4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Repression of hypoxia-inducible factor-1 contributes to increased mitochondrial reactive oxygen species production in diabetes

      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:

          Excessive production of mitochondrial reactive oxygen species (ROS) is a central mechanism for the development of diabetes complications. Recently, hypoxia has been identified to play an additional pathogenic role in diabetes. In this study, we hypothesized that ROS overproduction was secondary to the impaired responses to hypoxia due to the inhibition of hypoxia-inducible factor-1 (HIF-1) by hyperglycemia.

          Methods:

          The ROS levels were analyzed in the blood of healthy subjects and individuals with type 1 diabetes after exposure to hypoxia. The relation between HIF-1, glucose levels, ROS production and its functional consequences were analyzed in renal mIMCD-3 cells and in kidneys of mouse models of diabetes.

          Results:

          Exposure to hypoxia increased circulating ROS in subjects with diabetes, but not in subjects without diabetes. High glucose concentrations repressed HIF-1 both in hypoxic cells and in kidneys of animals with diabetes, through a HIF prolyl-hydroxylase (PHD)-dependent mechanism. The impaired HIF-1 signaling contributed to excess production of mitochondrial ROS through increased mitochondrial respiration that was mediated by Pyruvate dehydrogenase kinase 1 (PDK1). The restoration of HIF-1 function attenuated ROS overproduction despite persistent hyperglycemia, and conferred protection against apoptosis and renal injury in diabetes.

          Conclusions:

          We conclude that the repression of HIF-1 plays a central role in mitochondrial ROS overproduction in diabetes and is a potential therapeutic target for diabetic complications. These findings are timely since the first PHD inhibitor that can activate HIF-1 has been newly approved for clinical use.

          Funding:

          This work was supported by grants from the Swedish Research Council, Stockholm County Research Council, Stockholm Regional Research Foundation, Bert von Kantzows Foundation, Swedish Society of Medicine, Kung Gustaf V:s och Drottning Victorias Frimurarestifelse, Karolinska Institute’s Research Foundations, Strategic Research Programme in Diabetes, and Erling-Persson Family Foundation for S-B.C.; grants from the Swedish Research Council and Swedish Heart and Lung Foundation for T.A.S.; and ERC consolidator grant for M.M.

          Related collections

          Most cited references44

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

          Oxidative stress and diabetic complications.

          Oxidative stress plays a pivotal role in the development of diabetes complications, both microvascular and cardiovascular. The metabolic abnormalities of diabetes cause mitochondrial superoxide overproduction in endothelial cells of both large and small vessels, as well as in the myocardium. This increased superoxide production causes the activation of 5 major pathways involved in the pathogenesis of complications: polyol pathway flux, increased formation of AGEs (advanced glycation end products), increased expression of the receptor for AGEs and its activating ligands, activation of protein kinase C isoforms, and overactivity of the hexosamine pathway. It also directly inactivates 2 critical antiatherosclerotic enzymes, endothelial nitric oxide synthase and prostacyclin synthase. Through these pathways, increased intracellular reactive oxygen species (ROS) cause defective angiogenesis in response to ischemia, activate a number of proinflammatory pathways, and cause long-lasting epigenetic changes that drive persistent expression of proinflammatory genes after glycemia is normalized ("hyperglycemic memory"). Atherosclerosis and cardiomyopathy in type 2 diabetes are caused in part by pathway-selective insulin resistance, which increases mitochondrial ROS production from free fatty acids and by inactivation of antiatherosclerosis enzymes by ROS. Overexpression of superoxide dismutase in transgenic diabetic mice prevents diabetic retinopathy, nephropathy, and cardiomyopathy. The aim of this review is to highlight advances in understanding the role of metabolite-generated ROS in the development of diabetic complications.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia.

            Activation of glycolytic genes by HIF-1 is considered critical for metabolic adaptation to hypoxia through increased conversion of glucose to pyruvate and subsequently to lactate. We found that HIF-1 also actively suppresses metabolism through the tricarboxylic acid cycle (TCA) by directly trans-activating the gene encoding pyruvate dehydrogenase kinase 1 (PDK1). PDK1 inactivates the TCA cycle enzyme, pyruvate dehydrogenase (PDH), which converts pyruvate to acetyl-CoA. Forced PDK1 expression in hypoxic HIF-1alpha null cells increases ATP levels, attenuates hypoxic ROS generation, and rescues these cells from hypoxia-induced apoptosis. These studies reveal a hypoxia-induced metabolic switch that shunts glucose metabolites from the mitochondria to glycolysis to maintain ATP production and to prevent toxic ROS production.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing.

