,122]. A novel role for NR4A1 was observed in urinary tract infection (UTI) by uropathogenic E. coli (UPEC), where loss of NR4A1 in mice resulted in enhanced bacterial infection in the bladder [98] and CsnB decreased infection in wild-type mice and in cell culture, and this represents a novel mechanistic and therapeutic approach for treating UTI. Unilateral ureteral obstruction (UUO) in mice induced NR4A1 and enhanced fibrosis and interstitial kidney damage and CsnB enhanced this response, and similar enhancement was observed in a TGFβ-induced response in vitro [97]. The pro-fibrotic activity of CsnB contrasts with most other effects observed for this NR4A1 ligand. Moreover, another publication on UUO-induced renal damage reported that both NR4A1 and CsnB exhibited protective effects [94], though reasons for the differences between these studies are unclear. Moreover, CsnB also inhibited induced kidney fibrosis in a mouse model [104].

4.3. Effects of NR4A1 Ligands on Pulmonary Bone and CNS Damage

Pulmonary arterial hypertension (PAH) is a serious disease in humans and NR4A1 is downregulated in PASMCs in humans with PAH, and overexpression of NR4A1 in PASMCs blocked cell proliferation and migration [93]. LPS-induced pulmonary damage and activation of inflammatory factors in rats was inhibited by pretreatment with pterostilbene, which was shown to interact with NR4A1 [123]. Mouse model studies demonstrated that hypoxia-induced PAH-like symptoms were mitigated by treatment with CsnB. Influenza-induced adverse effects on pulmonary function are also ameliorated after treatment with CsnB, which acts on both lung tissue and macrophages, and this activity is associated with the induction of type I interferon [124]. Another study showed that 6-MP also inhibited progression of pulmonary hypertension, where NR4A1 induced bone morphogenic protein LCBMP signaling and decreased inflammation and proliferation in microvascular endothelial cells [125]. Inflammation associated with osteoarthritis in humans was accompanied by increased expression of NR4A1 and overexpression of several MMPs, COX-2, and iNOS [126]. In a rat model of osteoarthritis, CsnB inhibited the same set of inflammatory genes and protected against inflammation.
The role of NR4A2 and effects of NR4A2 ligands on different aspects of neuronal toxicities including learning and memory and Parkinson’s disease have been extensively investigated [52,127,128,129,130] whereas this is not the case for NR4A1 ligands. MPTP induced neurotoxic effects in a mouse model of Parkinson’s disease and is accompanied by induction of NFkB and progressive loss of dopaminergic neurons in the substantia nigra in mice [129]. Protection from the induced neurotoxic responses was observed after treatment with the NR4A1 ligand (1,1-bis(3′-indolyl)-1-(4-hydroxyphenyl)methane (DIM-4-OH) and the corresponding 4-chlorophenyl analog (DIM-4-CI), which is an NR4A2 ligand [130]. PC12 rat adrenal pheochromocytoma cells were used as a model of Parkinson’s disease for investigating the role of NR4A1 in modulating the effects of MPP+-induced inflammation (and oxidative stress) and induction of TNFα, MCP-1, IL-6, and NFkB. The loss of NR4A1 mimicked the effects of MPP+, and CsnB inhibited MPP+-induced inflammatory gene product formation and also decreased MPP+-induced oxidative stress in wild-type cells [39]. NR4A1 and CsnB also ameliorate induced inflammation in microglia and specifically protect dopaminergic neurons. A key mechanistic aspect associated with the NR4A1/CsnB-mediated response was due to inhibition of IKBα-phosphorylation [131]. In vivo studies in wild-type and NR4A1-KO mice showed that NR4A1 suppressed cytokine expression and NO production in microglia [132]. Moreover, the loss of NR4A1 enhanced experimental autoimmune encephalomyelitis in mice compared to wild-type mice, and in mice expressing NR4A1, treatment with CsnB inhibited the course of this disease model for multiple sclerosis. Carpenter and colleagues reported that in a mouse model, cocaine-activated expression of NR4A1 and several downstream genes including cocaine- and amphetamine-regulated transcript peptide (Cartpt) and mediated cocaine-induced behavior [133]. Moreover, CsnB activation of NR4A1 suppressed cocaine behavior, and it was suggested that NR4A1 ligands such as CsnB may be of therapeutic value for treating cocaine addiction.
Table 1. Effects of NR4A1 ligands on NR4A1-regulated responses and genes.
Table 1. Effects of NR4A1 ligands on NR4A1-regulated responses and genes.
Ligand [Ref]Tissue/Cell TypeResponse/Genes
CsnB [107]LiverPNLPLA3 I148M variant in human hepatic stellate cells increased dysfunction and decreased NR4A1 but this was increased by CsnB
CsnB [104]LiverNR4A1 inhibited TGFβ-induced fibrosis and CsnB enhanced inhibition (also skin, lung, kidney)
CsnB [99]LiverHomocysteine induced hepatic steatosis, which is blocked by CsnB
CsnB/6-MP [72]IntestineInduced smooth muscle cell phenotype enhanced in NR4A−/− mice; decreased by CsnB and 6-MP
CsnB/6-MP [73]IntestineTGFβ-induced fibrogenesis in myofibroblasts enhanced with loss of NR4A1: CsnB and 6-MP decreased response in wild-type cells
Csn [75]IntestineDSS-induced colitis inhibited by CsnB NR4A-TLR-1R via NR4A-TRAF6 interactions
GPApeptide [122]IntestineGPA inhibited NFkB activation and intestinal inflammation in DSS-induced colitis
CsnB [98]BladderNR4A1 protects against urinary tract infections and CsnB inhibits bacterial infection
CsnB [97]KidneyInduced unilateral ureteral obstruction in mouse UUO-induced kidney fibrosis and NR4A1 and CsnB enhanced the response
CsnB [94]KidneyNR4A1 is protective against UUO-induced fibrosis and CsnB is also protective
Pterostilbene [123]LungPterostilbene inhibited LPS-induced inflammation and damage in the rat lung in vivo
CsnB [93]LungNR4A1 downregulated in pulmonary arterial hypertension (PAH), CsnB inhibits symptoms in hypoxia-induced mouse model
CsnB [124]LungCsnB inhibits influenza-induced pulmonary damage and is associated with induction of type I interferon
6-MP [125]Lung6-MP inhibited progression of pulmonary hypertension and was associated with NR4A1 activation of bone morphogenic protein
CsnB [126]BoneNR4A1 elevated in osteoarthritis and CsnB inhibited IL-1β-induced inflammatory genes and decreased osteoarthritis in a rat model
CsnB [39]CNSCsnB inhibited MPP+-mediated inflammatory and oxidative stress genes in rat PC12 pheochromocytoma cells
CsnB [131]CNSCsnB inhibits inflammation/inflammatory genes and reverses MPTP-induced TH positive neurons and Iba-1 positive neurons
CsnB [132]CNS
(autoimmune)
NR4A1 suppressed cytokine production and NO in mouse microglial cells and NR4A1 alone and in combination inhibited the progress of AEA-induced disease in a model of multiple sclerosis
CsnB [133]CNSCocaine-induced NR4A1 and cartpl gene expression NR4A1/CsnB modify cocaine-induced behavior
CsnB [134]EndometrioticCsn inhibits TGFβ-induced fibrosis
DIM-4-OH and DIM-3-CI-4-OH-5-OCH3 [135]EndometrioticNR4A1 is pro-endometriotic and DIM compound inhibits mTOR, proliferation, and fibrosis
CsnB [136]CardiovascularLoss of NR4A1 in hypocholesterolemia enhances IL-6 and MCP-1 and CsnB inhibits cholesterol-induced IL-6 and MCP-1
CsnB [137]CardiovascularHypocholesterolemia induced platelet activation and thrombus inhibited by NR4A1 and CsnB further protects—due, in part, to cAMP phosphorylation of VASP
CsnB [138]CardiovascularNR4A1 decreases atherosclerotic responses and CsnB enhances their effects in mouse models of hypocholesterolemia
6-MP [139]CardiovascularNR4A1/6-MP protect from restenosis-induced neointima formation and inhibition of cell proliferation
Celastrol [140]CardiovascularDevelopment of carotid plaque in wild-type ApoE−/−/NR4A1-KO mice inhibited by NR4A1 and enhanced by celastrol by inhibiting bcat in macrophages
CsnB [141]CardiovascularCardiac allograft rejection in mice was decreased by NR4A1 and enhanced by CsnB by targeting by infiltrating CD4+ T cells and inducing Treg cell differentiation
CsnB [142]InflammationCsnB differentially inhibited LPS-induced NFkB in human macrophages
CsnB [143]InflammationCsnB inhibited NFkB-mediated inflammation in a mouse model of sepsis
CsnB [144]InflammationLy6Chigh monocytes contribute to a mouse model of arthritis and in the presence of Ly6Clow monocytes Csn inhibits progression of arthritis
AEA [145]
DIM-4-CI [145]
CsnB [145]
InflammationAEA is a dual NR4A1/2 ligand that inhibits IL-1β-induced cytokines (e.g., CCL2) in vascular smooth muscle cells, as do other ligands
TokinolideB [146]InflammationTokinolide induces NR4A1 nuclear export and inhibits inflammation in a mouse model of hepatitis through mitochondria autophagy pathways
Celastrol [147]InflammationInduces nuclear export of NR4A1, which interacts with TRAF2 to inhibit inflammatory signaling
6-MP [91]InflammationDecreased dendritic cell activation, decreased production of interferonƔ
CsnB [120]Metabolic disease /LiverCsnB elevated blood glucose levels and activated hepatic gluconeogenesis
TMPA [148]Metabolic disease /LiverTMPA decreased blood glucose levels and reversed insulin resistance
DIM-3,5 analogs [149]Metabolic disease (muscle)DIM-3,5 enhanced expression of GLUT4 and glycolytic genes and increased glucose uptake in muscle cells
CsnB [150]EyeInhibited subretinal fibrosis and macrophage-to-myoblast transition

4.4. Effects of NR4A1 Ligands on Endometriosis and Cardiovascular Damage

Endometriosis is also a highly inflammatory disease characterized by cell growth, migration, and fibrosis. Inhibition of NR4A1 expression in stromal cells increased TGFβ-induced fibrosis and in NR4A1-expressing cells CsnB inhibited fibrosis [134]. Similar results for NR4A1 were observed in epithelial and stromal endometriotic cells, where NR4A1 knockdown resulted in activation of mTOR signaling and induction of α-smooth muscle actin and related fibrotic genes. Moreover, treatment of these cells with bis-indole-derived NR4A1 ligands 1,1-bis(3′-indolyl)-1-(4-hydroxyphenyl)methane (DIM-4-OH) and the 3-chloro-4-hydroxy-5-methoxyphenyl analog (DIM-4-OH-3-CI-4-OCH3) inhibited cell proliferation, mTOR signaling, and fibrotic gene expression [135]. The results suggest that NR4A1 is a pro-endometriotic factor in endometriosis and NR4A1 ligands act as inverse agonists, and similar results have been observed in solid tumor-derived cells in culture and in vivo [14].
NR4A1 ligands play an important protective role in cardiovascular disease [95]. Higher expression of NR4A1 is observed in leukocytes from patients with hypocholesterolemia and in cell culture cholesterol induces NR4A1 and loss of the receptor enhances expression of inflammatory genes such as IL-6 and MCP-1 [136]. CsnB treatment inhibits expression of cholesterol-induced IL-6 and MCP-1. Using human NR4A1 expression data and platelets from patients with hypocholesterolemia and mouse models, it was demonstrated that loss of NR4A1 (in mice) enhanced thrombus formation, microvascular microthrombi obstruction, and platelet activation. In cells/tissue expressing NR4A1, CsnB inhibited activation of hypocholesterolemic platelets. The mechanisms of NR4A1/CsnB protective effects involved enhancement of cAMP levels and subsequent phosphorylation of vasodilator-stimulated phosphoprotein (VASP) [137]. NR4A1 also decreased atherosclerotic plaque formation in ApoE−/− mice maintained on a high-fat/high-cholesterol diet, and CsnB further enhanced the protective effects of NR4A1 in this model [138]. NR4A1 protects from restenosis, which can accompany coronary interventions and is associated with proliferation of smooth muscle cells and neointima formation [139] 6-MP also further inhibits neointima formation and this is accompanied by inhibition of PCNA, a marker of cell proliferation and induction of the cell cycle inhibitor p27Kip1 [139]. Using ApoE−/− and NR4A-KO mice, another study showed that carotid plaque formation was enhanced in NR4A1-deficient mice, and this was associated with enhanced macrophage-mediated inflammatory and oxidative responses [140]. These responses were ameliorated by NR4A1-dependent inhibition of Bcat1 expression. The NR4A1 ligand celastrol also stabilized formation of atherosclerotic plaques in mice. CsnB was also investigated for a possible role in modulating acute cardiac allograft rejection in a mouse model [141]. NR4A1 was primarily expressed in intragraft-infiltrating CD4+ T cells and associated with apoptosis, differentiation, and T cell dysfunction. In a mouse model, CsnB decreased the allograft rejection response by targeting Treg cells and CD4+ T cells.

