Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(5): e0252170, 2021.
Article in English | MEDLINE | ID: mdl-34043704

ABSTRACT

Seasonal influenza vaccines are often ineffective because they elicit strain-specific antibody responses to mutation-prone sites on the hemagglutinin (HA) head. Vaccines that provide long-lasting immunity to conserved epitopes are needed. Recently, we reported a nanoparticle-based vaccine platform produced by solid-phase peptide synthesis (SPPS) for targeting linear and helical protein-based epitopes. Here, we illustrate its potential for building broadly protective influenza vaccines. Targeting known epitopes in the HA stem, neuraminidase (NA) active site, and M2 ectodomain (M2e) conferred 50-75% survival against 5LD50 influenza B and H1N1 challenge; combining stem and M2e antigens increased survival to 90%. Additionally, protein sequence and structural information were employed in tandem to identify alternative epitopes that stimulate greater protection; we report three novel HA and NA sites that are highly conserved in type B viruses. One new target in the HA stem stimulated 100% survival, highlighting the value of this simple epitope discovery strategy. A candidate influenza B vaccine targeting two adjacent HA stem sites led to >104-fold reduction in pulmonary viral load. These studies describe a compelling platform for building vaccines that target conserved influenza epitopes.


Subject(s)
Antibodies, Viral/immunology , Epitopes/immunology , Influenza B virus/immunology , Influenza, Human , Orthomyxoviridae Infections , Animals , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
2.
Vaccine ; 38(33): 5256-5267, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32540272

ABSTRACT

Elderly individuals are highly susceptible to influenza virus (IAV) infection and respond poorly to influenza vaccines. Although the generally accepted correlate of protection following influenza vaccination is neutralizing antibody titers, cytotoxic T cell activity has been found to be a better correlate in the elderly. This suggests that vaccines designed to protect against influenza in the elderly should induce both humoral and cellular immunity. The co-induction of T cell immunity is additionally advantageous, as virus-specific T cells are frequently cross-reactive against different strains of IAV. Here, we tested the capacity of a synthetic TLR-4 adjuvant, SLA-SE (second-generation lipid adjuvant formulated in a squalene-based oil-in-water emulsion) to elicit T cell immunity to a recombinant influenza nucleoprotein (rNP), in both young and aged mice. IAV challenge of vaccinated mice resulted in a modest increase in the numbers of NP-specific CD4 and CD8 effector T cells in the spleen, but did not increase numbers of memory phenotype CD8 T cells generated following viral clearance (compared to control vaccinated mice). Cytotoxic activity of CD8, but not CD4 T cells was increased. In addition, SLA-SE adjuvanted vaccination specifically enhanced the production of NP-specific IgG2c antibodies in both young and aged mice. Although NP-specific antibodies are not neutralizing, they can cooperate with CD8 T cells and antigen-presenting cells to enhance protective immunity. Importantly, SLA-SE adjuvanted rNP-vaccination of aged mice resulted in significantly enhanced viral clearance. In addition, vaccination of aged mice resulted in enhanced survival after lethal challenge compared to control vaccination, that approached statistical significance. These data demonstrate the potential of SLA-SE adjuvanted rNP vaccines to (i) generate both cellular and humoral immunity to relatively conserved IAV proteins and (ii) elicit protective immunity to IAV in aged mice.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Antibodies, Viral , CD8-Positive T-Lymphocytes , Influenza, Human/prevention & control , Mice , Mice, Inbred BALB C , Nucleoproteins , Orthomyxoviridae Infections/prevention & control , Vaccination
3.
Diabetes Educ ; 46(3): 261-270, 2020 06.
Article in English | MEDLINE | ID: mdl-32228289

ABSTRACT

PURPOSE: The purpose of this study was to capture information on patient experiences and perspectives of group medical appointments (GMAs) and compare them to those attending individual appointments (IAs) with the diabetes education team (usual care) for managing type 2 diabetes. METHODS: Adults (N = 18; 61% male; 83% 50-70 years old ) with type 2 diabetes (or prediabetes) living in rural Saskatchewan were recruited to complete a semistructured interview on their experiences with GMAs or IAs. To be eligible to participate, individuals must have attended at least 2 GMAs or 2 IAs. Transcripts were coded and analyzed using content analysis. RESULTS: Overall, participants spoke highly of their respective appointment type. Results indicated that both appointment types positively influenced understanding of diabetes management, with the most notable difference being greater understanding of stress management in the GMAs. Participants identified several positive aspects of each appointment type, which included convenience, supportive and enjoyable, and informative for GMAs and time and tailored information for IAs. Participants provided some suggestions to improve diabetes related-care for their respective appointment type. CONCLUSIONS: Participants of GMAs and IAs for type 2 diabetes each reported unique strengths to their respective care plan and reported benefiting from their care.


