1
|
Park SW, Han MR. A pan-cancer analysis unveiling the function of NR4A family genes in tumor immune microenvironment, prognosis, and drug response. Genes Genomics 2024; 46:977-990. [PMID: 38976216 DOI: 10.1007/s13258-024-01539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND NR4A family genes play crucial roles in cancers. However, the role of NR4A family genes in cancers remains paradoxical as they promote or suppress tumorigenesis. OBJECTIVE We aimed to conduct comprehensive analyses of the association between the expression of NR4A family genes and tumor microenvironment (TME) based on bioinformatics methods. METHODS We collected RNA-seq data from 33 cancer types and 20 normal tissue sites from the TCGA and GTEx databases. Expression patterns of NR4A family genes and their associations with DNA methylation, miRNA, overall survival, drug responses, and tumor microenvironment were investigated. RESULTS Significant downregulation of all NR4A family genes was observed in 15 cancer types. DNA promoter methylation and expression of NR4A family genes were negatively correlated in five cancers. The expression of 10 miRNAs targeting NR4A family genes was negatively correlated with the expression of NR4A family genes. High expression of all NR4A family genes was associated with poor prognosis in stomach adenocarcinoma and increased expressions of NR4A2 and NR4A3 were associated with poor prognosis in adrenocortical carcinoma. In addition, we found an elevated expression of NR4A2, which enhances the response to various chemotherapeutic drugs, whereas NR4A3 decreases drug sensitivity. Interestingly, in breast cancer, NR4A3 was significantly associated with C2 (IFN-γ dominant), C3 (inflammatory), and C6 (TGF-β dominant) immune subtypes and infiltrated immune cell types, implying both oncogenic and tumor-suppressive functions of NR4A3 in breast cancer. CONCLUSION The NR4A family genes have the potential to serve as a diagnostic, prognostic, and immunological marker of human cancers.
Collapse
Affiliation(s)
- Seong-Woo Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Korea
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Korea.
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Korea.
| |
Collapse
|
2
|
Hashida R, Kawabata T. Structural Perspective of NR4A Nuclear Receptor Family and Their Potential Endogenous Ligands. Biol Pharm Bull 2024; 47:580-590. [PMID: 38432913 DOI: 10.1248/bpb.b23-00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
There are 48 nuclear receptors in the human genome, and many members of this superfamily have been implicated in human diseases. The NR4A nuclear receptor family consisting of three members, NR4A1, NR4A2, and NR4A3 (formerly annotated as Nur77, Nurr1, and NOR1, respectively), are still orphan receptors but exert pathological effects on immune-related and neurological diseases. We previously reported that prostaglandin A1 (PGA1) and prostaglandin A2 (PGA2) are potent activators of NR4A3, which bind directly to the ligand-binding domain (LBD) of the receptor. Recently, the co-crystallographic structures of NR4A2-LBD bound to PGA1 and PGA2 were reported, followed by reports of the neuroprotective effects of these possible endogenous ligands in mouse models of Parkinson's disease. Based on these structures, we modeled the binding structures of the other two members (NR4A1 and NR4A3) with these potential endogenous ligands using a template-based modeling method, and reviewed the similarity and diversity of ligand-binding mechanisms in the nuclear receptor family.
Collapse
Affiliation(s)
- Ryoichi Hashida
- Genox Research Inc
- Department of Microbiology, Matsumoto Dental University
| | - Takeshi Kawabata
- Department of Applied Information Sciences, Graduate School of Information Sciences, Tohoku University
| |
Collapse
|
3
|
Caba E, Sherman MD, Farizatto KLG, Alcira B, Wang HW, Giardina C, Shin DG, Sandefur CI, Bahr BA. Excitotoxic stimulation activates distinct pathogenic and protective expression signatures in the hippocampus. J Cell Mol Med 2021; 25:9011-9027. [PMID: 34414662 PMCID: PMC8435451 DOI: 10.1111/jcmm.16864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Excitotoxic events underlying ischaemic and traumatic brain injuries activate degenerative and protective pathways, particularly in the hippocampus. To understand opposing pathways that determine the brain's response to excitotoxicity, we used hippocampal explants, thereby eliminating systemic variables during a precise protocol of excitatory stimulation. N‐methyl‐d‐aspartate (NMDA) was applied for 20 min and total RNA isolated one and 24 h later for neurobiology‐specific microarrays. Distinct groups of genes exhibited early vs. delayed induction, with 63 genes exclusively reduced 24‐h post‐insult. Egr‐1 and NOR‐1 displayed biphasic transcriptional modulation: early induction followed by delayed suppression. Opposing events of NMDA‐induced genes linked to pathogenesis and cell survival constituted the early expression signature. Delayed degenerative indicators (up‐regulated pathogenic genes, down‐regulated pro‐survival genes) and opposing compensatory responses (down‐regulated pathogenic genes, up‐regulated pro‐survival genes) generated networks with temporal gene profiles mirroring coexpression network clustering. We then used the expression profiles to test whether NF‐κB, a potent transcription factor implicated in both degenerative and protective pathways, is involved in the opposing responses. The NF‐κB inhibitor MG‐132 indeed altered NMDA‐mediated transcriptional changes, revealing components of opposing expression signatures that converge on the single response element. Overall, this study identified counteracting avenues among the distinct responses to excitotoxicity, thereby suggesting multi‐target treatment strategies and implications for predictive medicine.
