Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2023, Vol. 17 Issue (1) : 143-155    https://doi.org/10.1007/s11684-022-0929-y
RESEARCH ARTICLE
RGS16 regulated by let-7c-5p promotes glioma progression by activating PI3K-AKT pathway
Chaochao Wang1,2, Hao Xue1,2, Rongrong Zhao1,2, Zhongzheng Sun2,3, Xiao Gao1,2, Yanhua Qi1,2, Huizhi Wang1,2, Jianye Xu1,2, Lin Deng1,2(), Gang Li1,2()
1. Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan 250012, China
2. Shandong Key Laboratory of Brain Function Remodeling, Jinan 250012, China
3. The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
 Download: PDF(8259 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Gliomas are the most common central nervous system tumours; they are highly aggressive and have a poor prognosis. RGS16 belongs to the regulator of G-protein signalling (RGS) protein family, which plays an important role in promoting various cancers, such as breast cancer, pancreatic cancer, and colorectal cancer. Moreover, previous studies confirmed that let-7c-5p, a well-known microRNA, can act as a tumour suppressor to regulate the progression of various tumours by inhibiting the expression of its target genes. However, whether RGS16 can promote the progression of glioma and whether it is regulated by miR let-7c-5p are still unknown. Here, we confirmed that RGS16 is upregulated in glioma tissues and that high expression of RGS16 is associated with poor survival. Ectopic deletion of RGS16 significantly suppressed glioma cell proliferation and migration both in vitro and in vivo. Moreover, RGS16 was validated as a direct target gene of miR let-7c-5p. The overexpression of miR let-7c-5p obviously downregulated the expression of RGS16, and knocking down miR let-7c-5p had the opposite effect. Thus, we suggest that the suppression of RGS16 by miR let-7c-5p can promote glioma progression and may serve as a potential prognostic biomarker and therapeutic target in glioma.

Keywords RGS16      let-7c-5p      glioma      proliferation      migration     
Corresponding Author(s): Lin Deng,Gang Li   
Just Accepted Date: 30 August 2022   Online First Date: 23 November 2022    Issue Date: 15 March 2023
 Cite this article:   
Chaochao Wang,Hao Xue,Rongrong Zhao, et al. RGS16 regulated by let-7c-5p promotes glioma progression by activating PI3K-AKT pathway[J]. Front. Med., 2023, 17(1): 143-155.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-022-0929-y
https://academic.hep.com.cn/fmd/EN/Y2023/V17/I1/143
Fig.1  Expression of RGS16 is upregulated in gliomas and is associated with tumor grade. (A) Quantification of RGS16 mRNA expression levels in LGG and GB tissues compared with normal brain tissues in TCGA. (B) IHC staining images for RGS16 in gliomas of various grades and normal brain specimens. Magnification, 200×, upper; 400×, lower. (C) Quantitative analysis of RGS16 staining scores in gliomas of various grades and normal brain specimens. Scores were presented as the median + interquartile range, significance was determined using one-way ANOVA of non-parametric test (Kruskal-Wallis). RGS, regulator of G-protein signaling; TCGA, The Cancer Genome Atlas; LGG, low grade glioma; GB, glioblastoma; WHO, World Health Organization.
Fig.2  Knockdown of RGS16 inhibits glioma cell proliferation. U87 and U251 cells transfected with RGS16 siRNA or NC were characterized. (A, B) Expression of RGS16 was downregulated in glioma cells at the mRNA and protein level. (C, D) CCK-8 assays revealed that knockdown of RGS16 reduced the proliferation of U87 and U251 glioma cells compared to transfection with NC. (E, F) Colony-formation assays showed that knockdown of RGS16 reduced colony numbers. (G, H) EdU staining performed 48 h after transfection showed fewer proliferating cells (DAPI staining) in cells with RGS16 knockdown compared with the NC-transfected cells. (I) Variations in protein expression levels were detected via Western blotting. GAPDH was used as the loading control. RGS, regulator of G-protein signaling; siRNA, small interfering RNA; NC, negative control; CCK-8, Cell Counting Kit-8.
