School of BioSciences - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 4 of 4
  • Item
    Thumbnail Image
    Development of Plasmodium-specific liver-resident memory CD8+ T cells after heat-killed sporozoite immunization in mice
    Ghilas, S ; Enders, MH ; May, R ; Holz, LE ; Fernandez-Ruiz, D ; Cozijnsen, A ; Mollard, V ; Cockburn, IA ; McFadden, G ; Heath, WR ; Beattie, L (WILEY, 2021-05)
    Malaria remains a major cause of mortality in the world and an efficient vaccine is the best chance of reducing the disease burden. Vaccination strategies for the liver stage of disease that utilise injection of live radiation-attenuated sporozoites (RAS) confer sterile immunity, which is mediated by CD8+ memory T cells, with liver-resident memory T cells (TRM ) being particularly important. We have previously described a TCR transgenic mouse, termed PbT-I, where all CD8+ T cells recognize a specific peptide from Plasmodium. PbT-I form liver TRM cells upon RAS injection and are capable of protecting mice against challenge infection. Here, we utilize this transgenic system to examine whether nonliving sporozoites, killed by heat treatment (HKS), could trigger the development of Plasmodium-specific liver TRM cells. We found that HKS vaccination induced the formation of memory CD8+ T cells in the spleen and liver, and importantly, liver TRM cells were fewer in number than that induced by RAS. Crucially, we showed the number of TRM cells was significantly higher when HKS were combined with the glycolipid α-galactosylceramide as an adjuvant. In the future, this work could lead to development of an antimalaria vaccination strategy that does not require live sporozoites, providing greater utility.
  • Item
    Thumbnail Image
    mRNA vaccine against malaria tailored for liver-resident memory T cells
    Ganley, M ; Holz, LE ; Minnell, JJ ; de Menezes, MN ; Burn, OK ; Poa, KCY ; Draper, SL ; English, K ; Chan, STS ; Anderson, RJ ; Compton, BJ ; Marshall, AJ ; Cozijnsen, A ; Chua, YC ; Ge, Z ; Farrand, KJ ; Mamum, JC ; Xu, C ; Cockburn, IA ; Yui, K ; Bertolino, P ; Gras, S ; Le Nours, J ; Rossjohn, J ; Fernandez-Ruiz, D ; McFadden, GI ; Ackerley, DF ; Painter, GF ; Hermans, IF ; Heath, WR (NATURE PORTFOLIO, 2023-09)
    Malaria is caused by Plasmodium species transmitted by Anopheles mosquitoes. Following a mosquito bite, Plasmodium sporozoites migrate from skin to liver, where extensive replication occurs, emerging later as merozoites that can infect red blood cells and cause symptoms of disease. As liver tissue-resident memory T cells (Trm cells) have recently been shown to control liver-stage infections, we embarked on a messenger RNA (mRNA)-based vaccine strategy to induce liver Trm cells to prevent malaria. Although a standard mRNA vaccine was unable to generate liver Trm or protect against challenge with Plasmodium berghei sporozoites in mice, addition of an agonist that recruits T cell help from type I natural killer T cells under mRNA-vaccination conditions resulted in significant generation of liver Trm cells and effective protection. Moreover, whereas previous exposure of mice to blood-stage infection impaired traditional vaccines based on attenuated sporozoites, mRNA vaccination was unaffected, underlining the potential for such a rational mRNA-based strategy in malaria-endemic regions.
  • Item
    Thumbnail Image
    6"-Modifed α-GalCer-peptide conjugate vaccine candidates protect against liver-stage malaria
    Meijlink, MA ; Chua, YC ; Chan, STS ; Anderson, RJ ; Rosenberg, MW ; Cozijnsen, A ; Mollard, V ; McFadden, G ; Draper, SL ; Holz, LE ; Hermans, IF ; Heath, WR ; Painter, GF ; Compton, BJ (ROYAL SOC CHEMISTRY, 2022-05-11)
    Self-adjuvanting vaccines consisting of peptide epitopes conjugated to immune adjuvants are a powerful way of generating antigen-specific immune responses. We previously showed that a Plasmodium-derived peptide conjugated to a rearranged form of α-galactosylceramide (α-GalCer) could stimulate liver-resident memory T (TRM) cells that were effective killers of liver-stage Plasmodium berghei ANKA (Pba)-infected cells. To investigate if similar or even superior TRM responses can be induced by modifying the α-GalCer adjuvant, we created new conjugate vaccine cadidates by attaching an immunogenic Plasmodium-derived peptide antigen to 6″-substituted α-GalCer analogues. Vaccine synthesis involved developing an efficient route to α-galactosylphytosphingosine (α-GalPhs), from which the prototypical iNKT cell agonist, α-GalCer, and its 6″-deoxy-6″-thio and -amino analogues were derived. Attaching a cathepsin B-cleavable linker to the 6″-modified α-GalCer created pro-adjuvants bearing a pendant ketone group available for peptide conjugation. Optimized reaction conditions were developed that allow for the efficient conjugation of peptide antigens to the pro-adjuvants via oxime ligation to create new glycolipid-peptide (GLP) conjugate vaccines. A single dose of the vaccine candidates induced acute NKT and Plasmodium-specific CD8+ T cell responses that generated potent hepatic TRM responses in mice. Our findings demonstrate that attaching antigenic peptides to 6″-modifed α-GalCer generates powerful self-adjuvanting conjugate vaccine candidates that could potentially control hepatotropic infections such as liver-stage malaria.
  • Item
    Thumbnail Image
    CD8+ T Cell Activation Leads to Constitutive Formation of Liver Tissue-Resident Memory T Cells that Seed a Large and Flexible Niche in the Liver
    Holz, LE ; Prier, JE ; Freestone, D ; Steiner, TM ; English, K ; Johnson, DN ; Mollard, V ; Cozijnsen, A ; Davey, GM ; Godfrey, D ; Yui, K ; Mackay, LK ; Lahoud, MH ; Caminschi, I ; McFadden, G ; Bertolino, P ; Fernandez-Ruiz, D ; Heath, WR (CELL PRESS, 2018-10-02)
    Liver tissue-resident memory T (Trm) cells migrate throughout the sinusoids and are capable of protecting against malaria sporozoite challenge. To gain an understanding of liver Trm cell development, we examined various conditions for their formation. Although liver Trm cells were found in naive mice, their presence was dictated by antigen specificity and required IL-15. Liver Trm cells also formed after adoptive transfer of in vitro-activated but not naive CD8+ T cells, indicating that activation was essential but that antigen presentation within the liver was not obligatory. These Trm cells patrolled the liver sinusoids with a half-life of 36 days and occupied a large niche that could be added to sequentially without effect on subsequent Trm cell cohorts. Together, our findings indicate that liver Trm cells form as a normal consequence of CD8+ T cell activation during essentially any infection but that inflammatory and antigenic signals preferentially tailor their development.