4.5. Effects of NR4A1 Ligands on Inflammation, Metabolic Diseases, and the Eye

Induced inflammatory responses are intimately linked to many NR4A-regulated responses in many tissues, as outlined in Table 1, and a few additional examples include the following: NR4A1 was more highly expressed in human macrophage cells cultured with macrophage colony-stimulating factor (M-MDM) compared to culture with granulocyte/macrophage-colony stimulating factor (GM-MDM). CsnB suppressed LPS-induced inflammatory responses (e.g., TNF and IL-6) in GM-MDMs and this was associated with decreased NFkB nuclear uptake; however, increased levels of IL-10 were also observed [142]. In addition, CsnB also inhibited sepsis in a mouse model, and this was associated with inhibition of NFkB and downstream inflammatory genes [143]. Results from NR4A1-KO mice indicate that inflammatory Ly6Chigh monocytes contribute to a mouse model of arthritis; in NR4A wild-type mice, CsnB increases levels of CD4+, CD25+, and FOXP3+ Treg cells in the presence of Ly6Clow monocytes to inhibit progression of arthritis [144]. A recent report showed that the endocannabinoid anandamide AEA interacts with NR4A1 and NR4A2 and is a dual receptor ligand. AEA inhibited IL-1β-induced cytokines such as CCL2 in vascular smooth muscle cells [145]. Similar results were observed for CsnB and DIM-4-CI, which have been characterized as NR4A1 and NR4A2 ligands, respectively. TokinolideB is a phthalide isolated from Angelica sinensis that binds NR4A1 [146]; like celastrol [147], tokinolide induces nuclear export of NR4A1 and exhibits anti-inflammatory activity in a mouse model of hepatitis. The effects involve mitochondrial interactions with TRAFα to induce autophagy and this results in inhibition of LPS-induced inflammation. 6-MP decreased dendritic cell activation and inhibited interferonƔ production by allogeneic T cells [91].
Results of NR4A1 knockdown or overexpression demonstrate that this receptor plays a key role in protection from metabolic diseases; studies on the effects of NR4A1 ligands and their influence on diabetes and related responses are limited. CsnB induces blood glucose levels and activates hepatic gluconeogenes [120] whereas another CsnB analog, ethy l2-[2,3,4-trimethoxy-6-(1-octanoyl)phenyl] acetate (TMPA), decreased blood glucose levels and reversed insulin resistance [147]. Thus two structurally related compounds (CsnB/TMPA) exhibit inverse activities on metabolic disease, and it is possible that the prodiabetic activity of CsnB may be due to other modes of action, such as enhancement of NR4A1 nuclear export. Another study reported that DIM-3,5 analogs were antidiabetic and induced GLUT4, glucose uptake, and glycolytic genes in C2C12 muscle cells [148]. A recent study reported another application of CsnB where subretinal fibrosis was inhibited by NR4A1 [149]. Knockdown of NR4A1 promoted macrophage-to-myoblast transition (MMT) whereas CsnB inhibited this pathway and protected from subretinal fibrosis [150]. These results show that for most responses NR4A1 ligands enhance the health-promoting effects of NR4A1 in cell culture and animal models, as illustrated in Figure 4.

5. NR4A1 as Nutrient Sensor for Health-Promoting Polyphenolics

5.1. Introduction

It has previously been hypothesized that NR4A1 is protective against aging [151] and the age-dependent expression of NR4A1, and its interactions with key genes involved in aging support this hypothesis. There is also extensive evidence showing the protective effects of NR4A1 expression against multiple responses associated with premature aging, including inhibition of inflammation, enhanced immune cell functions, and decreased autoimmunity and damaged cells from multiple organs and tissues. Moreover, NR4A1 ligands such as CsnB enhance the health-promoting responses and genes regulated by NR4A1. These observations suggest that since NR4A1 is a receptor it is possible that one of its functions could be as a receptor for health-promoting dietary compounds such as the polyphenolics in fruits, nuts, and vegetables. These compounds include flavonoids, resveratrol, and other polyphenolics that are known to be associated with decreased mortality and lower incidence of cardiovascular, metabolic, and neurotoxicity diseases such as dementia and Alzheimer’s and Parkinson’s disease [5,6,7,8,152,153,154].

5.2. Flavonoids and Resveratrol Bind NR4A1 and Inhibit Endometriosis

The linkage between polyphenolics and NR4A1 and their role in promoting health was first demonstrated in a study showing that the flavonoids quercetin and kaempferol bind NR4A1 and exhibit NR4A1-dependent transactivation [155]. Moreover, a direct comparison of the flavonoids with CDIM NR4A1 ligands showed that they exhibited comparable activity as inhibitors of endometriotic cell growth in both in vitro and in vivo models [135]. For example in endometriotic cells, the CDIM compounds and the flavonoids (quercetin/kaempferol) decreased NR4A1-dependent transactivation (using a GAL4-NR4A1 construct), inhibited endometriotic cell proliferation, induced apoptosis, and decreased mTOR signaling and fibrotic genes, including α-smooth muscle actin (αSMA), fibronectin (FN), connective tissue growth factor (CTGF), and collagen type 1α (COL1A1) [155]. These responses were also observed after knockdown of NR4A1 by RNA interference, demonstrating that like the CDIM compounds, quercetin and kaempferol acted as inverse NR4A1 agonists in endometriotic cell lines. Similar results were observed for these compounds in Rh30 rhabdomyosarcoma cells, where the flavonoids inhibited growth and induced apoptosis [156]. Subsequent studies showed that in addition to quercetin and kaempferol, 18 hydroxylated flavones also bound NR4A1, and this included several individual compounds found in fruits, nuts, and vegetables, including galangin, chrysin, luteolin, apigenin, and baicalein. KD binding values were variable but most of the compounds noted above exhibited KD’s < 10 µM, with the lowest values (0.36 µM) observed for galangin [157].

5.3. Quercetin Effects Mimic Those of Synthetic NR4A1 Ligands

The health-promoting effects of flavonoids have been extensively investigated and the number of citations (PubMed) for quercetin alone is >31,000. A comparison shows that the effects observed for quercetin mimic those reported for other NR4A1 ligands, suggesting a role for NR4A1 in mediating quercetin-induced responses (Figure 5). For example, numerous studies show that quercetin inhibits induced hepatic fibrosis and hepatic stellate cell activation [158,159,160,161,162] by modulating many of the same responses observed for NR4A1 ligands, and the effects of quercetin on kidney fibrosis [163,164,165] were also similar to those observed for NR4A1/ligands (Table 1). Quercetin also inhibits cyclophosphamide-induced urotoxicity and expression of inflammatory genes [166], and several studies reported antibacterial effects of quercetin [167]. Quercetin also inhibits induced neurotoxicity and its associated inflammation [168,169] and also lung damage by inhibiting fibrosis, oxidative stress, and inflammation and there is also evidence of a role for NRF2 [170,171,172,173]. Quercetin inhibits mouse models of intestinal inflammation [174]; for example, dietary quercetin inhibited DSS-induced intestinal damage by multiple pathways, including improve colonic permeability and inhibition of IL-6, TGFβ, and other inflammatory markers [175]. There are extensive studies on the protective effects of quercetin on most types of cardiovascular damage and diseases [176,177,178]. For example, quercetin inhibited LPS-induced sepsis through inhibition of multiple pro-inflammatory cytokines and NFkB [179]. Like NR4A1 and its ligands, quercetin also exhibited antidiabetic activity [180]. Quercetin induced GLUT4 and uptake of glucose in C2CI2 muscle cells and inhibited gluconeogenesis in liver cancer cells [181] and inhibited oxidative damage and potentiated insulin secretion in pancreatic β cells [182]. Quercetin inhibited inflammatory markers, enhanced M2 polarization in macrophages [183,184], and inhibited LPS-induced activation of dendritic cells, which was accompanied by inhibition of pro-inflammatory cytokines and chemokines [185]. These results (Figure 5) demonstrate that quercetin, an NR4A1 ligand, exhibits anti-inflammatory activities similar to those observed for CsnB (Table 1) and other synthetic NR4A1 ligands. Moreover, the same holds true for other flavonoids that bind NR4A1. A recent study showed that the flavonoid baicalein activated NR4A1 to inhibit ovalbumin-induced allergic rhinitis in mice [186] and it is likely that this response was also due, in part, to baicalein acting as an NR4A1 ligand [153]. It is also likely that NR4A1 plays a role in mediating some of the health-protective responses associated with quercetin, kaempferol, baicalein, and related compounds that bind NR4A1 [153].

5.4. Resveratrol Exhibits NR4A1 Ligand-like Activities

Many other health-promoting phenolics and their interactions with NR4A1 have not been investigated; however, a recent study showed that resveratrol, a polyphenolic found in grapes, red wine, peanuts, and chocolate also bound NR4A1 and inhibited NR4A1-mediated pro-oncogenic pathways in lung cancer cells [187]. Resveratrol, like quercetin and other polyphenolics, has been linked to multiple health benefits in humans [188]. Resveratrol induces many of the same responses observed for NR4A1 and its ligands in cell culture and animal models (Table 1) and is also associated with anti-aging effects [189,190]. For example, resveratrol inhibited intestinal fibrosis and intestinal barrier dysfunction [191,192], inhibited liver and kidney fibrosis [193,194,195,196], exhibited therapeutic value for treating diabetes [197,198], inhibited cardiovascular damage [199,200], protected against neurotoxicity [201,202], inhibited urinary tract infections and bladder damage [203,204,205,206], reversed lung damage [207,208], and modulated immune cell functions [209,210,211]. Moreover, resveratrol, quercetin, and other flavonoids have been extensively used in clinical trials to treat some of these health problems. Studies in humans demonstrate an association between consumption of polyphenolics and some protection against aging-related diseases in humans and this has also been demonstrated in cell culture and animal models [5,6,7,8,212,213,214]. NR4A1 and its ligands have been extensively characterized for their protective effects against stressors/inflammation-induced tissue damage. Based on the similarities observed for the protective effects of NR4A1 ligands (Table 1) and the newly characterized polyphenolic NR4A1 ligands (flavonoids and resveratrol), it is possible that NR4A1 may also act as a nutrient sensor and mediate some of effects of polyphenolics as “geroprotectors” [214].

6. Conclusions

This review demonstrates that the orphan nuclear receptor NR4A1 protects against aging and interacts with other gene products involved in the aging process. Results of NR4A1-KO mouse and cell culture studies show that NR4A1 protects against tissue damage, including inflammation, fibrosis, and barrier dysfunction, and this is consistent with initial studies on NR4A1 and other members of this subfamily as genes that are induced in response to stressors and inflammatory agents to ameliorate their effects [18,19]. The functional effects of synthetic and diet-derived polyphenolic NR4A1 ligands are similar; however their genomic pathways show some differences, which are not surprising for compounds that act through nuclear receptors. Nuclear receptor ligands exhibit differences due to several factors, including their unique structure-dependent interactions with receptors, which is accompanied by conformational differences in ligand–receptor structures. This suggests that NR4A1 ligands, synthetic or natural act, as selective NR4A1 modulators and this is consistent with some of the differences in their tissue-specific activities. Thus the synthetic NR4A1 ligands enhance the protective effects of NR4A1 and the discovery that flavonoids and resveratrol are also NR4A1 ligands suggests that the anti-aging and health benefits of these polyphenolics may be due, in part, to their activities as NR4A1 ligands. The linkage between polyphenolics and NR4A1 as a nutrient sensor for this class of “anti-aging” compounds is based primarily on a few studies. Further confirmation is required to delineate the contributions of NR4A1 in mediating the health-protective effects of dietary phenolics and other compounds and for the development of dietary supplements that will enhance these effects.

Funding

This research was funded by the Syd Kyle Chair endowment and the support from Texas A&M AgriLife Institute for Advancing Health Through Agriculture.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article. Material used in this review was obtained primarily from PubMed searches.

Acknowledgments

We would like to acknowledge Amber N. Meyer for her administrative and technical support as well as her writing and proof-reading assistance and the overall handling of the manuscript and the figures created by her. Permission has been obtained to acknowledge the aforementioned individual.