Fig.3  Knockdown of RGS16 reduces migration and invasion in glioma cells in vitro. (A–D) Cell migration/invasion assays were performed after RGS16 knockdown in glioma U87 and U251 cells, scale bar = 200 μm. Data are shown as the mean ± standard deviation. (E) Changes in protein expression levels were assessed by Western blotting and showed that MMP2 and MMP9 levels were lower in the si-RGS16 group compared with the NC group. (F) Western blotting assay showed that AKT2 and p-AKT levels were lower in the si-RGS16 group compared with the NC group. GAPDH was used as the internal control. (G, H) Wound-healing assays were performed on monolayers of U87 and U251 glioma cells after 48 h of transfection. Images were taken 0 and 24 h after the wounds were made, scale bar = 200 μm. RGS, regulator of G-protein signaling; siRNA, small interfering RNA; NC, negative control; p-, phospho.
Fig.4  Overexpression of miR-let-7c-5p inhibits glioma cell proliferation/migration/invasion ability. (A) Target site of let-7c-5p in the 3′-UTR of RGS16 was predicted by TargetScan. The mutant sequence used was identical to the wild-type sequence except for mutations at the 3′ end of the target site. (B) Relative expression of miR let-7c-5p in NBTs and GBM tissues. (C) U87 and U251 cells were transfected with let-7c-5p mimics or inhibitor; two independent scrambled sequence was used as the negative control. RT-qPCR assays proved that the expression of let-7c-5p was upregulated when transfected with mimics, while downregulated with inhibitor. (D) EdU staining showed fewer proliferating cells (DAPI staining) in the mimics group compared with the NC group and inhibitor group. (E) Cell migration/invasion assays were performed, and the mimics group exhibited the lowest amount of migrating/invading cells. (F) A bioluminescence imaging system was used to evaluate tumor development in each group as indicated. Representative bioluminescent images are shown. (G) Survival analysis for mice implanted with ov-let-7c-5p or NC. n = 5. (H,I) mRNA and protein levels of RGS16 were downregulated in glioma cells overexpressing let-7c-5p, while upregulated after let-7c-5p inhibitor was transfected. (J) Reporter constructs, including wild-type and mutant, were co-transfected with let-7c-5p mimics or NC for 48 h and luciferase activity was detected. (K) AGO2-dependent RNA immunoprecipitation (RIP) assay in U87 and U251 cells, vs. anti-IgG. The results are presented as the mean of the normalized luciferase intensity. RGS, regulator of G-protein signaling; UTR, untranslated region.
Fig.5  RGS16 is a target of let-7c-5p. (A) CCK8 assays showing the proliferation ability of GBM cells co-transfected with NC, mimics and ov-RGS16 as indicated, n = 3. (B) Representative EdU assays showing the proliferation ability of GBM cells co-transfected with NC, mimics and ov-RGS16 as indicated. Quantification histogram represented relative cell numbers, n = 3. (C) Wound-healing assays were performed on monolayers of GBM cells in each group. Images were taken 0 and 24 h after the wounds were made. (D) Representative Transwell migration and invasion assays showing the migration and invasion ability of GBM cells co-transfected with NC, mimics and ov-RGS16 as indicated. Quantification histogram represented relative cell numbers.
Fig.6  Knockdown of RGS16 inhibits tumorigenesis in vivo. Luciferase-labeled U87 cells were infected with sh-RGS16 or control. (A) Expression of RGS16 was downregulated after transfected with sh-RGS16. (B) A bioluminescence imaging system was used to evaluate tumor development in each group weekly. Representative bioluminescent images are shown. (C) Relative radiance is represented as the total flux value. (D) Survival analysis for mice implanted with sh-RGS16 or control. n = 5. (E) Relative mRNA levels of RGS16 were detected when mice died, tumors were taken out and RNA was extracted for RT-qPCR. (F) Hematoxylin and eosin staining of tumor and peripheral tissue border in the two groups of xenografts. (G–J) Immunohistochemical analysis of RGS16 and Ki-67 expression in sections from sacrificed mice. Magnification, 200×, upper; 400×, lower. Arrows indicate the tumor margin. sh, short hairpin; T, tumor, NBT, normal brain tissue.
