1
|
Ahuja P, Bi X, Ng CF, Tse MCL, Hang M, Pang BPS, Iu ECY, Chan WS, Ooi XC, Sun A, Herlea-Pana O, Liu Z, Yang X, Jiao B, Ma X, Wu KKL, Lee LTO, Cheng KKY, Lee CW, Chan CB. Src homology 3 domain binding kinase 1 protects against hepatic steatosis and insulin resistance through the Nur77-FGF21 pathway. Hepatology 2023; 77:213-229. [PMID: 35363898 DOI: 10.1002/hep.32501] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS Metabolism in the liver is dysregulated in obesity, contributing to various health problems including steatosis and insulin resistance. While the pathogenesis of lipid accumulation has been extensively studied, the protective mechanism against lipid challenge in the liver remains unclear. Here, we report that Src homology 3 domain binding kinase 1 (SBK1) is a regulator of hepatic lipid metabolism and systemic insulin sensitivity in response to obesity. APPROACH AND RESULTS Enhanced Sbk1 expression was found in the liver of high-fat diet (HFD)-induced obese mice and fatty acid (FA)-challenged hepatocytes. SBK1 knockdown in mouse liver cells augmented FA uptake and lipid accumulation. Similarly, liver-specific SBK1 knockout ( Lsko ) mice displayed more severe hepatosteatosis and higher expression of genes in FA uptake and lipogenesis than the Flox/Flox ( Fl/Fl ) control mice when fed the HFD. The HFD-fed Lsko mice also showed symptoms of hyperglycemia, poor systemic glucose tolerance, and lower insulin sensitivity than the Fl/Fl mice. On the other hand, hepatic Sbk1 overexpression alleviated the high-fructose diet-induced hepatosteatosis, hyperlipidemia, and hyperglycemia in mice. White adipose tissue browning was also observed in hepatic SBK1 -overexpressed mice. Moreover, we found that SBK1 was a positive regulator of FGF21 in the liver during energy surplus conditions. Mechanistically, SBK1 phosphorylates the orphan nuclear receptor 4A1 (Nur77) on serine 344 to promote hepatic FGF21 expression and inhibit the transcription of genes involved in lipid anabolism. CONCLUSIONS Collectively, our data suggest that SBK1 is a regulator of the metabolic adaption against obesity through the Nur77-FGF21 pathway.
Collapse
Affiliation(s)
- Palak Ahuja
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Xinyi Bi
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Chun Fai Ng
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | | | - Miaojia Hang
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Brian Pak Shing Pang
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Elsie Chit Yu Iu
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Wing Suen Chan
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Xin Ci Ooi
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Anqi Sun
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Oana Herlea-Pana
- Department of Physiology , The University of Oklahoma Health Sciences Center , Oklahoma City , Oklahoma , USA
| | - Zhixue Liu
- Center for Molecular & Translational Medicine , Georgia State University , Atlanta , Georgia , USA
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing , Key Laboratory of Drug Target and Screening Research , Institute of Materia Medica of Peking Union Medical College , Beijing , China
| | - Baowei Jiao
- State Key Laboratory of Genetic Resources and Evolution , Kunming Institute of Zoology , Chinese Academy of Sciences , Kunming , China
| | - Xin Ma
- Cancer Centre , Faculty of Health Sciences , University of Macau , Taipa, Macau , China
| | - Kelvin Ka Lok Wu
- Department of Health Technology and Informatics , The Hong Kong Polytechnic University , Hong Kong SAR , China
| | - Leo Tsz On Lee
- Cancer Centre , Faculty of Health Sciences , University of Macau , Taipa, Macau , China
- MOE Frontiers Science Center for Precision Oncology , University of Macau , Taipa, Macau , China
| | - Kenneth King Yip Cheng
- Department of Health Technology and Informatics , The Hong Kong Polytechnic University , Hong Kong SAR , China
| | - Chi Wai Lee
- School of Biomedical Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Chi Bun Chan
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
- State Key Laboratory of Pharmaceutical Biotechnology , The University of Hong Kong , Hong Kong SAR , China
| |
Collapse
|
2
|
hUMSCs Transplantation Regulates AMPK/NR4A1 Signaling Axis to Inhibit Ovarian Fibrosis in POI Rats. Stem Cell Rev Rep 2022:10.1007/s12015-022-10469-y. [PMID: 36307672 DOI: 10.1007/s12015-022-10469-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The mechanism of human Umbilical Cord Mesenchymal Stem Cells (hUMSCs) transplantation to improve ovarian function in the rats with Premature Ovarian Insufficiency (POI) is still unclear. The aim of this study is to investigate the signal axis mechanism that is involved in the ovarian function recovery of POI rats following hUMSCs transplantation. METHODS The rat model with POI was established by intraperitoneal injection of cisplatin. The hUMSCs were transplanted by caudal vein injection into POI rats. Hematoxylin-eosin (H&E) staining was performed to examine the morphology of rat ovarian tissue. Masson staining, Sirus red staining and immunofluorescence were used to observe the fibrosis extent of ovarian tissue. The levels of serum sex hormones and the expression of fibrosis related markers in ovarian tissues were measured by enzyme-linked immunosorbent assay (ELISA). The expression of NR4A1, Phospho-NR4A1 and AMP-activated protein kinase (AMPK) signaling in rat ovarian tissues was measured by immunohistochemistry and immunofluorescence. The role of AMPK/NR4A1 signaling axis in the regulation of ovarian function recovery in POI rats following hUMSCs transplantation was further investigated by adenovirus and siRNA intervention in isolated stromal cells. RESULTS The results showed that the hUMSCs transplantation significantly inhibited ovarian tissue fibrosis and restored the ovarian function in POI rats. The level of NR4A1 and AMPK expression in ovarian tissue of POI rats after hUMSCs transplantation was significantly increased compared with the control group. In the cultured ovarian stromal cells, the similar results were obtained on the expression of NR4A1 and its regulation on fibrosis related molecular markers in Cisplatin (CDDP) damaged stromal cells following hUMSCs supernatant treatment. Both hUMSCs supernatant treatment and the addition of AMPK inhibitors increased NR4A1 expression in stromal cells. And after NR4A1 molecular intervention, fibrosis-related indicators in stromal cells changed. The data suggests that the AMPK/NR4A1 signaling axis is involved in the ovarian function changes in POI rats following hUMSCs transplantation. CONCLUSION The data from this study indicate that the inhibition of tissue fibrosis and recovery of ovarian function is regulated by AMPK/NR4A1 signaling axis in POI rats following hUMSCs transplantation.
Collapse
|
3
|
Hiwa R, Brooks JF, Mueller JL, Nielsen HV, Zikherman J. NR4A nuclear receptors in T and B lymphocytes: Gatekeepers of immune tolerance . Immunol Rev 2022; 307:116-133. [PMID: 35174510 DOI: 10.1111/imr.13072] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/30/2022] [Indexed: 12/21/2022]
Abstract
Random VDJ recombination early in T and B cell development enables the adaptive immune system to recognize a vast array of evolving pathogens via antigen receptors. However, the potential of such randomly generated TCRs and BCRs to recognize and respond to self-antigens requires layers of tolerance mechanisms to mitigate the risk of life-threatening autoimmunity. Since they were originally cloned more than three decades ago, the NR4A family of nuclear hormone receptors have been implicated in many critical aspects of immune tolerance, including negative selection of thymocytes, peripheral T cell tolerance, regulatory T cells (Treg), and most recently in peripheral B cell tolerance. In this review, we discuss important insights from many laboratories as well as our own group into the function and mechanisms by which this small class of primary response genes promotes self-tolerance and immune homeostasis to balance the need for host defense against the inherent risks posed by the adaptive immune system.