Conflicts of Interest

The author declares no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AEAAnandamide
BVPBasal vascular permeability
CDIMsBis-indole compounds
CsnBCytosporoneB
CVHChronic vascular hyperpermeability
DIM-4-OH1,1-bis(3′-indolyl)-1-(4-hydroxyphenyl)methane
DIM-4-OH-3-Cl-5-OCH31,1-bis(3′-indolyl)-1-(3-chloro-4-hdroxy-5-methoxphenyl)methane
DSBsDouble-strand breaks
ECMExtracellular matrix
GPAGly-pro-ala
LPSLipopolysaccharide
MMTMacrophagetomyoblast transition
6-MP6-mexaptopurine
NR4ANuclear receptor 4A
PAHPulmonary arterial hypertension
PASMCsPulmonary artery smooth muscle cells
TMPAEthyl 2-[2,3,4-trimethoxy-6-(octanoyl)phenyl] acetate
UPECUropathogenic Escherichia coli
UTIUrinary tract infection
UUOUnilateral ureteral obstruction

References

  1. Ferrucci, L.; Gonzalez-Freire, M.; Fabbri, E.; Simonsick, E.; Tanaka, T.; Moore, Z.; Salimi, S.; Sierra, F.; de Cabo, R. Measuring biological aging in humans: A quest. Aging Cell 2020, 19, e13080. [Google Scholar] [CrossRef]
  2. Kareem, M.A.; Ashwini, A.; Sunil, T. The Role of the Exposome in Aging and Age-Related Diseases: A Comprehensive Review. J. Pharm. Bioallied Sci. 2025, 17 (Suppl. 1), S2–S5. [Google Scholar] [CrossRef]
  3. Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef] [PubMed]
  4. Childs, B.G.; Gluscevic, M.; Baker, D.J.; Laberge, R.M.; Marquess, D.; Dananberg, J.; van Deursen, J.M. Senescent cells: An emerging target for diseases of ageing. Nat. Rev. Drug Discov. 2017, 16, 718–735. [Google Scholar] [CrossRef]
  5. Davinelli, S.; Medoro, A.; Hu, F.B.; Scapagnini, G. Dietary polyphenols as geroprotective compounds: From Blue Zones to hallmarks of ageing. Ageing Res. Rev. 2025, 108, 102733. [Google Scholar] [CrossRef] [PubMed]
  6. Liu, Y.; Fang, M.; Tu, X.; Mo, X.; Zhang, L.; Yang, B.; Wang, F.; Kim, Y.B.; Huang, C.; Chen, L.; et al. Dietary Polyphenols as Anti-Aging Agents: Targeting the Hallmarks of Aging. Nutrients 2024, 16, 3305. [Google Scholar] [CrossRef] [PubMed]
  7. Wu, W.; Mi, Y.; Meng, Q.; Li, N.; Li, W.; Wang, P.; Hou, Y. Natural polyphenols as novel interventions for aging and age-related diseases: Exploring efficacy, mechanisms of action and implications for future research. Chin. Herb. Med. 2024, 17, 279–291. [Google Scholar] [CrossRef]
  8. Ristori, S.; Bertoni, G.; Bientinesi, E.; Monti, D. The Role of Nutraceuticals and Functional Foods in Mitigating Cellular Senescence and Its Related Aspects: A Key Strategy for Delaying or Preventing Aging and Neurodegenerative Disorders. Nutrients 2025, 17, 1837. [Google Scholar] [CrossRef]
  9. Min, M.; Egli, C.; Dulai, A.S.; Sivamani, R.K. Critical review of aging clocks and factors that may influence the pace of aging. Front. Aging 2024, 5, 1487260. [Google Scholar] [CrossRef]
  10. Liu, Y.; Weng, W.; Gao, R.; Liu, Y. New Insights for Cellular and Molecular Mechanisms of Aging and Aging-Related Diseases: Herbal Medicine as Potential Therapeutic Approach. Oxid. Med. Cell Longev. 2019, 2019, 4598167. [Google Scholar] [CrossRef]
  11. Burris, T.P.; de Vera, I.M.S.; Cote, I.; Flaveny, C.A.; Wanninayake, U.S.; Chatterjee, A.; Walker, J.K.; Steinauer, N.; Zhang, J.; Coons, L.A.; et al. International Union of Basic and Clinical Pharmacology CXIII: Nuclear Receptor Superfamily-Update 2023. Pharmacol. Rev. 2023, 75, 1233–1318. [Google Scholar] [CrossRef]
  12. Maxwell, M.A.; Muscat, G.E. The NR4A subgroup: Immediate early response genes with pleiotropic physiological roles. Nucl. Recept. Signal. 2006, 4, e002. [Google Scholar] [CrossRef]
  13. Shi, Y. Orphan nuclear receptors in drug discovery. Drug Discov. Today 2007, 12, 440–445. [Google Scholar] [CrossRef]
  14. Safe, S.; Karki, K. The Paradoxical Roles of Orphan Nuclear Receptor 4A (NR4A) in Cancer. Mol. Cancer Res. 2021, 19, 180–191. [Google Scholar] [CrossRef]
  15. Perlmann, T.; Jansson, L. A novel pathway for vitamin A signaling mediated by RXR heterodimerization with NGFI-B and NURR1. Genes. Dev. 1995, 9, 769–782. [Google Scholar] [CrossRef]
  16. Zetterström, R.H.; Solomin, L.; Mitsiadis, T.; Olson, L.; Perlmann, T. Retinoid X receptor heterodimerization and developmental expression distinguish the orphan nuclear receptors NGFI-B, Nurr1, and Nor1. Mol. Endocrinol. 1996, 10, 1656–1666. [Google Scholar] [CrossRef]
  17. Jiang, L.; Dai, S.; Li, J.; Liang, X.; Qu, L.; Chen, X.; Guo, M.; Chen, Z.; Chen, L.; Wei, H.; et al. Structural basis of binding of homodimers of the nuclear receptor NR4A2 to selective Nur-responsive DNA elements. J. Biol. Chem. 2019, 294, 19795–19803. [Google Scholar] [CrossRef]
  18. Pearen, M.A.; Muscat, G.E. Minireview: Nuclear hormone receptor 4A signaling: Implications for metabolic disease. Mol. Endocrinol. 2010, 24, 1891–1903. [Google Scholar] [CrossRef] [PubMed]
  19. Murphy, E.P.; Crean, D. Molecular Interactions between NR4A Orphan Nuclear Receptors and NF-κB Are Required for Appropriate Inflammatory Responses and Immune Cell Homeostasis. Biomolecules 2015, 5, 1302–1318. [Google Scholar] [CrossRef] [PubMed]
  20. Lith, S.C.; de Vries, C.J.M. Nuclear receptor Nur77: Its role in chronic inflammatory diseases. Essays Biochem. 2021, 65, 927–939. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, Y.; Li, N.; Guan, W.; Wang, D. Controversy and multiple roles of the solitary nucleus receptor Nur77 in disease and physiology. FASEB J. 2025, 39, e70468. [Google Scholar] [CrossRef]
  22. Hashida, R.; Kawabata, T. Structural Perspective of NR4A Nuclear Receptor Family and Their Potential Endogenous Ligands. Biol. Pharm. Bull. 2024, 47, 580–590. [Google Scholar] [CrossRef] [PubMed]
  23. Wu, L.; Chen, L. Characteristics of Nur77 and its ligands as potential anticancer compounds (Review). Mol. Med. Rep. 2018, 18, 4793–4801. [Google Scholar] [CrossRef] [PubMed]
  24. Safe, S. Natural products and synthetic analogs as selective orphan nuclear receptor 4A (NR4A) modulators. Histol. Histopathol. 2024, 39, 543–556. [Google Scholar] [CrossRef] [PubMed]
  25. Upadhyay, S.; Hailemariam, A.E.; Mariyam, F.; Hafiz, Z.; Martin, G.; Kothari, J.; Farkas, E.; Sivaram, G.; Bell, L.; Tjalkens, R.; et al. Bis-Indole Derivatives as Dual Nuclear Receptor 4A1 (NR4A1) and NR4A2 Ligands. Biomolecules 2024, 14, 284. [Google Scholar] [CrossRef]
  26. Chen, J.; Zhang, Z.; Liu, Y.; Huang, L.; Liu, Y.; Yang, D.; Bao, X.; Liu, P.; Ge, Y.; Li, Q.; et al. Progressive reduction of nuclear receptor Nr4a1 mediates age-dependent cognitive decline. Alzheimers Dement. 2024, 20, 3504–3524. [Google Scholar] [CrossRef]
  27. Ma, G.; Chen, F.; Liu, Y.; Zheng, L.; Jiang, X.; Tian, H.; Wang, X.; Song, X.; Yu, Y.; Wang, D. Nur77 ameliorates age-related renal tubulointerstitial fibrosis by suppressing the TGF-β/Smads signaling pathway. FASEB J. 2022, 36, e22124. [Google Scholar] [CrossRef]
  28. Yu, Y.; Song, X.; Wang, X.; Zheng, L.; Ma, G.; Liu, W.; Su, H.; Liu, X.; Liu, T.; Cao, L.; et al. Oxidative stress impairs the Nur77-SIRT1 axis resulting in a decline in organism homeostasis during aging. Aging Cell 2023, 22, e13812. [Google Scholar] [CrossRef]
  29. Zhang, T.; Ma, R.; Li, Z.; Liu, T.; Yang, S.; Li, N.; Wang, D. Nur77 alleviates cardiac fibrosis by upregulating GSK-3β transcription during aging. Eur. J. Pharmacol. 2024, 965, 176290. [Google Scholar] [CrossRef]
  30. Sundaresan, N.R.; Bindu, S.; Pillai, V.B.; Samant, S.; Pan, Y.; Huang, J.Y.; Gupta, M.; Nagalingam, R.S.; Wolfgeher, D.; Verdin, E.; et al. SIRT3 Blocks Aging-Associated Tissue Fibrosis in Mice by Deacetylating and Activating Glycogen Synthase Kinase 3β. Mol. Cell Biol. 2015, 36, 678–692. [Google Scholar] [CrossRef]
  31. Cortez-Toledo, O.; Schnair, C.; Sangngern, P.; Metzger, D.; Chao, L.C. Nur77 deletion impairs muscle growth during developmental myogenesis and muscle regeneration in mice. PLoS ONE 2017, 12, e0171268. [Google Scholar] [CrossRef]
  32. Yao, Y.; Wang, B.; Yu, K.; Song, J.; Wang, L.; Zhang, X.; Li, Y. Nur77 improves ovarian function in reproductive aging mice by activating mitophagy and inhibiting apoptosis. Reprod. Biol. Endocrinol. 2024, 22, 86. [Google Scholar] [CrossRef] [PubMed]
  33. Cui, L.; Bao, H.; Zhu, W.; Tang, Y.; Luo, Q.; Si, Y.; Fu, Q.; Jiang, Z. hUMSCs Transplantation Regulates AMPK/NR4A1 Signaling Axis to Inhibit Ovarian Fibrosis in POI Rats. Stem Cell Rev. Rep. 2023, 19, 1449–1465. [Google Scholar] [CrossRef] [PubMed]
  34. Luo, Q.; Tang, Y.; Jiang, Z.; Bao, H.; Fu, Q.; Zhang, H. hUCMSCs reduce theca interstitial cells apoptosis and restore ovarian function in premature ovarian insufficiency rats through regulating NR4A1-mediated mitochondrial mechanisms. Reprod. Biol. Endocrinol. 2022, 20, 125. [Google Scholar] [CrossRef]
  35. Malewicz, M.; Kadkhodaei, B.; Kee, N.; Volakakis, N.; Hellman, U.; Viktorsson, K.; Leung, C.Y.; Chen, B.; Lewensohn, R.; van Gent, D.C.; et al. Essential role for DNA-PK-mediated phosphorylation of NR4A nuclear orphan receptors in DNA double-strand break repair. Genes. Dev. 2011, 25, 2031–2040. [Google Scholar] [CrossRef] [PubMed]
  36. Munnur, D.; Somers, J.; Skalka, G.; Weston, R.; Jukes-Jones, R.; Bhogadia, M.; Dominguez, C.; Cain, K.; Ahel, I.; Malewicz, M. NR4A Nuclear Receptors Target Poly-ADP-Ribosylated DNA-PKcs Protein to Promote DNA Repair. Cell Rep. 2019, 26, 2028–2036.e6. [Google Scholar] [CrossRef]
  37. Wang, R.H.; He, J.P.; Su, M.L.; Luo, J.; Xu, M.; Du, X.D.; Chen, H.Z.; Wang, W.J.; Wang, Y.; Zhang, N.; et al. The orphan receptor TR3 participates in angiotensin II-induced cardiac hypertrophy by controlling mTOR signalling. EMBO Mol. Med. 2013, 5, 137–148. [Google Scholar] [CrossRef]
  38. Tontonoz, P.; Cortez-Toledo, O.; Wroblewski, K.; Hong, C.; Lim, L.; Carranza, R.; Conneely, O.; Metzger, D.; Chao, L.C. The orphan nuclear receptor Nur77 is a determinant of myofiber size and muscle mass in mice. Mol. Cell Biol. 2015, 35, 1125–1138. [Google Scholar] [CrossRef]
  39. Yan, J.; Huang, J.; Wu, J.; Fan, H.; Liu, A.; Qiao, L.; Shen, M.; Lai, X. Nur77 attenuates inflammatory responses and oxidative stress by inhibiting phosphorylated IκB-α in Parkinson’s disease cell model. Aging 2020, 12, 8107–8119. [Google Scholar] [CrossRef]
