Medical Biology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 6 of 6
  • Item
    Thumbnail Image
    Safety, infectivity and immunogenicity of a Chick for genetically attenuated blood-stage malaria updates vaccine
    Webster, R ; Sekuloski, S ; Odedra, A ; Woolley, S ; Jennings, H ; Amante, F ; Trenholme, KR ; Healer, J ; Cowman, AF ; Eriksson, EM ; Sathe, P ; Penington, J ; Blanch, AJ ; Dixon, MWA ; Tilley, L ; Duffy, MF ; Craig, A ; Storm, J ; Chan, J-A ; Evans, K ; Papenfuss, AT ; Schofield, L ; Griffin, P ; Barber, BE ; Andrew, D ; Boyle, MJ ; Rivera, FDL ; Engwerda, C ; McCarthy, JS (BMC, 2021-11-22)
    BACKGROUND: There is a clear need for novel approaches to malaria vaccine development. We aimed to develop a genetically attenuated blood-stage vaccine and test its safety, infectivity, and immunogenicity in healthy volunteers. Our approach was to target the gene encoding the knob-associated histidine-rich protein (KAHRP), which is responsible for the assembly of knob structures at the infected erythrocyte surface. Knobs are required for correct display of the polymorphic adhesion ligand P. falciparum erythrocyte membrane protein 1 (PfEMP1), a key virulence determinant encoded by a repertoire of var genes. METHODS: The gene encoding KAHRP was deleted from P. falciparum 3D7 and a master cell bank was produced in accordance with Good Manufacturing Practice. Eight malaria naïve males were intravenously inoculated (day 0) with 1800 (2 subjects), 1.8 × 105 (2 subjects), or 3 × 106 viable parasites (4 subjects). Parasitemia was measured using qPCR; immunogenicity was determined using standard assays. Parasites were rescued into culture for in vitro analyses (genome sequencing, cytoadhesion assays, scanning electron microscopy, var gene expression). RESULTS: None of the subjects who were administered with 1800 or 1.8 × 105 parasites developed parasitemia; 3/4 subjects administered 3× 106 parasites developed significant parasitemia, first detected on days 13, 18, and 22. One of these three subjects developed symptoms of malaria simultaneously with influenza B (day 17; 14,022 parasites/mL); one subject developed mild symptoms on day 28 (19,956 parasites/mL); and one subject remained asymptomatic up to day 35 (5046 parasites/mL). Parasitemia rapidly cleared with artemether/lumefantrine. Parasitemia induced a parasite-specific antibody and cell-mediated immune response. Parasites cultured ex vivo exhibited genotypic and phenotypic properties similar to inoculated parasites, although the var gene expression profile changed during growth in vivo. CONCLUSIONS: This study represents the first clinical investigation of a genetically attenuated blood-stage human malaria vaccine. A P. falciparum 3D7 kahrp- strain was tested in vivo and found to be immunogenic but can lead to patent parasitemia at high doses. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry (number: ACTRN12617000824369 ; date: 06 June 2017).
  • Item
    Thumbnail Image
    High-dimensional mass cytomet identifies T cell and B cell signatures predicting reduced risk of Plasmodium vivax malaria
    Ioannidis, LJ ; Pietrzak, HM ; Ly, A ; Utami, RA ; Eriksson, EM ; Studniberg, S ; Abeysekera, W ; Li-Wai-Suen, CSN ; Sheerin, D ; Healer, J ; Puspitasari, AM ; Apriyanti, D ; Coutrier, FN ; Poespoprodjo, JR ; Kenangalem, E ; Andries, B ; Prayoga, P ; Sariyanti, N ; Smyth, GK ; Trianty, L ; Cowman, AF ; Price, RN ; Noviyanti, R ; Hansen, DS (AMER SOC CLINICAL INVESTIGATION INC, 2021-07-22)
    IFN-γ-driven responses to malaria have been shown to modulate the development and function of T follicular helper (TFH) cells and memory B cells (MBCs), with conflicting evidence of their involvement in the induction of antibody responses required to achieve clinical immunity and their association with disease outcomes. Using high-dimensional single-cell mass cytometry, we identified distinct populations of TH1-polarized CD4+ T cells and MBCs expressing the TH1-defining transcription factor T-bet, associated with either increased or reduced risk of Plasmodium vivax (P. vivax) malaria, demonstrating that inflammatory responses to malaria are not universally detrimental for infection. Furthermore, we found that, whereas class-switched but not IgM+ MBCs were associated with a reduced risk of symptomatic malaria, populations of TH1 cells with a stem central memory phenotype, TH17 cells, and T regulatory cells were associated with protection from asymptomatic infection, suggesting that activation of cell-mediated immunity might also be required to control persistent P. vivax infection with low parasite burden.