Collapse
Affiliation(s)
- Ryosuke Hiwa
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
- Department of Rheumatology and Clinical Immunology, Kyoto University Hospital, Kyoto, Japan
| | - Jeremy F Brooks
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - James L Mueller
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - Hailyn V Nielsen
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| | - Julie Zikherman
- Division of Rheumatology, Department of Medicine, Rosalind Russell and Ephraim P. Engelman Arthritis Research Center, University of California, San Francisco, California, USA
| |
Collapse
|
4
|
di Martino O, Niu H, Hadwiger G, Ferris MA, Welch JS. Cytokine exposure mediates transcriptional activation of the orphan nuclear receptor Nur77 in hematopoietic cells. J Biol Chem 2021; 297:101240. [PMID: 34571009 PMCID: PMC8528724 DOI: 10.1016/j.jbc.2021.101240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 11/20/2022] Open
Abstract
The orphan nuclear receptor Nur77 is an immediate-early response gene that based on tissue and cell context is implicated in a plethora of cellular processes, including proliferation, differentiation, apoptosis, metabolism, and inflammation. Nur77 has a ligand-binding pocket that is obstructed by hydrophobic side groups. Naturally occurring, cell-endogenous ligands have not been identified, and Nur77 transcriptional activity is thought to be regulated through posttranslational modification and modulation of protein levels. To determine whether Nur77 is transcriptionally active in hematopoietic cells in vivo, we used an upstream activating sequence (UAS)-GFP transgenic reporter. We found that Nur77 is transcriptionally inactive in vivo in hematopoietic cells under basal conditions, but that activation occurs following cytokine exposure by G-CSF or IL-3. We also identified a series of serine residues required for cytokine-dependent transactivation of Nur77. Moreover, a kinase inhibitor library screen and proximity labeling-based mass spectrometry identified overlapping kinase pathways that physically interacted with Nur77 and whose inhibition abrogated cytokine-induced activation of Nur77. We determined that transcriptional activation of Nur77 by G-CSF or IL-3 requires functional JAK and mTor signaling since their inhibition leads to Nur77 transcriptional inactivation. Thus, intracellular cytokine signaling networks appear to regulate Nur77 transcriptional activity in mouse hematopoietic cells.
Collapse
Affiliation(s)
- Orsola di Martino
- Department of Internal Medicine, Washington University, St Louis, Missouri, USA
| | - Haixia Niu
- Department of Internal Medicine, Washington University, St Louis, Missouri, USA; Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Gayla Hadwiger
- Department of Internal Medicine, Washington University, St Louis, Missouri, USA
| | - Margaret A Ferris
- Department of Pediatrics, Washington University, St Louis, Missouri, USA
| | - John S Welch
- Department of Internal Medicine, Washington University, St Louis, Missouri, USA.
| |
Collapse
|
5
|
He L, Yuan L, Yu W, Sun Y, Jiang D, Wang X, Feng X, Wang Z, Xu J, Yang R, Zhang W, Feng H, Chen HZ, Zeng YA, Hui L, Wu Q, Zhang Y, Zhang L. A Regulation Loop between YAP and NR4A1 Balances Cell Proliferation and Apoptosis. Cell Rep 2020; 33:108284. [PMID: 33086070 DOI: 10.1016/j.celrep.2020.108284] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 07/31/2020] [Accepted: 09/25/2020] [Indexed: 01/03/2023] Open
Abstract
The Hippo signaling pathway maintains organ size and tissue homeostasis via orchestration of cell proliferation and apoptosis. How this pathway triggers cell apoptosis remains largely unexplored. Here, we identify NR4A1 as a target of the Hippo pathway that mediates the pro-apoptotic and anti-tumor effects of the Hippo pathway whereby YAP regulates the transcription, phosphorylation, and mitochondrial localization of NR4A1. NR4A1, in turn, functions as a feedback inhibitor of YAP to promote its degradation, thereby inhibiting the function of YAP during liver regeneration and tumorigenesis. Our studies elucidate a regulatory loop between NR4A1 and YAP to coordinate Hippo signaling activity during liver regeneration and tumorigenesis and highlight NR4A1 as a marker of Hippo signaling, as well as a therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Lingli He
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang Yuan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wentao Yu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yang Sun
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Jiang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaodong Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xue Feng
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zuoyun Wang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinjin Xu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Ruizeng Yang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenxiang Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua Feng
- Omics Core of Bio-Med Big Data Center, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hang-Zi Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lijian Hui
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiao Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Yonglong Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Lei Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Suzhou 215121, China.
| |
Collapse
|
6
|
NR4A nuclear receptors restrain B cell responses to antigen when second signals are absent or limiting. Nat Immunol 2020; 21:1267-1279. [PMID: 32868928 PMCID: PMC8081071 DOI: 10.1038/s41590-020-0765-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Abstract
Antigen stimulation (signal 1) triggers B cell proliferation, and primes B cells to recruit, engage, and respond to T cell help (signal 2). Failure to receive signal 2 within a defined time window results in B cell apoptosis, yet the mechanisms that enforce dependence upon co-stimulation are incompletely understood. Nr4a1-3 encode a small family of orphan nuclear receptors that are rapidly induced by B cell antigen receptor (BCR) stimulation. Here we showed that Nr4a1 and Nr4a3 play partially redundant roles to restrain B cell responses to antigen in the absence of co-stimulation, and do so in part by repressing expression of BATF and consequently MYC. The NR4A family also restrains B cell access to T cell help by repressing expression of the T cell chemokines CCL3 and CCL4, as well as CD86 and ICAM1. Such NR4A-mediated regulation plays a role specifically under conditions of competition for limiting T cell help.
Collapse
|
7
|
Zárraga-Granados G, Muciño-Hernández G, Sánchez-Carbente MR, Villamizar-Gálvez W, Peñas-Rincón A, Arredondo C, Andrés ME, Wood C, Covarrubias L, Castro-Obregón S. The nuclear receptor NR4A1 is regulated by SUMO modification to induce autophagic cell death. PLoS One 2020; 15:e0222072. [PMID: 32210435 PMCID: PMC7094859 DOI: 10.1371/journal.pone.0222072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/08/2020] [Indexed: 12/31/2022] Open
Abstract
NR4A is a nuclear receptor protein family whose members act as sensors of cellular environment and regulate multiple processes such as metabolism, proliferation, migration, apoptosis, and autophagy. Since the ligand binding domains of these receptors have no cavity for ligand interaction, their function is most likely regulated by protein abundance and post-translational modifications. In particular, NR4A1 is regulated by protein abundance, phosphorylation, and subcellular distribution (nuclear-cytoplasmic translocation), and acts both as a transcription factor and as a regulator of other interacting proteins. SUMOylation is a post-translational modification that can affect protein stability, transcriptional activity, alter protein-protein interactions and modify intracellular localization of target proteins. In the present study we evaluated the role of SUMOylation as a posttranslational modification that can regulate the activity of NR4A1 to induce autophagy-dependent cell death. We focused on a model potentially relevant for neuronal cell death and demonstrated that NR4A1 needs to be SUMOylated to induce autophagic cell death. We observed that a triple mutant in SUMOylation sites has reduced SUMOylation, increased transcriptional activity, altered intracellular distribution, and more importantly, its ability to induce autophagic cell death is impaired.
Collapse
Affiliation(s)
- Gabriela Zárraga-Granados
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Gabriel Muciño-Hernández
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - María R. Sánchez-Carbente
- Biotechnology Research Center, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Wendy Villamizar-Gálvez
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Ana Peñas-Rincón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
| | - Cristian Arredondo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María E. Andrés
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christopher Wood
- Laboratorio Nacional de Microscopía Avanzada, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Luis Covarrubias
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Susana Castro-Obregón
- División de Neurociencias, Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, México
- * E-mail:
| |
Collapse
|
8
|
Jin F, Li X, Deng Y, Timilshina M, Huang B, Kim DY, Chang JH, Ichinose H, Baek SH, Murakami M, Lee YJ, Chang HW. The orphan nuclear receptor NR4A1 promotes FcεRI-stimulated mast cell activation and anaphylaxis by counteracting the inhibitory LKB1/AMPK axis. Allergy 2019; 74:1145-1156. [PMID: 30565708 DOI: 10.1111/all.13702] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 10/26/2018] [Accepted: 11/05/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Nuclear receptor subfamily 4 group A member 1 (NR4A1), an orphan nuclear receptor, has been implicated in several biological events such as metabolism, apoptosis, and inflammation. Recent studies indicate a potential role for NR4A1 in mast cells, yet its role in allergic responses remains largely unknown. OBJECTIVES The aim of this study was to clarify the role of NR4A1 in mast cell activation and anaphylaxis. METHODS To evaluate the function of NR4A1 in mast cells, the impacts of siRNA knockdown, gene knockout, adenoviral overexpression, and pharmacological inhibition of NR4A1 on FcεRI signaling and effector functions in mouse bone marrow-derived mast cells (BMMCs) in vitro and on anaphylactic responses in vivo were evaluated. RESULTS Knockdown or knockout of NR4A1 markedly suppressed degranulation and lipid mediator production by FcεRI-crosslinked BMMCs, while its overexpression augmented these responses. Treatment with a NR4A1 antagonist also blocked mast cell activation to a similar extent as NR4A1 knockdown or knockout. Moreover, mast cell-specific NR4A1-deficient mice displayed dampened anaphylactic responses in vivo. Mechanistically, NR4A1 promoted FcεRI signaling by counteracting the liver kinase B1 (LKB1)/adenosine monophosphate-activated protein kinase (AMPK) axis. Following FcεRI crosslinking, NR4A1 bound to the LKB1/AMPK complex and sequestered it in the nucleus, thereby promoting FcεRI downstream signaling pathways. Silencing or knockout of LKB1/AMPK largely abrogated the effect of NR4A1 on mast cell activation. Additionally, NR4A1 facilitated spleen tyrosine kinase activation independently of LKB1/AMPK. CONCLUSIONS Nuclear receptor subfamily 4 group A member 1 positively regulates mast cell activation by antagonizing the LKB1-AMPK-dependent negative regulatory axis. This finding may provide a novel therapeutic strategy for the development of anti-allergic compounds.
