12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The lipid metabolism gene FTO influences breast cancer cell energy metabolism via the PI3K/AKT signaling pathway

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The present study assessed the effect of the lipid metabolism, fat mass and the obesity-associated gene (FTO), on energy metabolism of breast cancer cells. The human breast cancer cell lines, MCF-7 and MDA-MB-231, and HCC1937 human breast cells were studied. Real-time PCR was used to measure the levels of FTO mRNA from breast cancer cells and normal breast cells. MDA-MB-231 cells were transfected with miFTO inhibitor or inhibitor control, and cells were assessed for levels of lactic acid, ATP, pyruvate kinase activity, and hexokinase activity assay using specific kits. Western blot analysis was used to measure the levels of phosphatidylinositol 3-kinase (PI3K), p-PI3K, protein kinase B (Akt) and p-Akt in transfected breast cancer cells. The expression of FTO was significantly increased in MCF-7 and MDA-MB-231 cells compared with HCC1937 cells (P<0.01). The lactic acid content of breast cancer cells transfected with the miFTO inhibitor was significantly lower compared with cells transfected with the miFTO inhibitor control and nontransfected cells (P<0.05). The ATP content of breast cancer cells transfected with the miFTO inhibitor was significantly lower compared with the control group and inhibitor control group (P<0.05). The pyruvate kinase activity and hexokinase activity of breast cancer cells transfected with the miFTO inhibitor were significantly lower compared with the control group and inhibitor control group (P<0.01). Western blot analysis showed that after breast cancer cells were transfected with the miFTO inhibitor, the levels of PI3K, p-PI3K, Akt and p-Akt were significantly lower than in the control group and inhibitor control group. In conclusion, the FTO gene is overexpressed in breast cancer cells. Overexpression of the FTO gene can promote breast cancer cell glycolysis and the mechanism is related to the PI3K/AKT signaling pathway.

          Related collections

          Most cited references13

          • Record: found
          • Abstract: found
          • Article: not found

          Hexokinase II integrates energy metabolism and cellular protection: Akting on mitochondria and TORCing to autophagy.

          Accumulating evidence reveals that metabolic and cell survival pathways are closely related, sharing common signaling molecules. Hexokinase catalyzes the phosphorylation of glucose, the rate-limiting first step of glycolysis. Hexokinase II (HK-II) is a predominant isoform in insulin-sensitive tissues such as heart, skeletal muscle, and adipose tissues. It is also upregulated in many types of tumors associated with enhanced aerobic glycolysis in tumor cells, the Warburg effect. In addition to the fundamental role in glycolysis, HK-II is increasingly recognized as a component of a survival signaling nexus. This review summarizes recent advances in understanding the protective role of HK-II, controlling cellular growth, preventing mitochondrial death pathway and enhancing autophagy, with a particular focus on the interaction between HK-II and Akt/mTOR pathway to integrate metabolic status with the control of cell survival.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Stabilization of LKB1 and Akt by neddylation regulates energy metabolism in liver cancer

            The current view of cancer progression highlights that cancer cells must undergo through a post-translational regulation and metabolic reprogramming to progress in an unfriendly environment. In here, the importance of neddylation modification in liver cancer was investigated. We found that hepatic neddylation was specifically enriched in liver cancer patients with bad prognosis. In addition, the treatment with the neddylation inhibitor MLN4924 in Phb1-KO mice, an animal model of hepatocellular carcinoma showing elevated neddylation, reverted the malignant phenotype. Tumor cell death in vivo translating into liver tumor regression was associated with augmented phosphatidylcholine synthesis by the PEMT pathway, known as a liver-specific tumor suppressor, and restored mitochondrial function and TCA cycle flux. Otherwise, in protumoral hepatocytes, neddylation inhibition resulted in metabolic reprogramming rendering a decrease in oxidative phosphorylation and concomitant tumor cell apoptosis. Moreover, Akt and LKB1, hallmarks of proliferative metabolism, were altered in liver cancer being new targets of neddylation. Importantly, we show that neddylation-induced metabolic reprogramming and apoptosis were dependent on LKB1 and Akt stabilization. Overall, our results implicate neddylation/signaling/metabolism, partly mediated by LKB1 and Akt, in the development of liver cancer, paving the way for novel therapeutic approaches targeting neddylation in hepatocellular carcinoma.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Regulation of mitochondrial nutrient and energy metabolism by BCL-2 family proteins.

              Cells have evolved a highly integrated network of mechanisms to coordinate cellular survival/death, proliferation, differentiation, and repair with metabolic states. It is therefore not surprising that proteins with canonical roles in cell death/survival also modulate nutrient and energy metabolism and vice versa. The finding that many BCL-2 (B cell lymphoma 2) proteins reside at mitochondria or can translocate to this organelle has long motivated investigation into their involvement in normal mitochondrial physiology and metabolism. These endeavors have led to the discovery of homeostatic roles for BCL-2 proteins beyond apoptosis. We predominantly focus on recent findings that link select BCL-2 proteins to carbon substrate utilization at the level of mitochondrial fuel choice, electron transport, and metabolite import independent of their cell death regulatory function.
                Bookmark

                Author and article information

                Journal
                Oncol Lett
                Oncol Lett
                OL
                Oncology Letters
                D.A. Spandidos
                1792-1074
                1792-1082
                June 2017
                13 April 2017
                13 April 2017
                : 13
                : 6
                : 4685-4690
                Affiliations
                [1 ]Department of Metabolic Diseases and Nutritional Disorders, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
                [2 ]Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
                [3 ]Department of Respiratory Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
                [4 ]Department of Nursing, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
                [5 ]Department of Metabolic Diseases and Nutritional Disorders, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
                [6 ]Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
                Author notes
                Correspondence to: Dr Bingyin Shi, Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Street, Xi'an, Shaanxi 710061, P.R. China, E-mail: shi_by1@ 123456163.com
                Article
                OL-0-0-6038
                10.3892/ol.2017.6038
                5452952
                28599470
                5fdefac0-d6ff-4e57-8a49-ee5e67612cf8
                Copyright: © Liu et al.

                This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.

                History
                : 21 July 2016
                : 22 February 2017
                Categories
                Articles

                Oncology & Radiotherapy
                breast cancer,energy metabolism,fat mass and obesity-associated,phosphatidylinositol 3-kinase/protein kinase b

                Comments

                Comment on this article