5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Hypoxia‐inducible factor 2α drives hepatosteatosis through the fatty acid translocase CD36

      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

          Background & Aims

          Molecular mechanisms by which hypoxia might contribute to hepatosteatosis, the earliest stage in non‐alcoholic fatty liver disease (NAFLD) pathogenesis, remain still to be elucidated. We aimed to assess the impact of hypoxia‐inducible factor 2α (HIF2α) on the fatty acid translocase CD36 expression and function in vivo and in vitro.

          Methods

          CD36 expression and intracellular lipid content were determined in hypoxic hepatocytes, and in hypoxic CD36‐ or HIF2α ‐silenced human liver cells. Histological analysis, and HIF2α and CD36 expression were evaluated in livers from animals in which von Hippel‐Lindau ( Vhl) gene is inactivated (Vhl f/f‐deficient mice), or both Vhl and Hif2a are simultaneously inactivated (Vhl f/fHif2α /f‐deficient mice), and from 33 biopsy‐proven NAFLD patients and 18 subjects with histologically normal liver.

          Results

          In hypoxic hepatocytes, CD36 expression and intracellular lipid content were augmented. Noteworthy, CD36 knockdown significantly reduced lipid accumulation, and HIF2A gene silencing markedly reverted both hypoxia‐induced events in hypoxic liver cells. Moreover livers from Vhl f/f‐deficient mice showed histologic characteristics of non‐alcoholic steatohepatitis (NASH) and increased CD36 mRNA and protein amounts, whereas both significantly decreased and NASH features markedly ameliorated in Vhl f/fHif2α f/f‐deficient mice. In addition, both HIF2α and CD36 were significantly overexpressed within the liver of NAFLD patients and, interestingly, a significant positive correlation between hepatic transcript levels of CD36 and erythropoietin ( EPO), a HIF2α ‐dependent gene target, was observed in NAFLD patients.

          Conclusions

          This study provides evidence that HIF2α drives lipid accumulation in human hepatocytes by upregulating CD36 expression and function, and could contribute to hepatosteatosis setup.

          Related collections

          Most cited references17

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

          The palmitoylation machinery is a spatially organizing system for peripheral membrane proteins.

          Reversible S-palmitoylation of cysteine residues critically controls transient membrane tethering of peripheral membrane proteins. Little is known about how the palmitoylation machinery governs their defined localization and function. We monitored the spatially resolved reaction dynamics and substrate specificity of the core mammalian palmitoylation machinery using semisynthetic substrates. Palmitoylation is detectable only on the Golgi, whereas depalmitoylation occurs everywhere in the cell. The reactions are not stereoselective and lack any primary consensus sequence, demonstrating that substrate specificity is not essential for de-/repalmitoylation. Both palmitate attachment and removal require seconds to accomplish. This reaction topography and rapid kinetics allows the continuous redirection of mislocalized proteins via the post-Golgi sorting apparatus. Unidirectional secretion ensures the maintenance of a proper steady-state protein distribution between the Golgi and the plasma membrane, which are continuous with endosomes. This generic spatially organizing system differs from conventional receptor-mediated targeting mechanisms and efficiently counteracts entropy-driven redistribution of palmitoylated peripheral membrane proteins over all membranes. 2010 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            HIF2α acts as an mTORC1 activator through the amino acid carrier SLC7A5.

            The mammalian target of rapamycin (mTOR) pathway, which is essential for cell proliferation, is repressed in certain cell types in hypoxia. However, hypoxia-inducible factor 2α (HIF2α) can act as a proliferation-promoting factor in some biological settings. This paradoxical situation led us to study whether HIF2α has a specific effect on mTORC1 regulation. Here we show that activation of the HIF2α pathway increases mTORC1 activity by upregulating expression of the amino acid carrier SLC7A5. At the molecular level we also show that HIF2α binds to the Slc7a5 proximal promoter. Our findings identify a link between the oxygen-sensing HIF2α pathway and mTORC1 regulation, revealing the molecular basis of the tumor-promoting properties of HIF2α in von Hippel-Lindau-deficient cells. We also describe relevant physiological scenarios, including those that occur in liver and lung tissue, wherein HIF2α or low-oxygen tension drive mTORC1 activity and SLC7A5 expression. Copyright © 2012 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Oxygen sensors at the crossroad of metabolism.

