ONCOLOGY / BASIC RESEARCH
Nur77 knock-down suppresses glioma by regulating CXCR4
More details
Hide details
1
Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
2
Department of Neurosurgery, Suqian People’s Hospital Affiliated to Xuzhou Medical University, Nanjing Drum Tower Hospital Group, Suqian, Jingsu, China
Submission date: 2020-01-15
Final revision date: 2020-04-11
Acceptance date: 2020-04-15
Online publication date: 2020-05-30
Publication date: 2026-01-16
Corresponding author
Jing Zheng
Department
of Neurosurgery
Suqian People’s Hospital
Affiliated to Xuzhou
Medical University
Nanjing Drum Tower
Hospital Group
Suqian, Jingsu, China
Hongbin Ni
Department
of Neurosurgery
Drum Tower Hospital
School of Medicine
Nanjing University
Nanjing, Jiangsu, China
Arch Med Sci 2025;21(6):2572-2588
KEYWORDS
TOPICS
ABSTRACT
Introduction:
The aim of this study is to explain the effects and mechanisms of Nur77 in glioma development.
Material and methods:
Nur77 protein and gene expression were measured by immunohistochemistry and real-time quantitative polymerase chain reaction (RT-qPCR) in tissues from glioma and craniocerebral injury patients. The patients were divided into two groups according to median Nur77 gene expression and the correlations between clinical pathology, progression-free survival (PFS), overall survival (OS), and Nur77 gene expression were analysed. U257 and U87 cells were employed to assess the effects of Nur77 in cancer cell development by MTT, flow cytometry, and transwell and wound healing assays. Relative protein and gene expression were evaluated by Western blot assay and RT-qPCR, and PI3K nuclear volumes were evaluated by cellular immunofluorescence to explain the relevant mechanisms.
Results:
Compared with that in normal tissues, Nur77 protein and gene expression in glioma tissues was significantly up-regulated (p < 0.01); this up-regulation increased with increasing tumour stage (p < 0.01). Compared with the high Nur77 expression group, the low Nur expression group showed significantly improved PFS and OS (both p < 0.01). Nur77 gene expression appeared to be closely correlated with clinical pathology. In vitro studies of si-Nur77 transfection revealed a significant reduction in the cell viability of the si-Nur77 group, with increasing apoptosis rate (p < 0.001). Nur77 knock-down resulted in significant downregulation of cell invasion and wound healing rates in the si-Nur77 group (p < 0.01), with remarkable reductions in CXCR4 and PI3K.
Conclusions:
Nur77 is an important oncogene in glioma, and Nur77 expression is closely correlated with glioma clinical pathology, PFS, and OS. Nur77 knock-down exerts inhibitory effects on glioma by regulating CXCR4/PI3K.
REFERENCES (30)
1.
Louis DN, Perry A, Reifenberger G, et al. The 2016 Would Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol 2016; 131: 803-20.
2.
Ostrom QT, Gittleman H, Fulop J, et al. CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumor Diagnosed in the United States in 2008-2012. Neuro Oncol 2015; 17 Suppl 4: iv1-62.
3.
Lee SO, Li X, Hedrick E, et al. Diindolylmethane analogs bind NR4A1 and are NR4A1 antagonists in colon cancer cells. Mol Endocrinol 2014; 28: 1729-39.
4.
Hedrick E, Lee SO, Doddapaneni R, et al. Nuclear receptor 4A1 as a drug target for breast cancer chemotherapy. Endocr Relat Cancer 2015; 22: 831-40.
5.
Lacey A, Hedrick E, Li X, et al. Nuclear receptor 4A1 (NR4A1) as a drug target for treating rhabdomyosarcoma (RMS). Oncotarget 2016; 7: 31257-69.
6.
Kolluri SK, Bruey-Sedano N, Cao X, et al. Mitogenic effect of orphan receptor TR3 and its regulation by MEKK1 in lung cancer cells. Mol Cell Biol 2003; 23: 8651-67.
7.
Zeng H, Qin L, Zhao D, et al. Orphan nuclear receptor TR3/Nur77 regulates VEGF-A-induced angiogenesis through its transcriptional activity. J Exp Med 2006; 203: 719-29.
8.
Alexopoulou AN, Leao M, Caballero OL, et al. Dissecting the transcriptional networks underlying breast cancer: NR4A1 reduces the migration of normal and breast cancer cell lines. Breast Cancer Res 2010; 12: R51.
9.
Zhao BX, Chen HZ, Du XD, et al. Orphan receptor TR3 enhances p53 transactivation and represses DNA double-strand break repair in hepatoma cells under ionizing radiation. Mol Endocrinol 2011; 25: 1337-50.
10.
Xie L, Jiang F, Zhang X, et al. Honokiol sensitizes breast cancer cells to TNF-alpha induction of apoptosis by inhibiting Nur77 expression. Br J Pharmacol 2016; 173: 344-56.