Subject(s)
Appointments and Schedules , Diabetes Mellitus, Type 2/psychology , Diabetes Mellitus, Type 2/therapy , Shared Medical Appointments , Aged , Female , Humans , Male , Middle Aged , Qualitative Research , Saskatchewan
4.
NPJ Vaccines ; 4: 30, 2019.
Article in English | MEDLINE | ID: mdl-31341647

ABSTRACT

Nanoparticle-based delivery systems are being used to simplify and accelerate new vaccine development. Previously, we described the solid-phase synthesis of a 61-amino acid conjugate vaccine carrier comprising a α-helical domain followed by two universal T cell epitopes. Circular dichroism, analytical centrifugation, and dynamic light scattering indicate that this carrier forms coiled-coil nanoparticles. Here we expand the potential of this carrier by appending B cell epitopes to its amino acid sequence, thereby eliminating the need for traditional conjugation reactions. Peptides containing Tau or amyloid-ß epitopes at either terminus assemble into ~20 nm particles and induce antibody responses in outbred mice. Vaccine function was verified in three experiments. The first targeted gonadotropin-releasing hormone, a 10-amino acid neuropeptide that regulates sexual development. Induction of peak antibody titers in male mice stimulated a dramatic loss in fertility and marked testis degeneration. The second experiment generated antibodies to an epitope on the murine IgE heavy chain analogous to human IgE sequence recognized by omalizumab, the first monoclonal antibody approved for the treatment of allergic asthma. Like omalizumab, the anti-IgE antibodies in immunized mice reduced the concentrations of circulating free IgE and prevented IgE-induced anaphylaxis. Finally, a peptide containing the highly conserved Helix A epitope within the influenza hemagglutinin stem domain induced antibodies that successfully protected mice against a lethal H1N1 challenge. These results establish the utility of a new vaccine platform for eliciting prophylactic and therapeutic antibodies to linear and helical B cell epitopes.

5.
NPJ Vaccines ; 4: 1, 2019.
Article in English | MEDLINE | ID: mdl-30622742

ABSTRACT

Aluminum salts, developed almost a century ago, remain the most commonly used adjuvant for licensed human vaccines. Compared to more recently developed vaccine adjuvants, aluminum adjuvants such as Alhydrogel are heterogeneous in nature, consisting of 1-10 micrometer-sized aggregates of nanoparticle aluminum oxyhydroxide fibers. To determine whether the particle size and aggregated state of aluminum oxyhydroxide affects its adjuvant activity, we developed a scalable, top-down process to produce stable nanoparticles (nanoalum) from the clinical adjuvant Alhydrogel by including poly(acrylic acid) (PAA) polymer as a stabilizing agent. Surprisingly, the PAA:nanoalum adjuvant elicited a robust TH1 immune response characterized by antigen-specific CD4+ T cells expressing IFN-γ and TNF, as well as high IgG2 titers, whereas the parent Alhydrogel and PAA elicited modest TH2 immunity characterized by IgG1 antibodies. ASC, NLRP3 and the IL-18R were all essential for TH1 induction, indicating an essential role of the inflammasome in this adjuvant's activity. Compared to microparticle Alhydrogel this nanoalum adjuvant provided superior immunogenicity and increased protective efficacy against lethal influenza challenge. Therefore PAA:nanoalum represents a new class of alum adjuvant that preferentially enhances TH1 immunity to vaccine antigens. This adjuvant may be widely beneficial to vaccines for which TH1 immunity is important, including tuberculosis, pertussis, and malaria.