Collapse
Affiliation(s)
- Ebru Caba
- Vertex Pharmaceuticals, Cambridge, MA, USA.,Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, CT, USA
| | - Marcus D Sherman
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Britney Alcira
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Hsin-Wei Wang
- Bioinformatics and Biocomputing Institute, University of Connecticut, Storrs, CT, USA.,Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Dong-Guk Shin
- Bioinformatics and Biocomputing Institute, University of Connecticut, Storrs, CT, USA.,Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Conner I Sandefur
- Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Pharmacology and the Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.,Sandefur Modeling, Pittsboro, NC, USA
| | - Ben A Bahr
- Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, CT, USA.,Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Chemistry and Physics, University of North Carolina-Pembroke, Pembroke, NC, USA
| |
Collapse
|
4
|
Safe S, Karki K. The Paradoxical Roles of Orphan Nuclear Receptor 4A (NR4A) in Cancer. Mol Cancer Res 2020; 19:180-191. [PMID: 33106376 DOI: 10.1158/1541-7786.mcr-20-0707] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/22/2020] [Accepted: 10/19/2020] [Indexed: 11/16/2022]
Abstract
The three-orphan nuclear receptor 4A genes are induced by diverse stressors and stimuli, and there is increasing evidence that NR4A1 (Nur77), NR4A2 (Nurr1), and NR4A3 (Nor1) play an important role in maintaining cellular homeostasis and in pathophysiology. In blood-derived tumors (leukemias and lymphomas), NR4A expression is low and NR4A1-/-/NR4A3-/- double knockout mice rapidly develop acute myelocytic leukemia, suggesting that these receptors exhibit tumor suppressor activity. Treatment of leukemia and most lymphoma cells with drugs that induce expression of NR4A1and NR4A3 enhances apoptosis, and this represents a potential clinical application for treating this disease. In contrast, most solid tumor-derived cell lines express high levels of NR4A1 and NR4A2, and both receptors exhibit pro-oncogenic activities in solid tumors, whereas NR4A3 exhibits tumor-specific activities. Initial studies with retinoids and apoptosis-inducing agents demonstrated that their cytotoxic activity is NR4A1 dependent and involved drug-induced nuclear export of NR4A1 and formation of a mitochondrial proapoptotic NR4A1-bcl-2 complex. Drug-induced nuclear export of NR4A1 has been reported for many agents/biologics and involves interactions with multiple mitochondrial and extramitochondrial factors to induce apoptosis. Synthetic ligands for NR4A1, NR4A2, and NR4A3 have been identified, and among these compounds, bis-indole derived (CDIM) NR4A1 ligands primarily act on nuclear NR4A1 to inhibit NR4A1-regulated pro-oncogenic pathways/genes and similar results have been observed for CDIMs that bind NR4A2. Based on results of laboratory animal studies development of NR4A inducers (blood-derived cancers) and NR4A1/NR4A2 antagonists (solid tumors) may be promising for cancer therapy and also for enhancing immune surveillance.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| | - Keshav Karki
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| |
Collapse
|
5
|
Lipid-sensors, enigmatic-orphan and orphan nuclear receptors as therapeutic targets in breast-cancer. Oncotarget 2018; 7:42661-42682. [PMID: 26894976 PMCID: PMC5173165 DOI: 10.18632/oncotarget.7410] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/29/2016] [Indexed: 12/28/2022] Open
Abstract
Breast-cancer is heterogeneous and consists of various groups with different biological characteristics. Innovative pharmacological approaches accounting for this heterogeneity are needed. The forty eight human Nuclear-Hormone-Receptors are ligand-dependent transcription-factors and are classified into Endocrine-Receptors, Adopted-Orphan-Receptors (Lipid-sensors and Enigmatic-Orphans) and Orphan-receptors. Nuclear-Receptors represent ideal targets for the design/synthesis of pharmacological ligands. We provide an overview of the literature available on the expression and potential role played by Lipid-sensors, Enigmatic-Orphans and Orphan-Receptors in breast-cancer. The data are complemented by an analysis of the expression levels of each selected Nuclear-Receptor in the PAM50 breast-cancer groups, following re-elaboration of the data publicly available. The major aim is to support the idea that some of the Nuclear-Receptors represent largely unexploited therapeutic-targets in breast-cancer treatment/chemo-prevention. On the basis of our analysis, we conclude that the Lipid-Sensors, NR1C3, NR1H2 and NR1H3 are likely to be onco-suppressors in breast-cancer. The Enigmatic-Orphans, NR1F1 NR2A1 and NR3B3 as well as the Orphan-Receptors, NR0B1, NR0B2, NR1D1, NR2F1, NR2F2 and NR4A3 exert a similar action. These Nuclear-Receptors represent candidates for the development of therapeutic strategies aimed at increasing their expression or activating them in tumor cells. The group of Nuclear-Receptors endowed with potential oncogenic properties consists of the Lipid-Sensors, NR1C2 and NR1I2, the Enigmatic-Orphans, NR1F3, NR3B1 and NR5A2, as well as the Orphan-Receptors, NR2E1, NR2E3 and NR6A1. These oncogenic Nuclear-Receptors should be targeted with selective antagonists, reverse-agonists or agents/strategies capable of reducing their expression in breast-cancer cells.
Collapse
|
6
|
Deutsch AJ, Rinner B, Pichler M, Prochazka K, Pansy K, Bischof M, Fechter K, Hatzl S, Feichtinger J, Wenzl K, Frisch MT, Stiegelbauer V, Prokesch A, Krogsdam A, Sill H, Thallinger GG, Greinix HT, Wang C, Beham-Schmid C, Neumeister P. NR4A3 Suppresses Lymphomagenesis through Induction of Proapoptotic Genes. Cancer Res 2017; 77:2375-2386. [DOI: 10.1158/0008-5472.can-16-2320] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/12/2016] [Accepted: 02/22/2017] [Indexed: 11/16/2022]
|
7
|
Aesoy R, Clyne CD, Chand AL. Insights into Orphan Nuclear Receptors as Prognostic Markers and Novel Therapeutic Targets for Breast Cancer. Front Endocrinol (Lausanne) 2015; 6:115. [PMID: 26300846 PMCID: PMC4528200 DOI: 10.3389/fendo.2015.00115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/11/2015] [Indexed: 12/11/2022] Open
Abstract
There is emerging evidence asserting the importance of orphan nuclear receptors (ONRs) in cancer initiation and progression. In breast cancer, there is a lot unknown about ONRs in terms of their expression profile and their transcriptional targets in the various stages of tumor progression. With the classification of breast tumors into distinct molecular subtypes, we assess ONR expression in the different breast cancer subtypes and with patient outcomes. Complementing this, we review evidence implicating ONR-dependent molecular pathways in breast cancer progression to identify candidate ONRs as potential prognostic markers and/or as therapeutic targets.
Collapse
Affiliation(s)
- Reidun Aesoy
- Cancer Drug Discovery, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Colin D. Clyne
- Cancer Drug Discovery, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Ashwini L. Chand
- Cancer Drug Discovery, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- *Correspondence: Ashwini L. Chand,
| |
Collapse
|
8
|
Control of energy balance by hypothalamic gene circuitry involving two nuclear receptors, neuron-derived orphan receptor 1 and glucocorticoid receptor. Mol Cell Biol 2013; 33:3826-34. [PMID: 23897430 DOI: 10.1128/mcb.00385-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nuclear receptors (NRs) regulate diverse physiological processes, including the central nervous system control of energy balance. However, the molecular mechanisms for the central actions of NRs in energy balance remain relatively poorly defined. Here we report a hypothalamic gene network involving two NRs, neuron-derived orphan receptor 1 (NOR1) and glucocorticoid receptor (GR), which directs the regulated expression of orexigenic neuropeptides agouti-related peptide (AgRP) and neuropeptide Y (NPY) in response to peripheral signals. Our results suggest that the anorexigenic signal leptin induces NOR1 expression likely via the transcription factor cyclic AMP response element-binding protein (CREB), while the orexigenic signal glucocorticoid mobilizes GR to inhibit NOR1 expression by antagonizing the action of CREB. Also, NOR1 suppresses glucocorticoid-dependent expression of AgRP and NPY. Consistently, relative to wild-type mice, NOR1-null mice showed significantly higher levels of AgRP and NPY and were less responsive to leptin in decreasing the expression of AgRP and NPY. These results identify mutual antagonism between NOR1 and GR to be a key rheostat for peripheral metabolic signals to centrally control energy balance.