Collapse
Affiliation(s)
- Fansi Jin
- College of Pharmacy; Yeungnam University; Gyeongsan Korea
| | - Xian Li
- College of Pharmacy; Yeungnam University; Gyeongsan Korea
| | - Yifeng Deng
- College of Pharmacy; Yeungnam University; Gyeongsan Korea
| | | | - Bin Huang
- Department of Biochemistry and Molecular Biology; College of Medicine; Yeungnam University; Daegu Korea
| | - Dong-Young Kim
- College of Pharmacy; Yeungnam University; Gyeongsan Korea
| | - Jae-Hoon Chang
- College of Pharmacy; Yeungnam University; Gyeongsan Korea
| | - Hiroshi Ichinose
- School of Life Science and Technology; Tokyo Institute of Technology; Yokohama Japan
| | - Suk-Hwan Baek
- Department of Biochemistry and Molecular Biology; College of Medicine; Yeungnam University; Daegu Korea
| | - Makoto Murakami
- Laboratory of Microenvironmental Metabolic Health Sciences; Center for Disease Biology and Integrative Medicine; Graduate School of Medicine; The University of Tokyo; Hongo, Bunkyo-ku Japan
| | - Youn Ju Lee
- Department of Pharmacology; School of Medicine; Catholic University of Daegu; Daegu Korea
| | | |
Collapse
|
9
|
Peng J, Zhao S, Li Y, Niu G, Chen C, Ye T, Zhao D, Zeng H. DLL4 and Jagged1 are angiogenic targets of orphan nuclear receptor TR3/Nur77. Microvasc Res 2019; 124:67-75. [PMID: 30930165 DOI: 10.1016/j.mvr.2019.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/22/2019] [Accepted: 03/27/2019] [Indexed: 12/12/2022]
Abstract
Pathological angiogenesis is a hallmark of many diseases. Previously, we reported that orphan nuclear receptor TR3/Nur77 was a critical mediator of angiogenesis to regulate tumor growth and skin wound healing via regulating the expression of the junctional proteins and integrins. However, the molecular mechanism, by which TR3/Nur77 regulates angiogenesis is not completely understood. Here, we were the first to find that TR3/Nur77, via its various amino acid fragments, regulated the expression of DLL4 and Jagged 1 in cultured endothelial cells. DLL4 and Jagged1 mediated TR3/Nur77-induced angiogenic responses and signaling molecules, but not the expression of integrins. Instead, integrins regulated the expressions of DLL4 and Jagged1 induced by TR3/Nur77. Further, DLL4, Jagged1 and integrins α1, α2, β3 and β5 were regulated by TR3/Nur77 in animal sepsis models of lipopolysaccharide (LPS)-induced endotoxemia, and cecal ligation and puncture (CLP), in which, TR3/Nur77 expression was significantly and tranciently increased. Mouse survival rates were greatly increased in Nur77 knockout mice bearing both CLP and LPS models. The results elucidated a novel axis of VEGF/histamine ➔ TR3/Nur77 ➔ integrins ➔ DLL4/Jagged1 in angiogenesis, and demonstrated that TR3/Nur77 was an excellent target for sepsis. These studies supported our previous findings that TR3/Nur77 was an excellent therapeutic target, and further our understanding of the molecular mechanism, by which TR3/Nur77 regulated angiogenesis.
Collapse
Affiliation(s)
- Jin Peng
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Radiotherapy and Medical Oncology Department, Zhongnan Hospital, Wuhan University, Wuhan, PR China
| | - Shengqiang Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, PR China
| | - Yan Li
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, PR China
| | - Gengming Niu
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Chen Chen
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Surgery of Breast and Thyroid, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Taiyang Ye
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA; Department of Obstetrics & Gynecology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200127, PR China
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
10
|
The involvement of NR4A1 and NR4A2 in the regulation of the luteal function in rats. Acta Histochem 2018; 120:713-719. [PMID: 30097186 DOI: 10.1016/j.acthis.2018.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/29/2018] [Accepted: 07/30/2018] [Indexed: 11/23/2022]
Abstract
The nuclear receptor 4A (NR4A) members play important roles in cellular proliferation, differentiation and apoptosis. The current study first evaluate the expression of ovarian NR4A1 during different luteal stages in rats. Immature rats aged 28 days were treated with sequential Pregnant mare serum gonadotropin (PMSG) (D -2) / human chorionic gonadotropin (hCG) (D 0) to induce pseudopregnancy. Serum progesterone (P4) and ovarian expression of NR4A1 were detected by RIA and WB, respectively, at follicle stage (D 0), early (D 2), middle (D 7) and late (D 14 and D 20) luteal stages. To confirm the role of NR4A1 during the luteal regression, rats were treated with prostaglandin F2α analog (PGF) for 0-8 h on D 7 to detect the expressions of NR4A1 and NR4A2. RIA result showed that serum P4 reached highest level on D 7 and then declined. WB results showed that there were two types of NR4A1 (NR4A1-L and NR4A1-S) expressed in the ovary. The ovarian NR4A1-L decreased at the late luteal stage (D 20). However, the NR4A1-S increased at the late luteal stage (D 14). After PGF treatment on D 7, the expression of NR4A1-S increased which peaked at 0.5-1 h and then declined; while NR4A1-L expression did not change within 8 h. Real-time PCR results showed that the ovarian NR4A1 mRNA increased within 0.5 h, maintained high at 1 h and then declined. The NR4A2 mRNA expression exhibited a similar pattern to that of NR4A1 mRNA, though its abundance was not as high as NR4A1. IHC results revealed that NR4A1-L was expressed mainly in the cytoplasm of luteal steroidogenic cells, faintly expressed in the follicle theca cells, oocytes and the pericytes; while NR4A2 was primarily localized in the cytoplasm of luteal steroidogenic cells. In conclusion, all these results demonstrate that NR4A2 as well as NR4A1 might be involved in the luteal development and luteolysis in rats.
Collapse
|
11
|
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 2018; 122:22-33. [PMID: 30391133 DOI: 10.1016/j.mvr.2018.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/12/2018] [Accepted: 10/25/2018] [Indexed: 12/13/2022]
Abstract
Pathological angiogenesis is a hallmark of many diseases. Previously, we reported that orphan nuclear receptor TR3/Nur77 (human homolog, Nur77, mouse homolog) is a critical mediator of angiogenesis to regulate tumor growth and skin wound healing via down-regulating the expression of the junctional proteins and integrin β4. However, the molecular mechanism, by which TR3/Nur77 regulated angiogenesis, was still not completely understood. In this report by analyzing the integrin expression profile in endothelial cells, we found that the TR3/Nur77 expression highly increased the expression of integrins α1 and β5, decreased the expression of integrins α2 and β3, but had some or no effect on the expression of integrins αv, α3, α4, α5, α6, β1 and β7. In the angiogenic responses mediated by TR3/Nur77, integrin α1 regulated endothelial cell proliferation and adhesion, but not migration. Integrin β5 shRNA inhibited cell migration, but increased proliferation and adhesion. Integrin α2 regulated all of the endothelial cell proliferation, migration and adhesion. However, integrin β3 did not play any role in endothelial cell proliferation, migration and adhesion. TR3/Nur77 regulated the transcription of integrins α1, α2, β3 and β5, via various amino acid fragments within its transactivation domain and DNA binding domain. Furthermore, TR3/Nur77 regulated the integrin α1 promoter activity by directly interacting with a novel DNA element within the integrin α1 promoter. These studies furthered our understanding of the molecular mechanism by which TR3/Nur77 regulated angiogenesis, and supported our previous finding that TR3/Nur77 was an excellent therapeutic target for pathological angiogenesis. Therefore, targeting TR3/Nur77 inhibits several signaling pathways that are activated by various angiogenic factors.
