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

      Modelling heme-mediated brain injury associated with cerebral malaria in human brain cortical organoids

      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

          Human cerebral malaria (HCM), a severe encephalopathy associated with Plasmodium falciparum infection, has a 20–30% mortality rate and predominantly affects African children. The mechanisms mediating HCM-associated brain injury are difficult to study in human subjects, highlighting the urgent need for non-invasive ex vivo human models. HCM elevates the systemic levels of free heme, which damages the blood-brain barrier and neurons in distinct regions of the brain. We determined the effects of heme on induced pluripotent stem cells (iPSCs) and a three-dimensional cortical organoid system and assessed apoptosis and differentiation. We evaluated biomarkers associated with heme-induced brain injury, including a pro-inflammatory chemokine, CXCL-10, and its receptor, CXCR3, brain-derived neurotrophic factor (BDNF) and a receptor tyrosine-protein kinase, ERBB4, in the organoids. We then tested the neuroprotective effect of neuregulin-1 (NRG-1) against heme treatment in organoids. Neural stem and mature cells differentially expressed CXCL-10, CXCR3, BDNF and ERBB4 in the developing organoids and in response to heme-induced neuronal injury. The organoids underwent apoptosis and structural changes that were attenuated by NRG-1. Thus, cortical organoids can be used to model heme-induced cortical brain injury associated with HCM pathogenesis as well as for testing agents that reduce brain injury and neurological sequelae.

          Related collections

          Most cited references65

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

          Guided self-organization and cortical plate formation in human brain organoids

          Engineering human brain organoids with floating scaffolds enhances the maturity and reproducibility of cortical tissue structure.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen.

            In response to the current global health emergency posed by the Zika virus (ZIKV) outbreak and its link to microcephaly and other neurological conditions, we performed a drug repurposing screen of ∼6,000 compounds that included approved drugs, clinical trial drug candidates and pharmacologically active compounds; we identified compounds that either inhibit ZIKV infection or suppress infection-induced caspase-3 activity in different neural cells. A pan-caspase inhibitor, emricasan, inhibited ZIKV-induced increases in caspase-3 activity and protected human cortical neural progenitors in both monolayer and three-dimensional organoid cultures. Ten structurally unrelated inhibitors of cyclin-dependent kinases inhibited ZIKV replication. Niclosamide, a category B anthelmintic drug approved by the US Food and Drug Administration, also inhibited ZIKV replication. Finally, combination treatments using one compound from each category (neuroprotective and antiviral) further increased protection of human neural progenitors and astrocytes from ZIKV-induced cell death. Our results demonstrate the efficacy of this screening strategy and identify lead compounds for anti-ZIKV drug development.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Hemolysis-induced lethality involves inflammasome activation by heme.

              The increase of extracellular heme is a hallmark of hemolysis or extensive cell damage. Heme has prooxidant, cytotoxic, and inflammatory effects, playing a central role in the pathogenesis of malaria, sepsis, and sickle cell disease. However, the mechanisms by which heme is sensed by innate immune cells contributing to these diseases are not fully characterized. We found that heme, but not porphyrins without iron, activated LPS-primed macrophages promoting the processing of IL-1β dependent on nucleotide-binding domain and leucine rich repeat containing family, pyrin domain containing 3 (NLRP3). The activation of NLRP3 by heme required spleen tyrosine kinase, NADPH oxidase-2, mitochondrial reactive oxygen species, and K(+) efflux, whereas it was independent of heme internalization, lysosomal damage, ATP release, the purinergic receptor P2X7, and cell death. Importantly, our results indicated the participation of macrophages, NLRP3 inflammasome components, and IL-1R in the lethality caused by sterile hemolysis. Thus, understanding the molecular pathways affected by heme in innate immune cells might prove useful to identify new therapeutic targets for diseases that have heme release.
                Bookmark

                Author and article information

                Contributors
                aharbuzariu@msm.edu
                jstiles@msm.edu
                Journal
                Sci Rep
                Sci Rep
                Scientific Reports
                Nature Publishing Group UK (London )
                2045-2322
                16 December 2019
                16 December 2019
                2019
                : 9
                : 19162
                Affiliations
                [1 ]ISNI 0000 0001 2228 775X, GRID grid.9001.8, Department of Microbiology, Biochemistry and Immunology, , Morehouse School of Medicine, ; 720 Westview Dr, Atlanta, GA 30310 USA
                [2 ]ISNI 0000 0001 2097 4943, GRID grid.213917.f, Parker H. Petit Institute for Bioengineering and Bioscience, , Georgia Institute of Technology, ; 315 Ferst Drive, Atlanta, GA 30332 USA
                Article
                55631
                10.1038/s41598-019-55631-8
                6914785
                31844087
                b00e9670-a5dd-4f6a-aba3-7e7b91bd1655
                © The Author(s) 2019

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as 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. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 31 May 2019
                : 26 November 2019
                Funding
                Funded by: FundRef https://doi.org/10.13039/100000065, U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS);
                Award ID: R01NS091616
                Award ID: R01NS091616
                Award ID: R01NS091616
                Award ID: R01NS091616
                Award ID: R01NS091616
                Award Recipient :
                Funded by: Georgia Clinical and Translational Science Alliance (GCTSA; NIH/NCATS) Pilot Translational and Clinical Studies Program PTCS; NIH/RCMI RR033062 (G12)
                Funded by: U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
                Funded by: Georgia Clinical and Translational Science Alliance (GCTSA; NIH/NCATS) Pilot Translational and Clinical Studies Program PTCS; NIH/RCMI RR033062 (G12);
                Funded by: U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
                Funded by: U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
                Funded by: U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
                Categories
                Article
                Custom metadata
                © The Author(s) 2019

                Uncategorized
                induced pluripotent stem cells,malaria
                Uncategorized
                induced pluripotent stem cells, malaria

                Comments

                Comment on this article