Collapse
|
9
|
Zhu X, Walton RG, Tian L, Luo N, Ho SR, Fu Y, Garvey WT. Prostaglandin A2 enhances cellular insulin sensitivity via a mechanism that involves the orphan nuclear receptor NR4A3. Horm Metab Res 2013; 45:213-20. [PMID: 23104421 PMCID: PMC4116744 DOI: 10.1055/s-0032-1327619] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We have previously reported that members of the NR4A family of orphan nuclear receptors can augment insulin's ability to stimulate glucose transport in adipocytes. In the current study, we endeavored to test for an insulin-sensitizing effect in muscle cells and to identify a potential transactivator. Lentiviral constructs were used to engineer both hyperexpression and shRNA silencing of NR4A3 in C2C12 myocytes. The NR4A3 hyper-expression construct led to a significant increase in glucose transport rates in the presence of maximal insulin while the NR4A3 knock-down exhibited a significant reduction in insulin-stimulated glucose transport rates. Consistently, insulin-mediated AKT phosphorylation was increased by NR4A3 hyperexpression and decreased following shRNA NR4A3 suppression. Then, we examined effects of prostaglandin A2 (PGA2) on insulin action and NR4A3 transactivation. PGA2 augmented insulin-stimulated glucose uptake in C2C12 myocytes and AKT phosphorylation after 12-h treatment, without significant effects on basal transport or basal AKT phosphorylation. More importantly, we demonstrated that PGA2 led to a greater improvement in insulin-stimulated glucose rates in NR4A3 overexpressing C2C12 myocytes, when compared with Lac-Z controls stimulated with insulin and PGA2. Moreover, the sensitizing effect of PGA2 was significantly diminished in NR4A3 knockdown myocytes compared to scramble controls. These results show for the first time that: (i) PGA2 augments insulin action in myocytes as manifested by enhanced stimulation of glucose transport and AKT phosphorylation; and (ii) the insulin sensitizing effect is dependent upon the orphan nuclear receptor NR4A3.
Collapse
Affiliation(s)
- X Zhu
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294-3360, USA.
| | | | | | | | | | | | | |
Collapse
|
10
|
Mohan HM, Aherne CM, Rogers AC, Baird AW, Winter DC, Murphy EP. Molecular pathways: the role of NR4A orphan nuclear receptors in cancer. Clin Cancer Res 2012; 18:3223-8. [PMID: 22566377 DOI: 10.1158/1078-0432.ccr-11-2953] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nuclear receptors are of integral importance in carcinogenesis. Manipulation of classic ligand-activated nuclear receptors, such as estrogen receptor blockade in breast cancer, is an important established cancer therapy. Orphan nuclear receptors, such as nuclear family 4 subgroup A (NR4A) receptors, have no known natural ligand(s). These elusive receptors are increasingly recognized as molecular switches in cell survival and a molecular link between inflammation and cancer. NR4A receptors act as transcription factors, altering expression of downstream genes in apoptosis (Fas-ligand, TRAIL), proliferation, DNA repair, metabolism, cell migration, inflammation (interleukin-8), and angiogenesis (VEGF). NR4A receptors are modulated by multiple cell-signaling pathways, including protein kinase A/CREB, NF-κB, phosphoinositide 3-kinase/AKT, c-jun-NH(2)-kinase, Wnt, and mitogen-activated protein kinase pathways. NR4A receptor effects are context and tissue specific, influenced by their levels of expression, posttranslational modification, and interaction with other transcription factors (RXR, PPAR-Υ). The subcellular location of NR4A "nuclear receptors" is also important functionally; novel roles have been described in the cytoplasm where NR4A proteins act both indirectly and directly on the mitochondria to promote apoptosis via Bcl-2. NR4A receptors are implicated in a wide variety of malignancies, including breast, lung, colon, bladder, and prostate cancer; glioblastoma multiforme; sarcoma; and acute and/or chronic myeloid leukemia. NR4A receptors modulate response to conventional chemotherapy and represent an exciting frontier for chemotherapeutic intervention, as novel agents targeting NR4A receptors have now been developed. This review provides a concise clinical overview of current knowledge of NR4A signaling in cancer and the potential for therapeutic manipulation.
Collapse
Affiliation(s)
- Helen M Mohan
- UCD Veterinary Sciences Centre, University College Dublin, Department of Surgery, St Vincent's University Hospital, Elm Park, Dublin, Ireland.
| | | | | | | | | | | |
Collapse
|
11
|
Riggins RB, Mazzotta MM, Maniya OZ, Clarke R. Orphan nuclear receptors in breast cancer pathogenesis and therapeutic response. Endocr Relat Cancer 2010; 17:R213-31. [PMID: 20576803 PMCID: PMC3518023 DOI: 10.1677/erc-10-0058] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors comprise a large family of highly conserved transcription factors that regulate many key processes in normal and neoplastic tissues. Most nuclear receptors share a common, highly conserved domain structure that includes a carboxy-terminal ligand-binding domain. However, a subgroup of this gene family is known as the orphan nuclear receptors because to date there are no known natural ligands that regulate their activity. Many of the 25 nuclear receptors classified as orphan play critical roles in embryonic development, metabolism, and the regulation of circadian rhythm. Here, we review the emerging role(s) of orphan nuclear receptors in breast cancer, with a particular focus on two of the estrogen-related receptors (ERRalpha and ERRgamma) and several others implicated in clinical outcome and response or resistance to cytotoxic or endocrine therapies, including the chicken ovalbumin upstream promoter transcription factors, nerve growth factor-induced B, DAX-1, liver receptor homolog-1, and retinoic acid-related orphan receptor alpha. We also propose that a clearer understanding of the function of orphan nuclear receptors in mammary gland development and normal mammary tissues could significantly improve our ability to diagnose, treat, and prevent breast cancer.
