7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Dual PPARα/γ agonist aleglitazar confers stroke protection in a model of mild focal brain ischemia in mice

      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

          Abstract

          Peroxisome proliferator-activated receptors (PPARs) control the expression of genes involved in glucose homeostasis, lipid metabolism, inflammation, and cell differentiation. Here, we analyzed the effects of aleglitazar, a dual PPARα and PPARγ agonist with balanced affinity for either subtype, on subacute stroke outcome. Healthy young adult mice were subjected to transient 30 min middle cerebral artery occlusion (MCAo)/reperfusion. Daily treatment with aleglitazar was begun on the day of MCAo and continued until sacrifice. Blood glucose measurements and lipid profile did not differ between mice receiving aleglitazar and mice receiving vehicle after MCAo. Aleglitazar reduced the size of the ischemic lesion as assessed using NeuN immunohistochemistry on day 7. Sensorimotor performance on the rotarod was impaired during the first week after MCAo, an effect that was significantly attenuated by treatment with aleglitazar. Smaller lesion volume in mice treated with aleglitazar was accompanied by a decrease in mRNA transcription of IL-1β, Vcam-1, and Icam-1, suggesting that reduced proinflammatory signaling and reduced vascular inflammation in the ischemic hemisphere contribute to the beneficial effects of aleglitazar during the first week after stroke. Further experiments in primary murine microglia confirmed that aleglitazar reduces key aspects of microglia activation including NO production, release of proinflammatory cytokines, migration, and phagocytosis. In aggregate, a brief course of PPARα/γ agonist aleglitazar initiated post-event affords stroke protection and functional recovery in a model of mild brain ischemia. Our data underscores the theme of delayed injury processes such as neuroinflammation as promising therapeutic targets in stroke.

          Key messages

          • PPARα/γ agonist aleglitazar improves stroke outcome after transient brain ischemia.

          • Aleglitazar attenuates inflammatory responses in post-ischemic brain.

          • Aleglitazar reduces microglia migration, phagocytosis, and release of cytokines.

          • Beneficial effects of aleglitazar independent of glucose regulation.

          • Aleglitazar provides extended window of opportunity for stroke treatment.

          Related collections

          Most cited references42

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

          Effects of cerebral ischemia in mice deficient in neuronal nitric oxide synthase.

          The proposal that nitric oxide (NO) or its reactant products mediate toxicity in brain remains controversial in part because of the use of nonselective agents that block NO formation in neuronal, glial, and vascular compartments. In mutant mice deficient in neuronal NO synthase (NOS) activity, infarct volumes decreased significantly 24 and 72 hours after middle cerebral artery occlusion, and the neurological deficits were less than those in normal mice. This result could not be accounted for by differences in blood flow or vascular anatomy. However, infarct size in the mutant became larger after endothelial NOS inhibition by nitro-L-arginine administration. Hence, neuronal NO production appears to exacerbate acute ischemic injury, whereas vascular NO protects after middle cerebral artery occlusion. The data emphasize the importance of developing selective inhibitors of the neuronal isoform.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Phagocytosis executes delayed neuronal death after focal brain ischemia.

            Delayed neuronal loss and brain atrophy after cerebral ischemia contribute to stroke and dementia pathology, but the mechanisms are poorly understood. Phagocytic removal of neurons is generally assumed to be beneficial and to occur only after neuronal death. However, we report herein that inhibition of phagocytosis can prevent delayed loss and death of functional neurons after transient brain ischemia. Two phagocytic proteins, Mer receptor tyrosine kinase (MerTK) and Milk fat globule EGF-like factor 8 (MFG-E8), were transiently up-regulated by macrophages/microglia after focal brain ischemia in vivo. Strikingly, deficiency in either protein completely prevented long-term functional motor deficits after cerebral ischemia and strongly reduced brain atrophy as a result of inhibiting phagocytosis of neurons. Correspondingly, in vitro glutamate-stressed neurons reversibly exposed the "eat-me" signal phosphatidylserine, leading to their phagocytosis by microglia; this neuronal loss was prevented in the absence of microglia and reduced if microglia were genetically deficient in MerTK or MFG-E8, both of which mediate phosphatidylserine-recognition. Thus, phagocytosis of viable neurons contributes to brain pathology and, surprisingly, blocking this process is strongly beneficial. Therefore, inhibition of specific phagocytic pathways may present therapeutic targets for preventing delayed neuronal loss after transient cerebral ischemia.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Quantitative determination of glutamate mediated cortical neuronal injury in cell culture by lactate dehydrogenase efflux assay.

