9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      BioID Reveals Novel Proteins of the Plasmodium Parasitophorous Vacuole Membrane

      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

          Intracellular pathogens are often surrounded by a host-cell derived membrane. This membrane is modified by the pathogens to their own needs and is crucial for their intracellular lifestyle. In Plasmodium parasites, this membrane is referred to as the PVM and only a limited number of its proteins are known so far. Here, we applied in rodent P. berghei parasites a method called BioID, which is based on biotinylation of proximal and interacting proteins by the promiscuous biotin ligase BirA*, and demonstrated its usefulness in identification of novel PVM proteins.

          ABSTRACT

          During their development within the vertebrate host, Plasmodium parasites infect hepatocytes and red blood cells. Within these cells, parasites are surrounded by a parasitophorous vacuole membrane (PVM). The PVM plays an essential role for the interaction of parasites with their host cells; however, only a limited number of proteins of this membrane have been identified so far. This is partially because systematic proteomic analysis of the protein content of the PVM has been difficult in the past, due to difficulties encountered in attempts to separate the PVM from other membranes such as the parasite plasma membrane. In this study, we adapted the BioID technique to in vitro-cultivated Plasmodium berghei blood stage parasites and utilized the promiscuous biotin ligase BirA* fused to PVM-resident exported protein 1 to biotinylate proteins of the PVM. These we further processed by affinity purification, liquid chromatography-tandem mass spectrometry (LC-MS/MS), and label-free quantitation, leading to a list of 61 known and candidate PVM proteins. Seven proteins were analyzed further during blood and liver stage development. This resulted in the identification of three novel PVM proteins, which were the serine/threonine protein phosphatase UIS2 (PlasmoDB accession no. PBANKA_1328000) and two conserved Plasmodium proteins with unknown functions ( PBANKA_0519300 and PBANKA_0509000). In conclusion, our report expands the number of known PVM proteins and experimentally validates BioID as a powerful method to screen for novel constituents of specific cellular compartments in P. berghei.

          IMPORTANCE Intracellular pathogens are often surrounded by a host-cell derived membrane. This membrane is modified by the pathogens to their own needs and is crucial for their intracellular lifestyle. In Plasmodium parasites, this membrane is referred to as the PVM and only a limited number of its proteins are known so far. Here, we applied in rodent P. berghei parasites a method called BioID, which is based on biotinylation of proximal and interacting proteins by the promiscuous biotin ligase BirA*, and demonstrated its usefulness in identification of novel PVM proteins.

          Related collections

          Most cited references28

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

          A novel protein export machine in malaria parasites

          Several hundred malaria parasite proteins are exported beyond an encasing vacuole and into the cytosol of the host erythrocyte, a process that is key to the virulence and viability of the causative Plasmodium species. The trafficking machinery responsible for this export is unknown. Here, we identify a Plasmodium Translocon of EXported proteins (PTEX), which is located in the vacuole membrane. The PTEX complex is ATP-powered and comprises HSP101, which is a ClpA/B-like AAA+ ATPase of a type commonly associated with protein translocons, a novel protein termed PTEX150 and a known parasite protein EXP2. EXP2 is the potential channel as it is the membrane-associated component of the core PTEX complex. Two other proteins, a novel protein PTEX88 and a thioredoxin known as TRX2, were also identified as PTEX components. As a common portal for numerous crucial processes, this novel translocon offers an exciting new avenue for therapeutic intervention.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Genetically modified Plasmodium parasites as a protective experimental malaria vaccine.

            Malaria is a mosquito-borne disease that is transmitted by inoculation of the Plasmodium parasite sporozoite stage. Sporozoites invade hepatocytes, transform into liver stages, and subsequent liver-stage development ultimately results in release of pathogenic merozoites. Liver stages of the parasite are a prime target for malaria vaccines because they can be completely eliminated by sterilizing immune responses, thereby preventing malarial infection. Using expression profiling, we previously identified genes that are only expressed in the pre-erythrocytic stages of the parasite. Here, we show by reverse genetics that one identified gene, UIS3 (upregulated in infective sporozoites gene 3), is essential for early liver-stage development. uis3-deficient sporozoites infect hepatocytes but are unable to establish blood-stage infections in vivo, and thus do not lead to disease. Immunization with uis3-deficient sporozoites confers complete protection against infectious sporozoite challenge in a rodent malaria model. This protection is sustained and stage specific. Our findings demonstrate that a safe and effective, genetically attenuated whole-organism malaria vaccine is possible.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Plasmodium liver stage developmental arrest by depletion of a protein at the parasite-host interface.

              Plasmodium parasites of mammals, including the species that cause malaria in humans, infect the liver first and develop there into clinically silent liver stages. Liver stages grow and ultimately produce thousands of first-generation merozoites, which initiate the erythrocytic cycles causing malaria pathology. Here, we present a Plasmodium protein with a critical function for complete liver stage development. UIS4 (up-regulated in infective sporozoites gene 4) is expressed exclusively in infective sporozoites and developing liver stages, where it localizes to the parasitophorous vacuole membrane. Targeted gene disruption of UIS4 in the rodent model malaria parasite Plasmodium berghei generated knockout parasites that progress through the malaria life cycle until after hepatocyte invasion but are severely impaired in further liver stage development. Immunization with UIS4 knockout sporozoites completely protects mice against subsequent infectious WT sporozoite challenge. Genetically attenuated liver stages may thus induce immune responses, which inhibit subsequent infection of the liver with WT parasites.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                mSphere
                mSphere
                msph
                msph
                mSphere
                mSphere
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2379-5042
                24 January 2018
                Jan-Feb 2018
                : 3
                : 1
                : e00522-17
                Affiliations
                [a ]Institute of Cell Biology, University of Bern, Bern, Switzerland
                [b ]Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
                [c ]Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, Australia
                [d ]Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
                [e ]Department of Health Sciences and Technology, BMPP, ETH Zurich, Zurich, Switzerland
                University at Buffalo
                Author notes
                Address correspondence to Paul-Christian Burda, burda@ 123456bnitm.de .
                [*]

                Present address: Paul-Christian Burda, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.

                Citation Schnider CB, Bausch-Fluck D, Brühlmann F, Heussler VT, Burda P-C. 2018. BioID reveals novel proteins of the Plasmodium parasitophorous vacuole membrane. mSphere 3:e00522-17. https://doi.org/10.1128/mSphere.00522-17.

                Article
                mSphere00522-17
                10.1128/mSphere.00522-17
                5784244
                7a65473b-c29d-4ee1-9e3f-593a76d05b93
                Copyright © 2018 Schnider et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 2 November 2017
                : 22 December 2017
                Page count
                supplementary-material: 9, Figures: 6, Tables: 0, Equations: 0, References: 38, Pages: 13, Words: 8441
                Funding
                Funded by: EVIMalaR EU Consortium;
                Award Recipient :
                Funded by: Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (SNF), https://doi.org/10.13039/501100001711;
                Award ID: 310030_159519
                Award Recipient :
                Categories
                Research Article
                Host-Microbe Biology
                Custom metadata
                January/February 2018

                bioid,blood stage,liver stage,pvm,plasmodium,membranes
                bioid, blood stage, liver stage, pvm, plasmodium, membranes

                Comments

                Comment on this article