Microbiology & Immunology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 66
  • Item
    Thumbnail Image
    Systemic inflammatory response syndrome triggered by blood-borne pathogens induces prolonged dendritic cell paralysis and immunosuppression
    Ashayeripanah, M ; Vega-Ramos, J ; Fernandez-Ruiz, D ; Valikhani, S ; Lun, ATL ; White, JT ; Young, LJ ; Yaftiyan, A ; Zhan, Y ; Wakim, L ; Caminschi, I ; Lahoud, MH ; Lew, AM ; Shortman, K ; Smyth, GK ; Heath, WR ; Mintern, JD ; Roquilly, A ; Villadangos, JA (CELL PRESS, 2024-02-27)
    Blood-borne pathogens can cause systemic inflammatory response syndrome (SIRS) followed by protracted, potentially lethal immunosuppression. The mechanisms responsible for impaired immunity post-SIRS remain unclear. We show that SIRS triggered by pathogen mimics or malaria infection leads to functional paralysis of conventional dendritic cells (cDCs). Paralysis affects several generations of cDCs and impairs immunity for 3-4 weeks. Paralyzed cDCs display distinct transcriptomic and phenotypic signatures and show impaired capacity to capture and present antigens in vivo. They also display altered cytokine production patterns upon stimulation. The paralysis program is not initiated in the bone marrow but during final cDC differentiation in peripheral tissues under the influence of local secondary signals that persist after resolution of SIRS. Vaccination with monoclonal antibodies that target cDC receptors or blockade of transforming growth factor β partially overcomes paralysis and immunosuppression. This work provides insights into the mechanisms of paralysis and describes strategies to restore immunocompetence post-SIRS.
  • Item
    Thumbnail Image
    Display of Native Antigen on cDC1 That Have Spatial Access to Both T and B Cells Underlies Efficient Humoral Vaccination.
    Kato, Y ; Steiner, TM ; Park, H-Y ; Hitchcock, RO ; Zaid, A ; Hor, JL ; Devi, S ; Davey, GM ; Vremec, D ; Tullett, KM ; Tan, PS ; Ahmet, F ; Mueller, SN ; Alonso, S ; Tarlinton, DM ; Ploegh, HL ; Kaisho, T ; Beattie, L ; Manton, JH ; Fernandez-Ruiz, D ; Shortman, K ; Lahoud, MH ; Heath, WR ; Caminschi, I (American Association of Immunologists, 2020-10-01)
    Follicular dendritic cells and macrophages have been strongly implicated in presentation of native Ag to B cells. This property has also occasionally been attributed to conventional dendritic cells (cDC) but is generally masked by their essential role in T cell priming. cDC can be divided into two main subsets, cDC1 and cDC2, with recent evidence suggesting that cDC2 are primarily responsible for initiating B cell and T follicular helper responses. This conclusion is, however, at odds with evidence that targeting Ag to Clec9A (DNGR1), expressed by cDC1, induces strong humoral responses. In this study, we reveal that murine cDC1 interact extensively with B cells at the border of B cell follicles and, when Ag is targeted to Clec9A, can display native Ag for B cell activation. This leads to efficient induction of humoral immunity. Our findings indicate that surface display of native Ag on cDC with access to both T and B cells is key to efficient humoral vaccination.
  • 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
    The liver contains distinct interconnected networks of CX3CR1+ macrophages, XCR1+ type 1 and CD301a+ type 2 conventional dendritic cells embedded within portal tracts
    English, K ; Tan, SY ; Kwan, R ; Holz, LE ; Sierro, F ; McGuffog, C ; Kaisho, T ; Heath, WR ; MacDonald, KPA ; McCaughan, GW ; Bowen, DG ; Bertolino, P (WILEY, 2022-07)
    Portal tracts are key intrahepatic structures where leukocytes accumulate during immune responses. They contain the blood inflow, which includes portal blood from the gut, and lymphatic and biliary outflow of the liver, and as such represent a key interface for potential pathogen entry to the liver. Myeloid cells residing in the interstitium of the portal tract might play an important role in the surveillance or prevention of pathogen dissemination; however, the exact composition and localization of this population has not been explored fully. Our in-depth characterization of portal tract myeloid cells revealed that in addition to T lymphocytes, portal tracts contain a heterogeneous population of MHCIIhigh myeloid cells with potential antigen presenting cell (APC) function. These include a previously unreported subset of CSF1R-dependent CX3CR1+ macrophages that phenotypically and morphologically resemble liver capsular macrophages, as well as the two main dendritic cell subsets (cDC1 and cDC2). These cells are not randomly distributed, but each subset forms interconnected networks intertwined with specific components of the portal tract. The CX3CR1+ cells were preferentially detected along the outer border of the portal tracts, and also in the portal interstitium adjacent to the portal vein, bile duct, lymphatic vessels and hepatic artery. cDC1s abounded along the lymphatic vessels, while cDC2s mostly surrounded the biliary tree. The specific distributions of these discrete subsets predict that they may serve distinct functions in this compartment. Overall, our findings suggest that portal tracts and their embedded cellular networks of myeloid cells form a distinctive lymphoid compartment in the liver that has the potential to orchestrate immune responses in this organ.