1 GP Dunn, ML Rinne, J Wykosky, G Genovese, SN Quayle, IF Dunn, PK Agarwalla, MG Chheda, B Campos, A Wang, C Brennan, KL Ligon, F Furnari, WK Cavenee, RA Depinho, L Chin, WC Hahn. Emerging insights into the molecular and cellular basis of glioblastoma. Genes Dev 2012; 26(8): 756–784
https://doi.org/10.1101/gad.187922.112 pmid: 22508724
2 C Villa, C Miquel, D Mosses, M Bernier, AL Di Stefano. The 2016 World Health Organization classification of tumours of the central nervous system. Presse Med 2018; 47(11–12): e187–e200
https://doi.org/10.1016/j.lpm.2018.04.015 pmid: 30449638
3 F Bray, J Ferlay, I Soerjomataram, RL Siegel, LA Torre, A Jemal. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394–424
https://doi.org/10.3322/caac.21492 pmid: 30207593
4 J Chen, RM McKay, LF Parada. Malignant glioma: lessons from genomics, mouse models, and stem cells. Cell 2012; 149(1): 36–47
https://doi.org/10.1016/j.cell.2012.03.009 pmid: 22464322
5 EM Ross, TM Wilkie. GTPase-activating proteins for heterotrimeric G proteins: regulators of G protein signaling (RGS) and RGS-like proteins. Annu Rev Biochem 2000; 69(1): 795–827
https://doi.org/10.1146/annurev.biochem.69.1.795 pmid: 10966476
6 KM Druey, KJ Blumer, VH Kang, JH Kehrl. Inhibition of G-protein-mediated MAP kinase activation by a new mammalian gene family. Nature 1996; 379(6567): 742–746
https://doi.org/10.1038/379742a0 pmid: 8602223
7 G Liang, G Bansal, Z Xie, KM Druey. RGS16 inhibits breast cancer cell growth by mitigating phosphatidylinositol 3-kinase signaling. J Biol Chem 2009; 284(32): 21719–21727
https://doi.org/10.1074/jbc.M109.028407 pmid: 19509421
8 EN Johnson, TM Seasholtz, AA Waheed, B Kreutz, N Suzuki, T Kozasa, TL Jones, JH Brown, KM Druey. RGS16 inhibits signalling through the G alpha 13-Rho axis. Nat Cell Biol 2003; 5(12): 1095–1103
https://doi.org/10.1038/ncb1065 pmid: 14634662
9 M Berthebaud, C Rivière, P Jarrier, A Foudi, Y Zhang, D Compagno, A Galy, W Vainchenker, F Louache. RGS16 is a negative regulator of SDF-1-CXCR4 signaling in megakaryocytes. Blood 2005; 106(9): 2962–2968
https://doi.org/10.1182/blood-2005-02-0526 pmid: 15998835
10 MB Carper, J Denvir, G Boskovic, DA Primerano, PP Claudio. RGS16, a novel p53 and pRb cross-talk candidate inhibits migration and invasion of pancreatic cancer cells. Genes Cancer 2014; 5(11–12): 420–435
https://doi.org/10.18632/genesandcancer.43 pmid: 25568667
11 N Miyoshi, H Ishii, M Sekimoto, Y Doki, M Mori. RGS16 is a marker for prognosis in colorectal cancer. Ann Surg Oncol 2009; 16(12): 3507–3514
https://doi.org/10.1245/s10434-009-0690-3 pmid: 19760045
12 X Sun, C Charbonneau, L Wei, Q Chen, RM Terek. miR-181a targets RGS16 to promote chondrosarcoma growth, angiogenesis, and metastasis. Mol Cancer Res 2015; 13(9): 1347–1357
https://doi.org/10.1158/1541-7786.MCR-14-0697 pmid: 26013170
13 D Sayed, M Abdellatif. MicroRNAs in development and disease. Physiol Rev 2011; 91(3): 827–887
https://doi.org/10.1152/physrev.00006.2010 pmid: 21742789
14 GA Calin, CM Croce. MicroRNA signatures in human cancers. Nat Rev Cancer 2006; 6(11): 857–866
https://doi.org/10.1038/nrc1997 pmid: 17060945
15 X Fu, X Mao, Y Wang, X Ding, Y Li. Let-7c-5p inhibits cell proliferation and induces cell apoptosis by targeting ERCC6 in breast cancer. Oncol Rep 2017; 38(3): 1851–1856
https://doi.org/10.3892/or.2017.5839 pmid: 28731186