Collapse
Affiliation(s)
- Rebecca B. Riggins
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Mary M. Mazzotta
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Omar Z. Maniya
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Robert Clarke
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
- Department of Physiology and Biophysics, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| |
Collapse
|
12
|
The hypoxia-inducible factor 1/NOR-1 axis regulates the survival response of endothelial cells to hypoxia. Mol Cell Biol 2009; 29:5828-42. [PMID: 19720740 DOI: 10.1128/mcb.00945-09] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hypoxia induces apoptosis but also triggers adaptive mechanisms to ensure cell survival. Here we show that the prosurvival effects of hypoxia-inducible factor 1 (HIF-1) in endothelial cells are mediated by neuron-derived orphan receptor 1 (NOR-1). The overexpression of NOR-1 decreased the rate of endothelial cells undergoing apoptosis in cultures exposed to hypoxia, while the inhibition of NOR-1 increased cell apoptosis. Hypoxia upregulated NOR-1 mRNA levels in a time- and dose-dependent manner. Blocking antibodies against VEGF or SU5614 (a VEGF receptor 2 inhibitor) did not prevent hypoxia-induced NOR-1 expression, suggesting that NOR-1 is not induced by the autocrine secretion of VEGF in response to hypoxia. The reduction of HIF-1 alpha protein levels by small interfering RNAs, or by inhibitors of the phosphatidylinositol-3 kinase (PI3K)/Akt pathway or mTOR, significantly counteracted hypoxia-induced NOR-1 upregulation. Intracellular Ca(2+) was involved in hypoxia-induced PI3K/Akt activation and in the downstream NOR-1 upregulation. A hypoxia response element mediated the transcriptional activation of NOR-1 induced by hypoxia as we show by transient transfection and chromatin immunoprecipitation assays. Finally, the attenuation of NOR-1 expression reduced both basal and hypoxia-induced cIAP2 (cellular inhibitor of apoptosis protein 2) mRNA levels, while NOR-1 overexpression upregulated cIAP2. Therefore, NOR-1 is a downstream effector of HIF-1 signaling involved in the survival response of endothelial cells to hypoxia.
Collapse
|
13
|
Zhang T, Wang P, Ren H, Fan J, Wang G. NGFI-B nuclear orphan receptor Nurr1 interacts with p53 and suppresses its transcriptional activity. Mol Cancer Res 2009; 7:1408-15. [PMID: 19671681 DOI: 10.1158/1541-7786.mcr-08-0533] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nurr1 is a member of the NGFI-B nuclear orphan receptor family which includes two other members, Nur77 and Nor-1. Nurr1 is essential for the development and survival of dopaminergic neurons. It was reported that Nurr1 has antiapoptotic functions, however, the mechanisms by which Nurr1 mediates these effects remain unknown. Here, we show that overexpression of Nurr1 decreases Bax expression whereas knockdown of Nurr1 increases Bax expression. Nurr1 also interacts with p53 and represses its assembly. Furthermore, Nurr1 represses p53 transcriptional activity in interaction-dependent and dose-dependent manners. Moreover, Nurr1 protects cells from doxorubicin-induced apoptosis. These findings provide evidence that Nurr1 promotes cell survival through its interacting with and repressing p53, thus implicating that Nurr1 may play an important role in carcinogenesis and other diseases.
Collapse
Affiliation(s)
- Tao Zhang
- Laboratory of Molecular Neuropathology, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, People's Republic of China
| | | | | | | | | |
Collapse
|
14
|
Filion C, Motoi T, Olshen AB, Laé M, Emnett RJ, Gutmann DH, Perry A, Ladanyi M, Labelle Y. The EWSR1/NR4A3 fusion protein of extraskeletal myxoid chondrosarcoma activates the PPARG nuclear receptor gene. J Pathol 2009; 217:83-93. [PMID: 18855877 DOI: 10.1002/path.2445] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The NR4A3 nuclear receptor is implicated in the development of extraskeletal myxoid chondrosarcoma (EMC), primitive sarcoma unrelated to conventional chondrosarcomas, through a specific fusion with EWSR1 resulting in an aberrant fusion protein that is thought to disrupt the transcriptional regulation of specific target genes. We performed an expression microarray analysis of EMC tumours expressing the EWSR1/NR4A3 fusion protein, comparing their expression profiles to those of other sarcoma types. We thereby identified a set of genes significantly overexpressed in EMC relative to other sarcomas, including PPARG and NDRG2. Western blot or immunohistochemical analyses confirm that PPARG and NDRG2 are expressed in tumours positive for EWSR1/NR4A3. Bioinformatic analysis identified a DNA response element for EWSR1/NR4A3 in the PPARG promoter, and band-shift experiments and transient transfections indicate that EWSR1/NR4A3 can activate transcription through this element. Western blots further show that an isoform of the native NR4A3 receptor lacking the C-terminal domain is very highly expressed in tumours positive for EWSR1/NR4A3, and co-transfections of this isoform along with EWSR1/NR4A3 indicate that it may negatively regulate the activity of the fusion protein on the PPARG promoter. These results suggest that the overall expression of PPARG in EMC may be regulated in part by the balance between EWSR1/NR4A3 and NR4A3, and that PPARG may play a crucial role in the development of these tumours. The specific up-regulation of PPARG by EWSR1/NR4A3 may also have potential therapeutic implications.