11.
Yang H, Bushue N, Bu P, et al. Induction and intracellular localization of Nur77 dictate fenretinide-induced apoptosis of human liver cancer cells. Biochem Pharmacol 2010; 79: 948-54.
12.
Katagiri Y, Takeda K, Yu ZX, et al. Modulation of retinoid signaling through NGF-induced nuclear export of NGF1-B. Nat Cell Biol 2000; 2: 435-40.
13.
Okabe T, Takayanagi R, Adachi M, et al. Nur77, a member of the steroid receptor superfamily, antagonizes negative feedback of ACTH synthesis and secretion by glucocorticoid in pituitary corticotrope cells. J Endocrinol 1998; 156: 169-75.
14.
Li H, Kolluri SK, Gu J, et al. Cytochrome C release and apoptosis induced by mitochondrial targeting of nuclear receptor TR3. Science 2000; 289: 1159-64.
15.
Wu L, Chen L. Characteristics of Nur77 and its ligands as potential anticancer compounds (Review). Mol Med Rep 2018; 18: 4793-801.
16.
Lee SO, Abdelrahim M, Yoon K, et al. Inactivation of the orphan nuclear receptor TR3/Nur77 inhibits pancreatic cancer cell and tumor growth. Cancer Res 2010; 70: 6824-36.
17.
Kolluri SK, Bruey-Sedano N, Cao XH, et al. Mitogenic effect of orphan receptor TR3 and its regulation by MEKK1 in lung cancer cells. Mol Cell Biol 2003; 23: 8651-67.
18.
Wang JR, Gan WJ, Li XM, et al. Orphan nuclear receptor Nur77 promotes colorectal cancer invasion and metastasis by regulating MMP-9 and E-cadherin. Carcinogonesis 2014; 35: 2474-84.
19.
Kingsmore KM, Logsdon DK, Floyd DH, et al. Interstitial flow differentially increases patient-derived glioblastoma stem cell invasion via CXCR4, CXCL12, and CD44-mediated mechanisms. Integr Biol (Camb) 2016; 8: 1246-60.
20.
Pal J, Patil V, Mondal B, et al. Epigenetically silenced GNG4 inhibits SDF1alpha/CXCR4 signaling in mesenchymal glioblastoma. Genes Cancer 2016; 7: 136-47.
21.
Gravina GL, Mancini A, Colapietro A, et al. The novel CXCR4 antagonist, PRX177561, reduces tumor cell proliferation and accelerates cancer stem cell differentiation in glioblastoma preclinical models. Tumour Biol 2017; 39: 1010428317695528.
22.
Qin L, Kufareva I, Holden LG, et al. Structural biology. Crystal structure of the chemokine receptor CXCR4 in complex with a viral chemokine. Science 2015; 347: 1117-22.
23.
Ludwig H, Weisel K, Peltrucci MT, et al. Olapesed pegol, an anti-CXCL12/SDF-1 Spiegelmer, alone and with bortezomib-dexamethasone in relapsed/refractory multiple myeloma: a phase IIa study. Leukemia 2017; 31: 997-1000.
24.
Sleightholin RL, Neilsen BK, Li J, et al. Emerging roles of the CXCL12/CXCR4 axis in pancreatic cancer progression and therapy. Pharmacol Ther 2017; 179: 158-70.
25.
Zhou W, Guo S, Liu M, et al. Targeting CXCL12/CXCR4 Axis in tumor immunotherapy. Curr Med Chem 2019; 26: 3026-41.
26.
Yin X, Liu Z, Zhu P, et al. CXCL12/CXCR4 promotes proliferation, migration, and invasion of adamantinomatous craniopharyngiomas via PI3K/AKT signal pathway. J Cell Biochem 2019; 120: 9724-36.
27.
Han K, Li C, Zhang X, et al. DUXAP10 inhibition attenuates the proliferation and metastasis of hepatocellular carcinoma cells by regulation of the Wnt/beta-catenin and PI3K/Akt signaling pathways. Biosci Rep 2019; 39: BSR20181457.
28.
Wang X, Gao S, Xie F, et al. High expression of TCF12 contributes to gastric cancer development via being target regulated by miR-183 and activating PI3K/AKT pathway. J Cell Biochem 2019; 120: 13903-11.
29.
Zhu H, Diao S, Lim V, et al. FAM83D inhibits autophagy and promotes proliferation and invasion of ovarian cancer cells via PI3K/AKT/mTOR pathway. Acta Biochim Biophys Sin (Shanghai) 2019; 51: 509-26.
30.
Zhang L, Zhou J, Qin X, et al. Astragaloside IV inhibits the invasion and metastasis of SiHa cervical cancer cells via the TGF-beta1-mediated PI3K and MAPK pathways. Oncol Rep 2019; 41: 2975-86.