9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Curcumin Treatment in Combination with Glucose Restriction Inhibits Intracellular Alkalinization and Tumor Growth in Hepatoma Cells

      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

          Dysregulation of cellular energy metabolism is closely linked to cancer development and progression. Calorie or glucose restriction (CR or GR) inhibits energy-dependent pathways, including IGF-1/PI3K/Akt/mTOR, in cancer cells. However, alterations in proton dynamics and reversal of the pH gradient across the cell membrane, which results in intracellular alkalinization and extracellular acidification in cancer tissues, have emerged as important etiopathogenic factors. We measured glucose, lactate, and ATP production after GR, plant-derived CR-mimetic curcumin treatment, and curcumin plus GR in human hepatoma cells. Intracellular pH regulatory effects, in particular, protein–protein interactions within mTOR complex-1 and its structural change, were investigated. Curcumin treatment or GR mildly inhibited Na+/H+ exchanger-1 (NHE1). vATPase, monocarboxylate transporter (MCT)-1, and MCT4 level. Combination treatment with curcumin and GR further enhanced the inhibitory effects on these transporters and proton-extruding enzymes, with intracellular pH reduction. ATP and lactate production decreased according to pH change. Modeling of mTOR protein revealed structural changes upon treatments, and curcumin plus GR decreased binding of Raptor and GβL to mTOR, as well as of Rag A and Rag B to Raptor. Consequently, 4EBP1 phosphorylation was decreased and cell migration and proliferation were inhibited in a pH-dependent manner. Autophagy was increased by curcumin plus GR. In conclusion, curcumin treatment combined with GR may be a useful supportive approach for preventing intracellular alkalinization and cancer progression.

          Related collections

          Most cited references39

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

          Targeting lactate metabolism for cancer therapeutics.

          Lactate, once considered a waste product of glycolysis, has emerged as a critical regulator of cancer development, maintenance, and metastasis. Indeed, tumor lactate levels correlate with increased metastasis, tumor recurrence, and poor outcome. Lactate mediates cancer cell intrinsic effects on metabolism and has additional non-tumor cell autonomous effects that drive tumorigenesis. Tumor cells can metabolize lactate as an energy source and shuttle lactate to neighboring cancer cells, adjacent stroma, and vascular endothelial cells, which induces metabolic reprogramming. Lactate also plays roles in promoting tumor inflammation and in functioning as a signaling molecule that stimulates tumor angiogenesis. Here we review the mechanisms of lactate production and transport and highlight emerging evidence indicating that targeting lactate metabolism is a promising approach for cancer therapeutics.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            mTOR kinase structure, mechanism and regulation by the rapamycin-binding domain

            The mammalian target of rapamycin (mTOR), a phosphoinositide 3-kinase related protein kinase, controls cell growth in response to nutrients and growth factors and is frequently deregulated in cancer. Here we report co-crystal structures of a truncated mTOR-mLST8 complex with an ATP transition state mimic and with ATP-site inhibitors. The structures reveal an intrinsically active kinase conformation, with catalytic residues and mechanism remarkably similar to canonical protein kinases. The active site is highly recessed due to the FKBP12-Rapamycin binding (FRB) domain and an inhibitory helix protruding from the catalytic cleft. mTOR activating mutations map to the structural framework that holds these elements in place, indicating the kinase is controlled by restricted access. In vitro biochemistry indicates that the FRB domain acts as a gatekeeper, with its rapamycin-binding site interacting with substrates to grant them access to the restricted active site. FKBP12-rapamycin inhibits by directly blocking substrate recruitment and by further restricting active site access. The structures also reveal active site residues and conformational changes that underlie inhibitor potency and specificity.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              AMPK activators: mechanisms of action and physiological activities

              AMP-activated protein kinase (AMPK) is a central regulator of energy homeostasis, which coordinates metabolic pathways and thus balances nutrient supply with energy demand. Because of the favorable physiological outcomes of AMPK activation on metabolism, AMPK has been considered to be an important therapeutic target for controlling human diseases including metabolic syndrome and cancer. Thus, activators of AMPK may have potential as novel therapeutics for these diseases. In this review, we provide a comprehensive summary of both indirect and direct AMPK activators and their modes of action in relation to the structure of AMPK. We discuss the functional differences among isoform-specific AMPK complexes and their significance regarding the development of novel AMPK activators and the potential for combining different AMPK activators in the treatment of human disease.
                Bookmark

                Author and article information

                Journal
                Int J Mol Sci
                Int J Mol Sci
                ijms
                International Journal of Molecular Sciences
                MDPI
                1422-0067
                14 May 2019
                May 2019
                : 20
                : 10
                : 2375
                Affiliations
                [1 ]Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea; kswlab2015@ 123456gmail.com
                [2 ]Institute for Translational and Clinical Research, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Korea; minjicha619@ 123456gmail.com
                [3 ]KANT Science Research Institute, Incheon 22711, Korea; seulki1011@ 123456nate.com (S.-K.L.); jxh630@ 123456gmail.com (X.J.)
                [4 ]Department of Medical Science, Catholic Kwandong University College of Medicine, Gangneung 25601, Korea; songbw@ 123456gmail.com
                [5 ]Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea; jaemyun@ 123456yuhs.ac
                [6 ]Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
                [7 ]Department of Nuclear Medicine, Catholic Kwandong University International St. Mary’s Hospital, Incheon 22711, Korea
                Author notes
                [* ]Correspondence: jhpark5277@ 123456yuhs.ac (J.H.P.); jdlee@ 123456yuhs.ac (J.D.L.); Tel.: +82-2-2228-1815 (J.H.P.); +82-10-2829-1159 (J.D.L.)
                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0002-9962-0941
                Article
                ijms-20-02375
                10.3390/ijms20102375
                6566721
                31091659
                6159b794-5817-4f72-80c5-35c781099818
                © 2019 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 18 April 2019
                : 11 May 2019
                Categories
                Article

                Molecular biology
                hepatoma,intracellular ph,curcumin,glucose restriction,tumor suppression
                Molecular biology
                hepatoma, intracellular ph, curcumin, glucose restriction, tumor suppression

                Comments

                Comment on this article