6.
Eur J Immunol ; 49(2): 266-276, 2019 02.
Article in English | MEDLINE | ID: mdl-30548475

ABSTRACT

Influenza A annually infects 5-10% of the world's human population resulting in one million deaths. Influenza causes annual epidemics and reinfects previously exposed individuals because of antigenic drift in the glycoprotein hemagglutinin. Due to antigenic drift, the immune system is simultaneously exposed to novel and conserved parts of the influenza virus via vaccination and/or infection throughout life. Preexisting immunity has long been known to augment subsequent hemagglutination inhibitory antibody (hAb) responses. However, the preexisting immunological contributors that influence hAb responses are not understood. Therefore, we adapted and developed sequential infection and immunization mouse models using drifted influenza strains to show that MHC Class II haplotype and T-cell reactivity influences subsequent hAb responses. We found that CB6F1 mice infected with A/CA followed by immunization with A/PR8 have increased hAb responses to A/PR8 compared to C57BL/6 mice. Increased hAb responses in CB6F1 mice were CD4+  T-cell and B-cell dependent and corresponded to increased germinal center A/PR8-specific B and T-follicular helper cells. These results suggest conserved MHC Class II restricted epitopes within HA are essential for B cells to respond to drifting influenza and could be leveraged to boost hAb responses.


Subject(s)
Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Immunization , Immunologic Memory , Influenza A virus/immunology , Animals , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Female , Mice
7.
NPJ Vaccines ; 3: 39, 2018.
Article in English | MEDLINE | ID: mdl-30302281

ABSTRACT

Members of the Flaviviridae family are the leading causes of mosquito-borne viral disease worldwide. While dengue virus is the most prevalent, the recent Zika virus outbreak in the Americas triggered a WHO public health emergency, and yellow fever and West Nile viruses (WNV) continue to cause regional epidemics. Given the sporadic nature of flaviviral epidemics both temporally and geographically, there is an urgent need for vaccines that can rapidly provide effective immunity. Protection from flaviviral infection is correlated with antibodies to the viral envelope (E) protein, which encodes receptor binding and fusion functions. TLR agonist adjuvants represent a promising tool to enhance the protective capacity of flavivirus vaccines through dose and dosage reduction and broadening of antiviral antibody responses. This study investigates the ability to improve the immunogenicity and protective capacity of a promising clinical-stage WNV recombinant E-protein vaccine (WN-80E) using a novel combination adjuvant, which contains a potent TLR-4 agonist and the saponin QS21 in a liposomal formulation (SLA-LSQ). Here, we show that, in combination with WN-80E, optimized SLA-LSQ is capable of inducing long-lasting immune responses in preclinical models that provide sterilizing protection from WNV challenge, reducing viral titers following WNV challenge to undetectable levels in Syrian hamsters. We have investigated potential mechanisms of action by examining the antibody repertoire generated post-immunization. SLA-LSQ induced a more diverse antibody response to WNV recombinant E-protein antigen than less protective adjuvants. Collectively, these studies identify an adjuvant formulation that enhances the protective capacity of recombinant flavivirus vaccines.

8.
Mol Ther ; 26(10): 2507-2522, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30078765

ABSTRACT

Since the first demonstration of in vivo gene expression from an injected RNA molecule almost two decades ago,1 the field of RNA-based therapeutics is now taking significant strides, with many cancer and infectious disease targets entering clinical trials.2 Critical to this success has been advances in the knowledge and application of delivery formulations. Currently, various lipid nanoparticle (LNP) platforms are at the forefront,3 but the encapsulation approach underpinning LNP formulations offsets the synthetic and rapid-response nature of RNA vaccines.4 Second, limited stability of LNP formulated RNA precludes stockpiling for pandemic readiness.5 Here, we show the development of a two-vialed approach wherein the delivery formulation, a highly stable nanostructured lipid carrier (NLC), can be manufactured and stockpiled separate from the target RNA, which is admixed prior to administration. Furthermore, specific physicochemical modifications to the NLC modulate immune responses, either enhancing or diminishing neutralizing antibody responses. We have combined this approach with a replicating viral RNA (rvRNA) encoding Zika virus (ZIKV) antigens and demonstrated a single dose as low as 10 ng can completely protect mice against a lethal ZIKV challenge, representing what might be the most potent approach to date of any Zika vaccine.


