10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Lomerizine inhibits LPS-mediated neuroinflammation and tau hyperphosphorylation by modulating NLRP3, DYRK1A, and GSK3α/β

      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

          Introduction

          Lomerizine is a calcium channel blocker that crosses the blood–brain barrier and is used clinically in the treatment of migraines. However, whether lomerizine is beneficial in modulating neuroinflammatory responses has not been tested yet.

          Methods

          To assess the potential of lomerizine for repurposing as a treatment for neuroinflammation, we investigated the effects of lomerizine on LPS-induced proinflammatory responses in BV2 microglial cells, Alzheimer’s disease (AD) excitatory neurons differentiated from induced pluripotent stem cells (iPSCs), and in LPS-treated wild type mice.

          Results

          In BV2 microglial cells, lomerizine pretreatment significantly reduced LPS-evoked proinflammatory cytokine and NLRP3 mRNA levels. Similarly, lomerizine pretreatment significantly suppressed the increases in Iba-1, GFAP, proinflammatory cytokine and NLRP3 expression induced by LPS in wild-type mice. In addition, lomerizine posttreatment significantly decreased LPS-stimulated proinflammatory cytokine and SOD2 mRNA levels in BV2 microglial cells and/or wild-type mice. In LPS-treated wild-type mice and AD excitatory neurons differentiated from iPSCs, lomerizine pretreatment ameliorated tau hyperphosphorylation. Finally, lomerizine abolished the LPS-mediated activation of GSK3α/β and upregulation of DYRK1A, which is responsible for tau hyperphosphorylation, in wild-type mice.

          Discussion

          These data suggest that lomerizine attenuates LPS-mediated neuroinflammatory responses and tau hyperphosphorylation and is a potential drug for neuroinflammation- or tauopathy-associated diseases.

          Related collections

          Most cited references62

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

          Cytokines, inflammation, and pain.

            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors

            The NLRP3 inflammasome is a multimeric protein complex that initiates an inflammatory form of cell death and triggers the release of proinflammatory cytokines IL-1β and IL-18. The NLRP3 inflammasome has been implicated in a wide range of diseases, including Alzheimer’s disease, Prion diseases, type 2 diabetes, and some infectious diseases. It has been found that a variety of stimuli including danger-associated molecular patterns (DAMPs, such as silica and uric acid crystals) and pathogen-associated molecular patterns (PAMPs) can activate NLRP3 inflammasome, but the specific regulatory mechanisms of NLRP3 inflammasome activation remain unclear. Understanding the mechanisms of NLRP3 activation will enable the development of its specific inhibitors to treat NLRP3-related diseases. In this review, we summarize current understanding of the regulatory mechanisms of NLRP3 inflammasome activation as well as inhibitors that specifically and directly target NLRP3.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes

              Neuroinflammation is associated with neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. Microglia and astrocytes are key regulators of inflammatory responses in the central nervous system. The activation of microglia and astrocytes is heterogeneous and traditionally categorized as neurotoxic (M1-phenotype microglia and A1-phenotype astrocytes) or neuroprotective (M2-phenotype microglia and A2-phenotype astrocytes). However, this dichotomized classification may not reflect the various phenotypes of microglia and astrocytes. The relationship between these activated glial cells is also very complicated, and the phenotypic distribution can change, based on the progression of neurodegenerative diseases. A better understanding of the roles of microglia and astrocytes in neurodegenerative diseases is essential for developing effective therapies. In this review, we discuss the roles of inflammatory response in neurodegenerative diseases, focusing on the contributions of microglia and astrocytes and their relationship. In addition, we discuss biomarkers to measure neuroinflammation and studies on therapeutic drugs that can modulate neuroinflammation.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                26 June 2023
                2023
                : 14
                : 1150940
                Affiliations
                [1] 1 Department of Neural Development and Disease, Korea Brain Research Institute (KBRI) , Daegu, Republic of Korea
                [2] 2 Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology , Daegu, Republic of Korea
                Author notes

                Edited by: Cheorl-Ho Kim, Sungkyunkwan University, Republic of Korea

                Reviewed by: Matteo Stravalaci, Humanitas University, Italy; Gustavo Pedraza-Alva, Universidad Nacional Autónoma de México, Mexico

                *Correspondence: Jinsoo Seo, jsseo@ 123456dgist.ac.kr ; Hyang-Sook Hoe, sookhoe72@ 123456kbri.re.kr

                †These authors contributed equally to this work

                Article
                10.3389/fimmu.2023.1150940
                10331167
                764d0eec-7277-4fcb-95ab-1b1eb4fbb551
                Copyright © 2023 Park, Hwang, Lee, Jang, Jeong, Cho, Seo and Hoe

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 25 January 2023
                : 05 June 2023
                Page count
                Figures: 9, Tables: 2, Equations: 0, References: 62, Pages: 19, Words: 8530
                Funding
                Funded by: Korea Brain Research Institute , doi 10.13039/100018749;
                Funded by: Daegu Gyeongbuk Institute of Science and Technology , doi 10.13039/501100010274;
                This work was supported by the KBRI basic research program through KBRI funded by the Ministry of Science, ICT & Future Planning (grant numbers 23-BR-02-03, 23-BR-03-05, 23-BR-05-02, and 23-BR-03-01, H-SH) and the DGIST R&D program of the Ministry of Science and ICT (22-CoE-BT-01, JS).
                Categories
                Immunology
                Original Research
                Custom metadata
                Molecular Innate Immunity

                Immunology
                lomerizine,neuroinflammation,tauopathy,alzheimer’s disease,dyrk1a,nlrp3
                Immunology
                lomerizine, neuroinflammation, tauopathy, alzheimer’s disease, dyrk1a, nlrp3

                Comments

                Comment on this article