              Multicellular organisms initiate adaptive responses when oxygen (O(2)) availability decreases, but the underlying mechanism of O(2) sensing remains elusive. We find that functionality of complex III of the mitochondrial electron transport chain (ETC) is required for the hypoxic stabilization of HIF-1 alpha and HIF-2 alpha and that an increase in reactive oxygen species (ROS) links this complex to HIF-alpha stabilization. Using RNAi to suppress expression of the Rieske iron-sulfur protein of complex III, hypoxia-induced HIF-1 alpha stabilization is attenuated, and ROS production, measured using a novel ROS-sensitive FRET probe, is decreased. These results demonstrate that mitochondria function as O(2) sensors and signal hypoxic HIF-1 alpha and HIF-2 alpha stabilization by releasing ROS to the cytosol.
                Bookmark

                Author and article information

                Contributors
                Role: Reviewing Editor
                Role: Senior Editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                15 February 2022
                2022
                : 11
                : e70714
                Affiliations
                [1 ] Department of Molecular Medicine and Surgery, Karolinska Institutet Stockholm Sweden
                [2 ] Centre for Inherited Cardiovascular Diseases, IRCCS Foundation University Hospital Policlinico San Matteo Pavia Italy
                [3 ] Folkälsan Research Center, Folkälsan Institute of Genetics, University of Helsinki Helsinki Finland
                [4 ] Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB); Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, Katholieke Universiteit (KU) Leuven Leuven Belgium
                [5 ] Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, Katholieke Universiteit (KU) Leuven; Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Vlaams Instituut voor Biotechnologie (VIB) Leuven Belgium
                [6 ] Dipartimento di Biochimica, Università di Bologna Bologna Italy
                [7 ] Department of Medical Cell Biology, Uppsala University Uppsala Sweden
                [8 ] Department of Endocrinology and Diabetes, Karolinska University Hospital Stockholm Sweden
                [9 ] Center for Diabetes, Academic Specialist Centrum Stockholm Sweden
                University of Pennsylvania United States
                Icahn School of Medicine at Mount Sinai United States
                University of Pennsylvania United States
                University of Pennsylvania United States
                University of Pennsylvania United States
                Author notes
                [†]

                These authors contributed equally to this work.

                [‡]

                These authors also contributed equally to this work.

                [§]

                Department of Nephrology, GuangDong Second Traditional Chinese Medicine Hospital, GuangZhou, China.

                Author information
                https://orcid.org/0000-0002-2648-1119
                https://orcid.org/0000-0002-2492-0923
                https://orcid.org/0000-0001-7625-1103
                https://orcid.org/0000-0001-7961-1821
                https://orcid.org/0000-0001-5597-2593
                https://orcid.org/0000-0002-6914-3902
                Article
                70714
                10.7554/eLife.70714
                8846593
                35164902
                7231d7fa-2c41-4653-99a4-ec14a8f7f211
                © 2022, Zheng et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 26 May 2021
                : 27 January 2022
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100004359, Vetenskapsrådet;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100004348, Stockholms Läns Landsting;
                Award Recipient :
                Funded by: Stockholm Regional Research Foundation;
                Award Recipient :
                Funded by: Bert von Kantzows Foundation;
                Award Recipient :
                Funded by: Swedish Society of Medicine;
                Award Recipient :
                Funded by: Kung Gustaf V:s och Drottning Victorias Frimurarestifelse;
                Award Recipient :
                Funded by: Karolinska Institute's Research Foundations;
                Award Recipient :
                Funded by: Strategic Research Programme in Diabetes;
                Award Recipient :
                Funded by: Erling-Persson Family Foundation;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100004359, Vetenskapsrådet;
                Award ID: 2020-01645
                Award Recipient :
                Funded by: Swedish Heart and Lung Foundation;
                Award ID: 20210431
                Award Recipient :
                Funded by: ERC consolidator grant;
                Award ID: 773208
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Cell Biology
                Medicine
                Custom metadata
                The repression of HIF-1 plays a central role in mitochondrial reactive oxygen species (ROS) overproduction and subsequent tissue damage in diabetes, which is mediated by increased mitochondrial respiration, making HIF-1 signaling an attractive therapeutic target for diabetes complications.

                Life sciences
                diabetes,hypoxia,hypoxia-inducible factor-1,reactive oxygen species,mitochondria,diabetic complications,human,mouse

                Comments

                Comment on this article