24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      NRF2 deficiency replicates transcriptomic changes in Alzheimer's patients and worsens APP and TAU pathology

      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

          Failure to translate successful neuroprotective preclinical data to a clinical setting in Alzheimer's disease (AD) indicates that amyloidopathy and tauopathy alone provide an incomplete view of disease. We have tested here the relevance of additional homeostatic deviations that result from loss of activity of transcription factor NRF2, a crucial regulator of multiple stress responses whose activity declines with ageing. A transcriptomic analysis demonstrated that NRF2-KO mouse brains reproduce 7 and 10 of the most dysregulated pathways of human ageing and AD brains, respectively. Then, we generated a mouse that combines amyloidopathy and tauopathy with either wild type (AT-NRF2-WT) or NRF2-deficiency (AT-NRF2-KO). AT-NRF2-KO brains presented increased markers of oxidative stress and neuroinflammation as well as higher levels of insoluble phosphorylated-TAU and Aβ*56 compared to AT-NRF2-WT mice. Young adult AT-NRF2-KO mice exhibited deficits in spatial learning and memory and reduced long term potentiation in the perforant pathway. This study demonstrates the relevance of normal homeostatic responses that decline with ageing, such as NRF2 activity, in the protection against proteotoxic, inflammatory and oxidative stress and provide a new strategy to fight AD.

          Graphical abstract

          Highlights

          • NRF2-lack mimics many transcriptomic alterations of elderly and AD patients.

          • APP/TAU-induced oxidative and inflammatory stress is exacerbated by NRF2-deficiency.

          • APP/TAU-induced proteinopathy is exacerbated by NRF2-deficiency.

          • APP/TAU-induced deficits in cognition are aggravated by NRF2-deficiency.

          Related collections

          Most cited references28

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

          DAVID: Database for Annotation, Visualization, and Integrated Discovery.

          Functional annotation of differentially expressed genes is a necessary and critical step in the analysis of microarray data. The distributed nature of biological knowledge frequently requires researchers to navigate through numerous web-accessible databases gathering information one gene at a time. A more judicious approach is to provide query-based access to an integrated database that disseminates biologically rich information across large datasets and displays graphic summaries of functional information. Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) addresses this need via four web-based analysis modules: 1) Annotation Tool - rapidly appends descriptive data from several public databases to lists of genes; 2) GoCharts - assigns genes to Gene Ontology functional categories based on user selected classifications and term specificity level; 3) KeggCharts - assigns genes to KEGG metabolic processes and enables users to view genes in the context of biochemical pathway maps; and 4) DomainCharts - groups genes according to PFAM conserved protein domains. Analysis results and graphical displays remain dynamically linked to primary data and external data repositories, thereby furnishing in-depth as well as broad-based data coverage. The functionality provided by DAVID accelerates the analysis of genome-scale datasets by facilitating the transition from data collection to biological meaning.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Gene regulation and DNA damage in the ageing human brain.

            The ageing of the human brain is a cause of cognitive decline in the elderly and the major risk factor for Alzheimer's disease. The time in life when brain ageing begins is undefined. Here we show that transcriptional profiling of the human frontal cortex from individuals ranging from 26 to 106 years of age defines a set of genes with reduced expression after age 40. These genes play central roles in synaptic plasticity, vesicular transport and mitochondrial function. This is followed by induction of stress response, antioxidant and DNA repair genes. DNA damage is markedly increased in the promoters of genes with reduced expression in the aged cortex. Moreover, these gene promoters are selectively damaged by oxidative stress in cultured human neurons, and show reduced base-excision DNA repair. Thus, DNA damage may reduce the expression of selectively vulnerable genes involved in learning, memory and neuronal survival, initiating a programme of brain ageing that starts early in adult life.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Decline in transcriptional activity of Nrf2 causes age-related loss of glutathione synthesis, which is reversible with lipoic acid.

              Glutathione (GSH) significantly declines in the aging rat liver. Because GSH levels are partly a reflection of its synthetic capacity, we measured the levels and activity of gamma-glutamylcysteine ligase (GCL), the rate-controlling enzyme in GSH synthesis. With age, both the catalytic (GCLC) and modulatory (GCLM) subunits of GCL decreased by 47% and 52%, respectively (P < 0.005). Concomitant with lower subunit levels, GCL activity also declined by 53% (P < 0.05). Because nuclear factor erythroid2-related factor 2 (Nrf2) governs basal and inducible GCLC and GCLM expression by means of the antioxidant response element (ARE), we hypothesized that aging results in dysregulation of Nrf2-mediated GCL expression. We observed an approximately 50% age-related loss in total (P < 0.001) and nuclear (P < 0.0001) Nrf2 levels, which suggests attenuation in Nrf2-dependent gene transcription. By using gel-shift and supershift assays, a marked reduction in Nrf2/ARE binding in old vs. young rats was noted. To determine whether the constitutive loss of Nrf2 transcriptional activity also affects the inducible nature of Nrf2 nuclear translocation, old rats were treated with (R)-alpha-lipoic acid (LA; 40 mg/kg i.p. up to 48 h), a disulfide compound shown to induce Nrf2 activation in vitro and improve GSH levels in vivo. LA administration increased nuclear Nrf2 levels in old rats after 12 h. LA also induced Nrf2 binding to the ARE, and, consequently, higher GCLC levels and GCL activity were observed 24 h after LA injection. Thus, the age-related loss in GSH synthesis may be caused by dysregulation of ARE-mediated gene expression, but chemoprotective agents, like LA, can attenuate this loss.
                Bookmark

                Author and article information

                Contributors
                Journal
                Redox Biol
                Redox Biol
                Redox Biology
                Elsevier
                2213-2317
                05 July 2017
                October 2017
                05 July 2017
                : 13
                : 444-451
                Affiliations
                [a ]Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII. Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC. Instituto de Investigación Sanitaria La Paz (IdiPaz), and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
                [b ]Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain. Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC. Instituto de Investigación Sanitaria La Paz (IdiPaz); and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
                [c ]Department of Anatomy, Histology and Neuroscience, Autonomous University of Madrid, Madrid, Spain
                [d ]King's College London, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Camberwell, London, UK
                [e ]Experimental Genetics Group-LEGTEGG, Department of Human Genetics, KU Leuven, Leuven, Belgium
                [f ]Department of Psychiatry, Warneford Hospital, University of Oxford, OX3 7JX UK
                [g ]Cellular and Molecular Medicine Department, Radiobiology Laboratory, "Victor Babes" National Institute of Pathology, Bucharest, Romania
                [h ]Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
                Author notes
                [* ]Corresponding author. airojo@ 123456iib.uam.es
                [** ]Corresponding author at: Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII. Instituto de Investigaciones Biomédicas “Alberto Sols” UAM-CSIC. Instituto de Investigación Sanitaria La Paz (IdiPaz), and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain. antonio.cuadrado@ 123456uam.es
                Article
                S2213-2317(17)30399-3
                10.1016/j.redox.2017.07.006
                5508523
                28704727
                aa5a68a7-0b4d-4661-9191-8becabf5774f
                © 2017 The Authors

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

                History
                : 31 May 2017
                : 28 June 2017
                : 3 July 2017
                Categories
                Research Paper

                nrf2,alzheimer's disease,transcriptomics,proteotoxicity,neuroinflammation,oxidative stress

                Comments

                Comment on this article