              Aerobic organisms developed mechanisms to protect themselves against a shortage of oxygen (O(2)). Recent studies reveal that O(2) sensors, belonging to the novel class of 2-oxoglutarate dependent iron(ii)-dioxygenases, have more important roles in metabolism than anticipated. Here, we provide a "metabolo-centric" overview of the role of the PHD/FIH members of this family in metabolism, in particular on how they regulate O(2) supply and consumption, energy compensation and conservation, O(2) conformance and hypoxia tolerance, redox and pH homeostasis, and other vital metabolic processes with implications in health and disease. These insights may offer novel opportunities for the treatment of ischemic diseases.
                Bookmark

                Author and article information

                Contributors
                garciamonzon@hotmail.com , aguedagr.phd@gmail.com
                garciamonzon@hotmail.com , aguedagr.phd@gmail.com
                Journal
                Liver Int
                Liver Int
                10.1111/(ISSN)1478-3231
                LIV
                Liver International
                John Wiley and Sons Inc. (Hoboken )
                1478-3223
                1478-3231
                10 June 2020
                October 2020
                : 40
                : 10 ( doiID: 10.1111/liv.v40.10 )
                : 2553-2567
                Affiliations
                [ 1 ] Unidad de Investigación Hospital Universitario Santa Cristina Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa Madrid Spain
                [ 2 ] Unidad de Investigación Hospital Universitario Santa Cristina Instituto de Investigación Sanitaria del Hospital Universitario de La Princesa Universidad Autónoma de Madrid Madrid Spain
                [ 3 ] Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
                [ 4 ] Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD) Madrid Spain
                [ 5 ]Present address: Dpto. Ciencias Básicas de la Salud Universidad Rey Juan Carlos Alcorcón Spain
                Author notes
                [*] [* ] Correspondence

                Carmelo García‐Monzón and Águeda González‐Rodríguez, C/Maestro Vives 2, 28009 Madrid, Spain.

                Emails: garciamonzon@ 123456hotmail.com ; aguedagr.phd@ 123456gmail.com .

                Author information
                https://orcid.org/0000-0002-2118-8706
                https://orcid.org/0000-0001-6428-6210
                Article
                LIV14519
                10.1111/liv.14519
                7539965
                32432822
                adb7da9f-7ef1-48e7-afec-27b5d867182f
                © 2020 The Authors. Liver International published by John Wiley & Sons Ltd

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 10 July 2019
                : 11 May 2020
                : 12 May 2020
                Page count
                Figures: 6, Tables: 2, Pages: 15, Words: 7113
                Funding
                Funded by: Instituto de Salud Carlos III , open-funder-registry 10.13039/501100004587;
                Award ID: CIBERCV
                Award ID: CIBEREHD
                Award ID: CP14/00181
                Award ID: PI13/01299
                Award ID: PI16/00823
                Award ID: PI17/00535
                Award ID: PI19/00123
                Funded by: Fundación Francisco Cobos
                Funded by: Miniisterio de Economía y Competitividad
                Award ID: SAF2016‐76815
                Funded by: TV3 Marató
                Award ID: 534/C/2016
                Categories
                Original Article
                Experimental Hepatology
                Custom metadata
                2.0
                October 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.9.2 mode:remove_FC converted:07.10.2020

                Gastroenterology & Hepatology
                cd36,hif2α,hypoxia,nafld,steatosis
                Gastroenterology & Hepatology
                cd36, hif2α, hypoxia, nafld, steatosis

                Comments

                Comment on this article