Collapse
Affiliation(s)
- Taiyang Ye
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Obstetrics & Gynecology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200127, PR China
| | - Jin Peng
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Radiotherapy and Medical Oncology Department, Zhongnan Hospital, Wuhan University, Wuhan, PR China
| | - Xin Liu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Beijing Traditional Chinese Medicine Hospital, Capital Medical University, Beijing, PR China
| | - Shiqiang Hou
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Gengming Niu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Yan Li
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, PR China
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
12
|
Scholtysek C, Ipseiz N, Böhm C, Krishnacoumar B, Stenzel M, Czerwinski T, Palumbo-Zerr K, Rothe T, Weidner D, Klej A, Stoll C, Distler J, Tuckermann J, Herrmann M, Fabry B, Goldmann WH, Schett G, Krönke G. NR4A1 Regulates Motility of Osteoclast Precursors and Serves as Target for the Modulation of Systemic Bone Turnover. J Bone Miner Res 2018; 33:2035-2047. [PMID: 29949664 DOI: 10.1002/jbmr.3533] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/29/2018] [Accepted: 06/21/2018] [Indexed: 11/10/2022]
Abstract
NR4A1 (Nur77 or NGFI-B), an orphan member of the nuclear receptor superfamily, has been identified as a key regulator of the differentiation and function of myeloid, lymphoid, and mesenchymal cells. The detailed role of NR4A1 in bone biology is incompletely understood. Here, we report a role for NR4A1 as novel factor controlling the migration and recruitment of osteoclast precursors during bone remodeling. Myeloid-specific but not osteoblast-specific deletion of NR4A1 resulted in osteopenia due to an increase in the number of bone-lining osteoclasts. Although NR4A1-deficient osteoclast precursors displayed a regular differentiation into mature osteoclasts, they showed a hyper-motile phenotype that was largely dependent on increased osteopontin expression, suggesting that expression of NR4A1 negatively controlled osteopontin-mediated recruitment of osteoclast precursors to the trabecular bone. Pharmacological activation of NR4A1, in turn, inhibited osteopontin expression and osteopontin-dependent migration of osteoclast precursors resulted in reduced abundance of bone-resorbing osteoclasts in vivo as well as in an ameliorated bone loss after ovariectomy in mice. This study identifies NR4A1 as a crucial player in the regulation of osteoclast biology and bone remodeling and highlights this nuclear receptor as a promising target for therapeutic intervention during the treatment of osteoporosis. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Carina Scholtysek
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Natacha Ipseiz
- School of Medicine, University of Cardiff, Cardiff, Wales
| | - Christina Böhm
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Brenda Krishnacoumar
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Martin Stenzel
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tina Czerwinski
- Department of Biophysics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Katrin Palumbo-Zerr
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Tobias Rothe
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Daniela Weidner
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alexandra Klej
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Cornelia Stoll
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jörg Distler
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ben Fabry
- Department of Biophysics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang H Goldmann
- Department of Biophysics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3, University of Erlangen-Nuremberg, Erlangen, Germany.,Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
13
|
Fechter K, Feichtinger J, Prochazka K, Unterluggauer JJ, Pansy K, Steinbauer E, Pichler M, Haybaeck J, Prokesch A, Greinix HT, Beham-Schmid C, Neumeister P, Thallinger GG, Deutsch AJA. Cytoplasmic location of NR4A1 in aggressive lymphomas is associated with a favourable cancer specific survival. Sci Rep 2018; 8:14528. [PMID: 30266952 PMCID: PMC6162226 DOI: 10.1038/s41598-018-32972-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023] Open
Abstract
The nuclear orphan receptor NR4A1 functions as tumour suppressor in aggressive lymphomas by pro-apoptotic genomic and non-genomic effects. Here, we immunohistochemically studied the clinico-pathological relevance of NR4A1 protein expression patterns in a cohort of 60 diffuse large B cell lymphoma (DLBCL) patients and non-neoplastic lymph nodes. We observed a significant association between high cytoplasmic NR4A1 and favourable cancer-specific survival and the germinal centre B cell-like subtype, respectively. Moreover, the percentage of lymphoma cells exhibiting cytoplasmic NR4A1 significantly correlated to those showing cleaved caspase 3. Complementary, functional profiling using gene set enrichment of Reactome pathways based on publicly available microarray data was applied to determine pathways potentially implicated in cytoplasmic localization of NR4A1 and validated by means of semi quantitative real-time PCR. The pathway analysis revealed changes in the ERK1/2 pathway, and this was corroborated by the finding that high cytoplasmic NR4A1 was associated with higher expression of ERK1/2 targets in our cohort. These data indicate that high cytoplasmic NR4A1 is associated with a favourable lymphoma-specific survival and highlights the importance of NR4A1 expression patterns as potential prognostic marker for risk assessment in aggressive lymphomas.
Collapse
MESH Headings
- Aged
- Cohort Studies
- Cytoplasm/genetics
- Cytoplasm/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lymphoma, Large B-Cell, Diffuse/epidemiology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Nuclear Receptor Subfamily 4, Group A, Member 1/analysis
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Survival Analysis
Collapse
Affiliation(s)
- Karoline Fechter
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Julia Feichtinger
- Institute of Computational Biotechnology, Graz University of Technology, Graz, Austria
- BioTechMed Omics Center Graz, Graz, Austria
| | - Katharina Prochazka
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | | | - Katrin Pansy
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | | | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Johannes Haybaeck
- Institute of Pathology, Medical University Graz, Graz, Austria
- Department of Pathology, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Institute of Pathology, Medical University Innsbruck, Innsbruck, Austria
| | - Andreas Prokesch
- Institute of Cell Biology, Histology and Embryology, Medical University Graz, Graz, Austria
| | - Hildegard T Greinix
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | | | - Peter Neumeister
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Gerhard G Thallinger
- Institute of Computational Biotechnology, Graz University of Technology, Graz, Austria.
- BioTechMed Omics Center Graz, Graz, Austria.
| | - Alexander J A Deutsch
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria.
| |
Collapse
|
14
|
Requirement of novel amino acid fragments of orphan nuclear receptor TR3/Nur77 for its functions in angiogenesis. Oncotarget 2016; 6:24261-76. [PMID: 26155943 PMCID: PMC4695184 DOI: 10.18632/oncotarget.4637] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/05/2015] [Indexed: 01/08/2023] Open
Abstract
Pathological angiogenesis is a hallmark of many diseases. We demonstrated that TR3/Nur77 is an excellent target for pro-angiogenesis and anti-angiogenesis therapies. Here, we report that TR3 transcriptionally regulates endothelial cell migration, permeability and the formation of actin stress fibers that is independent of RhoA GTPase. 1) Amino acid residues 344-GRR-346 and de-phosphorylation of amino acid residue serine 351 in the DNA binding domain, and 2) phosphorylation of amino acid residues in the 41-61 amino acid fragment of the transactivation domain, of TR3 are required for its induction of the formation of actin stress fibers, cell proliferation, migration and permeability. The 41-61 amino acid fragment contains one of the three potential protein interaction motifs in the transactivation domain of TR3, predicted by computational modeling and analysis. These studies further our understanding of the molecular mechanism, by which TR3 regulates angiogenesis, identify novel therapeutic targeted sites of TR3, and set the foundation for the development of high-throughput screening assays to identify compounds targeting TR3/Nur77 for pro-angiogenesis and anti-angiogenesis therapies.
Collapse
|
15
|
Niu G, Ye T, Qin L, Bourbon PM, Chang C, Zhao S, Li Y, Zhou L, Cui P, Rabinovitz I, Mercurio AM, Zhao D, Zeng H. Orphan nuclear receptor TR3/Nur77 improves wound healing by upregulating the expression of integrin β4. FASEB J 2014; 29:131-40. [PMID: 25326539 DOI: 10.1096/fj.14-257550] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tissue repair/wound healing, in which angiogenesis plays an important role, is a critical step in many diseases including chronic wound, myocardial infarction, stroke, cancer, and inflammation. Recently, we were the first to report that orphan nuclear receptor TR3/Nur77 is a critical mediator of angiogenesis and its associated microvessel permeability. Tumor growth and angiogenesis induced by VEGF-A, histamine, and serotonin are almost completely inhibited in Nur77 knockout mice. However, it is not known whether TR3/Nur77 plays any roles in wound healing. In these studies, skin wound-healing assay was performed in 3 types of genetically modified mice having various Nur77 activities. We found that ectopic induction of Nur77 in endothelial cells of mice is sufficient to improve skin wound healing. Although skin wound healing in Nur77 knockout mice is comparable to the wild-type control mice, the process is significantly delayed in the EC-Nur77-DN mice, in which a dominant negative Nur77 mutant is inducibly and specifically expressed in mouse endothelial cells. By a loss-of-function assay, we elucidate a novel feed-forward signaling pathway, integrin β4 → PI3K → Akt → FAK, by which TR3 mediates HUVEC migration. Furthermore, TR3/Nur77 regulates the expression of integrin β4 by targeting its promoter activity. In conclusion, expression of TR3/Nur77 improves wound healing by targeting integrin β4. TR3/Nur77 is a potential candidate for proangiogenic therapy. The results further suggest that TR3/Nur77 is required for pathologic angiogenesis but not for developmental/physiologic angiogenesis and that Nur77 and its family members play a redundant role in normal skin wound healing.
