18 research outputs found

    Therapeutic DNA vaccine induces broad T cell responses in the gut and sustained protection from viral rebound and AIDS in SIV-infected rhesus macaques.

    Get PDF
    Immunotherapies that induce durable immune control of chronic HIV infection may eliminate the need for life-long dependence on drugs. We investigated a DNA vaccine formulated with a novel genetic adjuvant that stimulates immune responses in the blood and gut for the ability to improve therapy in rhesus macaques chronically infected with SIV. Using the SIV-macaque model for AIDS, we show that epidermal co-delivery of plasmids expressing SIV Gag, RT, Nef and Env, and the mucosal adjuvant, heat-labile E. coli enterotoxin (LT), during antiretroviral therapy (ART) induced a substantial 2-4-log fold reduction in mean virus burden in both the gut and blood when compared to unvaccinated controls and provided durable protection from viral rebound and disease progression after the drug was discontinued. This effect was associated with significant increases in IFN-Ξ³ T cell responses in both the blood and gut and SIV-specific CD8+ T cells with dual TNF-Ξ± and cytolytic effector functions in the blood. Importantly, a broader specificity in the T cell response seen in the gut, but not the blood, significantly correlated with a reduction in virus production in mucosal tissues and a lower virus burden in plasma. We conclude that immunizing with vaccines that induce immune responses in mucosal gut tissue could reduce residual viral reservoirs during drug therapy and improve long-term treatment of HIV infection in humans

    GM-CSF Increases Mucosal and Systemic Immunogenicity of an H1N1 Influenza DNA Vaccine Administered into the Epidermis of Non-Human Primates

    Get PDF
    Background: The recent H5N1 avian and H1N1 swine-origin influenza virus outbreaks reaffirm that the threat of a worldwide influenza pandemic is both real and ever-present. Vaccination is still considered the best strategy for protection against influenza virus infection but a significant challenge is to identify new vaccine approaches that offer accelerated production, broader protection against drifted and shifted strains, and the capacity to elicit anti-viral immune responses in the respiratory tract at the site of viral entry. As a safe alternative to live attenuated vaccines, the mucosal and systemic immunogenicity of an H1N1 influenza (A/New Caledonia/20/99) HA DNA vaccine administered by particle-mediated epidermal delivery (PMED or gene gun) was analyzed in rhesus macaques. Methodology/Principal Findings: Macaques were immunized at weeks 0, 8, and 16 using a disposable single-shot particlemediated delivery device designed for clinical use that delivers plasmid DNA directly into cells of the epidermis. Significant levels of hemagglutination inhibiting (HI) antibodies and cytokine-secreting HA-specific T cells were observed in the periphery of macaques following 1-3 doses of the PMED HA DNA vaccine. In addition, HA DNA vaccination induced detectable levels of HA-specific mucosal antibodies and T cells in the lung and gut-associated lymphoid tissues of vaccinated macaques. Importantly, co-delivery of a DNA encoding the rhesus macaque GM-CSF gene was found to significantly enhance both the systemic and mucosal immunogenicity of the HA DNA vaccine. Conclusions/Significance: These results provide strong support for the development of a particle-mediated epidermal DNA vaccine for protection against respiratory pathogens such as influenza and demonstrate, for the first time, the ability of skindelivered GM-CSF to serve as an effective mucosal adjuvant for vaccine induction of immune responses in the gut and respiratory tract. Β© 2010 Loudon et al

    Erythroid mitochondrial retention triggers myeloid-dependent type I interferon in human SLE.

    No full text
    Emerging evidence supports that mitochondrial dysfunction contributes to systemic lupus erythematosus (SLE) pathogenesis. Here we show that programmed mitochondrial removal, a hallmark of mammalian erythropoiesis, is defective in SLE. Specifically, we demonstrate that during human erythroid cell maturation, a hypoxia-inducible factor (HIF)-mediated metabolic switch is responsible for the activation of the ubiquitin-proteasome system (UPS), which precedes and is necessary for the autophagic removal of mitochondria. A defect in this pathway leads to accumulation of red blood cells (RBCs) carrying mitochondria (Mit

    The GM-CSF genetic adjuvant increases both the magnitude and breadth of mucosal T cell responses elicited in the lungs and guts following PMED HA DNA vaccination.

    No full text
    <p>HA-specific T cell responses in the (<b>A</b>) lung and (<b>B</b>) gut mucosa of macaques were determined by IFN-Ξ³ ELISpot assay 4–11 weeks after the final vaccination with HA DNA (solid bars) or HA DNA+GM-CSF (hatched bars). Individual bars represent the peak number of HA-specific IFN-Ξ³ T cells detected in the jejunum and lung tissue of individual animals 4–11 weeks after the final vaccination. (<b>C</b>) Breadth of the IFN-Ξ³ T cell response in PBMC, lung, and gut. HA-specific T cell responses in the indicated tissues were measured using a standard IFN-Ξ³ ELISpot assay with 6 individual pools of overlapping peptides (11 amino acid overlaps, 103 15-mers per pool) comprising the entire amino acid sequence of the influenza A/New Caledonia/20/99 HA protein. The percent contribution of each peptide-pool specific response to the total response was determined by dividing the mean number of IFN-Ξ³ spot forming cells (SFC) measured against each individual peptide pool by the sum of the response against all peptide pools. Results represent the average of 2 time-points tested after the 3<sup>rd</sup> DNA dose (weeks 19 and 23). *Measurement below positive threshold level for the assay.</p

    Induction of mucosal antibody responses in the respiratory tract of PMED HA DNA vaccinated macaques.