  • Item
    Thumbnail Image
    4D analysis of malaria parasite invasion offers insights into erythrocyte membrane remodeling and parasitophorous vacuole formation
    Geoghegan, ND ; Evelyn, C ; Whitehead, LW ; Pasternak, M ; McDonald, P ; Triglia, T ; Marapana, DS ; Kempe, D ; Thompson, JK ; Mlodzianoski, MJ ; Healer, J ; Biro, M ; Cowman, AF ; Rogers, KL (NATURE RESEARCH, 2021-06-15)
    Host membrane remodeling is indispensable for viruses, bacteria, and parasites, to subvert the membrane barrier and obtain entry into cells. The malaria parasite Plasmodium spp. induces biophysical and molecular changes to the erythrocyte membrane through the ordered secretion of its apical organelles. To understand this process and address the debate regarding how the parasitophorous vacuole membrane (PVM) is formed, we developed an approach using lattice light-sheet microscopy, which enables the parasite interaction with the host cell membrane to be tracked and characterized during invasion. Our results show that the PVM is predominantly formed from the erythrocyte membrane, which undergoes biophysical changes as it is remodeled across all stages of invasion, from pre-invasion through to PVM sealing. This approach enables a functional interrogation of parasite-derived lipids and proteins in PVM biogenesis and echinocytosis during Plasmodium falciparum invasion and promises to yield mechanistic insights regarding how this is more generally orchestrated by other intracellular pathogens.
  • Item
    Thumbnail Image
    Dual Plasmepsin-Targeting Antimalarial Agents Disrupt Multiple Stages of the Malaria Parasite Life Cycle
    Favuzza, P ; Ruiz, MDL ; Thompson, JK ; Triglia, T ; Ngo, A ; Steel, RWJ ; Vavrek, M ; Christensen, J ; Healer, J ; Boyce, C ; Guo, Z ; Hu, M ; Khan, T ; Murgolo, N ; Zhao, L ; Penington, JS ; Reaksudsan, K ; Jarman, K ; Dietrich, MH ; Richardson, L ; Guo, K-Y ; Lopaticki, S ; Tham, W-H ; Rottmann, M ; Papenfuss, T ; Robbins, JA ; Boddey, JA ; Sleebs, BE ; Sabroux, HJ ; McCauley, JA ; Olsen, DB ; Cowman, AF (CELL PRESS, 2020-04-08)
    Artemisin combination therapy (ACT) is the main treatment option for malaria, which is caused by the intracellular parasite Plasmodium. However, increased resistance to ACT highlights the importance of finding new drugs. Recently, the aspartic proteases Plasmepsin IX and X (PMIX and PMX) were identified as promising drug targets. In this study, we describe dual inhibitors of PMIX and PMX, including WM382, that block multiple stages of the Plasmodium life cycle. We demonstrate that PMX is a master modulator of merozoite invasion and direct maturation of proteins required for invasion, parasite development, and egress. Oral administration of WM382 cured mice of P. berghei and prevented blood infection from the liver. In addition, WM382 was efficacious against P. falciparum asexual infection in humanized mice and prevented transmission to mosquitoes. Selection of resistant P. falciparum in vitro was not achievable. Together, these show that dual PMIX and PMX inhibitors are promising candidates for malaria treatment and prevention.