  • Item
    Thumbnail Image
    Marginal zone B cells acquire dendritic cell functions by trogocytosis
    Schriek, P ; Ching, AC ; Moily, NS ; Moffat, J ; Beattie, L ; Steiner, TM ; Hosking, LM ; Thurman, JM ; Holers, VM ; Ishido, S ; Lahoud, MH ; Caminschi, I ; Heath, WR ; Mintern, JD ; Villadangos, JA (AMER ASSOC ADVANCEMENT SCIENCE, 2022-02-11)
    Marginal zone (MZ) B cells produce broad-spectrum antibodies that protect against infection early in life. In some instances, antibody production requires MZ B cells to display pathogen antigens bound to major histocompatibility complex class II (MHC II) molecules to T cells. We describe the trogocytic acquisition of these molecules from conventional dendritic cells (cDCs). Complement component 3 (C3) binds to murine and human MHC II on cDCs. MZ B cells recognize C3 with complement receptor 2 (CR2) and trogocytose the MHC II-C3 complexes, which become exposed on their cell surface. The ubiquitin ligase MARCH1 limits the number of MHC II-C3 complexes displayed on cDCs to prevent their elimination through excessive trogocytosis. Capture of C3 by MHC II thus enables the transfer of cDC-like properties to MZ B cells.
  • Item
    No Preview Available
    Complexing CpG adjuvants with cationic liposomes enhances vaccine-induced formation of liver TRM cells
    Valencia-Hernandez, AM ; Zillinger, T ; Ge, Z ; Tan, PS ; Cozijnsen, A ; McFadden, GI ; Lahoud, MH ; Caminschi, I ; Barchet, W ; Heath, WR ; Fernandez-Ruiz, D (ELSEVIER SCI LTD, 2023-01-27)
    Tissue resident memory T cells (TRM cells) can provide effective tissue surveillance and can respond rapidly to infection. Vaccination strategies aimed at generating TRM cells have shown promise against a range of pathogens. We have previously shown that the choice of adjuvant critically influences CD8+ TRM cell formation in the liver. However, the range of adjuvants tested was limited. Here, we assessed the ability of a broad range of adjuvants stimulating membrane (TLR4), endosomal (TLR3, TLR7 and TLR9) and cytosolic (cGAS, RIG-I) pathogen recognition receptors for their capacity to induce CD8+ TRM formation in a subunit vaccination model. We show that CpG oligodeoxynucleotides (ODN) remain the most efficient inducers of liver TRM cells among all adjuvants tested. Moreover, their combination with the cationic liposome DOTAP further enhances the potency, particularly of the class B ODN CpG 1668 and the human TLR9 ligand CpG 2006 (CpG 7909). This study informs the design of efficient liver TRM-based vaccines for their potential translation.
  • Item
    No Preview Available
    The Batman and Robin of liver-stage immunity to malaria
    Heath, WR ; Holz, LE ; Fernandez-Ruiz, D (CELL PRESS, 2022-01)
    Malaria parasites replicate within the liver shortly after infection. This stage can be controlled by CD8 T cells, but which subsets undertake this function is unclear. Lefebvre et al. now elegantly show that effector memory T (TEM) cells are avid participants, working as a dynamic duo with liver tissue-resident memory T (TRM) cells to combat infection.
  • Item
    No Preview Available
    Interplay between liver and blood stages of Plasmodium infection dictates malaria severity via y8 T cells and IL-17-promoted stress erythropoiesis
    Chora, AF ; Marques, S ; Goncalves, JL ; Lima, P ; da Costa, DG ; Fernandez-Ruiz, D ; Marreiros, MI ; Ruivo, P ; Carvalho, T ; Ribeiro, RM ; Serre, K ; Heath, WR ; Silva-Santos, B ; Tate, AT ; Mota, MM (CELL PRESS, 2023-03-14)
    Plasmodium replicates within the liver prior to reaching the bloodstream and infecting red blood cells. Because clinical manifestations of malaria only arise during the blood stage of infection, a perception exists that liver infection does not impact disease pathology. By developing a murine model where the liver and blood stages of infection are uncoupled, we showed that the integration of signals from both stages dictated mortality outcomes. This dichotomy relied on liver stage-dependent activation of Vγ4+ γδ T cells. Subsequent blood stage parasite loads dictated their cytokine profiles, where low parasite loads preferentially expanded IL-17-producing γδ T cells. IL-17 drove extra-medullary erythropoiesis and concomitant reticulocytosis, which protected mice from lethal experimental cerebral malaria (ECM). Adoptive transfer of erythroid precursors could rescue mice from ECM. Modeling of γδ T cell dynamics suggests that this protective mechanism may be key for the establishment of naturally acquired malaria immunity among frequently exposed individuals.
  • 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.