              Measurement of lactate dehydrogenase (LDH) activity released to the extracellular bathing media has been found to be a simple yet quantitative method for assessing glutamate mediated central neuronal cell injury in cortical cell culture. Extracellular LDH is both chemically and biologically stable; the magnitude of LDH efflux in the cultures correlates in a linear fashion with the number of neurons damaged by glutamate exposure.
                Bookmark

                Author and article information

                Contributors
                karen.gertz@charite.de
                Journal
                J Mol Med (Berl)
                J. Mol. Med
                Journal of Molecular Medicine (Berlin, Germany)
                Springer Berlin Heidelberg (Berlin/Heidelberg )
                0946-2716
                1432-1440
                30 May 2019
                30 May 2019
                2019
                : 97
                : 8
                : 1127-1138
                Affiliations
                [1 ]GRID grid.484013.a, Charité – Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, , Berlin Institute of Health, Klinik und Hochschulambulanz für Neurologie und Centrum für Schlaganfallforschung Berlin (CSB), ; Charité Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany
                [2 ]ISNI 0000 0004 0374 1269, GRID grid.417570.0, Present Address: pRED, Pharma Research & Early Development, , F. Hoffmann-La Roche AG, ; Strekin AG, Basel, Switzerland
                [3 ]ISNI 0000 0000 8517 9062, GRID grid.411339.d, Klinik und Poliklinik für Kardiologie, , Universitätsklinikum Leipzig, ; 04103 Leipzig, Germany
                [4 ]ISNI 0000 0004 5937 5237, GRID grid.452396.f, DZHK (German Center for Cardiovascular Research), ; 10115 Berlin, Germany
                [5 ]ISNI 0000 0004 0438 0426, GRID grid.424247.3, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), ; 10117 Berlin, Germany
                [6 ]ISNI 0000 0004 1936 8411, GRID grid.9918.9, College of Life Sciences, , University of Leicester, and Leicestershire Partnership NHS Trust, ; Leicester, UK
                Article
                1801
                10.1007/s00109-019-01801-0
                6647083
                31147725
                90117a19-620a-43b0-8768-af49a58e600c
                © The Author(s) 2019

                Open access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

                History
                : 14 August 2018
                : 17 May 2019
                : 21 May 2019
                Funding
                Funded by: DFG
                Award ID: Exc257
                Award ID: KR 2956/4-1
                Award ID: GE 2576/2-1
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100002347, Bundesministerium für Bildung und Forschung;
                Award ID: CSB
                Award Recipient :
                Funded by: DZNE
                Award ID: n.a.
                Award Recipient :
                Funded by: DZHK
                Award ID: n.a.
                Award Recipient :
                Funded by: Corona Foundation
                Award ID: n.a.
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100007013, F. Hoffmann-La Roche;
                Award ID: n.a.
                Award Recipient :
                Categories
                Original Article
                Custom metadata
                © Springer-Verlag GmbH Germany, part of Springer Nature 2019

                Molecular medicine
                aleglitazar,microglia,pparα,pparγ,neuroinflammation,stroke
                Molecular medicine
                aleglitazar, microglia, pparα, pparγ, neuroinflammation, stroke

                Comments

                Comment on this article