    No full text
    <p>Bronchoalveolar lavage fluid (BALF) and tracheal swab samples were collected from macaques prior to immunization and again at 4 weeks following each boosting dose of the HA DNA vaccine, ± rhGM-CSF. IgG antibody responses in BALF (<b>A</b>) and IgA antibody responses in tracheal swabs (<b>B</b>) against the A/New Caledonia/20/99 hemagglutinin protein were detected by ELISA. Data are reported as the O.D. measured at 450 nm for each sample, diluted 1∢20 in PBS, from individual immunized animals.</p

    Particle-mediated DNA vaccine delivery into the epidermis of rhesus macaques using the disposable commercial prototype ND10 device.

    No full text
    <p>Plasmid DNA encoding (<b>A</b>) the gene for influenza A/New Caledonia/20/99 haemagglutinin (HA) was administered alone or in combination with (<b>B</b>) a plasmid encoding the gene for rhesus macaque granulocyte-macrophage colony stimulating factor (rhGM-CSF) co-formulated onto 1–3 Β΅M gold particles. (<b>C</b>, <b>D</b>) The ND10 delivery device consists of a cassette containing 2.0 Β΅g plasmid (1.8Β΅g HA DNA+0.2Β΅g GM-CSF DNA) coated onto 1.0 mg of gold particles, a safety catch that is released when the device is held firmly against the skin surface, and an actuation button that breaks the tip off a gas microcylinder and releases helium at high pressure. Release of the helium ruptures the cassette membrane, entrains the DNA-coated gold particles into the helium jet, and propels them directly into cells in the skin. (<b>E</b>) Vaccinations were targeted to the skin located on the upper inner thigh adjacent to the inguinal lymph node. Immediately following vaccination, vaccination sites are easily visualized as red (erythema) targets in the skin. (<b>F</b>) The erythema is transient (24 hours) and vaccination sites faded but were still discernible at 8 weeks post-vaccination. (<b>G</b>) Shown is gold particle penetration into the epidermal and dermal skin layers in a representative histological cross-section of a skin biopsy collected 10 minutes after ND10 delivery.</p

    Generation of multifunctional T cells in the peripheral blood and lungs of macaques following PMED HA DNA vaccination.

    No full text
    <p>Intracellular cytokine staining was performed on BALF collected from both vaccine groups at 4 weeks after the first boost (panels on left), as well as PBMCs from both vaccine groups collected 4 weeks after the second boost (panels on right) to measure the expression of IFN-Ξ³, TNF-Ξ±, and IL-2 by CD4+ and CD8+ T cells after stimulation with overlapping peptides derived from the A/New Caledonia/20/99 HA protein. Bar charts show the mean total percentage (+/βˆ’ SEM) of CD4+ and CD8+ T cells in the lungs (<b>A</b>) and blood (<b>B</b>) of animals from the unadjuvanted (black bars) and GM-CSF adjuvanted (hatched bars) found to express IFN-Ξ³, TNF-Ξ±, or IL-2 following HA peptide stimulation. Indicated P values were determined using the Mann-Whitney U test (two-tailed). Stacked bar charts show the mean proportion of cells producing IFN-Ξ³ (black), TNF-Ξ± (gray), or IL-2 (hatched) to the total HA-specific CD4+ and CD8+ T cell response detected in the lung (<b>C</b>, <b>E</b>) and blood (<b>D</b>, <b>F</b>) of animals from the unadjuvanted and GM-CSF adjuvanted vaccine groups. Pie charts show the proportion of HA-specific CD4+ and CD8+ T cells in the lung (<b>G</b>) and blood (<b>H</b>) from both vaccine groups positive for the different combinations of one, two, or three cytokines.</p

    LT-adjuvanted DNA vaccine increases T cells with multiple effector functions in the blood.

    No full text
    <p>SIV-specific T cells with 1–4 cytokine (IFN-Ξ³, TNF-Ξ±, IL-2) or cytolytic (CD107a) effector functions were measured in PBMC 14 weeks after ART was withdrawn (week 58) by flow cytometry following <i>in vitro</i> stimulation with overlapping peptide pools derived from genes included in the DNA vaccine (RT, Nef, Gag, Env). Boolean gating was performed to identify the total frequency of CD4+ and CD8+ T cells in the blood of ART responders with any one of 1–4 effector functions. (<b>A</b>) Cumulative mean (Β±SEM) frequency of SIV-specific CD4+ and CD8+ T cells expressing IFN-Ξ³, TNF-Ξ±, IL-2 or CD107a. Cumulative mean frequency (Β± SEM) of SIV-specific (<b>B</b>) CD4+ and (<b>C</b>) CD8+ T cells co-expressing any combination of 2–4 effector functions. Mean (Β± SEM) frequency of SIV-specific (<b>D</b>) CD4+ and (<b>E</b>) CD8+ T cells expressing the indicated combination of dual effector functions.</p
    corecore