Subject(s)
Antigens, Viral/administration & dosage , Lipids/administration & dosage , Nanoparticles/administration & dosage , Zika Virus Infection/therapy , Animals , Antigens, Viral/genetics , Disease Models, Animal , Drug Delivery Systems , Humans , Lipids/chemistry , Mice , Nanoparticles/chemistry , RNA, Viral/genetics , RNA, Viral/immunology , Virus Replication/drug effects , Zika Virus/genetics , Zika Virus/pathogenicity , Zika Virus Infection/genetics , Zika Virus Infection/virology
9.
J Clin Invest ; 128(9): 4163-4178, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29999501

ABSTRACT

Germinal centers (GCs) are major sites of clonal B cell expansion and generation of long-lived, high-affinity antibody responses to pathogens. Signaling through TLRs on B cells promotes many aspects of GC B cell responses, including affinity maturation, class switching, and differentiation into long-lived memory and plasma cells. A major challenge for effective vaccination is identifying strategies to specifically promote GC B cell responses. Here, we have identified a mechanism of regulation of GC B cell TLR signaling, mediated by αv integrins and noncanonical autophagy. Using B cell-specific αv-KO mice, we show that loss of αv-mediated TLR regulation increased GC B cell expansion, somatic hypermutation, class switching, and generation of long-lived plasma cells after immunization with virus-like particles (VLPs) or antigens associated with TLR ligand adjuvants. Furthermore, targeting αv-mediated regulation increased the magnitude and breadth of antibody responses to influenza virus vaccination. These data therefore identify a mechanism of regulation of GC B cells that can be targeted to enhance antibody responses to vaccination.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Integrin alphaV/immunology , Animals , Autophagy/immunology , Female , Germinal Center/cytology , Immunization , Immunoglobulin Class Switching , Immunoglobulin G/blood , Immunologic Memory , Influenza A virus/immunology , Integrin alphaV/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/immunology , Signal Transduction/immunology , Somatic Hypermutation, Immunoglobulin , Toll-Like Receptors/immunology , Vaccines, Virus-Like Particle/immunology
10.
Front Immunol ; 9: 295, 2018.
Article in English | MEDLINE | ID: mdl-29515589

ABSTRACT

Elderly people are at high risk for influenza-related morbidity and mortality due to progressive immunosenescence. While toll-like receptor (TLR) agonist containing adjuvants, and other adjuvants, have been shown to enhance influenza vaccine-induced protective responses, the mechanisms underlying how these adjuvanted vaccines could benefit the elderly remain elusive. Here, we show that a split H1N1 influenza vaccine (sH1N1) combined with a TLR4 agonist, glucopyranosyl lipid adjuvant formulated in a stable oil-in-water emulsion (GLA-SE), boosts IgG2c:IgG1 ratios, enhances hemagglutination inhibition (HAI) titers, and increases protection in aged mice. We find that all adjuvanted sH1N1 vaccines tested were able to protect both young and aged mice from lethal A/H1N1/California/4/2009 virus challenge after two immunizations compared to vaccine alone. We show that GLA-SE combined with sH1N1, however, also provides enhanced protection from morbidity in aged mice given one immunization (based on change in weight percentage). While the GLA-SE-adjuvanted sH1N1 vaccine promotes the generation of cytokine-producing T helper 1 cells, germinal center B cells, and long-lived bone marrow plasma cells in young mice, these responses were muted in aged mice. Differential in vitro responses, dependent on age, were also observed from mouse-derived bone marrow-derived dendritic cells and lung homogenates following stimulation with adjuvants, including GLA-SE. Besides enhanced HAI titers, additional protective factors elicited with sH1N1 + GLA-SE in young mice were observed, including (a) rapid reduction of viral titers in the lung, (b) prevention of excessive lung inflammation, and (c) homeostatic maintenance of alveolar macrophages (AMs) following H1N1 infection. Collectively, our results provide insight into mechanisms of adjuvant-mediated immune protection in the young and elderly.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Aged , Animals , Antibodies, Viral/blood , Cells, Cultured , Dendritic Cells/virology , Female , Glucosides/pharmacology , Glucosides/therapeutic use , Humans , Immunity , Immunization , Lipid A/pharmacology , Lipid A/therapeutic use , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/agonists
11.
Eur J Immunol ; 46(12): 2719-2729, 2016 12.
Article in English | MEDLINE | ID: mdl-27701733

ABSTRACT

The contribution of B cells to immunity against many infectious diseases is unquestionably important and well characterized. Here, we sought to determine the role of B cells in the induction of T-helper 1 (TH 1) CD4+ T cells upon vaccination with a tuberculosis (TB) antigen combined with a TLR4 agonist. We used B-cell deficient mice (µMT-/- ), tetramer-positive CD4+ T cells, markers of memory "precursor" effector cells (MPECs), and T-cell adoptive transfers and demonstrated that the early antigen-specific cytokine-producing TH 1 responses are unaffected in the absence of B cells, however MPEC induction is strongly impaired resulting in a deficiency of the memory TH 1 response in µMT-/- mice. We further show that antigen-presentation by B cells is necessary for their role in MPEC generation using B-cell adoptive transfers from wt or MHC class II knock-out mice into µMT-/- mice. Our study challenges the view that B-cell deficiency exclusively alters the TH 1 response at memory time-points. Collectively, our results provide new insights on the multifaceted roles of B cells that will have a high impact on vaccine development against several pathogens including those requiring TH 1 cell-mediated immunity.


Subject(s)
Antigen Presentation , B-Lymphocytes/physiology , Immunologic Factors/immunology , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Adoptive Transfer , Animals , B-Lymphocytes/transplantation , Cell Differentiation , Cells, Cultured , Humans , Immunoglobulin mu-Chains/genetics , Immunologic Memory , Lymphocyte Activation , Mice , Mice, Knockout , T-Lymphocyte Subsets/transplantation , Toll-Like Receptor 4/agonists , Tuberculosis/prevention & control
12.
Toxins (Basel) ; 3(9): 1131-45, 2011 09.
Article in English | MEDLINE | ID: mdl-22069759

ABSTRACT

The entry of ricin toxin into macrophages and certain other cell types in the spleen and liver results in toxin-induced inflammation, tissue damage and organ failure. It has been proposed that uptake of ricin into macrophages is facilitated by the mannose receptor (MR; CD206), a C-type lectin known to recognize the oligosaccharide side chains on ricin's A (RTA) and B (RTB) subunits. In this study, we confirmed that the MR does indeed promote ricin binding, uptake and killing of monocytes in vitro. To assess the role of MR in the pathogenesis of ricin in vivo, MR knockout (MR(-/-)) mice were challenged with the equivalent of 2.5× or 5× LD(50) of ricin by intraperitoneal injection. We found that MR(-/-) mice were significantly more susceptible to toxin-induced death than their age-matched, wild-type control counterparts. These data are consistent with a role for the MR in scavenging and degradation of ricin, not facilitating its uptake and toxicity in vivo.


Subject(s)
Immunity, Innate/drug effects , Lectins, C-Type/physiology , Mannose-Binding Lectins/physiology , Receptors, Cell Surface/physiology , Ricin/toxicity , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cytokines/immunology , Female , Humans , Leukocytes/drug effects , Macrophages/drug effects , Male , Mannose Receptor , Mice , Mice, Knockout , Ricin/blood , Ricin/pharmacokinetics
13.
Vision Res ; 50(1): 36-45, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19819251

ABSTRACT

People with central-field loss must use peripheral vision for reading. Previous studies have shown that reading performance in peripheral vision can improve with extensive practice on a trigram letter-recognition task. The present study compared training on this task with training on two other character-based tasks (lexical-decision and Rapid Serial Visual Presentation (RSVP) reading) which might plausibly produce more improvement in peripheral reading speed. Twenty-eight normally-sighted young adults were trained at 10 degrees in the lower visual field in a pre/post design. All three training methods produced significant improvements in reading speed, with average gains of 39% for lexical-decision, 54% for trigram letter-recognition, and 72% for RSVP training. Although the RSVP training was most effective, the lexical-decision task has the advantage of easy self administration making it more practical for home-based training.


Subject(s)
Patient Education as Topic/methods , Reading , Visual Fields/physiology , Visual Perception/physiology , Adult , Female , Humans , Male , Task Performance and Analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...