Unknown

Dataset Information

0

KPNA2 promotes metabolic reprogramming in glioblastomas by regulation of c-myc.


ABSTRACT: BACKGROUND:Cancer cells maintain energy metabolism mainly by glycolysis, even under sufficient oxygen conditions. It gives cancer cells better growth advantages under complicated internal environment. KPNA2 is a novel oncogene that has received much attention in recent years, but the exact mechanisms of KPNA2 in tumorigenesis and progression are largely unknown. Especially its potential roles in the metabolic transformation of tumors still remain to be explored. METHODS:The expressions of KPNA2 in glioblastoma and normal human brain samples were analyzed by immunohistochemical analysis. The activities of key enzymes in glycolysis, the production of lactate acid and glucose uptake were investigated by colorimetry. GLUT-1 expression was measured by flow cytometry. CCK8 was used to examine the cell viability in vitro, and the xenograft models in nude mice were established to explore the roles of KPNA2 in vivo. In addition, Co-IP, subcellular fractionation, western blot, immunofluorescence and luciferase assay were used to investigate the internal connection between KPNA2, c-myc and E2F1. RESULTS:In the present study, we found that KPNA2 was highly expressed in the glioma compared to the normal brain tissues. Level of KPNA2 was an independent predictor of prognosis in the glioma patients. Knockdown of KPNA2 in the glioblastoma cell lines U87 and U251 decreased deoxyglucose uptake, activities of the key glycolytic enzymes and lactate production. The level of oxidative phosphorylation (OXPHOS) was moderately decreased. Additioanlly, tumor proliferation and invasiveness were concomitantly downregulated. We have identified c-myc as a potential mediator of KPNA2. Aberrant expression of KPNA2 significantly changed the subcellular distribution of c-myc as well as its expression level. E2F1, another key cargo protein of KPNA2, was further identified to play a potential role in regulating the transcription of c-myc by KPNA2. CONCLUSIONS:Our findings suggested that KPNA2, a potential tumor oncogene, performs its function in part via regulating cellular metabolism through c-myc signaling axis. It would provide a possible explanation for Warburg effect and thus offer a new perspective to the roles of KPNA2 in gliomagenesis.

SUBMITTER: Li J 

PROVIDER: S-EPMC6097452 | biostudies-literature | 2018 Aug

REPOSITORIES: biostudies-literature

altmetric image

Publications

KPNA2 promotes metabolic reprogramming in glioblastomas by regulation of c-myc.

Li Jie J   Liu Qian Q   Liu Zihao Z   Xia Qian Q   Zhang Zihao Z   Zhang Rui R   Gao Taihong T   Gu Guangyan G   Wang Yanan Y   Wang Dan D   Chen Xiuyang X   Yang Yihang Y   He Dong D   Xin Tao T  

Journal of experimental & clinical cancer research : CR 20180816 1


<h4>Background</h4>Cancer cells maintain energy metabolism mainly by glycolysis, even under sufficient oxygen conditions. It gives cancer cells better growth advantages under complicated internal environment. KPNA2 is a novel oncogene that has received much attention in recent years, but the exact mechanisms of KPNA2 in tumorigenesis and progression are largely unknown. Especially its potential roles in the metabolic transformation of tumors still remain to be explored.<h4>Methods</h4>The expres  ...[more]

Similar Datasets

| S-EPMC5216711 | biostudies-literature
| S-EPMC6636331 | biostudies-literature
| S-EPMC4326605 | biostudies-literature
| S-EPMC5308762 | biostudies-literature
| S-EPMC3248798 | biostudies-literature
| S-EPMC3818456 | biostudies-literature
| S-EPMC4418882 | biostudies-literature
| S-EPMC5082756 | biostudies-literature