Collapse
Affiliation(s)
- C Filion
- Human and Molecular Genetic Research Unit, Saint-François d'Assise Hospital Research Center, CHUQ, Quebec, Qc, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Pearen MA, Myers SA, Raichur S, Ryall JG, Lynch GS, Muscat GEO. The orphan nuclear receptor, NOR-1, a target of beta-adrenergic signaling, regulates gene expression that controls oxidative metabolism in skeletal muscle. Endocrinology 2008; 149:2853-65. [PMID: 18325999 DOI: 10.1210/en.2007-1202] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
beta 1-3-Adrenoreceptor (AR)-deficient mice are unable to regulate energy expenditure and develop diet-induced obesity on a high-fat diet. We determined previously that beta2-AR agonist treatment activated expression of the mRNA encoding the orphan nuclear receptor, NOR-1, in muscle cells and plantaris muscle. Here we show that beta2-AR agonist treatment significantly and transiently activated the expression of NOR-1 (and the other members of the NR4A subgroup) in slow-twitch oxidative soleus muscle and fast-twitch glycolytic tibialis anterior muscle. The activation induced by beta-adrenergic signaling is consistent with the involvement of protein kinase A, MAPK, and phosphorylation of cAMP response element-binding protein. Stable cell lines transfected with a silent interfering RNA targeting NOR-1 displayed decreased palmitate oxidation and lactate accumulation. In concordance with these observations, ATP production in the NOR-1 silent interfering RNA (but not control)-transfected cells was resistant to (azide-mediated) inhibition of oxidative metabolism and expressed significantly higher levels of hypoxia inducible factor-1alpha. In addition, we observed the repression of genes that promote fatty acid oxidation (peroxisomal proliferator-activated receptor-gamma coactivator-1alpha/beta and lipin-1alpha) and trichloroacetic acid cycle-mediated carbohydrate (pyruvate) oxidation [pyruvate dehydrogenase phosphatase 1 regulatory and catalytic subunits (pyruvate dehydrogenase phosphatases-1r and -c)]. Furthermore, we observed that beta2-AR agonist administration in mouse skeletal muscle induced the expression of genes that activate fatty acid oxidation and modulate pyruvate use, including PGC-1alpha, lipin-1alpha, FOXO1, and PDK4. Finally, we demonstrate that NOR-1 is recruited to the lipin-1alpha and PDK-4 promoters, and this is consistent with NOR-1-mediated regulation of these genes. In conclusion, NOR-1 is necessary for oxidative metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Michael A Pearen
- Institute for Molecular Bioscience, The University of Queensland, Queensland 4072, Australia
| | | | | | | | | | | |
Collapse
|
16
|
Martorell L, Martínez-González J, Crespo J, Calvayrac O, Badimon L. Neuron-derived orphan receptor-1 (NOR-1) is induced by thrombin and mediates vascular endothelial cell growth. J Thromb Haemost 2007; 5:1766-73. [PMID: 17596136 DOI: 10.1111/j.1538-7836.2007.02627.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND AIM Neuron-derived orphan receptor-1 (NOR-1) is a transcription factor overexpressed in human atherosclerotic plaques that is involved in vascular smooth muscle cell (VSMC) proliferation. The aim of this study was to analyze the role of NOR-1 in thrombin-induced endothelial cell growth. RESULTS Thrombin induced an early and transient up-regulation of NOR-1 in human umbilical vein endothelial cells (HUVEC). NOR-1 up-regulation by thrombin is dependent on multiple pathways, including cytosolic Ca(2+), activation of protein kinase C (PKC), mitogen-activated protein kinase (MAPK) pathways [both extracellular-regulated kinase (ERK) and p38 MAPK], and downstream activation of cAMP response element binding protein (CREB). The critical role of CREB in the induction of NOR-1 by thrombin was demonstrated using a dominant-negative of CREB. By site-direct mutagenesis we identified two CRE sites present at -79 and -53 bp in the NOR-1 promoter involved in the up-regulation of NOR-1 by thrombin. Inhibition of thrombin receptor PAR-1 abolished CREB activation, NOR-1 up-regulation and DNA synthesis (used as an index of cell proliferation). TRAP-6 mimicked both NOR-1 up-regulation and CREB activation induced by thrombin, while PPACK (an irreversible thrombin inhibitor) prevented such an effect. Direct inhibition of thrombin-induced NOR-1 up-regulation, using antisense oligonucleotides or siRNA against NOR-1, reduced DNA synthesis and endothelial cell re-growth after injury in an in vitro model of wound repair. CONCLUSIONS These results indicate that NOR-1 up-regulation plays a key role in thrombin-induced endothelial cell growth. Strategies aimed to block NOR-1 could be useful to prevent vascular effects triggered by thrombin.
Collapse
Affiliation(s)
- L Martorell
- Centro de Investigación Cardiovascular, CSIC/ICCC, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | |
Collapse
|
17
|
Pearen MA, Ryall JG, Maxwell MA, Ohkura N, Lynch GS, Muscat GEO. The orphan nuclear receptor, NOR-1, is a target of beta-adrenergic signaling in skeletal muscle. Endocrinology 2006; 147:5217-27. [PMID: 16901967 DOI: 10.1210/en.2006-0447] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
beta-Adrenergic receptor (beta-AR) agonists induce Nur77 mRNA expression in the C2C12 skeletal muscle cell culture model and elicit skeletal muscle hypertrophy. We previously demonstrated that Nur77 (NR4A1) is involved in lipolysis and gene expression associated with the regulation of lipid homeostasis. Subsequently it was demonstrated by another group that beta-AR agonists and cold exposure-induced Nur77 expression in brown adipocytes and brown adipose tissue, respectively. Moreover, NOR-1 (NR4A3) was hyperinduced by cold exposure in the nur77(-/-) animal model. These studies underscored the importance of understanding the role of NOR-1 in skeletal muscle. In this context we observed 30-480 min of beta-AR agonist treatment significantly and transiently increased expression of the orphan nuclear receptor NOR-1 in both mouse skeletal muscle tissue (plantaris) and C2C12 skeletal muscle cells. Specific beta(2)- and beta(3)-AR agonists had similar effects as the pan-agonist and were blocked by the beta-AR antagonist propranolol. Moreover, in agreement with these observations, isoprenaline also significantly increased the activity of the NOR-1 promoter. Stable exogenous expression of a NOR-1 small interfering RNA (but not the negative control small interfering RNA) in skeletal muscle cells significantly repressed endogenous NOR-1 mRNA expression and led to changes in the expression of genes involved in the control of lipid use and muscle mass underscored by a dramatic increase in myostatin mRNA expression. Concordantly the myostatin promoter was repressed by NOR-1 expression. In conclusion, NOR-1 is highly responsive to beta-adrenergic signaling and regulates the expression of genes controlling fatty acid use and muscle mass.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cells, Cultured
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Homeostasis
- Ion Channels/physiology
- Isoproterenol/pharmacology
- Lipid Metabolism
- Mice
- Mitochondrial Proteins/physiology
- Muscle Development
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- Myostatin
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Promoter Regions, Genetic
- RNA, Messenger/analysis
- RNA, Small Interfering/pharmacology
- Receptors, Adrenergic, beta/physiology
- Receptors, Cytoplasmic and Nuclear/physiology
- Receptors, Steroid/genetics
- Receptors, Steroid/physiology
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/physiology
- Signal Transduction/physiology
- Transcription Factors/physiology
- Transforming Growth Factor beta/genetics
- Uncoupling Protein 2
- Uncoupling Protein 3
Collapse
Affiliation(s)
- Michael A Pearen
- Institute for Molecular Bioscience, Division of Molecular Genetics and Development, University of Queensland, St. Lucia, Queensland 4072, Australia
| | | | | | | | | | | |
Collapse
|
18
|
Nomiyama T, Nakamachi T, Gizard F, Heywood EB, Jones KL, Ohkura N, Kawamori R, Conneely OM, Bruemmer D. The NR4A orphan nuclear receptor NOR1 is induced by platelet-derived growth factor and mediates vascular smooth muscle cell proliferation. J Biol Chem 2006; 281:33467-76. [PMID: 16945922 PMCID: PMC1829169 DOI: 10.1074/jbc.m603436200] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Members of the nuclear hormone receptor superfamily function as key transcriptional regulators of inflammation and proliferation in cardiovascular diseases. In addition to the ligand-dependent peroxisome proliferator-activated receptors and liver X receptors, this family of transcription factors includes a large number of orphan receptors, and their role in vascular diseases remains to be investigated. The neuron-derived orphan receptor-1 (NOR1) belongs to the ligand-independent NR4A subfamily, which has been implicated in cell proliferation, differentiation, and apoptosis. In this study, we demonstrate NOR1 expression in vascular smooth muscle cells (SMC) of human atherosclerotic lesions. In response to mitogenic stimulation with platelet-derived growth factor (PDGF), SMC rapidly express NOR1 through an ERK-MAPK-dependent signaling pathway. 5'-deletion analysis, site-directed mutagenesis, and transactivation experiments demonstrate that PDGF-induced NOR1 expression is mediated through a cAMP-response element-binding protein (CREB)-dependent transactivation of the NOR1 promoter. Consequently, short interfering RNA-mediated depletion of CREB abolished PDGF-induced NOR1 expression in SMC. Furthermore, PDGF induced Ser-133 phosphorylation of CREB and subsequent binding to the CRE sites of the endogenous NOR1 promoter. Functional analysis demonstrated that PDGF induces NOR1 transactivation of its consensus NGFI-B-response elements (NBRE) in SMC. We finally demonstrate that SMC isolated from NOR1-deficient mice exhibit decreased cell proliferation and characterize cyclin D1 and D2 as NOR1 target genes in SMC. These experiments indicate that PDGF-induced NOR1 transcription in SMC is mediated through CREB-dependent transactivation of the NOR1 promoter and further demonstrate that NOR1 functions as a key transcriptional regulator of SMC proliferation.