16 M Huang, X Gong. Let-7c inhibits the proliferation, invasion, and migration of glioma cells via targeting E2F5. Oncol Res 2018; 26(7): 1103–1111
https://doi.org/10.3727/096504018X15164123839400 pmid: 29362021
17 PY Wen, S Kesari. Malignant gliomas in adults. N Engl J Med 2008; 359(5): 492–507
https://doi.org/10.1056/NEJMra0708126 pmid: 18669428
18 R Huang, G Li, Z Zhao, F Zeng, K Zhang, Y Liu, K Wang, H Hu. RGS16 promotes glioma progression and serves as a prognostic factor. CNS Neurosci Ther 2020; 26(8): 791–803
https://doi.org/10.1111/cns.13382 pmid: 32319728
19 I Chu, J Sun, A Arnaout, H Kahn, W Hanna, S Narod, P Sun, CK Tan, L Hengst, J Slingerland. p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2. Cell 2007; 128(2): 281–294
https://doi.org/10.1016/j.cell.2006.11.049 pmid: 17254967
20 G Choe, S Horvath, TF Cloughesy, K Crosby, D Seligson, A Palotie, L Inge, BL Smith, CL Sawyers, PS Mischel. Analysis of the phosphatidylinositol 3′-kinase signaling pathway in glioblastoma patients in vivo. Cancer Res 2003; 63(11): 2742–2746
pmid: 12782577
21 L Wang, ZG Zhang, RL Zhang, SR Gregg, A Hozeska-Solgot, Y LeTourneau, Y Wang, M Chopp. Matrix metalloproteinase 2 (MMP2) and MMP9 secreted by erythropoietin-activated endothelial cells promote neural progenitor cell migration. J Neurosci 2006; 26(22): 5996–6003
https://doi.org/10.1523/JNEUROSCI.5380-05.2006 pmid: 16738242
22 QW Fan, C Cheng, C Hackett, M Feldman, BT Houseman, T Nicolaides, D Haas-Kogan, CD James, SA Oakes, J Debnath, KM Shokat, WA Weiss. Akt and autophagy cooperate to promote survival of drug-resistant glioma. Sci Signal 2010; 3(147): ra81
https://doi.org/10.1126/scisignal.2001017 pmid: 21062993
23 Y Suzuki, K Shirai, K Oka, A Mobaraki, Y Yoshida, SE Noda, M Okamoto, Y Suzuki, J Itoh, H Itoh, S Ishiuchi, T Nakano. Higher pAkt expression predicts a significant worse prognosis in glioblastomas. J Radiat Res (Tokyo) 2010; 51(3): 343–348
https://doi.org/10.1269/jrr.09109 pmid: 20410674
24 G Choe, S Horvath, TF Cloughesy, K Crosby, D Seligson, A Palotie, L Inge, BL Smith, CL Sawyers, PS Mischel. Analysis of the phosphatidylinositol 3′-kinase signaling pathway in glioblastoma patients in vivo. Cancer Res 2003; 63(11): 2742–2746
pmid: 12782577
25 H Mure, K Matsuzaki, KT Kitazato, Y Mizobuchi, K Kuwayama, T Kageji, S Nagahiro. Akt2 and Akt3 play a pivotal role in malignant gliomas. Neuro-oncol 2010; 12(3): 221–232
https://doi.org/10.1093/neuonc/nop026 pmid: 20167810
26 DF Quail, JA Joyce. The microenvironmental landscape of brain tumors. Cancer Cell 2017; 31(3): 326–341
https://doi.org/10.1016/j.ccell.2017.02.009 pmid: 28292436
27 MD Jansson, AH Lund. MicroRNA and cancer. Mol Oncol 2012; 6(6): 590–610
https://doi.org/10.1016/j.molonc.2012.09.006 pmid: 23102669
28 A Esquela-Kerscher, FJ Slack. Oncomirs—microRNAs with a role in cancer. Nat Rev Cancer 2006; 6(4): 259–269
https://doi.org/10.1038/nrc1840 pmid: 16557279
29 M Piwecka, K Rolle, A Belter, AM Barciszewska, M Żywicki, M Michalak, S Nowak, MZ Naskręt-Barciszewska, J Barciszewski. Comprehensive analysis of microRNA expression profile in malignant glioma tissues. Mol Oncol 2015; 9(7): 1324–1340
https://doi.org/10.1016/j.molonc.2015.03.007 pmid: 25864039
[1] FMD-22006-OF-LG_suppl_1 Download
[1] Shuzhen Guan, Lin Feng, Jinrui Wei, Guizhen Wang, Lichuan Wu. Knockdown of RFC4 inhibits the cell proliferation of nasopharyngeal carcinoma in vitro and in vivo[J]. Front. Med., 2023, 17(1): 132-142.