Collapse
Affiliation(s)
- Gengming Niu
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Taiyang Ye
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Obstetrics & Gynecology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, People's Republic of China
| | | | | | - Cheng Chang
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shengqiang Zhao
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Gastroenterology, Provincial Hospital Affiliated to Shandong University, Ji-nan, People's Republic of China
| | - Yan Li
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Gastroenterology, Provincial Hospital Affiliated to Shandong University, Ji-nan, People's Republic of China
| | - Lei Zhou
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Departments of Hepatobiliary Surgery and General Surgery, the First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Pengfei Cui
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of General Surgery, Pancreatic Disease Institute, Union Hospital, Huazhong University of Science & Technology (HUST), Wuhan, People's Republic of China
| | - Issac Rabinovitz
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Department of Pathology
| | - Arthur M Mercurio
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Molecular and Vascular Biology, Department of Medicine and
| |
Collapse
|
16
|
Eger G, Papadopoulos N, Lennartsson J, Heldin CH. NR4A1 promotes PDGF-BB-induced cell colony formation in soft agar. PLoS One 2014; 9:e109047. [PMID: 25269081 PMCID: PMC4182636 DOI: 10.1371/journal.pone.0109047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/30/2014] [Indexed: 01/04/2023] Open
Abstract
The fibroblast mitogen platelet-derived growth factor -BB (PDGF-BB) induces a transient expression of the orphan nuclear receptor NR4A1 (also named Nur77, TR3 or NGFIB). The aim of the present study was to investigate the pathways through which NR4A1 is induced by PDGF-BB and its functional role. We demonstrate that in PDGF-BB stimulated NIH3T3 cells, the MEK1/2 inhibitor CI-1040 strongly represses NR4A1 expression, whereas Erk5 downregulation delays the expression, but does not block it. Moreover, we report that treatment with the NF-κB inhibitor BAY11-7082 suppresses NR4A1 mRNA and protein expression. The majority of NR4A1 in NIH3T3 was found to be localized in the cytoplasm and only a fraction was translocated to the nucleus after continued PDGF-BB treatment. Silencing NR4A1 slightly increased the proliferation rate of NIH3T3 cells; however, it did not affect the chemotactic or survival abilities conferred by PDGF-BB. Moreover, overexpression of NR4A1 promoted anchorage-independent growth of NIH3T3 cells and the glioblastoma cell lines U-105MG and U-251MG. Thus, whereas NR4A1, induced by PDGF-BB, suppresses cell growth on a solid surface, it increases anchorage-independent growth.
Collapse
MESH Headings
- Agar
- Animals
- Becaplermin
- Benzamides/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Chemotaxis/drug effects
- Gene Expression Regulation
- Humans
- MAP Kinase Kinase 1/antagonists & inhibitors
- MAP Kinase Kinase 1/genetics
- MAP Kinase Kinase 1/metabolism
- MAP Kinase Kinase 2/antagonists & inhibitors
- MAP Kinase Kinase 2/genetics
- MAP Kinase Kinase 2/metabolism
- Mice
- Mitogen-Activated Protein Kinase 7/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 7/genetics
- Mitogen-Activated Protein Kinase 7/metabolism
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/genetics
- NF-kappa B/metabolism
- NIH 3T3 Cells
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neuroglia/pathology
- Nitriles/pharmacology
- Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-sis/pharmacology
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Sulfones/pharmacology
Collapse
Affiliation(s)
- Glenda Eger
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
| | | | - Johan Lennartsson
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
- * E-mail:
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Murphy SR, Oslund KL, Hyde DM, Miller LA, Van Winkle LS, Schelegle ES. Ozone-induced airway epithelial cell death, the neurokinin-1 receptor pathway, and the postnatal developing lung. Am J Physiol Lung Cell Mol Physiol 2014; 307:L471-81. [PMID: 25063800 DOI: 10.1152/ajplung.00324.2013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Children are uniquely susceptible to ozone because airway and lung growth continue for an extensive period after birth. Early-life exposure of the rhesus monkey to repeated ozone cycles results in region-specific disrupted airway/lung growth, but the mediators and mechanisms are poorly understood. Substance P (SP), neurokinin-1 receptor (NK-1R); and nuclear receptor Nur77 (NR4A1) are signaling pathway components involved in ozone-induced cell death. We hypothesize that acute ozone (AO) exposure during postnatal airway development disrupts SP/NK-1R/Nur77 pathway expression and that these changes correlate with increased ozone-induced cell death. Our objectives were to 1) spatially define the normal development of the SP/NK-1R/Nur77 pathway in conducting airways; 2) compare how postnatal age modulates responses to AO exposure; and 3) determine how concomitant, episodic ozone exposure modifies age-specific acute responses. Male infant rhesus monkeys were assigned at age 1 mo to two age groups, 2 or 6 mo, and then to one of three exposure subgroups: filtered air (FA), FA+AO (AO: 8 h/day × 2 days), or episodic biweekly ozone exposure cycles (EAO: 8 h/day × 5 days/14-day cycle+AO). O3 = 0.5 ppm. We found that 1) ozone increases SP/NK-1R/Nur77 pathway expression in conducting airways, 2) an ozone exposure cycle (5 days/cycle) delivered early at age 2 mo resulted in an airway that was hypersensitive to AO exposure at the end of 2 mo, and 3) continued episodic exposure (11 cycles) resulted in an airway that was hyposensitive to AO exposure at 6 mo. These observations collectively associate with greater overall inflammation and epithelial cell death, particularly in early postnatal (2 mo), distal airways.
Collapse
Affiliation(s)
- Shannon R Murphy
- Center for Health and the Environment, University of California-Davis, Davis, California
| | - Karen L Oslund
- California National Primate Research Center, University of California-Davis, Davis, California; and
| | - Dallas M Hyde
- California National Primate Research Center, University of California-Davis, Davis, California; and School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California-Davis, Davis, California
| | - Lisa A Miller
- California National Primate Research Center, University of California-Davis, Davis, California; and School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California-Davis, Davis, California
| | - Laura S Van Winkle
- Center for Health and the Environment, University of California-Davis, Davis, California; School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California-Davis, Davis, California
| | - Edward S Schelegle
- California National Primate Research Center, University of California-Davis, Davis, California; and School of Veterinary Medicine, Department of Anatomy, Physiology and Cell Biology, University of California-Davis, Davis, California
| |
Collapse
|
18
|
Sokanovic SJ, Janjic MM, Stojkov NJ, Baburski AZ, Bjelic MM, Andric SA, Kostic TS. Age related changes of cAMP and MAPK signaling in Leydig cells of Wistar rats. Exp Gerontol 2014; 58:19-29. [PMID: 25019473 DOI: 10.1016/j.exger.2014.07.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/09/2014] [Accepted: 07/11/2014] [Indexed: 12/14/2022]
Abstract
Here, we chronologically analyzed age-associated changes of cAMP- and MAPK-signaling in Leydig cells (LCs) in relation with decreased testosterone (T) production. In Wistar rats, decreased serum T observed in 12 to 24-month-old rats was not related to decreased serum LH concentration but to reduced luteinizing hormone receptor (Lhr/LHR) and time-coordinated reduction of steroidogenic gene expression (decreased Cyp11a1, Cyp17a1 in 12-month-old rats followed by decreased Star/StAR, Hsd3b/HSD3B, Hsd17b4, and increased Cyp19a1 later in life). The predecessors of age-related changes noted in LCs from 6 to 12-month-old rats were increased level of soluble adenylate cyclase (Adcy/AC) 10, increased JNK phosphorylation but suppressed P38 MAPK. At approximately the same time changed mRNA abundance for transcription factors important for steroidogenesis was detected (increased Nur77 and decreased Sf1, Dax1). Aging caused biphasic expression pattern of ERK1/2 and Nur77: increased in 12-month but decreased in LCs from 24-month-old rats. Further, decreased basal cAMP level observed from 12 to 24th month coincidence with increased expression of cAMP-specific phosphodiesterase (Pde)4a, Pde4b and regulatory subunit of protein kinase A (Prkar/PKAR). Exposing of senescent LCs to permeable cAMP-analog improved transcription of Sf1, Nur77, Star, Cyp11a1,Cyp17a1, but without effect on aging pattern of Dax1, Pde4a/b, Prkar2a, Lhr and MAPK genes. Collectively, results indicated that age-related LC dysfunction is accompanied with changes in MAPK and cAMP signaling and coordinated reduction in the expression of many of the genes that participate in T synthesis. The predecessors of aged-related changes are increased ratio of pJNK/JNK, AC10 and decreased P38 level in LCs from 6-month-old rats.