Collapse
MESH Headings
- Animals
- Atherosclerosis/metabolism
- Cell Proliferation
- Cells, Cultured
- Cyclic AMP Response Element-Binding Protein/genetics
- Cyclic AMP Response Element-Binding Protein/metabolism
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Gene Expression Regulation
- Humans
- MAP Kinase Signaling System
- Mitogen-Activated Protein Kinases/metabolism
- Molecular Sequence Data
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Phosphoserine/metabolism
- Platelet-Derived Growth Factor/pharmacology
- Promoter Regions, Genetic/genetics
- Protein Binding
- RNA, Messenger/genetics
- Rats
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- Response Elements
- Transcription, Genetic/genetics
- Transcriptional Activation/genetics
Collapse
Affiliation(s)
- Takashi Nomiyama
- From the Division of Endocrinology and Molecular Medicine, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Takafumi Nakamachi
- From the Division of Endocrinology and Molecular Medicine, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Florence Gizard
- From the Division of Endocrinology and Molecular Medicine, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Elizabeth B. Heywood
- From the Division of Endocrinology and Molecular Medicine, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Karrie L. Jones
- From the Division of Endocrinology and Molecular Medicine, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Naganari Ohkura
- National Cancer Center Research Institute, Tumor Endocrinology Project, Tokyo 104-0045, Japan
| | - Ryuzo Kawamori
- Department of Medicine, Metabolism, and Endocrinology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Orla M. Conneely
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Dennis Bruemmer
- From the Division of Endocrinology and Molecular Medicine, University of Kentucky College of Medicine, Lexington, Kentucky 40536
- To whom correspondence should be addressed: Wethington Health Sciences Bldg., Rm. 575, 900 South Limestone St., Lexington, KY 40536-0200. Tel.: 859-323-4933(ext.81418);Fax:859-257-3646;E-mail:
| |
Collapse
|
19
|
Maxwell MA, Muscat GEO. The NR4A subgroup: immediate early response genes with pleiotropic physiological roles. NUCLEAR RECEPTOR SIGNALING 2006; 4:e002. [PMID: 16604165 PMCID: PMC1402209 DOI: 10.1621/nrs.04002] [Citation(s) in RCA: 331] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2005] [Accepted: 12/20/2005] [Indexed: 12/20/2022]
Abstract
The nuclear hormone receptor (NR) superfamily includes the orphan NR4A subgroup, comprised of Nur77 (NR4A1), Nurr1 (NR4A2) and NOR-1 (NR4A3). These NRs are classified as early response genes, are induced by a diverse range of signals, including fatty acids, stress, growth factors, cytokines, peptide hormones, phorbol esters, neurotransmitters, and physical stimuli (for example magnetic fields, shear stress). The ability to sense and rapidly respond to changes in the cellular environment thus appears to be a hallmark of this subfamily. The members of the NR4A subgroup are well conserved in the DNA binding domain (~91-95%) and the C-terminal ligand-binding domain (~60%), but are divergent in the N-terminal AB region. These receptors bind as monomers, homodimers and heterodimers with RXRs (to mediate retinoid signaling) to different permutations of the canonical NR binding motif. The NR4A subgroup activates gene expression in a constitutive ligand-independent manner. NR4A-mediated trans-activation (LBD) involves unusually active N-terminal AF-1 domains that mediate coactivator recruitment. Moreover, the NR4A receptors encode atypical LBDs and AF-2 domains. For example, the LBDs contain no cavity due to bulky hydrophobic residue side chains, and lack the classical coactivator-binding cleft constituted by helices 3, 4 and 12. However, a hydrophobic patch exists between helices 11 and 12, that encodes a novel cofactor interface that modulates transcriptional activity. In line with the pleiotropic physiological stimuli that induce the NR4A subgroup, these orphan NRs have been implicated in cell cycle regulation (and apoptosis), neurological disease, steroidogenesis, inflammation, carcinogenesis and atherogenesis.
Collapse
Affiliation(s)
- Megan A Maxwell
- Institute for Molecular Bioscience, Division of Molecular Genetics and Development, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | | |
Collapse
|
20
|
Rius J, Martínez-González J, Crespo J, Badimon L. NOR-1 is involved in VEGF-induced endothelial cell growth. Atherosclerosis 2006; 184:276-82. [PMID: 15949808 DOI: 10.1016/j.atherosclerosis.2005.04.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2005] [Revised: 03/23/2005] [Accepted: 04/27/2005] [Indexed: 11/22/2022]
Abstract
Neuron-derived orphan receptor-1 (NOR-1) is a transcription factor over-expressed in human atherosclerotic plaques that is involved in vascular smooth muscle cell proliferation. The aim of this study was to analyze whether NOR-1 plays a role in vascular endothelial growth factor (VEGF) induced endothelial cell growth. VEGF induced an early and transient up-regulation of NOR-1 in human umbilical vein endothelial cells (HUVEC). NOR-1 up-regulation by VEGF is processed through VEGF receptor-2 (VEGFR-2) and involves different signaling pathways including increase in cytosolic Ca(2+), activation of protein kinase C and mitogen-activated protein kinase (MAPK) pathways (both extracellular-signaling regulated kinase [ERK] and p38 MAPK). VEGF induced CREB activation (phosphorylation in Ser(133)). In transfection assays, a dominant-negative of CREB inhibited NOR-1 promoter activity, while mutation of the three CRE sites in the NOR-1 promoter abolished VEGF-induced NOR-1 promoter activity. Antisense oligonucleotides against NOR-1 inhibited VEGF-induced endothelial cell growth (reduced DNA synthesis, and inhibited cell cycle progression and endothelial cell wound repair after mechanical injury). These results indicate that NOR-1 could be a key transcription factor regulating endothelial cell growth induced by VEGF.
Collapse
Affiliation(s)
- Jordi Rius
- Centro de Investigación Cardiovascular, CSIC/ICCC, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret # 167, Barcelona 8025, Spain
| | | | | | | |
Collapse
|
21
|
Darragh J, Soloaga A, Beardmore V, Wingate A, Wiggin G, Peggie M, Arthur J. MSKs are required for the transcription of the nuclear orphan receptors Nur77, Nurr1 and Nor1 downstream of MAPK signalling. Biochem J 2006; 390:749-59. [PMID: 15910281 PMCID: PMC1199668 DOI: 10.1042/bj20050196] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
MSK (mitogen- and stress-activated protein kinase) 1 and MSK2 are kinases activated downstream of either the ERK (extracellular-signal-regulated kinase) 1/2 or p38 MAPK (mitogen-activated protein kinase) pathways in vivo and are required for the phosphorylation of CREB (cAMP response element-binding protein) and histone H3. Here we show that the MSKs are involved in regulating the transcription of the immediate early gene Nur77. Stimulation of mouse embryonic fibroblasts with PMA, EGF (epidermal growth factor), TNF (tumour necrosis factor) or anisomycin resulted in induction of the Nur77 mRNA. The induction of Nur77 by TNF and anisomycin was abolished in MSK1/2 double-knockout cells, whereas induction was significantly reduced in response to PMA or EGF. The MSK responsive elements were mapped to two AP (activator protein)-1-like elements in the Nur77 promoter. The induction of Nur77 was also blocked by A-CREB, suggesting that MSKs control Nur77 transcription by phosphorylating CREB bound to the two AP-1-like elements. Consistent with the decrease in Nur77 mRNA levels in the MSK1/2-knockout cells, it was also found that MSKs were required for the induction of Nur77 protein by PMA and TNF. MSKs were also found to be required for the transcription of two genes related to Nur77, Nurr1 and Nor1, which were also transcribed in a CREB- or ATF1 (activating transcription factor-1)-dependent manner. Downstream of anisomycin signalling, a second ERK-dependent pathway, independent of MSK and CREB, was also required for the transcription of Nurr1 and Nor1.
Collapse
MESH Headings
- Animals
- Anisomycin
- CREB-Binding Protein/metabolism
- Cells, Cultured
- DNA-Binding Proteins/genetics
- Epidermal Growth Factor
- Fibroblasts
- MAP Kinase Signaling System
- Mice
- Mice, Knockout
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Nerve Tissue Proteins/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Nuclear Receptor Subfamily 4, Group A, Member 2
- Promoter Regions, Genetic
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Steroid/genetics
- Receptors, Thyroid Hormone/genetics
- Ribosomal Protein S6 Kinases/genetics
- Ribosomal Protein S6 Kinases/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/genetics
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Tetradecanoylphorbol Acetate
- Transcription Factors/genetics
- Transcription, Genetic
- Tumor Necrosis Factor-alpha
Collapse
Affiliation(s)
- Joanne Darragh
- MRC Protein Phosphorylation Unit, Faculty of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | - Ana Soloaga
- MRC Protein Phosphorylation Unit, Faculty of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | - Victoria A. Beardmore
- MRC Protein Phosphorylation Unit, Faculty of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | - Andrew D. Wingate
- MRC Protein Phosphorylation Unit, Faculty of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | - Giselle R. Wiggin
- MRC Protein Phosphorylation Unit, Faculty of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | - Mark Peggie
- MRC Protein Phosphorylation Unit, Faculty of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, U.K
| | | |
Collapse
|
22
|
Pirih FQ, Aghaloo TL, Bezouglaia O, Nervina JM, Tetradis S. Parathyroid hormone induces the NR4A family of nuclear orphan receptors in vivo. Biochem Biophys Res Commun 2005; 332:494-503. [PMID: 15910753 DOI: 10.1016/j.bbrc.2005.04.132] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Accepted: 04/26/2005] [Indexed: 11/29/2022]
Abstract
Parathyroid hormone (PTH) has both anabolic and catabolic effects on bone metabolism, although the molecular mechanisms mediating these effects are largely unknown. Among the transcription factors induced by PTH in osteoblasts are the nerve growth factor-inducible factor B (NR4A; NGFI-B) family of orphan nuclear receptors: Nurr1, Nur77, and NOR-1. PTH induces NR4A members through the cAMP-protein kinase A (PKA) pathway in vitro. We report here that PTH rapidly and transiently induced expression of all three NR4A genes in PTH-target tissues in vivo. In calvaria, long bones, and kidneys, NR4A induction was maximal 0.5-1 h after a single intraperitoneal (i.p.) injection of 80 microg/kg PTH. Nur77 demonstrated the highest expression, followed, in order, by Nurr1 and NOR-1. In calvaria and long bone, PTH-induced expression of each NR4A gene was detectable at 10 microg/kg i.p. with maximum induction at 40-80 microg/kg. PTH (3-34) did not induce NR4A mRNA levels in calvaria, long bone, and kidney in vivo, confirming our in vitro results that NR4A genes are induced primarily through the cAMP-PKA pathway. The magnitude of PTH-induced NR4A expression was comparable in vivo and in vitro. However, NR4A mRNA levels peaked and returned to baseline faster in vivo. Both in vivo and in vitro, PTH induced NR4A pre-mRNA levels suggesting that induction of these genes is, at least in part, through activation of mRNA synthesis. The in vivo induction of the NR4A family members by PTH suggests their involvement in, at least some, PTH-induced changes in bone metabolism.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cells, Cultured
- DNA-Binding Proteins/metabolism
- Dose-Response Relationship, Drug
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/physiology
- Mice
- Nerve Tissue Proteins/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Nuclear Receptor Subfamily 4, Group A, Member 2
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Parathyroid Hormone/pharmacology
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Flavia Q Pirih
- Division of Diagnostic and Surgical Sciences, UCLA School of Dentistry, Los Angeles, CA 90095, USA.
| | | | | | | | | |
Collapse
|
23
|
Wansa KDSA, Harris JM, Yan G, Ordentlich P, Muscat GEO. The AF-1 domain of the orphan nuclear receptor NOR-1 mediates trans-activation, coactivator recruitment, and activation by the purine anti-metabolite 6-mercaptopurine. J Biol Chem 2003; 278:24776-90. [PMID: 12709428 DOI: 10.1074/jbc.m300088200] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NOR-1/NR4A3 is an "orphan member" of the nuclear hormone receptor superfamily. NOR-1 and its close relatives Nurr1 and Nur77 are members of the NR4A subgroup of nuclear receptors. Members of the NR4A subgroup are induced through multiple signal transduction pathways. They have been implicated in cell proliferation, differentiation, T-cell apoptosis, chondrosarcomas, neurological disorders, inflammation, and atherogenesis. However, the mechanism of transcriptional activation, coactivator recruitment, and agonist-mediated activation remain obscure. Hence, we examined the molecular basis of NOR-1-mediated activation. We observed that NOR-1 trans-activates gene expression in a cell- and target-specific manner; moreover, it operates in an activation function (AF)-1-dependent manner. The N-terminal AF-1 domain delimited to between amino acids 1 and 112, preferentially recruits the steroid receptor coactivator (SRC). Furthermore, SRC-2 modulates the activity of the AF-1 domain but not the C-terminal ligand binding domain (LBD). Homology modeling indicated that the NOR-1 LBD was substantially different from that of hRORbeta, a closely related AF-2-dependent receptor. In particular, the hydrophobic cleft characteristic of nuclear receptors was replaced with a very hydrophilic surface with a distinct topology. This observation may account for the inability of this nuclear receptor LBD to efficiently mediate cofactor recruitment and transcriptional activation. In contrast, the N-terminal AF-1 is necessary for cofactor recruitment and can independently conscript coactivators. Finally, we demonstrate that the purine anti-metabolite 6-mercaptopurine, a widely used antineoplastic and anti-inflammatory drug, activates NOR-1 in an AF-1-dependent manner. Additional 6-mercaptopurine analogs all efficiently activated NOR-1, suggesting that the signaling pathways that modulate proliferation via inhibition of de novo purine and/or nucleic acid biosynthesis are involved in the regulation NR4A activity. We hypothesize that the NR4A subgroup mediates the genotoxic stress response and suggest that this subgroup may function as sensors that respond to genotoxicity.
Collapse
|
24
|
Maltais A, Filion C, Labelle Y. The AF2 domain of the orphan nuclear receptor TEC is essential for the transcriptional activity of the oncogenic fusion protein EWS/TEC. Cancer Lett 2002; 183:87-94. [PMID: 12049818 DOI: 10.1016/s0304-3835(02)00104-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The EWS/TEC fusion protein encoded by the t(9:22) chromosomal translocation in human extraskeletal myxoid chondrosarcoma tumors is thought to participate in the tumoral process at least in part by deregulating the expression of specific target genes involved in the control of cell proliferation. In this work we show that the activation function-2 (AF2) domain of TEC is essential for the transcriptional activity of the EWS/TEC fusion protein. Significantly, deleting only the last 15 amino acids of the fusion protein, which contains 949 amino acids in its full form, results in a loss of over 70% of its transcriptional activity in transfected human chondrocyte cell lines. Point mutation analyses indicate that within the AF2 domain, amino acid residues I939, D940 and F943 all play a crucial role in the activity of EWS/TEC. Comparable results were obtained with the native TEC receptor. These results suggest that EWS/TEC interacts at least in part with the same transcriptional coactivators as the native TEC receptor, and that these coactivators may be involved in the tumoral process leading to human chondrosarcoma tumors.
Collapse
MESH Headings
- 3T3 Cells
- Amino Acid Sequence
- Animals
- Binding Sites
- Bone Neoplasms/genetics
- Chondrosarcoma/genetics
- Chromosomes, Human, Pair 22
- Chromosomes, Human, Pair 9
- Cloning, Molecular
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/genetics
- Gene Expression Regulation, Neoplastic
- Heterogeneous-Nuclear Ribonucleoproteins
- Humans
- Mice
- Molecular Sequence Data
- Nerve Tissue Proteins
- Nuclear Proteins/chemistry
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- RNA-Binding Protein EWS
- RNA-Binding Proteins/chemistry
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Receptors, Cytoplasmic and Nuclear
- Receptors, Steroid
- Receptors, Thyroid Hormone
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/metabolism
- Ribonucleoproteins/genetics
- Sequence Alignment
- Sequence Homology, Amino Acid
- Transcription, Genetic
- Transfection
- Translocation, Genetic
Collapse
Affiliation(s)
- Annie Maltais
- Unité de Recherche en Génétique Humaine et Moléculaire, Pavillon Saint-François d'Assise, CHUQ, 10 rue de l'Espinay, G1L 3L5, Québec, QC, Canada
| | | | | |
Collapse
|
25
|
Ohkubo T, Sugawara Y, Sasaki K, Maruyama K, Ohkura N, Makuuchi M. Early induction of nerve growth factor-induced genes after liver resection-reperfusion injury. J Hepatol 2002; 36:210-7. [PMID: 11830332 DOI: 10.1016/s0168-8278(01)00258-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND/AIMS Nur-related factor 1 (Nurr1) has been implicated in liver generation after hepatectomy. We hypothesized that the genes in the nerve growth factor-induced gene B (NGFI-B) family was induced in liver ischemia-reperfusion injury. METHODS Expression of the NGFI-B family genes was examined by the reverse transcription-polymerase chain reaction in rat and human livers. In situ hybridization was performed to check the localization of the NGFI-B gene in rat liver. Expression of phospho-Ser-133-specific cyclic adenosine-3' :5'-monophosphate response element binding (pCREB) protein was examined by Western blot analysis and gel shift assay, since the promoter region of the NGFI-B family genes contains CRE. RESULTS The expression of the NGFI-B family genes were recognized within 30 min after ischemia-reperfusion in rat liver, which was augmented by cycloheximide injection. In human specimens, the NGFI-B family genes expression was stronger than that before ischemic insult. pCREB protein was detected in the rat liver sampled 15 min after reperfusion. Gel shift assay suggested that CREB bound to neuron-derived orphan receptor gene in rat liver cells. CONCLUSIONS We recognized the early induction of the NGFI-B family genes after ischemia-reperfusion injury in rat and human livers. A pathway via CREB may be responsible for the induction.
Collapse
Affiliation(s)
- Takao Ohkubo
- Hepatobiliary Pancreatic Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | | | |
Collapse
|