  • Item
    Thumbnail Image
    Development and application of a high-throughput screening assay for identification of small molecule inhibitors of the P. falciparum reticulocyte binding-like homologue 5 protein
    Sleebs, BE ; Jarman, KE ; Frolich, S ; Wong, W ; Healer, J ; Dai, W ; Lucet, IS ; Wilson, DW ; Cowman, AF (ELSEVIER SCI LTD, 2020-12)
    The P. falciparum parasite, responsible for the disease in humans known as malaria, must invade erythrocytes to provide an environment for self-replication and survival. For invasion to occur, the parasite must engage several ligands on the host erythrocyte surface to enable adhesion, tight junction formation and entry. Critical interactions include binding of erythrocyte binding-like ligands and reticulocyte binding-like homologues (Rhs) to the surface of the host erythrocyte. The reticulocyte binding-like homologue 5 (Rh5) is the only member of this family that is essential for invasion and it binds to the basigin host receptor. The essential nature of Rh5 makes it an important vaccine target, however to date, Rh5 has not been targeted by small molecule intervention. Here, we describe the development of a high-throughput screening assay to identify small molecules which interfere with the Rh5-basigin interaction. To validate the utility of this assay we screened a known drug library and the Medicines for Malaria Box and demonstrated the reproducibility and robustness of the assay for high-throughput screening purposes. The screen of the known drug library identified the known leukotriene antagonist, pranlukast. We used pranlukast as a model inhibitor in a post screening evaluation cascade. We procured and synthesised analogues of pranlukast to assist in the hit confirmation process and show which structural moieties of pranlukast attenuate the Rh5 - basigin interaction. Evaluation of pranlukast analogues against P. falciparum in a viability assay and a schizont rupture assay show the parasite activity was not consistent with the biochemical inhibition of Rh5, questioning the developability of pranlukast as an antimalarial. The high-throughput assay developed from this work has the capacity to screen large collections of small molecules to discover inhibitors of P. falciparum Rh5 for future development of invasion inhibitory antimalarials.
  • Item
    Thumbnail Image
    Pan-active imidazolopiperazine antimalarials target the Plasmodium falciparum intracellular secretory pathway
    LaMonte, GM ; Rocamora, F ; Marapana, DS ; Gnadig, NF ; Ottilie, S ; Luth, MR ; Worgall, TS ; Goldgof, GM ; Mohunlal, R ; Kumar, TRS ; Thompson, JK ; Vigil, E ; Yang, J ; Hutson, D ; Johnson, T ; Huang, J ; Williams, RM ; Zou, BY ; Cheung, AL ; Kumar, P ; Egan, TJ ; Lee, MCS ; Siegel, D ; Cowman, AF ; Fidock, DA ; Winzeler, EA (NATURE PUBLISHING GROUP, 2020-04-14)
    A promising new compound class for treating human malaria is the imidazolopiperazines (IZP) class. IZP compounds KAF156 (Ganaplacide) and GNF179 are effective against Plasmodium symptomatic asexual blood-stage infections, and are able to prevent transmission and block infection in animal models. But despite the identification of resistance mechanisms in P. falciparum, the mode of action of IZPs remains unknown. To investigate, we here combine in vitro evolution and genome analysis in Saccharomyces cerevisiae with molecular, metabolomic, and chemogenomic methods in P. falciparum. Our findings reveal that IZP-resistant S. cerevisiae clones carry mutations in genes involved in Endoplasmic Reticulum (ER)-based lipid homeostasis and autophagy. In Plasmodium, IZPs inhibit protein trafficking, block the establishment of new permeation pathways, and cause ER expansion. Our data highlight a mechanism for blocking parasite development that is distinct from those of standard compounds used to treat malaria, and demonstrate the potential of IZPs for studying ER-dependent protein processing.