Collapse
Affiliation(s)
- S J Sokanovic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - M M Janjic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - N J Stojkov
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - A Z Baburski
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - M M Bjelic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - S A Andric
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - T S Kostic
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia.
| |
Collapse
|
19
|
Sudeshna T, Anand K, Medhamurthy R. Analysis of 20alpha-hydroxysteroid dehydrogenase expression in the corpus luteum of the buffalo cow: effect of prostaglandin F2-alpha treatment on circulating 20alpha-hydroxyprogesterone levels. Reprod Biol Endocrinol 2013; 11:111. [PMID: 24330451 PMCID: PMC3878843 DOI: 10.1186/1477-7827-11-111] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 12/09/2013] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND During female reproductive cycles, a rapid fall in circulating progesterone (P4) levels is one of the earliest events that occur during induced luteolysis in mammals. In rodents, it is well recognized that during luteolysis, P4 is catabolized to its inactive metabolite, 20alpha-hydroxyprogesterone (20alpha-OHP) by the action of 20alpha-hydroxysteroid dehydrogenase (20alpha-HSD) enzyme and involves transcription factor, Nur77. Studies have been carried out to examine expression of 20alpha-HSD and its activity in the corpus luteum (CL) of buffalo cow. METHODS The expression of 20alpha-HSD across different bovine tissues along with CL was examined by qPCR analysis. Circulating P4 levels were monitored before and during PGF2alpha treatment. Expression of 20alpha-HSD and Nur77 mRNA was determined in CL at different time points post PGF2alpha treatment in buffalo cows. The chromatographic separation of P4 and its metabolite, 20alpha-OHP, in rat and buffalo cow serum samples were performed on reverse phase HPLC system. To further support the findings, 20alpha-HSD enzyme activity was quantitated in cytosolic fraction of CL of both rat and buffalo cow. RESULTS Circulating P4 concentration declined rapidly in response to PGF2alpha treatment. HPLC analysis of serum samples did not reveal changes in circulating 20alpha-OHP levels in buffalo cows but serum from pseudo pregnant rats receiving PGF2alpha treatment showed an increased 20alpha-OHP level at 24 h post treatment with accompanying decrease in P4 concentration. qPCR expression of 20alpha-HSD in CL from control and PGF2alpha-treated buffalo cows showed higher expression at 3 and 18 h post treatment, but its specific activity was not altered at different time points post PGF2alpha treatment. The Nur77 expression increased several fold 3 h post PGF2alpha treatment similar to the increased expression observed in the PGF2alpha-treated pseudo pregnant rats which perhaps suggest initiation of activation of apoptotic pathways in response to PGF2alpha treatment. CONCLUSIONS The results taken together suggest that synthesis of P4 appears to be primarily affected by PGF2alpha treatment in buffalo cows in contrast to increased metabolism of P4 in rodents.
Collapse
Affiliation(s)
- Tripathy Sudeshna
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Kumarasamy Anand
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Rudraiah Medhamurthy
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
20
|
Abstract
A growing body of evidence suggests that a subset of orphan nuclear receptors are amplified and prognostic for some human cancers. However, the specific roles of these orphan nuclear receptors in tumor progression and their utility as drug targets are not fully understood. In this review, we summarize recent progress in elucidating the direct and indirect involvement of orphan nuclear receptors in cancer as well as their therapeutic potential in a variety of human cancers. Furthermore, we contrast the role of orphan nuclear receptors in cancer with the known roles of estrogen receptor and androgen receptor in hormone-dependent cancers.
Collapse
Affiliation(s)
- Sung Hee Baek
- School of Biological Sciences, Creative Research Initiative Center for Chromatin Dynamics, Seoul National University, Seoul 151-742, South Korea;
| | | |
Collapse
|
21
|
Hu YW, Zheng L, Wang Q, Zhong TY, Yu X, Bao J, Cao NN, Li B, Si-Tu B. Vascular endothelial growth factor downregulates apolipoprotein M expression by inhibiting Foxa2 in a Nur77-dependent manner. Rejuvenation Res 2013; 15:423-34. [PMID: 22877565 DOI: 10.1089/rej.2011.1295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE We aimed to investigate whether vascular endothelial growth factor (VEGF) influences apolipoprotein M (ApoM) expression and pre-β-high-density lipoprotin (HDL) formation, and whether forkhead box A2 (Foxa2) and Nur77 are involved in this process. METHODS AND RESULTS We analyzed the serum VEGF concentrations of 264 adults who underwent a medical checkup and found that VEGF concentration was positively correlated with serum triglyceride, total cholesterol, LDL cholesterol (LDL-C), very-low-density lipoprotein cholesterol (VLDL-C), and ApoB concentrations, but was negatively correlated with serum high-density lipoprotein cholesterol (HDL-C) and ApoM concentrations. We further investigated the effects of VEGF on ApoM expression and pre-β-HDL formation, and the mechanisms responsible, in HepG2 cells and mouse primary hepatocytes. VEGF markedly downregulated ApoM expression and pre-β-HDL formation. At the same time, expression of Foxa2 was also inhibited, whereas expression of Nur77 was increased by treatment with VEGF. Furthermore, small interfering (si) RNA knockdown of Foxa2 made the downregulation of VEGF on ApoM expression and pre-β-HDL formation even more obvious. In addition, siRNA knockdown of Nur77 significantly compensated for the inhibitory effect of VEGF on Foxa2 expression, whereas the Nur77 agonist cytosporone B led to the downregulation of Foxa2 expression more significantly than VEGF. Moreover, overexpression of a Nur77 transgene in C57BL/6 mice resulted in decreased serum ApoM and pre-β-HDL levels, whereas si-Nur77-treated mice displayed upregulated serum ApoM and pre-β-HDL levels. CONCLUSION These results provide evidence that VEGF may first downregulate expression of Foxa2 by enhancing Nur77 activity and then decrease expression of ApoM and pre-β-HDL formation. Therefore, our study may be useful in understanding the critical effect of VEGF in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
García-Yagüe ÁJ, Rada P, Rojo AI, Lastres-Becker I, Cuadrado A. Nuclear import and export signals control the subcellular localization of Nurr1 protein in response to oxidative stress. J Biol Chem 2013; 288:5506-17. [PMID: 23283970 PMCID: PMC3581400 DOI: 10.1074/jbc.m112.439190] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 12/28/2012] [Indexed: 11/06/2022] Open
Abstract
Orphan receptor Nurr1 participates in the acquisition and maintenance of the dopaminergic cell phenotype, modulation of inflammation, and cytoprotection, but little is known about its regulation. In this study, we report that Nurr1 contains a bipartite nuclear localization signal (NLS) within its DNA binding domain and two leucine-rich nuclear export signals (NES) in its ligand binding domain. Together, these signals regulate Nurr1 shuttling in and out of the nucleus. Immunofluorescence and immunoblot analysis revealed that Nurr1 is mostly nuclear. A Nurr1 mutant lacking the NLS failed to enter the nucleus. The Nurr1 NLS sequence, when fused to green fluorescent protein, led to nuclear accumulation of this chimeric protein, indicating that this sequence was sufficient to direct nuclear localization of Nurr1. Furthermore, two NES were characterized in the ligand binding domain, whose deletion caused Nurr1 to accumulate predominantly in the nucleus. The Nurr1 NES was sensitive to CRM1 and could function as an independent export signal when fused to green fluorescent protein. Sodium arsenite, an agent that induces oxidative stress, promoted nuclear export of ectopically expressed Nurr1 in HEK293T cells, and the antioxidant N-acetylcysteine rescued from this effect. Similarly, in dopaminergic MN9D cells, arsenite induced the export of endogenous Nurr1, resulting in the loss of expression of Nurr1-dependent genes. This study illustrates that Nurr1 shuttling between the cytosol and nucleus is controlled by specific nuclear import and export signals and that oxidative stress can unbalance the distribution of Nurr1 to favor its cytosolic accumulation.
Collapse
Affiliation(s)
- Ángel Juan García-Yagüe
- From the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid
- the Departamento de Bioquímica e Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid
- the Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, and
| | - Patricia Rada
- From the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid
- the Departamento de Bioquímica e Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid
- the Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, and
| | - Ana I. Rojo
- From the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid
- the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Isabel Lastres-Becker
- From the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid
- the Departamento de Bioquímica e Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid
- the Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, and
| | - Antonio Cuadrado
- From the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid
- the Departamento de Bioquímica e Instituto de Investigaciones Biomédicas “Alberto Sols,” Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid
- the Instituto de Investigación Sanitaria La Paz (IDIPAZ), Madrid, and
| |
Collapse
|
23
|
Bouzas-Rodríguez J, Zárraga-Granados G, Sánchez-Carbente MDR, Rodríguez-Valentín R, Gracida X, Anell-Rendón D, Covarrubias L, Castro-Obregón S. The nuclear receptor NR4A1 induces a form of cell death dependent on autophagy in mammalian cells. PLoS One 2012; 7:e46422. [PMID: 23071566 PMCID: PMC3465341 DOI: 10.1371/journal.pone.0046422] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/29/2012] [Indexed: 11/18/2022] Open
Abstract
The control of cell death is a biological process essential for proper development, and for preventing devastating pathologies like cancer and neurodegeneration. On the other hand, autophagy regulation is essential for protein and organelle degradation, and its dysfunction is associated with overlapping pathologies like cancer and neurodegeneration, but also for microbial infection and aging. In the present report we show that two evolutionarily unrelated receptors--Neurokinin 1 Receptor (NK(1)R,) a G-protein coupled receptor, and Insulin-like Growth Factor 1 Receptor (IGF1R), a tyrosine kinase receptor--both induce non-apoptotic cell death with autophagic features and requiring the activity of the autophagic core machinery proteins PI3K-III, Beclin-1 and Atg7. Remarkably, this form of cell death occurs in apoptosis-competent cells. The signal transduction pathways engaged by these receptors both converged on the activation of the nuclear receptor NR4A1, which has previously been shown to play a critical role in some paradigms of apoptosis and in NK(1)R-induced cell death. The activity of NR4A1 was necessary for IGF1R-induced cell death, as well as for a canonical model of cell death by autophagy induced by the presence of a pan-caspase inhibitor, suggesting that NR4A1 is a general modulator of this kind of cell death. During cell death by autophagy, NR4A1 was transcriptionally competent, even though a fraction of it was present in the cytoplasm. Interestingly, NR4A1 interacts with the tumor suppressor p53 but not with Beclin-1 complex. Therefore the mechanism to promote cell death by autophagy might involve regulation of gene expression, as well as protein interactions. Understanding the molecular basis of autophagy and cell death mediation by NR4A1, should provide novel insights and targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jimena Bouzas-Rodríguez
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Gabriela Zárraga-Granados
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Maria del Rayo Sánchez-Carbente
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Rocío Rodríguez-Valentín
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Xicotencatl Gracida
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Dámaris Anell-Rendón
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Luis Covarrubias
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Susana Castro-Obregón
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
- * E-mail:
| |
Collapse
|
24
|
Boldingh Debernard KA, Mathisen GH, Paulsen RE. Differences in NGFI-B, Nurr1, and NOR-1 expression and nucleocytoplasmic translocation in glutamate-treated neurons. Neurochem Int 2012; 61:79-88. [PMID: 22525717 DOI: 10.1016/j.neuint.2012.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 03/21/2012] [Accepted: 04/05/2012] [Indexed: 11/30/2022]
Abstract
NGFI-B (NR4A1, Nur77 or TR3) together with Nurr1 (NR4A2) and NOR-1 (NR4A3) constitute the NR4A subgroup of orphan nuclear receptors. They play critical roles in proliferation, differentiation, survival and apoptosis in different cell types, including neurons, immature T-cells, and different cancer cells. As ligand-independent and constitutively active receptors, the diverse biological activities of NGFI-B, Nurr1 and NOR-1 depend on their levels of expression, post-translational modifications and subcellular localization. Nuclear localization of the NR4A proteins leads to transcriptional activity, whereas NGFI-B and recently also NOR-1 have been shown to induce apoptosis by a more direct mechanism when localized at mitochondria. In the present study we investigated mRNA expression and subcellular translocation of the NR4A proteins during glutamate excitotoxicity in rat cerebellar granule neurons. NGFI-B and Nurr1 mRNA, but not NOR-1 mRNA, were induced by treatments associated with calcium influx, although their regulation seemed to be different. NR4A(gfp) fusion proteins showed a predominant nuclear localization in untreated cells. After glutamate treatment NGFI-B(gfp) translocated to cytosol and mitochondria within a few hours, whereas Nurr1(gfp) translocation was delayed, and NOR-1(gfp) mainly stayed in the nucleus. Subcellular targeting of NGFI-B seems to be tightly regulated, as a single mutation of threonine 142 altered NGFI-B(gfp) localization. Differences in expression and subcellular translocation of NGFI-B, Nurr1, and NOR-1 may reflect different functions in neurons in glutamate excitotoxicity.
Collapse
|
25
|
Strøm BO, Paulsen RE. Apoptosis inducer NGFI-B is degraded by the proteasome and stabilized by treatment with EGF. Biochem Biophys Res Commun 2012; 417:1292-7. [PMID: 22234305 DOI: 10.1016/j.bbrc.2011.12.132] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 12/25/2011] [Indexed: 11/26/2022]
Abstract
NGFI-B is a nuclear receptor and immediate early gene that is upregulated in many different tumour cell lines. As it is involved in cell death and survival, it has been suggested as a target for anti-cancer drugs. The protein level of NGFI-B is important for its functions and may be regulated through induction or stabilization. NGFI-B protein stability was studied using the protein synthesis inhibitor cycloheximide in CV1 cells transiently transfected with NGFI-B. Inhibiting the proteasome with MG132 stabilized NGFI-B, indicating that the proteasome is responsible for break-down of NGFI-B, as it is for many nuclear receptors. In order to determine regions responsible for the break-down of NGFI-B two N-terminal regions with high PEST-scores were deleted. Deletion of amino acids 122-195 containing a PEST-sequence which includes an ERK2 phosphorylation target, gave a more stable protein. In addition, treatment of the cells with the ERK2 activator EGF increased the stability of wild type NGFI-B. We then tested whether a mutation at threonine 142 influenced the stability of NGFI-B. We found that the phosphorylation-mimicking mutant NGFI-B T142E had an increased stability, while the non-phosphorylable mutant (T142A) showed similar stability to the wild type. Thus, EGF-stimulation of cells may be a mechanism for priming the cells for effects of NGFI-B by increasing its stability.
Collapse
Affiliation(s)
- Bjørn O Strøm
- Department of Pharmaceutical Biosciences, University of Oslo, P.O. Box 1068 Blindern, N-0316 Oslo, Norway
| | | |
Collapse
|
26
|
Brion L, Maloberti PM, Gomez NV, Poderoso C, Gorostizaga AB, Mori Sequeiros Garcia MM, Acquier AB, Cooke M, Mendez CF, Podesta EJ, Paz C. MAPK phosphatase-1 (MKP-1) expression is up-regulated by hCG/cAMP and modulates steroidogenesis in MA-10 Leydig cells. Endocrinology 2011; 152:2665-77. [PMID: 21558315 DOI: 10.1210/en.2011-0021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
MAP kinases (MAPKs), such as ERK1/2, exert profound effects on a variety of physiological processes. In steroidogenic cells, ERK1/2 are involved in the expression and activation of steroidogenic acute regulatory protein, which plays a central role in the regulation of steroidogenesis. In MA-10 Leydig cells, LH and chorionic gonadotropin (CG) trigger transient ERK1/2 activation via protein kinase A, although the events that lead to ERK1/2 inactivation are not fully described. Here, we describe the hormonal regulation of MAPK phosphatase-1 (MKP-1), an enzyme that inactivates MAPKs, in MA-10 cells. In our experiments, human CG (hCG)/cAMP stimulation rapidly and transiently increased MKP-1 mRNA levels by a transcriptional action. This effect was accompanied by an increase in protein levels in both nuclear and mitochondrial compartments. In cells transiently expressing flag-MKP-1 protein, hCG/cAMP promoted the accumulation of the recombinant protein in a time-dependent manner (10-fold at 1 h). Moreover, hCG/cAMP triggered ERK1/2-dependent MKP-1 phosphorylation. The blockade of cAMP-induced MAPK kinase/ERK activation abated MKP-1 phosphorylation but only partially reduced flag-MKP-1 protein accumulation. Together, these results suggest that hCG regulates MKP-1 at transcriptional and posttranslational level, protein phosphorylation being one of the mechanisms involved in this regulation. Our study also demonstrates that MKP-1 overexpression reduces the effects of cAMP on ERK1/2 phosphorylation, steroidogenic acute regulatory gene promoter activity, mRNA levels, and steroidogenesis, whereas MKP-1 down-regulation by small interfering RNA produces opposite effects. In summary, our data demonstrate that hCG regulates MKP-1 expression at multiple stages as a negative feedback regulatory mechanism to modulate the hormonal action on ERK1/2 activity and steroidogenesis.
Collapse
Affiliation(s)
- Laura Brion
- Institute of Molecular Research in Hormonal, Neurodegenerative and Oncological Diseases, Department of Biochemistry, School of Medicine, University of Buenos Aires, Paraguay 2155, 5th Floor, C1121ABG Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yang H, Nie Y, Li Y, Wan YJY. ERK1/2 deactivation enhances cytoplasmic Nur77 expression level and improves the apoptotic effect of fenretinide in human liver cancer cells. Biochem Pharmacol 2011; 81:910-6. [PMID: 21241664 DOI: 10.1016/j.bcp.2011.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 01/06/2011] [Accepted: 01/06/2011] [Indexed: 01/06/2023]
Abstract
Fenretinide, a synthetic retinoid, is a promising anticancer agent based on many in vitro, animal, and chemoprevention clinical trial studies. However, cells such as HepG2 human liver cancer cells are resistant to the apoptotic effect of fenretinide. Previously, we have shown that fenretinide-induced apoptosis is Nur77 dependent, and the sensitivity of the cancer cells to fenretinide-induced apoptosis is positively associated with cytoplasmic enrichment of Nur77. The goal of current study was to identify means to modulate nuclear export of Nur77 in order to improve the efficacy of fenretinide. Fenretinide treatment deactivated ERK1/2 in Huh7 cells, but activated ERK1/2 in HepG2 cells, which was positively associated with the sensitivity of cells to the apoptotic effect of fenretinide. Neither fenretinide nor ERK1/2 inhibitor PD98059 alone could affect the survival of HepG2 cells, but the combination of both induced cell death and increased caspase 3/7 activity. In fenretinide sensitive Huh7 cells, activation of ERK1/2 by epidermal growth factor (EGF) prevented fenretinide-induced cell death and caspase 3/7 induction. In addition, modulation of ERK1/2 changed the intracellular localization of Nur77. Fenretinide/PD98059-induced cell death of HepG2 cell was positively associated with induction and cytoplasmic location as well as mitochondria enrichment of Nur77. The effect was specific for ERK1/2 because other mitogen activated protein kinases such as P38, Akt, and JNK did not have correlated changes in their phosphorylation levels. Taken together, the current study demonstrates that ERK1/2-modulated Nur77 intracellular location dictates the efficacy of fenretinide-induced apoptosis.
Collapse
Affiliation(s)
- Hui Yang
- Department of Gastroenterology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | | | | | | |
Collapse
|
28
|
Kang SA, Na H, Kang HJ, Kim SH, Lee MH, Lee MO. Regulation of Nur77 protein turnover through acetylation and deacetylation induced by p300 and HDAC1. Biochem Pharmacol 2010; 80:867-73. [PMID: 20438716 DOI: 10.1016/j.bcp.2010.04.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Revised: 04/24/2010] [Accepted: 04/26/2010] [Indexed: 10/19/2022]
Abstract
Although the roles of Nur77, an orphan member of the nuclear hormone receptor superfamily, in the control of cellular proliferation, apoptosis, inflammation, and glucose metabolism, are well recognized, the molecular mechanism regulating the activity and expression of Nur77 is not fully understood. Acetylation of transcription factors has emerged recently as a major post-translational modification that regulates protein stability and transcriptional activity. Here, we examined whether Nur77 is acetylated, and we characterized potential associated factors. First, Nur77 was found to be an acetylated protein when examined by immunoprecipitation and western blotting using acetyl protein-specific antibodies. Second, expression of p300, which possesses histone acetyltransferase activity, enhanced the acetylation and protein stability of Nur77. Treatment with a histone deacetylase (HDAC) inhibitor, trichostatin A, also increased Nur77 acetylation. Among the several types of HDACs, HDAC1 was found as the major enzyme affecting protein level of Nur77. HDAC1 decreased the acetylation level, protein level, and transcriptional activity of Nur77. Interestingly, overexpression of Nur77 induced expression of both p300 and HDAC1. Finally, the expression of Nur77 increased along with that of p300, but decreased with induction of HDAC1 after treatment with epithelial growth factor, nerve growth factor, or 6-mercaptopurine, suggesting that the self-control of the acetylation status contributes to the transient induction of Nur77 protein. Taken together, these results demonstrate that acetylation of Nur77 is modulated by p300 and HDAC1, and suggest that acetylation is an important post-translational modification for the rapid turnover of Nur77 protein.
Collapse
Affiliation(s)
- Shin-Ae Kang
- College of Pharmacy, Bio-MAX Institute, and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | | | | | | | |
Collapse
|
29
|
Liver X receptor is a regulator of orphan nuclear receptor NOR-1 gene transcription in adipocytes. Int J Obes (Lond) 2009; 33:519-24. [PMID: 19238156 DOI: 10.1038/ijo.2009.32] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The liver X receptors (LXRs) are ligand-activated nuclear transcription factors that have been shown to play major roles in lipid, glucose and cholesterol metabolism. Recently, members of the NR4A orphan nuclear receptor family have also been shown to regulate the expression of important genes in metabolically active tissues such as liver, adipose and skeletal muscle. Here, we investigated the role of LXRs to regulate the expression of the nuclear receptor NOR-1 (neuron-derived orphan receptor-1) in adipocytes. APPROACH White and brown adipose tissues from wild-type, LXRalpha-/-- and LXRalpha:beta-deficient mice were collected from animals at room temperature or following cold exposure to measure NOR-1 mRNA. The expression of NOR-1 and its promoter activity in response to LXR ligands were determined in cultured primary brown adipocytes or mouse embryo fibroblasts derived from wild-type or LXRalpha-/- mice differentiated into adipocytes. RESULTS In LXRalpha-/-- and LXRalpha:beta-deficient adipocytes, basal levels of NOR-1 were significantly reduced while retaining an equivalent proportional induction by beta-adrenergic agonists. This reduced basal expression of NOR-1 in adipose tissue from LXR-deficient mice is a cell-autonomous event as it was also preserved in adipocytes differentiated from mouse embryo fibroblasts derived from these mice. In cultured primary brown adipocytes or cell lines, the expression of NOR-1 increased in response to an LXR agonist. A DNA sequence element (DR-4) capable of binding LXRs was found at -997 bp of the NOR-1 promoter, which was shown to be functional by promoter reporter gene activity, gel shift and chromatin immunoprecipitation assays. CONCLUSION These data describe a new role for LXR to regulate NOR-1 gene expression in adipocytes and demonstrate that these two nuclear receptors have an interdependent regulatory relationship, in addition to each being involved in the control of metabolic fuel usage.
Collapse
|
30
|
O'Kane M, Markham T, McEvoy AN, Fearon U, Veale DJ, FitzGerald O, Kirby B, Murphy EP. Increased Expression of the Orphan Nuclear Receptor NURR1 in Psoriasis and Modulation following TNF-α Inhibition. J Invest Dermatol 2008; 128:300-10. [PMID: 17671512 DOI: 10.1038/sj.jid.5701023] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The orphan nuclear receptor NURR1 belongs to the NR4A subfamily of transcription factors which are emerging as important mediators of cytokine and growth factor signaling. The transcriptional function of these ligand-independent and constitutively active receptors is controlled at the level of expression and nuclear localization. This study examines the expression of NURR1 in psoriasis and biological effects on this receptor following inhibition of tumor necrosis factor-alpha (TNF-alpha) signaling. We report increased expression of NURR1 mRNA and protein in involved psoriasis skin compared with uninvolved and normal skin, which correlates significantly (P=0.0055) with clinical measures of the psoriasis area and severity index. Enhanced NURR1 expression localizes to both nucleus and cytoplasm of cells of involved epidermis, blood vessels, and inflammatory infiltrates, in contrast to predominant cytoplasmic distribution in uninvolved and normal skin. Endogenous NURR1 levels are rapidly and selectively increased in response to proinflammatory agonists and growth factors in normal dermal endothelial cells. Following TNF-alpha inhibition with infliximab or etanercept, NURR1 mRNA and protein levels in involved skin are significantly decreased and cytoplasmic distribution is restored. These findings establish the aberrant expression and distribution of NURR1 in psoriasis and suggest that clinical benefits of TNF-alpha inhibition may be mediated through altered NURR1 activity.
Collapse
Affiliation(s)
- Marina O'Kane
- College of Life Sciences, UCD Veterinary Sciences Centre, Dublin, Ireland
| | | | | | | | | | | | | | | |
Collapse
|