  40. Liang, C.Q.; Zhou, D.C.; Peng, W.T.; Chen, W.Y.; Wu, H.Y.; Zhou, Y.M.; Gu, W.L.; Park, K.S.; Zhao, H.; Pi, L.Q.; et al. FoxO3 restricts liver regeneration by suppressing the proliferation of hepatocytes. npj Regen. Med. 2022, 7, 33. [Google Scholar] [CrossRef]
  41. Liu, D.; Jia, H.; Holmes, D.I.; Stannard, A.; Zachary, I. Vascular endothelial growth factor-regulated gene expression in endothelial cells: KDR-mediated induction of Egr3 and the related nuclear receptors Nur77, Nurr1, and Nor1. Arter. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 2002–2007. [Google Scholar] [CrossRef]
  42. Zhao, D.; Qin, L.; Bourbon, P.M.; James, L.; Dvorak, H.F.; Zeng, H. Orphan nuclear transcription factor TR3/Nur77 regulates microvessel permeability by targeting endothelial nitric oxide synthase and destabilizing endothelial junctions. Proc. Natl. Acad. Sci. USA 2011, 108, 12066–12071. [Google Scholar] [CrossRef] [PubMed]
  43. Qin, L.; Zhao, D.; Xu, J.; Ren, X.; Terwilliger, E.F.; Parangi, S.; Lawler, J.; Dvorak, H.F.; Zeng, H. The vascular permeabilizing factors histamine and serotonin induce angiogenesis through TR3/Nur77 and subsequently truncate it through thrombospondin-1. Blood 2013, 121, 2154–2164. [Google Scholar] [CrossRef]
  44. Medzikovic, L.; Schumacher, C.A.; Verkerk, A.O.; van Deel, E.D.; Wolswinkel, R.; van der Made, I.; Bleeker, N.; Cakici, D.; van den Hoogenhof, M.M.; Meggouh, F.; et al. Orphan nuclear receptor Nur77 affects cardiomyocyte calcium homeostasis and adverse cardiac remodelling. Sci. Rep. 2015, 5, 15404. [Google Scholar] [CrossRef] [PubMed]
  45. Medzikovic, L.; van Roomen, C.; Baartscheer, A.; van Loenen, P.B.; de Vos, J.; Bakker, E.N.T.P.; Koenis, D.S.; Damanafshan, A.; Creemers, E.E.; Arkenbout, E.K.; et al. Nur77 protects against adverse cardiac remodelling by limiting neuropeptide Y signalling in the sympathoadrenal-cardiac axis. Cardiovasc. Res. 2018, 114, 1617–1628. [Google Scholar] [CrossRef] [PubMed]
  46. Hilgendorf, I.; Gerhardt, L.M.; Tan, T.C.; Winter, C.; Holderried, T.A.; Chousterman, B.G.; Iwamoto, Y.; Liao, R.; Zirlik, A.; Scherer-Crosbie, M.; et al. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ. Res. 2014, 114, 1611–1622. [Google Scholar] [CrossRef]
  47. Hanna, R.N.; Shaked, I.; Hubbeling, H.G.; Punt, J.A.; Wu, R.; Herrley, E.; Zaugg, C.; Pei, H.; Geissmann, F.; Ley, K.; et al. NR4A1 (Nur77) deletion polarizes macrophages toward an inflammatory phenotype and increases atherosclerosis. Circ. Res. 2012, 110, 416–427. [Google Scholar] [CrossRef]
  48. Hanna, R.N.; Carlin, L.M.; Hubbeling, H.G.; Nackiewicz, D.; Green, A.M.; Punt, J.A.; Geissmann, F.; Hedrick, C.C. The transcription factor NR4A1 (Nur77) controls bone marrow differentiation and the survival of Ly6C- monocytes. Nat. Immunol. 2011, 12, 778–785. [Google Scholar] [CrossRef]
  49. Hamers, A.A.; Hanna, R.N.; Nowyhed, H.; Hedrick, C.C.; de Vries, C.J. NR4A nuclear receptors in immunity and atherosclerosis. Curr. Opin. Lipidol. 2013, 24, 381–385. [Google Scholar] [CrossRef]
  50. Hawk, J.D.; Bookout, A.L.; Poplawski, S.G.; Bridi, M.; Rao, A.J.; Sulewski, M.E.; Kroener, B.T.; Manglesdorf, D.J.; Abel, T. NR4A nuclear receptors support memory enhancement by histone deacetylase inhibitors. J. Clin. Invest. 2012, 122, 3593–3602. [Google Scholar] [CrossRef]
  51. Bridi, M.S.; Abel, T. The NR4A orphan nuclear receptors mediate transcription-dependent hippocampal synaptic plasticity. Neurobiol. Learn. Mem. 2013, 105, 151–158. [Google Scholar] [CrossRef]
  52. Chatterjee, S.; Walsh, E.N.; Yan, A.L.; Giese, K.P.; Safe, S.; Abel, T. Pharmacological activation of Nr4a rescues age-associated memory decline. Neurobiol. Aging 2020, 85, 140–144. [Google Scholar] [CrossRef] [PubMed]
  53. Chatterjee, S.; Bahl, E.; Mukherjee, U.; Walsh, E.N.; Shetty, M.S.; Yan, A.L.; Vanrobaeys, Y.; Lederman, J.D.; Giese, K.P.; Michaelson, J.; et al. Endoplasmic reticulum chaperone genes encode effectors of long-term memory. Sci. Adv. 2022, 8, eabm6063. [Google Scholar] [CrossRef]
  54. Huang, M.; Pieraut, S.; Cao, J.; de Souza Polli, F.; Roncace, V.; Shen, G.; Ramos-Medina, C.; Lee, H.; Maximov, A. Nr4a1 regulates cell-specific transcriptional programs in inhibitory GABAergic interneurons. Neuron 2024, 112, 2031–2044.e7. [Google Scholar] [CrossRef]
  55. McNulty, S.E.; Barrett, R.M.; Vogel-Ciernia, A.; Malvaez, M.; Hernandez, N.; Davatolhagh, M.F.; Matheos, D.P.; Schiffman, A.; Wood, M.A. Differential roles for Nr4a1 and Nr4a2 in object location vs. object recognition long-term memory. Learn. Mem. 2012, 19, 588–592. [Google Scholar] [CrossRef]
  56. Liu, P.; Chen, Y.; Zhang, Z.; Yuan, Z.; Sun, J.G.; Xia, S.; Cao, X.; Chen, J.; Zhang, C.J.; Chen, Y.; et al. Noncanonical contribution of microglial transcription factor NR4A1 to post-stroke recovery through TNF mRNA destabilization. PLoS Biol. 2023, 21, e3002199. [Google Scholar] [CrossRef] [PubMed]
  57. Nagai, S.; Ikeda, K.; Horie-Inoue, K.; Takeda, S.; Inoue, S. Estrogen signaling increases nuclear receptor subfamily 4 group A member 1 expression and energy production in skeletal muscle cells. Endocr. J. 2018, 65, 1209–1218. [Google Scholar] [CrossRef]
  58. Liu, Y.; Liu, N.; Yu, Y.; Wang, D. Nr4a1 promotes cell adhesion and fusion by regulating ZEB1 transcript levels in myoblasts. Biochem. Biophys. Res. Commun. 2021, 556, 127–133. [Google Scholar] [CrossRef] [PubMed]
  59. Kunst, S.; Wolloscheck, T.; Kelleher, D.K.; Wolfrum, U.; Sargsyan, S.A.; Iuvone, P.M.; Baba, K.; Tosini, G.; Spessert, R. Pgc-1α and Nr4a1 Are Target Genes of Circadian Melatonin and Dopamine Release in Murine Retina. Invest. Ophthalmol. Vis. Sci. 2015, 56, 6084–6094. [Google Scholar] [CrossRef]
  60. Tessem, J.S.; Moss, L.G.; Chao, L.C.; Arlotto, M.; Lu, D.; Jensen, M.V.; Stephens, S.B.; Tontonoz, P.; Hohmeier, H.E.; Newgard, C.B. Nkx6.1 regulates islet β-cell proliferation via Nr4a1 and Nr4a3 nuclear receptors. Proc. Natl. Acad. Sci. USA 2014, 111, 5242–5247. [Google Scholar] [CrossRef]
  61. Reynolds, M.S.; Hancock, C.R.; Ray, J.D.; Kener, K.B.; Draney, C.; Garland, K.; Hardman, J.; Bikman, B.T.; Tessem, J.S. β-Cell deletion of Nr4a1 and Nr4a3 nuclear receptors impedes mitochondrial respiration and insulin secretion. Am. J. Physiol. Endocrinol. Metab. 2016, 311, E186–E201. [Google Scholar] [CrossRef]
  62. Chao, L.C.; Wroblewski, K.; Ilkayeva, O.R.; Stevens, R.D.; Bain, J.; Meyer, G.A.; Schenk, S.; Martinez, L.; Vergnes, L.; Narkar, V.A.; et al. Skeletal muscle Nur77 expression enhances oxidative metabolism and substrate utilization. J. Lipid Res. 2012, 53, 2610–2619. [Google Scholar] [CrossRef]
  63. Chao, L.C.; Wroblewski, K.; Zhang, Z.; Pei, L.; Vergnes, L.; Ilkayeva, O.R.; Ding, S.Y.; Reue, K.; Watt, M.J.; Newgard, C.B.; et al. Insulin resistance and altered systemic glucose metabolism in mice lacking Nur77. Diabetes 2009, 58, 2788–2796. [Google Scholar] [CrossRef]
  64. Chao, L.C.; Zhang, Z.; Pei, L.; Saito, T.; Tontonoz, P.; Pilch, P.F. Nur77 coordinately regulates expression of genes linked to glucose metabolism in skeletal muscle. Mol. Endocrinol. 2007, 21, 2152–2163. [Google Scholar] [CrossRef] [PubMed]
  65. Maxwell, M.A.; Cleasby, M.E.; Harding, A.; Stark, A.; Cooney, G.J.; Muscat, G.E. Nur77 regulates lipolysis in skeletal muscle cells. Evidence for cross-talk between the beta-adrenergic and an orphan nuclear hormone receptor pathway. J. Biol. Chem. 2005, 280, 12573–12584. [Google Scholar] [CrossRef] [PubMed]
  66. Kanzleiter, T.; Preston, E.; Wilks, D.; Ho, B.; Benrick, A.; Reznick, J.; Heilbronn, L.K.; Turner, N.; Cooney, G.J. Overexpression of the orphan receptor Nur77 alters glucose metabolism in rat muscle cells and rat muscle in vivo. Diabetologia 2010, 53, 1174–1183. [Google Scholar] [CrossRef] [PubMed]
  67. Pei, L.; Waki, H.; Vaitheesvaran, B.; Wilpitz, D.C.; Kurland, I.J.; Tontonoz, P. NR4A orphan nuclear receptors are transcriptional regulators of hepatic glucose metabolism. Nat. Med. 2006, 12, 1048–1055. [Google Scholar] [CrossRef]
  68. Chao, L.C.; Bensinger, S.J.; Villanueva, C.J.; Wroblewski, K.; Tontonoz, P. Inhibition of adipocyte differentiation by Nur77, Nurr1, and Nor1. Mol. Endocrinol. 2008, 22, 2596–2608. [Google Scholar] [CrossRef]
  69. Duszka, K.; Bogner-Strauss, J.G.; Hackl, H.; Rieder, D.; Neuhold, C.; Prokesch, A.; Trajanoski, Z.; Krogsdam, A.M. Nr4a1 is required for fasting-induced down-regulation of Pparγ2 in white adipose tissue. Mol. Endocrinol. 2013, 27, 135–149. [Google Scholar] [CrossRef]
  70. Chen, Y.; Wu, R.; Chen, H.Z.; Xiao, Q.; Wang, W.J.; He, J.P.; Li, X.X.; Yu, X.W.; Li, L.; Wang, P.; et al. Enhancement of hypothalamic STAT3 acetylation by nuclear receptor Nur77 dictates leptin sensitivity. Diabetes 2015, 64, 2069–2081. [Google Scholar] [CrossRef]
  71. Perez-Sieira, S.; Martinez, G.; Porteiro, B.; Lopez, M.; Vidal, A.; Nogueiras, R.; Dieguez, C. Female Nur77-deficient mice show increased susceptibility to diet-induced obesity. PLoS ONE 2013, 8, e53836. [Google Scholar] [CrossRef]
  72. Szczepanski, H.E.; Flannigan, K.L.; Mainoli, B.; Alston, L.; Baruta, G.M.; Lee, J.W.; Venu, V.K.P.; Shearer, J.; Dufour, A.; Hirota, S.A. NR4A1 modulates intestinal smooth muscle cell phenotype and dampens inflammation-associated intestinal remodeling. FASEB J. 2022, 36, e22609. [Google Scholar] [CrossRef]
  73. Pulakazhi Venu, V.K.; Alston, L.; Iftinca, M.; Tsai, Y.C.; Stephens, M.; Warriyar, K.V.V.; Rehal, S.; Hudson, G.; Szczepanski, H.; von der Weid, P.Y.; et al. Nr4A1 modulates inflammation-associated intestinal fibrosis and dampens fibrogenic signaling in myofibroblasts. Am. J. Physiol. Gastrointest. Liver Physiol. 2021, 321, G280–G297. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, H.; Li, X.M.; Wang, J.R.; Gan, W.J.; Jiang, F.Q.; Liu, Y.; Zhang, X.D.; He, X.S.; Zhao, Y.Y.; Lu, X.X.; et al. NUR77 exerts a protective effect against inflammatory bowel disease by negatively regulating the TRAF6/TLR-IL-1R signalling axis. J. Pathol. 2016, 238, 457–469. [Google Scholar] [CrossRef]