[2] Ronghui Yang, Guoguang Ying, Binghui Li. Potential of electron transfer and its application in dictating routes of biochemical processes associated with metabolic reprogramming[J]. Front. Med., 2021, 15(5): 679-692.
[3] Zhaoshi Bao, Yongzhi Wang, Qiangwei Wang, Shengyu Fang, Xia Shan, Jiguang Wang, Tao Jiang. Intratumor heterogeneity, microenvironment, and mechanisms of drug resistance in glioma recurrence and evolution[J]. Front. Med., 2021, 15(4): 551-561.
[4] Jiahui Zhang, Yanan Yin, Jiahui Wang, Jingjing Zhang, Hua Liu, Weiwei Feng, Wen Yang, Bruce Zetter, Yingjie Xu. Prohibitin regulates mTOR pathway via interaction with FKBP8[J]. Front. Med., 2021, 15(3): 448-459.
[5] Jing Ma, Shiyu Chen, Lili Hao, Wei Sheng, Weicheng Chen, Xiaojing Ma, Bowen Zhang, Duan Ma, Guoying Huang. Long non-coding RNA SAP30-2:1 is downregulated in congenital heart disease and regulates cell proliferation by targeting HAND2[J]. Front. Med., 2021, 15(1): 91-100.
[6] Jiajia Hu, Wenbin Shen, Qian Qu, Xiaochun Fei, Ying Miao, Xinyun Huang, Jiajun Liu, Yingli Wu, Biao Li. NES1/KLK10 and hNIS gene therapy enhanced iodine-131 internal radiation in PC3 proliferation inhibition[J]. Front. Med., 2019, 13(6): 646-657.
[7] Xiaodong Duan, Daizhi Peng, Yilan Zhang, Yalan Huang, Xiao Liu, Ruifu Li, Xin Zhou, Jing Liu. Sub-cytotoxic concentrations of ionic silver promote the proliferation of human keratinocytes by inducing the production of reactive oxygen species[J]. Front. Med., 2018, 12(3): 289-300.
[8] Xiaoyu Wang,Yuxuan Gao,Haigang Shi,Na Liu,Wei Zhang,Hongbo Li. Influence of the intensity and loading time of direct current electric field on the directional migration of rat bone marrow mesenchymal stem cells[J]. Front. Med., 2016, 10(3): 286-296.
[9] Lan Wang,Jueheng Wu,Jie Yuan,Xun Zhu,Hongmei Wu,Mengfeng Li. Midline2 is overexpressed and a prognostic indicator in human breast cancer and promotes breast cancer cell proliferation in vitro and in vivo[J]. Front. Med., 2016, 10(1): 41-51.
[10] Feng Gu,Yongjie Ma,Jiao Zhang,Fengxia Qin,Li Fu. Function of Slit/Robo signaling in breast cancer[J]. Front. Med., 2015, 9(4): 431-436.
[11] Yinyin Xie,Yuanliang Zhang,Lu Jiang,Mengmeng Zhang,Zhiwei Chen,Dan Liu,Qiuhua Huang. Disabled homolog 2 is required for migration and invasion of prostate cancer cells[J]. Front. Med., 2015, 9(3): 312-321.
[12] Runlin Shi,Haibing Xiao,Tao Yang,Lei Chang,Yuanfeng Tian,Bolin Wu,Hua Xu. Effects of miR-200c on the migration and invasion abilities of human prostate cancer Du145 cells and the corresponding mechanism[J]. Front. Med., 2014, 8(4): 456-463.
[13] Chuanbao Zhang, Zhaoshi Bao, Wei Zhang, Tao Jiang. Progress on molecular biomarkers and classification of malignant gliomas[J]. Front Med, 2013, 7(2): 150-156.
[14] Li LI, Jianxin JIANG. Regulatory factors of mesenchymal stem cell migration into injured tissues and their signal transduction mechanisms[J]. Front Med, 2011, 5(1): 33-39.
[15] Xiaowei GONG MD, PhD, Xiaoyan MING MD, Xu WANG MM, Daan WANG MD, Peng DENG MM, Yong JIANG MD, PhD, Aihua LIU MD, PhD, . Effect of PRAK gene knockout on the proliferation of mouse embryonic fibroblasts[J]. Front. Med., 2009, 3(4): 379-383.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed