27 research outputs found

    Memory Th1 Cells Are Protective in Invasive Staphylococcus aureus Infection

    Get PDF
    Mechanisms of protective immunity to Staphylococcus aureus infection in humans remain elusive. While the importance of cellular immunity has been shown in mice, T cell responses in humans have not been characterised. Using a murine model of recurrent S. aureus peritonitis, we demonstrated that prior exposure to S. aureus enhanced IFNγ responses upon subsequent infection, while adoptive transfer of S. aureus antigen-specific Th1 cells was protective in naïve mice. Translating these findings, we found that S. aureus antigen-specific Th1 cells were also significantly expanded during human S. aureus bloodstream infection (BSI). These Th1 cells were CD45RO+, indicative of a memory phenotype. Thus, exposure to S. aureus induces memory Th1 cells in mice and humans, identifying Th1 cells as potential S. aureus vaccine targets. Consequently, we developed a model vaccine comprising staphylococcal clumping factor A, which we demonstrate to be an effective human T cell antigen, combined with the Th1-driving adjuvant CpG. This novel Th1-inducing vaccine conferred significant protection during S. aureus infection in mice. This study notably advances our understanding of S. aureus cellular immunity, and demonstrates for the first time that a correlate of S. aureus protective immunity identified in mice may be relevant in humans

    Non-canonical inflammasome activation mediates the adjuvanticity of nanoparticles

    Get PDF
    The non-canonical inflammasome sensor caspase-11 and gasdermin D (GSDMD) drive inflammation and pyroptosis, a type of immunogenic cell death that favors cell-mediated immunity (CMI) in cancer, infection, and autoimmunity. Here we show that caspase-11 and GSDMD are required for CD8+ and Th1 responses induced by nanoparticulate vaccine adjuvants. We demonstrate that nanoparticle-induced reactive oxygen species (ROS) are size dependent and essential for CMI, and we identify 50- to 60-nm nanoparticles as optimal inducers of ROS, GSDMD activation, and Th1 and CD8+ responses. We reveal a division of labor for IL-1 and IL-18, where IL-1 supports Th1 and IL-18 promotes CD8+ responses. Exploiting size as a key attribute, we demonstrate that biodegradable poly-lactic co-glycolic acid nanoparticles are potent CMI-inducing adjuvants. Our work implicates ROS and the non-canonical inflammasome in the mode of action of polymeric nanoparticulate adjuvants and establishes adjuvant size as a key design principle for vaccines against cancer and intracellular pathogens

    Endogenous Oils Derived From Human Adipocytes Are Potent Adjuvants That Promote IL-1α–Dependent Inflammation

    No full text
    Obesity is characterized by chronic inflammation associated with neutrophil and M1 macrophage infiltration into white adipose tissue. However, the mechanisms underlying this process remain largely unknown. Based on the ability of oil-based adjuvants to induce immune responses, we hypothesized that endogenous oils derived from necrotic adipocytes may function as an immunological "danger signal." Here we show that endogenous oils of human origin are potent adjuvants, enhancing antibody responses to a level comparable to Freund's incomplete adjuvant. The endogenous oils were capable of promoting interleukin (IL)-1a-dependent recruitment of neutrophils and M1-like macrophages, while simultaneously diminishing M2-like macrophages. We found that endogenous oils from subcutaneous and omental adipocytes, and from healthy and unhealthy obese individuals, promoted comparable inflammatory responses. Furthermore, we also confirmed that white adipocytes in visceral fat of metabolically unhealthy obese (MUO) individuals are significantly larger than those in metabolically healthy obese individuals. Since adipocyte size is positively correlated with adipocyte death, we propose that endogenous oils have a higher propensity to be released from hypertrophied visceral fat in MUO individuals and that this is the key factor in driving inflammation. In summary, this study shows that adipocytes contain a potent oil adjuvant which drives IL-1α-dependent proinflammatory responses in vivo. © 2014 by the American Diabetes Association

    Multivalent Presentation of MPL by Porous Silicon Microparticles Favors T Helper 1 Polarization Enhancing the Anti-Tumor Efficacy of Doxorubicin Nanoliposomes

    No full text
    <div><p>Porous silicon (pSi) microparticles, in diverse sizes and shapes, can be functionalized to present pathogen-associated molecular patterns that activate dendritic cells. Intraperitoneal injection of MPL-adsorbed pSi microparticles, in contrast to free MPL, resulted in the induction of local inflammation, reflected in the recruitment of neutrophils, eosinophils and proinflammatory monocytes, and the depletion of resident macrophages and mast cells at the injection site. Injection of microparticle-bound MPL resulted in enhanced secretion of the T helper 1 associated cytokines IFN-γ and TNF-α by peritoneal exudate and lymph node cells in response to secondary stimuli while decreasing the anti-inflammatory cytokine IL-10. MPL-pSi microparticles independently exhibited anti-tumor effects and enhanced tumor suppression by low dose doxorubicin nanoliposomes. Intravascular injection of the MPL-bound microparticles increased serum IL-1β levels, which was blocked by the IL-1 receptor antagonist Anakinra. The microparticles also potentiated tumor infiltration by dendritic cells, cytotoxic T lymphocytes, and F4/80<sup>+</sup> macrophages, however, a specific reduction was observed in CD204<sup>+</sup> macrophages.</p></div

    Combined chemo and MPL-pSi therapy inhibits cellular proliferation and stimulates tumor infiltration by immune cells which is partly IL-1 dependent.

    No full text
    <p>a) Excised tumors from mice treated with low dose DOX-NPs with or without pSi-MPL microparticles were stained with antibodies specific for Ki67 (red), CD8 (red), F4/80 (green), CD204 (red), or 33D1 (red). Nuclei were stained with Prolong Gold Antifade Reagent with DAPI (blue). b) The percentage of the population comprised of each cell type is shown graphically as the mean of 3–4 regions in randomly selected tissues representing at least two animals per group. *p<0.05; **p<0.01. c) Plasma IL-1β level six hr after intravenous injection of free MPL or 5×10<sup>8</sup> control or MPL-pSi microparticles (10 µg MPL equivalents; n = 3/group; ***p<0.001; *p<0.05). d) Effect of anakinra on MPL-pSi microparticle-induced changes in plasma IL-1β six hr after injection (30 mg, 3×/week; *p<0.05; n = 3/group). e) Tumor growth of mice injected with MPL-pSi microparticles (weekly as indicated by arrows) with and without anakinra (30 mg, 3×/week; n = 3/group). Control verses MPL-pSi, *p<0.05; **p<0.01; ***p<0.001.</p

    Injection of pSi microparticles with the TLR4 agonist MPL induces a synergistic recruitment of inflammatory cells.

    No full text
    <p>Female wild-type C57BL/6 mice were injected with PBS, MPL (50 µg/mouse), pSi microparticles (5×10<sup>8</sup>) or MPL plus pSi microparticles. The mice were sacrificed 24 hr later and PECs were isolated. PECs were stained with various markers to identify cell populations in the peritoneum at this time point, specifically the number of neutrophils (a; CD11b<sup>+</sup> SiglecF<sup>−</sup> Gr-1<sup>+</sup>), M1-like macrophages (b; CD11b<sup>+</sup> SiglecF<sup>−</sup> F4/80<sup>+</sup> ), M2-like macrophages (c; CD11b<sup>high</sup> F4/80<sup>high</sup>), eosinophils (d; Gr-1<sup>−</sup> ckit<sup>−</sup> SiglecF<sup>−</sup>) and mast cells (e; ckit<sup>+</sup> SiglecF<sup>−</sup>). Indicated variable versus MPL+pSi, *p<0.05, ***p<0.001, n = 3/group.</p

    pSi microparticles are immunomodulatory both <i>in vitro</i> and <i>in vivo</i>.

    No full text
    <p>a) Fluorescent microparticle uptake by human monocytes (THP-1 cells) was measured by flow cytometry at select time points after adding microparticles to the cell culture media (top) or after settling of the microparticles on the cell surface by incubation of cells and microparticles on ice for 60 min (bottom) prior to adding warm media (37°C) and transferring to an incubator (n = 3/group). B-D) Wildtype (WT) (b, c) or NLRP3<sup>−/−</sup> (d) BMDCs were primed with 1 ng/ml (b) or 10 ng/ml (c, d) LPS, followed one hr later with 40 particles/cell using pSi microparticles of varying geometries. After 24 hours, supernatants were collected and IL-1β concentrations were determined by ELISA (***p<0.001). E-G) Female C57BL/6 mice were injected i.p. with PBS, Alum or D10.2, D10.4, D25.4 and RS18.4.4 pSi microparticles (0.3 mg/mouse) and the mice were sacrificed after 24 hr. PECs were isolated and stained with various markers to identify cell populations in the peritoneum, specifically the number of neutrophils (e; CD11b<sup>+</sup> Gr1<sup>high</sup> F4/80<sup>−</sup>), eosinophils (f; Gr1<sup>−</sup> ckit<sup>−</sup> SiglecF<sup>+</sup>) and resident macrophages (g; CD11b<sup>high</sup> F4/80<sup>high</sup>). **p<0.01 compared to PBS group, n = 3/group.</p
    corecore