  75. Zeng, H.; Qin, L.; Zhao, D.; Tan, X.; Manseau, E.J.; Van Hoang, M.; Senger, D.R.; Brown, L.F.; Nagy, J.A.; Dvorak, H.F. Orphan nuclear receptor TR3/Nur77 regulates VEGF-A-induced angiogenesis through its transcriptional activity. J. Exp. Med. 2006, 203, 719–729. [Google Scholar] [CrossRef]
  76. Niu, G.; Ye, T.; Qin, L.; Bourbon, P.M.; Chang, C.; Zhao, S.; Li, Y.; Zhou, L.; Cui, P.; Rabinovitz, I.; et al. Orphan nuclear receptor TR3/Nur77 improves wound healing by upregulating the expression of integrin β4. FASEB J. 2015, 29, 131–140. [Google Scholar] [CrossRef]
  77. Ye, T.; Peng, J.; Liu, X.; Hou, S.; Niu, G.; Li, Y.; Zeng, H.; Zhao, D. Orphan nuclear receptor TR3/Nur77 differentially regulates the expression of integrins in angiogenesis. Microvasc. Res. 2019, 122, 22–33. [Google Scholar] [CrossRef] [PubMed]
  78. Hamers, A.A.; Argmann, C.; Moerland, P.D.; Koenis, D.S.; Marinković, G.; Sokolović, M.; de Vos, A.F.; de Vries, C.J.; van Tiel, C.M. Nur77-deficiency in bone marrow-derived macrophages modulates inflammatory responses, extracellular matrix homeostasis, phagocytosis and tolerance. BMC Genom. 2016, 17, 162. [Google Scholar] [CrossRef] [PubMed]
  79. Koenis, D.S.; Medzikovic, L.; Vos, M.; Beldman, T.J.; van Loenen, P.B.; van Tiel, C.M.; Hamers, A.A.J.; Otermin Rubio, I.; de Waard, V.; de Vries, C.J.M. Nur77 variants solely comprising the amino-terminal domain activate hypoxia-inducible factor-1α and affect bone marrow homeostasis in mice and humans. J. Biol. Chem. 2018, 293, 15070–15083. [Google Scholar] [CrossRef]
  80. Liebmann, M.; Hucke, S.; Koch, K.; Eschborn, M.; Ghelman, J.; Chasan, A.I.; Glander, S.; Schädlich, M.; Kuhlencord, M.; Daber, N.M.; et al. Nur77 serves as a molecular brake of the metabolic switch during T cell activation to restrict autoimmunity. Proc. Natl. Acad. Sci. USA 2018, 115, E8017–E8026. [Google Scholar] [CrossRef]
  81. Shaked, I.; Hanna, R.N.; Shaked, H.; Chodaczek, G.; Nowyhed, H.N.; Tweet, G.; Tacke, R.; Basat, A.B.; Mikulski, Z.; Togher, S.; et al. Transcription factor Nr4a1 couples sympathetic and inflammatory cues in CNS-recruited macrophages to limit neuroinflammation. Nat. Immunol. 2015, 16, 1228–1234. [Google Scholar] [CrossRef]
  82. Wang, L.M.; Zhang, Y.; Li, X.; Zhang, M.L.; Zhu, L.; Zhang, G.X.; Xu, Y.M. Nr4a1 plays a crucial modulatory role in Th1/Th17 cell responses and CNS autoimmunity. Brain Behav. Immun. 2018, 68, 44–55. [Google Scholar] [CrossRef]
  83. Han, X.; Xu, T.; Ding, C.; Wang, D.; Yao, G.; Chen, H.; Fang, Q.; Hu, G.; Sun, L. Neuronal NR4A1 deficiency drives complement-coordinated synaptic stripping by microglia in a mouse model of lupus. Signal Transduct. Target. Ther. 2022, 7, 50, Erratum in Signal Transduct. Target. Ther. 2022, 7, 328. https://doi.org/10.1038/s41392-022-01155-z. [Google Scholar] [CrossRef]
  84. Sekiya, T. Comparison Between Nr4a Transcription Factor Regulation and Function in Lymphoid and Tumor Treg Cells. Front. Immunol. 2022, 13, 866339. [Google Scholar] [CrossRef]
  85. Sekiya, T.; Nakatsukasa, H.; Lu, Q.; Yoshimura, A. Roles of transcription factors and epigenetic modifications in differentiation and maintenance of regulatory T cells. Microbes Infect. 2016, 18, 378–386. [Google Scholar] [CrossRef] [PubMed]
  86. Sekiya, T.; Kondo, T.; Shichita, T.; Morita, R.; Ichinose, H.; Yoshimura, A. Suppression of Th2 and Tfh immune reactions by Nr4a receptors in mature T reg cells. J. Exp. Med. 2015, 212, 1623–1640. [Google Scholar] [CrossRef] [PubMed]
  87. Sekiya, T.; Kashiwagi, I.; Inoue, N.; Morita, R.; Hori, S.; Waldmann, H.; Rudensky, A.Y.; Ichinose, H.; Metzger, D.; Chambon, P.; et al. The nuclear orphan receptor Nr4a2 induces Foxp3 and regulates differentiation of CD4+ T cells. Nat. Commun. 2011, 2, 269. [Google Scholar] [CrossRef]
  88. Sekiya, T.; Kashiwagi, I.; Yoshida, R.; Fukaya, T.; Morita, R.; Kimura, A.; Ichinose, H.; Metzger, D.; Chambon, P.; Yoshimura, A. Nr4a receptors are essential for thymic regulatory T cell development and immune homeostasis. Nat. Immunol. 2013, 14, 230–237. [Google Scholar] [CrossRef]
  89. Tan, C.; Hiwa, R.; Mueller, J.L.; Vykunta, V.; Hibiya, K.; Noviski, M.; Huizar, J.; Brooks, J.F.; Garcia, J.; Heyn, C.; et al. NR4A nuclear receptors restrain B cell responses to antigen when second signals are absent or limiting. Nat. Immunol. 2020, 21, 1267–1279. [Google Scholar] [CrossRef] [PubMed]
  90. Hiwa, R.; Brooks, J.F.; Mueller, J.L.; Nielsen, H.V.; Zikherman, J. NR4A nuclear receptors in T and B lymphocytes: Gatekeepers of immune tolerance. Immunol. Rev. 2022, 307, 116–133. [Google Scholar] [CrossRef]
  91. Tel-Karthaus, N.; Kers-Rebel, E.D.; Looman, M.W.; Ichinose, H.; de Vries, C.J.; Ansems, M. Nuclear Receptor Nur77 Deficiency Alters Dendritic Cell Function. Front. Immunol. 2018, 9, 1797. [Google Scholar] [CrossRef]
  92. Boulet, S.; Le Corre, L.; Odagiu, L.; Labrecque, N. Role of NR4A family members in myeloid cells and leukemia. Curr. Res. Immunol. 2022, 3, 23–36. [Google Scholar] [CrossRef]
  93. Nie, X.; Tan, J.; Dai, Y.; Mao, W.; Chen, Y.; Qin, G.; Li, G.; Shen, C.; Zhao, J.; Chen, J. Nur77 downregulation triggers pulmonary artery smooth muscle cell proliferation and migration in mice with hypoxic pulmonary hypertension via the Axin2-β-catenin signaling pathway. Vasc. Pharmacol. 2016, 87, 230–241. [Google Scholar] [CrossRef]
  94. Wang, H.; Wei, Z.; Xu, C.; Fang, F.; Wang, Z.; Zhong, Y.; Wang, X. Nuclear receptor 4A1 ameliorates UUO-induced renal fibrosis by inhibiting the PI3K/AKT pathway. Sci. Rep. 2024, 14, 24787. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, H.; Fang, F.; Zhang, M.; Xu, C.; Liu, J.; Gao, L.; Zhao, C.; Wang, Z.; Zhong, Y.; Wang, X. Nuclear receptor 4A1 ameliorates renal fibrosis by inhibiting vascular endothelial growth factor A induced angiogenesis in UUO rats. Biochim. Biophys. Acta Mol. Cell Res. 2024, 1871, 119813. [Google Scholar] [CrossRef]
  96. Westbrook, L.; Johnson, A.C.; Regner, K.R.; Williams, J.M.; Mattson, D.L.; Kyle, P.B.; Henegar, J.R.; Garrett, M.R. Genetic susceptibility and loss of Nr4a1 enhances macrophage-mediated renal injury in CKD. J. Am. Soc. Nephrol. 2014, 25, 2499–2510. [Google Scholar] [CrossRef] [PubMed]
  97. Tao, Y.; Tang, C.; Wei, J.; Shan, Y.; Fang, X.; Li, Y. Nr4a1 promotes renal interstitial fibrosis by regulating the p38 MAPK phosphorylation. Mol. Med. 2023, 29, 63. [Google Scholar] [CrossRef] [PubMed]
  98. Collins, C.A.; Waller, C.; Batourina, E.; Kumar, L.; Mendelsohn, C.L.; Gilbert, N.M. Nur77 protects the bladder urothelium from intracellular bacterial infection. Nat. Commun. 2024, 15, 8308. [Google Scholar] [CrossRef]
  99. Liang, H.; Xie, X.; Song, X.; Huang, M.; Su, T.; Chang, X.; Liang, B.; Huang, D. Orphan nuclear receptor NR4A1 suppresses hyperhomocysteinemia-induced hepatic steatosis in vitro and in vivo. FEBS Lett. 2019, 593, 1061–1071. [Google Scholar] [CrossRef]
  100. Ma, T.; Huang, W.; Ding, Y.; Ji, R.; Ge, S.; Liu, Q.; Liu, Y.; Chen, J.; Yan, Y.; Lu, S.; et al. AIBP protects drug-induced liver injury by inhibiting MAPK-mediated NR4A1 expression. iScience 2024, 27, 110873. [Google Scholar] [CrossRef]
  101. Cao, J.; Xu, T.; Zhou, C.; Wang, S.; Jiang, B.; Wu, K.; Ma, L. NR4A1 knockdown confers hepatoprotection against ischaemia-reperfusion injury by suppressing TGFβ1 via inhibition of CYR61/NF-κB in mouse hepatocytes. J. Cell Mol. Med. 2021, 25, 5099–5112. [Google Scholar] [CrossRef]
  102. Sheng, M.; Weng, Y.; Cao, Y.; Zhang, C.; Lin, Y.; Yu, W. Caspase 6/NR4A1/SOX9 signaling axis regulates hepatic inflammation and pyroptosis in ischemia-stressed fatty liver. Cell Death Discov. 2023, 9, 106. [Google Scholar] [CrossRef]
  103. Zheng, Y.; Tao, Y.; Zhan, X.; Wu, Q. Nuclear receptor 4A1 (NR4A1) silencing protects hepatocyte against hypoxia-reperfusion injury in vitro by activating liver kinase B1 (LKB1)/AMP-activated protein kinase (AMPK) signaling. Bioengineered 2022, 13, 8349–8359. [Google Scholar] [CrossRef]
  104. Palumbo-Zerr, K.; Zerr, P.; Distler, A.; Fliehr, J.; Mancuso, R.; Huang, J.; Mielenz, D.; Tomcik, M.; Fürnrohr, B.G.; Scholtysek, C.; et al. Orphan nuclear receptor NR4A1 regulates transforming growth factor-β signaling and fibrosis. Nat. Med. 2015, 21, 150–158. [Google Scholar] [CrossRef]
  105. Huang, Q.; Xu, J.; Ge, Y.; Shi, Y.; Wang, F.; Zhu, M. NR4A1 inhibits the epithelial-mesenchymal transition of hepatic stellate cells: Involvement of TGF-β-SMAD2/3/4-ZEB signaling. Open Life Sci. 2022, 17, 447–454. [Google Scholar] [CrossRef]
  106. Fuchs, C.D.; Claudel, T.; Mlitz, V.; Riva, A.; Menz, M.; Brusilovskaya, K.; Haller, F.; Baumgartner, M.; Königshofer, P.; Unger, L.W.; et al. GLP-2 Improves Hepatic Inflammation and Fibrosis in Mdr2-/- Mice Via Activation of NR4a1/Nur77 in Hepatic Stellate Cells and Intestinal FXR Signaling. Cell Mol. Gastroenterol. Hepatol. 2023, 16, 847–856. [Google Scholar] [CrossRef]
  107. Caon, E.; Martins, M.; Hodgetts, H.; Blanken, L.; Vilia, M.G.; Levi, A.; Thanapirom, K.; Al-Akkad, W.; Abu-Hanna, J.; Baselli, G.; et al. Exploring the impact of the PNPLA3 I148M variant on primary human hepatic stellate cells using 3D extracellular matrix models. J. Hepatol. 2024, 80, 941–956. [Google Scholar] [CrossRef] [PubMed]
  108. Lu, L.; Jang, S.; Zhu, J.; Qin, Q.; Sun, L.; Sun, J. Nur77 mitigates endothelial dysfunction through activation of both nitric oxide production and anti-oxidant pathways. Redox Biol. 2024, 70, 103056. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  109. Yu, Y.; Cai, Z.; Cui, M.; Nie, P.; Sun, Z.; Sun, S.; Chu, S.; Wang, X.; Hu, L.; Yi, J.; et al. The orphan nuclear receptor Nur77 inhibits low shear stress-induced carotid artery remodeling in mice. Int. J. Mol. Med. 2015, 36, 1547–1555. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  110. Wu, Z.; Bai, Y.; Chang, C.; Jiao, Y.; Chen, Q.; Guo, Z. Metformin attenuates myocardial ischemia/reperfusion-induced ferroptosis through the upregulation of Nur77-mediated IDH1. Biochim. Biophys. Acta Mol. Cell Res. 2025, 1872, 119934. [Google Scholar] [CrossRef] [PubMed]
