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

      mTOR Signaling and SREBP Activity Increase FADS2 Expression and Can Activate Sapienate Biosynthesis

      brief-report

      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.

          Summary

          Cancer cells display an increased plasticity in their lipid metabolism, which includes the conversion of palmitate to sapienate via the enzyme fatty acid desaturase 2 (FADS2). We find that FADS2 expression correlates with mammalian target of rapamycin (mTOR) signaling and sterol regulatory element-binding protein 1 (SREBP-1) activity across multiple cancer types and is prognostic in some cancer types. Accordingly, activating mTOR signaling by deleting tuberous sclerosis complex 2 ( Tsc2) or overexpression of SREBP-1/2 is sufficient to increase FADS2 mRNA expression and sapienate metabolism in mouse embryonic fibroblasts (MEFs) and U87 glioblastoma cells, respectively. Conversely, inhibiting mTOR signaling decreases FADS2 expression and sapienate biosynthesis in MEFs with Tsc2 deletion, HUH7 hepatocellular carcinoma cells, and orthotopic HUH7 liver xenografts. In conclusion, we show that mTOR signaling and SREBP activity are sufficient to activate sapienate metabolism by increasing FADS2 expression. Consequently, targeting mTOR signaling can reduce sapienate metabolism in vivo.

          Graphical Abstract

          Highlights

          • FADS2 expression is prognostic in some cancers

          • FADS2 is a target of SREBP-1 and SREBP-2

          • SREBP activity and mTOR signaling regulate FADS2-mediated sapienate metabolism

          • Torin1 treatment reduces FADS2 expression and sapienate metabolism in xenografts

          Abstract

          Triki et al. report that FADS2 expression is prognostic in some cancers and that FADS2-mediated sapienate metabolism is regulated by mTOR signaling. Mechanistically, FADS2 is a target of SREBP-1/2. Inhibition of mTOR or SREBP reduces FADS2 expression and sapienate metabolism in cancer cells and liver xenografts.

          Related collections

          Most cited references26

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

          SREBP Activity Is Regulated by mTORC1 and Contributes to Akt-Dependent Cell Growth

          Summary Cell growth (accumulation of mass) needs to be coordinated with metabolic processes that are required for the synthesis of macromolecules. The PI3-kinase/Akt signaling pathway induces cell growth via activation of complex 1 of the target of rapamycin (TORC1). Here we show that Akt-dependent lipogenesis requires mTORC1 activity. Furthermore, nuclear accumulation of the mature form of the sterol responsive element binding protein (SREBP1) and expression of SREBP target genes was blocked by the mTORC1 inhibitor rapamycin. We also show that silencing of SREBP blocks Akt-dependent lipogenesis and attenuates the increase in cell size in response to Akt activation in vitro. Silencing of dSREBP in flies caused a reduction in cell and organ size and blocked the induction of cell growth by dPI3K. Our results suggest that the PI3K/Akt/TOR pathway regulates protein and lipid biosynthesis in an orchestrated manner and that both processes are required for cell growth.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A Pan-Cancer Proteogenomic Atlas of PI3K/AKT/mTOR Pathway Alterations.

            Molecular alterations involving the PI3K/AKT/mTOR pathway (including mutation, copy number, protein, or RNA) were examined across 11,219 human cancers representing 32 major types. Within specific mutated genes, frequency, mutation hotspot residues, in silico predictions, and functional assays were all informative in distinguishing the subset of genetic variants more likely to have functional relevance. Multiple oncogenic pathways including PI3K/AKT/mTOR converged on similar sets of downstream transcriptional targets. In addition to mutation, structural variations and partial copy losses involving PTEN and STK11 showed evidence for having functional relevance. A substantial fraction of cancers showed high mTOR pathway activity without an associated canonical genetic or genomic alteration, including cancers harboring IDH1 or VHL mutations, suggesting multiple mechanisms for pathway activation.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              SIRT1 deacetylates and inhibits SREBP-1C activity in regulation of hepatic lipid metabolism.

              The SIRT1 deacetylase inhibits fat synthesis and stimulates fat oxidation in response to fasting, but the underlying mechanisms remain unclear. Here we report that SREBP-1c, a key lipogenic activator, is an in vivo target of SIRT1. SIRT1 interaction with SREBP-1c was increased during fasting and decreased upon feeding, and consistently, SREBP-1c acetylation levels were decreased during fasting in mouse liver. Acetylated SREBP-1c levels were also increased in HepG2 cells treated with insulin and glucose to mimic feeding conditions, and down-regulation of p300 by siRNA decreased the acetylation. Depletion of hepatic SIRT1 by adenoviral siRNA increased acetylation of SREBP-1c with increased lipogenic gene expression. Tandem mass spectrometry and mutagenesis studies revealed that SREBP-1c is acetylated by p300 at Lys-289 and Lys-309. Mechanistic studies using acetylation-defective mutants showed that SIRT1 deacetylates and inhibits SREBP-1c transactivation by decreasing its stability and its occupancy at the lipogenic genes. Remarkably, SREBP-1c acetylation levels were elevated in diet-induced obese mice, and hepatic overexpression of SIRT1 or treatment with resveratrol, a SIRT1 activator, daily for 1 week decreased acetylated SREBP-1c levels with beneficial functional outcomes. These results demonstrate an intriguing connection between elevated SREBP-1c acetylation and increased lipogenic gene expression, suggesting that abnormally elevated SREBP-1c acetylation increases SREBP-1c lipogenic activity in obese mice. Reducing acetylation of SREBP-1c by targeting SIRT1 may be useful for treating metabolic disorders, including fatty liver, obesity, and type II diabetes.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Rep
                Cell Rep
                Cell Reports
                Cell Press
                2211-1247
                23 June 2020
                23 June 2020
                23 June 2020
                : 31
                : 12
                : 107806
                Affiliations
                [1 ]Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
                [2 ]Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
                [3 ]Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Im Neuenheimer Feld 581, 69120 Heidelberg, Germany
                [4 ]Faculty of Biosciences, University of Heidelberg, 69120 Heidelberg, Germany
                [5 ]Biochemistry and Molecular Biology, Theodor-Boveri-Institute, Biocenter, Am Hubland, 97074 Würzburg, Germany
                [6 ]Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Thalkirchner Strasse 36, 80337 Munich, Germany
                [7 ]German Cancer Consortium (DKTK), partner site Munich, 80337 Munich, Germany
                [8 ]Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
                [9 ]Institute of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
                Author notes
                []Corresponding author sarah-maria.fendt@ 123456kuleuven.vib.be
                [10]

                Lead Contact

                Article
                S2211-1247(20)30787-7 107806
                10.1016/j.celrep.2020.107806
                7326293
                32579932
                baaa71e7-8f75-4a49-b30a-6bd605ed99e1
                © 2020 The Author(s)

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 28 October 2019
                : 3 April 2020
                : 2 June 2020
                Categories
                Article

                Cell biology
                sapienate,fads2,mtor,hepatocellular carcinoma,glioblastoma,cancer,srebp,scd1,palmitoleate,fatty acid metabolism,palmitate

                Comments

                Comment on this article