  111. Kang, Q.; Chai, W.; Min, J.; Qu, X. Yin Yang 1 suppresses apoptosis and oxidative stress injury in SH-SY5Y cells by facilitating NR4A1 expression. J. Neurogenet. 2023, 37, 115–123. [Google Scholar] [CrossRef] [PubMed]
  112. Widiapradja, A.; Connery, H.; Bullock, M.; Kasparian, A.O.; Clifton-Bligh, R.; Levick, S.P. The orphan nuclear receptor Nr4a1 contributes to interstitial cardiac fibrosis via modulation of cardiac fibroblast and macrophage phenotype. Cell Mol. Life Sci. 2024, 81, 484. [Google Scholar] [CrossRef] [PubMed]
  113. Jeanneteau, F.; Barrère, C.; Vos, M.; De Vries, C.J.M.; Rouillard, C.; Levesque, D.; Dromard, Y.; Moisan, M.P.; Duric, V.; Franklin, T.C.; et al. The Stress-Induced Transcription Factor NR4A1 Adjusts Mitochondrial Function and Synapse Number in Prefrontal Cortex. J. Neurosci. 2018, 38, 1335–1350. [Google Scholar] [CrossRef]
  114. Murphy, E.P.; Crean, D. NR4A1-3 nuclear receptor activity and immune cell dysregulation in rheumatic diseases. Front. Med. 2022, 9, 874182. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, C.; Zhang, B.; Zhang, X.; Sun, G.; Sun, X. Targeting Orphan Nuclear Receptors NR4As for Energy Homeostasis and Diabetes. Front. Pharmacol. 2020, 11, 587457. [Google Scholar] [CrossRef]
  116. Wang, Z.; Benoit, G.; Liu, J.; Prasad, S.; Aarnisalo, P.; Liu, X.; Xu, H.; Walker, N.P.; Perlmann, T. Structure and function of Nurr1 identifies a class of ligand-independent nuclear receptors. Nature 2003, 423, 555–560. [Google Scholar] [CrossRef]
  117. Jordan, V.C. Antiestrogens and selective estrogen receptor modulators as multifunctional medicines. 1. Receptor interactions. J. Med. Chem. 2003, 46, 883–908. [Google Scholar] [CrossRef]
  118. Jordan, V.C. A century of deciphering the control mechanisms of sex steroid action in breast and prostate cancer: The origins of targeted therapy and chemoprevention. Cancer Res. 2009, 69, 1243–1254. [Google Scholar] [CrossRef]
  119. Emons, G.; Mustea, A.; Tempfer, C. Tamoxifen and Endometrial Cancer: A Janus-Headed Drug. Cancers 2020, 12, 2535. [Google Scholar] [CrossRef]
  120. Zhan, Y.; Du, X.; Chen, H.; Liu, J.; Zhao, B.; Huang, D.; Li, G.; Xu, Q.; Zhang, M.; Weimer, B.C.; et al. Cytosporone B is an agonist for nuclear orphan receptor Nur77. Nat. Chem. Biol. 2008, 4, 548–556. [Google Scholar] [CrossRef] [PubMed]
  121. Liu, J.J.; Zeng, H.N.; Zhang, L.R.; Zhan, Y.Y.; Chen, Y.; Wang, Y.; Wang, J.; Xiang, S.H.; Liu, W.J.; Wang, W.J.; et al. A unique pharmacophore for activation of the nuclear orphan receptor Nur77 in vivo and in vitro. Cancer Res. 2010, 70, 3628–3637. [Google Scholar] [CrossRef]
  122. Deng, Z.; Zheng, L.; Xie, X.; Wei, H.; Peng, J. GPA peptide enhances Nur77 expression in intestinal epithelial cells to exert a protective effect against DSS-induced colitis. FASEB J. 2020, 34, 15364–15378. [Google Scholar] [CrossRef]
  123. Li, Y.; Wang, S.M.; Li, X.; Lv, C.J.; Peng, L.Y.; Yu, X.F.; Song, Y.J.; Wang, C.J. Pterostilbene pre-treatment reduces LPS-induced acute lung injury through activating NR4A1. Pharm. Biol. 2022, 60, 394–403. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  124. Egarnes, B.; Blanchet, M.R.; Gosselin, J. Treatment with the NR4A1 agonist cytosporone B controls influenza virus infection and improves pulmonary function in infected mice. PLoS ONE 2017, 12, e0186639. [Google Scholar] [CrossRef]
  125. Kurakula, K.; Sun, X.Q.; Happé, C.; da Silva Goncalves Bos, D.; Szulcek, R.; Schalij, I.; Wiesmeijer, K.C.; Lodder, K.; Tu, L.; Guignabert, C.; et al. Prevention of progression of pulmonary hypertension by the Nur77 agonist 6-mercaptopurine: Role of BMP signalling. Eur. Respir. J. 2019, 54, 1802400. [Google Scholar] [CrossRef]
  126. Xiong, Y.; Ran, J.; Xu, L.; Tong, Z.; Adel Abdo, M.S.; Ma, C.; Xu, K.; He, Y.; Wu, Z.; Chen, Z.; et al. Reactivation of NR4A1 Restrains Chondrocyte Inflammation and Ameliorates Osteoarthritis in Rats. Front. Cell Dev. Biol. 2020, 8, 158. [Google Scholar] [CrossRef] [PubMed]
  127. De Miranda, B.R.; Miller, J.A.; Hansen, R.J.; Lunghofer, P.J.; Safe, S.; Gustafson, D.L.; Colagiovanni, D.; Tjalkens, R.B. Neuroprotective efficacy and pharmacokinetic behavior of novel anti-inflammatory para-phenyl substituted diindolylmethanes in a mouse model of Parkinson’s disease. J. Pharmacol. Exp. Ther. 2013, 345, 125–138, Erratum in J. Pharmacol. Exp. Ther. 2019, 369, 66. [Google Scholar] [CrossRef] [PubMed]
  128. Bridi, M.S.; Hawk, J.D.; Chatterjee, S.; Safe, S.; Abel, T. Pharmacological Activators of the NR4A Nuclear Receptors Enhance LTP in a CREB/CBP-Dependent Manner. Neuropsychopharmacology 2017, 42, 1243–1253. [Google Scholar] [CrossRef]
  129. Hammond, S.L.; Safe, S.; Tjalkens, R.B. A novel synthetic activator of Nurr1 induces dopaminergic gene expression and protects against 6-hydroxydopamine neurotoxicity in vitro. Neurosci. Lett. 2015, 607, 83–89. [Google Scholar] [CrossRef] [PubMed]
  130. Lee, S.O.; Li, X.; Hedrick, E.; Jin, U.H.; Tjalkens, R.B.; Backos, D.S.; Li, L.; Zhang, Y.; Wu, Q.; Safe, S. Diindolylmethane analogs bind NR4A1 and are NR4A1 antagonists in colon cancer cells. Mol. Endocrinol. 2014, 28, 1729–1739. [Google Scholar] [CrossRef] [PubMed]
  131. Liu, T.Y.; Yang, X.Y.; Zheng, L.T.; Wang, G.H.; Zhen, X.C. Activation of Nur77 in microglia attenuates proinflammatory mediators production and protects dopaminergic neurons from inflammation-induced cell death. J. Neurochem. 2017, 140, 589–604. [Google Scholar] [CrossRef]
  132. Rothe, T.; Ipseiz, N.; Faas, M.; Lang, S.; Perez-Branguli, F.; Metzger, D.; Ichinose, H.; Winner, B.; Schett, G.; Krönke, G. The Nuclear Receptor Nr4a1 Acts as a Microglia Rheostat and Serves as a Therapeutic Target in Autoimmune-Driven Central Nervous System Inflammation. J. Immunol. 2017, 198, 3878–3885. [Google Scholar] [CrossRef]
  133. Carpenter, M.D.; Hu, Q.; Bond, A.M.; Lombroso, S.I.; Czarnecki, K.S.; Lim, C.J.; Song, H.; Wimmer, M.E.; Pierce, R.C.; Heller, E.A. Nr4a1 suppresses cocaine-induced behavior via epigenetic regulation of homeostatic target genes. Nat. Commun. 2020, 11, 504. [Google Scholar] [CrossRef] [PubMed]
  134. Zeng, X.; Yue, Z.; Gao, Y.; Jiang, G.; Zeng, F.; Shao, Y.; Huang, J.; Yin, M.; Li, Y. NR4A1 is Involved in Fibrogenesis in Ovarian Endometriosis. Cell Physiol. Biochem. 2018, 46, 1078–1090. [Google Scholar] [CrossRef]
  135. Mohankumar, K.; Li, X.; Sung, N.; Cho, Y.J.; Han, S.J.; Safe, S. Bis-Indole-Derived Nuclear Receptor 4A1 (NR4A1, Nur77) Ligands as Inhibitors of Endometriosis. Endocrinology 2020, 161, bqaa027. [Google Scholar] [CrossRef] [PubMed]
  136. Xie, X.; Song, X.; Yuan, S.; Cai, H.; Chen, Y.; Chang, X.; Liang, B.; Huang, D. Histone acetylation regulates orphan nuclear receptor NR4A1 expression in hypercholesterolaemia. Clin. Sci. 2015, 129, 1151–1161. [Google Scholar] [CrossRef]
  137. Liu, W.; Li, G.; Shi, J.; Gao, Y.; Fang, P.; Zhao, Y.; Zhong, F.; Guo, X.; Lyu, Y.; Da, X.; et al. NR4A1 Acts as a Novel Regulator of Platelet Activation and Thrombus Formation. Circ. Res. 2025, 136, 809–826. [Google Scholar] [CrossRef] [PubMed]
  138. Hu, Y.W.; Zhang, P.; Yang, J.Y.; Huang, J.L.; Ma, X.; Li, S.F.; Zhao, J.Y.; Hu, Y.R.; Wang, Y.C.; Gao, J.J.; et al. Nur77 decreases atherosclerosis progression in apoE(-/-) mice fed a high-fat/high-cholesterol diet. PLoS ONE 2014, 9, e87313. [Google Scholar] [CrossRef]
  139. Pires, N.M.; Pols, T.W.; de Vries, M.R.; van Tiel, C.M.; Bonta, P.I.; Vos, M.; Arkenbout, E.K.; Pannekoek, H.; Jukema, J.W.; Quax, P.H.; et al. Activation of nuclear receptor Nur77 by 6-mercaptopurine protects against neointima formation. Circulation 2007, 115, 493–500. [Google Scholar] [CrossRef]
  140. Chen, L.; Shi, Y.; Xiao, D.; Huang, Y.; Jiang, Y.; Liang, M.; Liang, F.; Xue, J.; Chen, H.; Liu, Z.; et al. NR4A1 deficiency promotes carotid plaque vulnerability by activating integrated stress response via targeting Bcat1. Cell Mol. Life Sci. 2025, 82, 91. [Google Scholar] [CrossRef]
  141. Ding, X.; Le, S.; Wang, K.; Su, Y.; Chen, S.; Wu, C.; Chen, J.; Chen, S.; Zhang, A.; Xia, J. Cytosporone B (Csn-B), an NR4A1 agonist, attenuates acute cardiac allograft rejection by inducing differential apoptosis of CD4+T cells. Int. Immunopharmacol. 2022, 104, 108521. [Google Scholar] [CrossRef] [PubMed]
  142. Patiño-Martínez, E.; Solís-Barbosa, M.A.; Santana, E.; González-Domínguez, E.; Segovia-Gamboa, N.C.; Meraz-Ríos, M.A.; Córdova, E.J.; Valdés, J.; Corbí, Á.L.; Sánchez-Torres, C. The Nurr7 agonist Cytosporone B differentially regulates inflammatory responses in human polarized macrophages. Immunobiology 2022, 227, 152299. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, Q.; Huang, Y.; Gong, C.; Tang, Y.; Xiong, J.; Wang, D.; Liu, X. Dexmedetomidine attenuates inflammation and organ injury partially by upregulating Nur77 in sepsis. Immun. Inflamm. Dis. 2023, 11, e883. [Google Scholar] [CrossRef]
  144. Brunet, A.; LeBel, M.; Egarnes, B.; Paquet-Bouchard, C.; Lessard, A.J.; Brown, J.P.; Gosselin, J. NR4A1-dependent Ly6Clow monocytes contribute to reducing joint inflammation in arthritic mice through Treg cells. Eur. J. Immunol. 2016, 46, 2789–2800. [Google Scholar] [CrossRef]
  145. Teichmann, T.; Pflüger-Müller, B.; Giménez, V.M.M.; Sailer, F.; Dirks, H.; Zehr, S.; Warwick, T.; Brettner, F.; Munoz-Tello, P.; Zimmer, A.; et al. The endocannabinoid anandamide mediates anti-inflammatory effects through activation of NR4A nuclear receptors. Br. J. Pharmacol. 2025, 182, 1164–1182. [Google Scholar] [CrossRef]
  146. Xia, Y.; Chen, H.; Qin, J.; Zhang, W.; Gao, H.; Long, X.; He, H.; Zhang, L.; Zhang, C.; Cao, C.; et al. The phthalide compound tokinolide B from Angelica sinensis exerts anti-inflammatory effects through Nur77 binding. Phytomedicine 2024, 133, 155925. [Google Scholar] [CrossRef] [PubMed]
  147. Hu, M.; Luo, Q.; Alitongbieke, G.; Chong, S.; Xu, C.; Xie, L.; Chen, X.; Zhang, D.; Zhou, Y.; Wang, Z.; et al. Celastrol-Induced Nur77 Interaction with TRAF2 Alleviates Inflammation by Promoting Mitochondrial Ubiquitination and Autophagy. Mol. Cell 2017, 66, 141–153.e6. [Google Scholar] [CrossRef]
  148. Zhan, Y.Y.; Chen, Y.; Zhang, Q.; Zhuang, J.J.; Tian, M.; Chen, H.Z.; Zhang, L.R.; Zhang, H.K.; He, J.P.; Wang, W.J.; et al. The orphan nuclear receptor Nur77 regulates LKB1 localization and activates AMPK. Nat. Chem. Biol. 2012, 8, 897–904. [Google Scholar] [CrossRef]
  149. Mohankumar, K.; Lee, J.; Wu, C.S.; Sun, Y.; Safe, S. Bis-Indole-Derived NR4A1 Ligands and Metformin Exhibit NR4A1-Dependent Glucose Metabolism and Uptake in C2C12 Cells. Endocrinology 2018, 159, 1950–1963. [Google Scholar] [CrossRef]
  150. Yang, R.; Zong, T.; Wang, N.; Wang, F.; Su, Y. NR4A1 Alleviates Subretinal Fibrosis by Inhibiting Macrophage to Myofibroblast Transition. Invest. Ophthalmol. Vis. Sci. 2025, 66, 47. [Google Scholar] [CrossRef]
  151. Paillasse, M.R.; de Medina, P. The NR4A nuclear receptors as potential targets for anti-aging interventions. Med. Hypotheses 2015, 84, 135–140. [Google Scholar] [CrossRef]
  152. Hano, C.; Tungmunnithum, D. Plant Polyphenols, more than Just Simple Natural Antioxidants: Oxidative Stress, Aging and Age-Related Diseases. Medicines 2020, 7, 26. [Google Scholar] [CrossRef]
  153. Khoso, M.A.; Liu, H.; Zhao, T.; Zhao, W.; Huang, Q.; Sun, Z.; Dinislam, K.; Chen, C.; Kong, L.; Zhang, Y.; et al. Impact of plant-derived antioxidants on heart aging: A mechanistic outlook. Front. Pharmacol. 2025, 16, 1524584. [Google Scholar] [CrossRef]
  154. Nakadate, K.; Ito, N.; Kawakami, K.; Yamazaki, N. Anti-Inflammatory Actions of Plant-Derived Compounds and Prevention of Chronic Diseases: From Molecular Mechanisms to Applications. Int. J. Mol. Sci. 2025, 26, 5206. [Google Scholar] [CrossRef]
  155. Zhang, L.; Mohankumar, K.; Martin, G.; Mariyam, F.; Park, Y.; Han, S.J.; Safe, S. Flavonoids Quercetin and Kaempferol Are NR4A1 Antagonists and Suppress Endometriosis in Female Mice. Endocrinology 2023, 164, bqad133. [Google Scholar] [CrossRef]
  156. Shrestha, R.; Mohankumar, K.; Martin, G.; Hailemariam, A.; Lee, S.O.; Jin, U.H.; Burghardt, R.; Safe, S. Flavonoids kaempferol and quercetin are nuclear receptor 4A1 (NR4A1, Nur77) ligands and inhibit rhabdomyosarcoma cell and tumor growth. J. Exp. Clin. Cancer Res. 2021, 40, 392. [Google Scholar] [CrossRef] [PubMed]
  157. Lee, M.; Upadhyay, S.; Mariyam, F.; Martin, G.; Hailemariam, A.; Lee, K.; Jayaraman, A.; Chapkin, R.S.; Lee, S.O.; Safe, S. Flavone and Hydroxyflavones Are Ligands That Bind the Orphan Nuclear Receptor 4A1 (NR4A1). Int. J. Mol. Sci. 2023, 24, 8152. [Google Scholar] [CrossRef]
  158. Wang, R.; Zhang, H.; Wang, Y.; Song, F.; Yuan, Y. Inhibitory effects of quercetin on the progression of liver fibrosis through the regulation of NF-κB/IκBα, p38 MAPK, and Bcl-2/Bax signaling. Int. Immunopharmacol. 2017, 47, 126–133. [Google Scholar] [CrossRef] [PubMed]
  159. Wu, L.; Zhang, Q.; Mo, W.; Feng, J.; Li, S.; Li, J.; Liu, T.; Xu, S.; Wang, W.; Lu, X.; et al. Quercetin prevents hepatic fibrosis by inhibiting hepatic stellate cell activation and reducing autophagy via the TGF-β1/Smads and PI3K/Akt pathways. Sci. Rep. 2017, 7, 9289. [Google Scholar] [CrossRef]
  160. Chen, X.; Wang, Y.; Wan, J.; Dou, X.; Zhang, C.; Sun, M.; Ye, F. Quercetin alleviates liver fibrosis via regulating glycolysis of liver sinusoidal endothelial cells and neutrophil infiltration. Biomol. Biomed. 2024, 24, 1806–1815. [Google Scholar] [CrossRef] [PubMed]
  161. Shakerian, E.; Akbari, R.; Mohammadtaghvaei, N.; Mohammadi Gahrooie, M.; Afarin, R. Quercetin Reduces Hepatic Fibrogenesis by Inhibiting TGF-β/Smad3 Signaling Pathway in LX-2 Cell Line. Jundishapur J. Nat. Pharm. Prod. 2021, 17, e113484. [Google Scholar] [CrossRef]
  162. Guo, X.; Li, Y.; Wang, W.; Wang, L.; Hu, S.; Xiao, X.; Hu, C.; Dai, Y.; Zhang, Y.; Li, Z.; et al. The construction of preclinical evidence for the treatment of liver fibrosis with quercetin: A systematic review and meta-analysis. Phytother. Res. 2022, 36, 3774–3791. [Google Scholar] [CrossRef]
  163. Ren, J.; Li, J.; Liu, X.; Feng, Y.; Gui, Y.; Yang, J.; He, W.; Dai, C. Quercetin Inhibits Fibroblast Activation and Kidney Fibrosis Involving the Suppression of Mammalian Target of Rapamycin and β-catenin Signaling. Sci. Rep. 2016, 6, 23968. [Google Scholar] [CrossRef]
  164. Liu, X.; Sun, N.; Mo, N.; Lu, S.; Song, E.; Ren, C.; Li, Z. Quercetin inhibits kidney fibrosis and the epithelial to mesenchymal transition of the renal tubular system involving suppression of the Sonic Hedgehog signaling pathway. Food Funct. 2019, 10, 3782–3797. [Google Scholar] [CrossRef]
  165. Cao, Y.; Hu, J.; Sui, J.; Jiang, L.; Cong, Y.; Ren, G. Quercetin is able to alleviate TGF-β-induced fibrosis in renal tubular epithelial cells by suppressing miR-21. Exp. Ther. Med. 2018, 16, 2442–2448. [Google Scholar] [CrossRef] [PubMed]
  166. Sherif, I.O. Uroprotective mechanism of quercetin against cyclophosphamide-induced urotoxicity: Effect on oxidative stress and inflammatory markers. J. Cell Biochem. 2018, 119, 7441–7448. [Google Scholar] [CrossRef] [PubMed]
  167. Maisto, M.; Iannuzzo, F.; Novellino, E.; Schiano, E.; Piccolo, V.; Tenore, G.C. Natural Polyphenols for Prevention and Treatment of Urinary Tract Infections. Int. J. Mol. Sci. 2023, 24, 3277. [Google Scholar] [CrossRef]
  168. Nguyen, T.L.A.; Bhattacharya, D. Antimicrobial Activity of Quercetin: An Approach to Its Mechanistic Principle. Molecules 2022, 27, 2494. [Google Scholar] [CrossRef]
  169. Han, X.; Xu, T.; Fang, Q.; Zhang, H.; Yue, L.; Hu, G.; Sun, L. Quercetin hinders microglial activation to alleviate neurotoxicity via the interplay between NLRP3 inflammasome and mitophagy. Redox Biol. 2021, 44, 102010. [Google Scholar] [CrossRef] [PubMed]
  170. Zhang, Z.J.; Cheang, L.C.; Wang, M.W.; Lee, S.M. Quercetin exerts a neuroprotective effect through inhibition of the iNOS/NO system and pro-inflammation gene expression in PC12 cells and in zebrafish. Int. J. Mol. Med. 2011, 27, 195–203. [Google Scholar] [CrossRef]
  171. Geng, F.; Zhao, L.; Cai, Y.; Zhao, Y.; Jin, F.; Li, Y.; Li, T.; Yang, X.; Li, S.; Gao, X.; et al. Quercetin Alleviates Pulmonary Fibrosis in Silicotic Mice by Inhibiting Macrophage Transition and TGF-β-SMAD2/3 Pathway. Curr. Issues Mol. Biol. 2023, 45, 3087–3101. [Google Scholar] [CrossRef] [PubMed]
  172. Huang, R.; Zhong, T.; Wu, H. Quercetin protects against lipopolysaccharide-induced acute lung injury in rats through suppression of inflammation and oxidative stress. Arch. Med. Sci. 2015, 11, 427–432. [Google Scholar] [CrossRef]
  173. Lv, P.; Han, P.; Cui, Y.; Chen, Q.; Cao, W. Quercetin attenuates inflammation in LPS-induced lung epithelial cells via the NRF2 signaling pathway. Immun. Inflamm. Dis. 2024, 12, e1185. [Google Scholar] [CrossRef] [PubMed]
  174. Li, M.; Yun, G. Quercetin: A Potential Drug Candidate for Inflammatory Bowel Disease. In Quercetin-Effects on Human Health; IntechOpen: London, UK, 2024. [Google Scholar] [CrossRef]
  175. Dong, Y.; Lei, J.; Zhang, B. Dietary Quercetin Alleviated DSS-induced Colitis in Mice Through Several Possible Pathways by Transcriptome Analysis. Curr. Pharm. Biotechnol. 2020, 21, 1666–1673. [Google Scholar] [CrossRef] [PubMed]
  176. Zhang, W.; Zheng, Y.; Yan, F.; Dong, M.; Ren, Y. Research progress of quercetin in cardiovascular disease. Front. Cardiovasc. Med. 2023, 10, 1203713. [Google Scholar] [CrossRef]
  177. Espírito-Santo, D.A.; Cordeiro, G.S.; Santos, L.S.; Silva, R.T.; Pereira, M.U.; Matos, R.J.B.; Boaventura, G.T.; Barreto-Medeiros, J.M. Cardioprotective effect of the quercetin on cardiovascular remodeling and atherosclerosis in rodents fed a high-fat diet: A systematic review. Chem. Biol. Interact. 2023, 384, 110700. [Google Scholar] [CrossRef]
  178. Arabi, S.M.; Shahraki Jazinaki, M.; Chambari, M.; Bahrami, L.S.; Maleki, M.; Sukhorukov, V.N.; Sahebkar, A. The effects of Quercetin supplementation on cardiometabolic outcomes: An umbrella review of meta-analyses of randomized controlled trials. Phytother. Res. 2023, 37, 5080–5091. [Google Scholar] [CrossRef]
  179. Wei, X.; Meng, X.; Yuan, Y.; Shen, F.; Li, C.; Yang, J. Quercetin exerts cardiovascular protective effects in LPS-induced dysfunction in vivo by regulating inflammatory cytokine expression, NF-κB phosphorylation, and caspase activity. Mol. Cell Biochem. 2018, 446, 43–52. [Google Scholar] [CrossRef]
  180. Bule, M.; Abdurahman, A.; Nikfar, S.; Abdollahi, M.; Amini, M. Antidiabetic effect of quercetin: A systematic review and meta-analysis of animal studies. Food Chem. Toxicol. 2019, 125, 494–502. [Google Scholar] [CrossRef]
  181. Eid, H.M.; Nachar, A.; Thong, F.; Sweeney, G.; Haddad, P.S. The molecular basis of the antidiabetic action of quercetin in cultured skeletal muscle cells and hepatocytes. Pharmacogn. Mag. 2015, 11, 74–81. [Google Scholar] [CrossRef]
  182. Youl, E.; Bardy, G.; Magous, R.; Cros, G.; Sejalon, F.; Virsolvy, A.; Richard, S.; Quignard, J.F.; Gross, R.; Petit, P.; et al. Quercetin potentiates insulin secretion and protects INS-1 pancreatic β-cells against oxidative damage via the ERK1/2 pathway. Br. J. Pharmacol. 2010, 161, 799–814. [Google Scholar] [CrossRef]
  183. Kim, Y.J.; Park, W. Anti-Inflammatory Effect of Quercetin on RAW 264.7 Mouse Macrophages Induced with Polyinosinic-Polycytidylic Acid. Molecules 2016, 21, 450. [Google Scholar] [CrossRef] [PubMed]
  184. Tsai, C.F.; Chen, G.W.; Chen, Y.C.; Shen, C.K.; Lu, D.Y.; Yang, L.Y.; Chen, J.H.; Yeh, W.L. Regulatory Effects of Quercetin on M1/M2 Macrophage Polarization and Oxidative/Antioxidative Balance. Nutrients 2021, 14, 67. [Google Scholar] [CrossRef] [PubMed]
  185. Huang, R.Y.; Yu, Y.L.; Cheng, W.C.; OuYang, C.N.; Fu, E.; Chu, C.L. Immunosuppressive effect of quercetin on dendritic cell activation and function. J. Immunol. 2010, 184, 6815–6821. [Google Scholar] [CrossRef] [PubMed]
  186. Xu, Y.; Xu, L.; Jian, X.; Wang, Q.; Li, Z.; Ge, H. Baicalein attenuates ovalbumin-induced allergic rhinitis through the activation of nuclear receptor subfamily 4 group a member 1. Immunol. Res. 2025, 73, 32. [Google Scholar] [CrossRef] [PubMed]
  187. Zhang, L.; Martin, G.; Mohankumar, K.; Hampton, J.T.; Liu, W.R.; Safe, S. Resveratrol binds nuclear receptor 4A1 (NR4A1) and acts as an NR4A1 antagonist in lung cancer cells. Mol. Pharmacol. 2022, 102, 80–91. [Google Scholar] [CrossRef]
  188. Ren, Z.Q.; Zheng, S.Y.; Sun, Z.; Luo, Y.; Wang, Y.T.; Yi, P.; Li, Y.S.; Huang, C.; Xiao, W.F. Resveratrol: Molecular Mechanisms, Health Benefits, and Potential Adverse Effects. MedComm (2020) 2025, 6, e70252. [Google Scholar] [CrossRef]
  189. Zhou, D.D.; Cheng, J.; Li, J.; Wu, S.X.; Xiong, R.G.; Huang, S.Y.; Cheung, P.C.; Li, H.B. Resveratrol and Its Analogues: Anti-ageing Effects and Underlying Mechanisms. Subcell. Biochem. 2024, 107, 183–203. [Google Scholar] [CrossRef]
  190. Zhou, D.D.; Luo, M.; Huang, S.Y.; Saimaiti, A.; Shang, A.; Gan, R.Y.; Li, H.B. Effects and Mechanisms of Resveratrol on Aging and Age-Related Diseases. Oxid. Med. Cell Longev. 2021, 2021, 9932218. [Google Scholar] [CrossRef]
  191. Xu, X.; Ocansey, D.K.W.; Pei, B.; Zhang, Y.; Wang, N.; Wang, Z.; Mao, F. Resveratrol alleviates DSS-induced IBD in mice by regulating the intestinal microbiota-macrophage-arginine metabolism axis. Eur. J. Med. Res. 2023, 28, 319. [Google Scholar] [CrossRef]
  192. Zhang, B.; Zhang, Y.; Liu, X.; Yin, J.; Li, X.; Zhang, X.; Xing, X.; Wang, J.; Wang, S. Differential Protective Effect of Resveratrol and Its Microbial Metabolites on Intestinal Barrier Dysfunction is Mediated by the AMPK Pathway. J. Agric. Food Chem. 2022, 70, 11301–11313. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, H.; Sun, Q.; Xu, T.; Hong, L.; Fu, R.; Wu, J.; Ding, J. Resveratrol attenuates the progress of liver fibrosis via the Akt/nuclear factor-κB pathways. Mol. Med. Rep. 2016, 13, 224–230. [Google Scholar] [CrossRef]
  194. Zhu, L.; Mou, Q.; Wang, Y.; Zhu, Z.; Cheng, M. Resveratrol contributes to the inhibition of liver fibrosis by inducing autophagy via the microRNA 20a mediated activation of the PTEN/PI3K/AKT signaling pathway. Int. J. Mol. Med. 2020, 46, 2035–2046. [Google Scholar] [CrossRef] [PubMed]
  195. Liang, J.; Tian, S.; Han, J.; Xiong, P. Resveratrol as a therapeutic agent for renal fibrosis induced by unilateral ureteral obstruction. Ren. Fail. 2014, 36, 285–291. [Google Scholar] [CrossRef]
  196. He, T.; Xiong, J.; Nie, L.; Yu, Y.; Guan, X.; Xu, X.; Xiao, T.; Yang, K.; Liu, L.; Zhang, D.; et al. Resveratrol inhibits renal interstitial fibrosis in diabetic nephropathy by regulating AMPK/NOX4/ROS pathway. J. Mol. Med. 2016, 94, 1359–1371. [Google Scholar] [CrossRef] [PubMed]
  197. Yonamine, C.Y.; Pinheiro-Machado, E.; Michalani, M.L.; Alves-Wagner, A.B.; Esteves, J.V.; Freitas, H.S.; Machado, U.F. Resveratrol Improves Glycemic Control in Type 2 Diabetic Obese Mice by Regulating Glucose Transporter Expression in Skeletal Muscle and Liver. Molecules 2017, 22, 1180. [Google Scholar] [CrossRef]
  198. Delpino, F.M.; Figueiredo, L.M. Resveratrol supplementation and type 2 diabetes: A systematic review and meta-analysis. Crit. Rev. Food Sci. Nutr. 2022, 62, 4465–4480. [Google Scholar] [CrossRef]
  199. Teimouri, M.; Homayouni-Tabrizi, M.; Rajabian, A.; Amiri, H.; Hosseini, H. Anti-inflammatory effects of resveratrol in patients with cardiovascular disease: A systematic review and meta-analysis of randomized controlled trials. Complement. Ther. Med. 2022, 70, 102863. [Google Scholar] [CrossRef]
  200. Hao, H.D.; He, L.R. Mechanisms of cardiovascular protection by resveratrol. J. Med. Food 2004, 7, 290–298. [Google Scholar] [CrossRef]
  201. Latronico, T.; Rossano, R.; Miniero, D.V.; Casalino, E.; Liuzzi, G.M. Neuroprotective Effect of Resveratrol against Manganese-Induced Oxidative Stress and Matrix Metalloproteinase-9 in an “In Vivo” Model of Neurotoxicity. Int. J. Mol. Sci. 2024, 25, 2142. [Google Scholar] [CrossRef]
  202. Zhang, F.; Shi, J.S.; Zhou, H.; Wilson, B.; Hong, J.S.; Gao, H.M. Resveratrol protects dopamine neurons against lipopolysaccharide-induced neurotoxicity through its anti-inflammatory actions. Mol. Pharmacol. 2010, 78, 466–477, Erratum in Mol. Pharmacol. 2010, 78, 981. [Google Scholar] [CrossRef]
  203. ElFeky, D.S.; Kassem, A.A.; Moustafa, M.A.; Assiri, H.; El-Mahdy, A.M. Suppression of virulence factors of uropathogenic Escherichia coli by Trans-resveratrol and design of nanoemulgel. BMC Microbiol. 2024, 24, 412. [Google Scholar] [CrossRef]
  204. Calmasini, F.B.; Silva, F.H.; Alexandre, E.C.; Antunes, E. Efficacy of resveratrol in male urogenital tract dysfunctions: An evaluation of pre-clinical data. Nutr. Res. Rev. 2023, 36, 86–97. [Google Scholar] [CrossRef] [PubMed]
  205. Zeng, H.; He, Y.; Yu, Y.; Zhang, J.; Zeng, X.; Gong, F.; Liu, Q.; Yang, B. Resveratrol improves prostate fibrosis during progression of urinary dysfunction in chronic prostatitis by mast cell suppression. Mol. Med. Rep. 2018, 17, 918–924. [Google Scholar] [CrossRef]
  206. Li, W.; Luo, R.; Liu, Z.; Li, X.; Zhang, C.; Huang, J.; Wang, Z.; Chen, J.; Ding, H.; Zhou, X.; et al. Anti-inflammatory effects of resveratrol in treating interstitial cystitis/bladder pain syndrome: A multi-faceted approach integrating network pharmacology, molecular docking, and experimental validation. Mol. Divers. 2025, 29, 2489–2497. [Google Scholar] [CrossRef] [PubMed]
  207. Wang, Z.; Li, X.; Chen, H.; Han, L.; Ji, X.; Wang, Q.; Wei, L.; Miu, Y.; Wang, J.; Mao, J.; et al. Resveratrol alleviates bleomycin-induced pulmonary fibrosis via suppressing HIF-1α and NF-κB expression. Aging 2021, 13, 4605–4616. [Google Scholar] [CrossRef]
  208. Jiang, L.; Zhang, L.; Kang, K.; Fei, D.; Gong, R.; Cao, Y.; Pan, S.; Zhao, M.; Zhao, M. Resveratrol ameliorates LPS-induced acute lung injury via NLRP3 inflammasome modulation. Biomed. Pharmacother. 2016, 84, 130–138. [Google Scholar] [CrossRef] [PubMed]
  209. Noh, K.T.; Cho, J.; Chun, S.H.; Jang, J.H.; Cha, G.S.; Jung, I.D.; Jang, D.D.; Park, Y.M. Resveratrol regulates naïve CD 8+ T-cell proliferation by upregulating IFN-γ-induced tryptophanyl-tRNA synthetase expression. BMB Rep. 2015, 48, 283–288. [Google Scholar] [CrossRef]
  210. Svajger, U.; Obermajer, N.; Jeras, M. Dendritic cells treated with resveratrol during differentiation from monocytes gain substantial tolerogenic properties upon activation. Immunology 2010, 129, 525–535. [Google Scholar] [CrossRef]
  211. Silva, A.M.; Oliveira, M.I.; Sette, L.; Almeida, C.R.; Oliveira, M.J.; Barbosa, M.A.; Santos, S.G. Resveratrol as a natural anti-tumor necrosis factor-α molecule: Implications to dendritic cells and their crosstalk with mesenchymal stromal cells. PLoS ONE 2014, 9, e91406. [Google Scholar] [CrossRef]
  212. Moka, M.K.; George, M.; Sriram, D.K. Advancing Longevity: Exploring Antiaging Pharmaceuticals in Contemporary Clinical Trials Amid Aging Dynamics. Rejuvenation Res. 2024, 27, 220–233. [Google Scholar] [CrossRef] [PubMed]
  213. Shah, M.A.; Faheem, H.I.; Hamid, A.; Yousaf, R.; Haris, M.; Saleem, U.; Shah, G.M.; Alhasani, R.H.; Althobaiti, N.A.; Alsharif, I.; et al. The entrancing role of dietary polyphenols against the most frequent aging-associated diseases. Med. Res. Rev. 2024, 44, 235–274. [Google Scholar] [CrossRef] [PubMed]
  214. Proshkina, E.; Koval, L.; Platonova, E.; Golubev, D.; Ulyasheva, N.; Babak, T.; Shaposhnikov, M.; Moskalev, A. Polyphenols as Potential Geroprotectors. Antioxid. Redox Signal. 2024, 40, 564–593. [Google Scholar] [CrossRef] [PubMed]
Figure 1. NR4A subfamily of NRs. (A) Domain structure of NR4A shows the percentage (%) of similarity between NR4A1, NR4A2, and NR4A3 in 3 domains. (B) Protection interactions of NR4A1 and other ligands with DNA response elements.
Figure 1. NR4A subfamily of NRs. (A) Domain structure of NR4A shows the percentage (%) of similarity between NR4A1, NR4A2, and NR4A3 in 3 domains. (B) Protection interactions of NR4A1 and other ligands with DNA response elements.
Nutrients 17 02709 g001
Figure 2. Examples of synthetic and natural product-derived chemicals that bind NR4A1 [22,23,24].
Figure 2. Examples of synthetic and natural product-derived chemicals that bind NR4A1 [22,23,24].
Nutrients 17 02709 g002
Figure 3. Interactions of NR4A1 with various genes/gene products associated with aging.
Figure 3. Interactions of NR4A1 with various genes/gene products associated with aging.
Nutrients 17 02709 g003
Figure 4. Proposed model for NR4A1 as a nutrient sensor that plays a constitutive and ligand-induced role in preventing tissue damage. NR4A1 may also contribute to the overall health benefits of dietary polyphenolics.
Figure 4. Proposed model for NR4A1 as a nutrient sensor that plays a constitutive and ligand-induced role in preventing tissue damage. NR4A1 may also contribute to the overall health benefits of dietary polyphenolics.
Nutrients 17 02709 g004
Figure 5. Health-promoting effects of quercetin in various cell culture and laboratory animal models and their similarity to other synthetic NR4A1 ligands (Table 1).
Figure 5. Health-promoting effects of quercetin in various cell culture and laboratory animal models and their similarity to other synthetic NR4A1 ligands (Table 1).
Nutrients 17 02709 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Safe, S. NR4A1 Acts as a Nutrient Sensor That Inhibits the Effects of Aging. Nutrients 2025, 17, 2709. https://doi.org/10.3390/nu17162709

AMA Style

Safe S. NR4A1 Acts as a Nutrient Sensor That Inhibits the Effects of Aging. Nutrients. 2025; 17(16):2709. https://doi.org/10.3390/nu17162709

Chicago/Turabian Style

Safe, Stephen. 2025. "NR4A1 Acts as a Nutrient Sensor That Inhibits the Effects of Aging" Nutrients 17, no. 16: 2709. https://doi.org/10.3390/nu17162709

APA Style

Safe, S. (2025). NR4A1 Acts as a Nutrient Sensor That Inhibits the Effects of Aging. Nutrients, 17(16), 2709. https://doi.org/10.3390/nu17162709

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop