Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Influenza Other Respir Viruses ; 16(3): 562-567, 2022 05.
Article in English | MEDLINE | ID: mdl-34859584

ABSTRACT

Individuals with type 2 diabetes mellitus experience high rates of influenza virus infection and complications. We compared the magnitude and duration of serologic response to trivalent influenza vaccine in adults aged 50-80 with and without type 2 diabetes mellitus. Serologic response to influenza vaccination was similar in both groups: greater fold-increases in antibody titer occurred among participants with lower pre-vaccination antibody titers. Waning of antibody titers was not influenced by diabetes status.


Subject(s)
Diabetes Mellitus, Type 2 , Influenza Vaccines , Influenza, Human , Aged , Aged, 80 and over , Antibodies, Viral , Hemagglutination Inhibition Tests , Humans , Influenza A Virus, H3N2 Subtype , Influenza, Human/prevention & control , Middle Aged , Vaccines, Inactivated
2.
Nat Commun ; 12(1): 1222, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619272

ABSTRACT

Programmed death-ligand 1 is a glycoprotein expressed on antigen presenting cells, hepatocytes, and tumors which upon interaction with programmed death-1, results in inhibition of antigen-specific T cell responses. Here, we report a mechanism of inhibiting programmed death-ligand 1 through small molecule-induced dimerization and internalization. This represents a mechanism of checkpoint inhibition, which differentiates from anti-programmed death-ligand 1 antibodies which function through molecular disruption of the programmed death 1 interaction. Testing of programmed death ligand 1 small molecule inhibition in a humanized mouse model of colorectal cancer results in a significant reduction in tumor size and promotes T cell proliferation. In addition, antigen-specific T and B cell responses from patients with chronic hepatitis B infection are significantly elevated upon programmed death ligand 1 small molecule inhibitor treatment. Taken together, these data identify a mechanism of small molecule-induced programmed death ligand 1 internalization with potential therapeutic implications in oncology and chronic viral infections.


Subject(s)
B7-H1 Antigen/metabolism , Endocytosis , Immune Checkpoint Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , CHO Cells , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Cricetulus , Disease Models, Animal , Female , Hepatitis B virus/drug effects , Humans , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Protein Multimerization/drug effects , Small Molecule Libraries/chemistry
3.
4.
Sci Rep ; 10(1): 1835, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32020034

ABSTRACT

Chronic hepatitis B (CHB) infection functional cure is defined as sustained loss of HBsAg and several therapeutic strategies are in clinical development designed to pharmacologically reduce serum HBsAg, break immune tolerance, and increase functional cure rates. However, little is known about pre-treatment HBsAg levels as an indicator of HBV immune potential. Here, we compared the phenotypes and HBV-specific response of lymphocytes in CHB patients stratified by serum HBsAg levels <500 (HBslo) or >50,000 IU/ml (HBshi) using immunological assays (flow cytometry, ICS, ELISPOT). HBshi patients had significantly higher expression of inhibitory PD-1 on CD4+ T cells, particularly among TEMRA subset, and higher FcRL5 expression on B cells. Upon HBcAg(core) or HBsAg(env)-stimulation, 85% and 60% of HBslo patients had IFNγ+TNFα+ and IFNγ+ IL2+ CD4+ T cell responses respectively, in comparison to 33% and 13% of HBshi patients. Checkpoint blockade with αPD-1 improved HBV-specific CD4+ T cell function only in HBslo patients. HBsAg-specific antibody-secreting cells (ASCs) response was not different between these groups, yet αPD-1 treatment resulted in significantly higher fold change in ASCs among patients with HBsAg <100 IU/ml compared to patients with HBsAg >5,000 IU/ml. Thus, serum HBsAg correlates with inhibitory receptor expression, HBV-specific CD4+ T cell responses, and augmentation by checkpoint blockade.


Subject(s)
B-Lymphocytes/immunology , Hepatitis B Surface Antigens/blood , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , T-Lymphocytes/immunology , Biomarkers/blood , Flow Cytometry , Hepatitis B, Chronic/blood , Humans , Programmed Cell Death 1 Receptor/metabolism
5.
Hepat Oncol ; 8(1): HEP35, 2020 Dec 29.
Article in English | MEDLINE | ID: mdl-33680431

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a major public health challenge on the global scale. Affecting hundreds of millions worldwide, HBV is a leading risk factor for hepatocellular carcinoma (HCC). Clinical outcomes from chronic HBV infection are varied and appear to be influenced by a complex and dysregulated host immune response. In turn, much attention has been given to the immunologic response to HBV in an effort to identify host factors that lead to the development of HCC. However, the role of nonimmunologic host factors, such as chronic stress, in HBV-related HCC is poorly defined. Indeed, a growing appreciation for the effects of stress on chronic liver diseases raises the question of its role in chronic HBV infection. In this light, the present review will untangle the roles of key host factors in HBV-related HCC with an emphasis on chronic stress as a viable contributor. First discussed is the interplay of stress, inflammation and chronic liver disease. The host immune response's role as a driver of HBV-related HCC is then reviewed, allowing for a close exploration of the effects of stress on immune function in chronic hepatitis B and as a potential risk factor for HBV-related HCC.

6.
J Infect Dis ; 220(5): 743-751, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31045222

ABSTRACT

BACKGROUND: Human immunodeficiency virus (HIV)-infected persons are at a higher risk of severe influenza. Although we have shown that a standard-dose intradermal influenza vaccine versus a standard-dose intramuscular influenza vaccine does not result in differences in hemagglutination-inhibition titers in this population, a comprehensive examination of cell-mediated immune responses remains lacking. METHODS: Serological, antigen-specific B-cell, and interleukin 2-, interferon γ-, and tumor necrosis factor α-secreting T-cell responses were assessed in 79 HIV-infected men and 79 HIV-uninfected men. RESULTS: The route of vaccination did not affect the immunoglobulin A and immunoglobulin G (IgG) plasmablast or memory B-cell response, although these were severely impaired in the group with a CD4+ T-cell count of <200 cells/µL. The frequencies of IgG memory B cells measured on day 28 after vaccination were highest in the HIV-uninfected group, followed by the group with a CD4+ T-cell count of ≥200 cells/µL and the group with a CD4+ T-cell count of <200 cells/µL. The route of vaccination did not affect the CD4+ or CD8+ T-cell responses measured at various times after vaccination. CONCLUSIONS: The route of vaccination had no effect on antibody responses, antibody avidity, T-cell responses, or B-cell responses in HIV-infected or HIV-uninfected subjects. With the serological and cellular immune responses to influenza vaccination being impaired in HIV-infected individuals with a CD4+ T-cell count of <200 cells/µL, passive immunization strategies need to be explored to protect this population. CLINICAL TRIALS REGISTRATION: NCT01538940.


Subject(s)
HIV Infections/immunology , Immunity, Cellular/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza Vaccines/standards , Influenza, Human/prevention & control , Adult , Antibodies, Viral/immunology , Antibody Formation , B-Lymphocytes/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , HIV Infections/complications , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunoglobulin A , Immunoglobulin G , Influenza A Virus, H1N1 Subtype/immunology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Male , Middle Aged , Thailand , Tumor Necrosis Factor-alpha/metabolism , Vaccination
7.
Sci Rep ; 8(1): 6112, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29666412

ABSTRACT

Influenza virus causes widespread, yearly epidemics by accumulating surface protein mutations to escape neutralizing antibodies established from prior exposure. In contrast to antibody epitopes, T cell mediated immunity targets influenza epitopes that are more highly conserved and have potential for cross-protection. The extent of T cell cross-reactivity between a diverse array of contemporary and historical influenza strains was investigated in ferrets challenged with 2009 pandemic H1N1 influenza or the seasonal H3N2 strain, A/Perth/16/2009. Post-challenge cell-mediated immune responses demonstrated extensive cross-reactivity with a wide variety of contemporary and historical influenza A strains as well as influenza B. Responses in peripheral blood were undetectable by 36d post-challenge, but cross-reactivity persisted in spleen. The strongest responses targeted peptides from the NP protein and demonstrated cross-reactivity in both the CD4+ and CD8+ T cell populations. Cross-reactive CD4+ T cells also targeted HA and NA epitopes, while cross-reactive CD8+ T cells targeted internal M1, NS2, and PA. T cell epitopes demonstrated extensive cross-reactivity between diverse influenza strains in outbred animals, with NP implicated as a significant antigenic target demonstrating extensive cross-reactivity for both CD4+ and CD8+ T cells.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Ferrets/virology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Animals , Cross Reactions , Disease Models, Animal , Ferrets/immunology , Immunity, Cellular , Male , Seasons
8.
Vaccine ; 35(25): 3318-3325, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28499553

ABSTRACT

Sporadic, yet frequent human infections with avian H5N1 influenza A viruses continue to pose a potential pandemic threat. Poor immunogenicity of unadjuvanted H5N1 vaccines warrants developing novel adjuvants and formulations as well as alternate delivery systems to improve their immunogenicity and efficacy. Here, we show that Protollin, a nasal adjuvant composed of Neisseria meningitides outer membrane proteins non-covalently linked to Shigella flexneri 2a lipopolysaccharide, is a potent nasal adjuvant for an inactivated split virion H5N1 clade 1 A/Viet Nam1203/2004 (A/VN/1203/04) vaccine in a mouse model. Protollin-adjuvanted vaccines elicited enhanced serum protective hemagglutination inhibition titers, mucosal IgA responses, and H5N1-specific cell-mediated immunity that resulted in complete protection against a lethal challenge with a homologous virus as well as a heterologous clade 2 virus A/Indonesia/05/2005 (A/IN/05/05). Detailed analysis of adaptive immunity revealed that Protollin increased the frequency of lymphoid- as well as local tissue-resident antibody-secreting cells, local germinal center reaction of B cells, broad-spectrum of CD4 T cell response. Our findings suggest that nasal delivery of H5N1 vaccine with Protollin adjuvant can overcome the poor immunogenicity of H5N1 vaccines, induce both cellular and humoral immune responses, enhance protection against challenge with clade 1 and clade 2 H5N1 viruses and achieve significant antigen dose-sparing.


Subject(s)
Adjuvants, Immunologic , Cysteine Endopeptidases/immunology , Immunity, Mucosal , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Lipopolysaccharides/immunology , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Viral/blood , Antibody-Producing Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Drug Combinations , Hemagglutination Inhibition Tests , Immunity, Cellular , Immunity, Humoral , Immunogenicity, Vaccine , Immunoglobulin A/biosynthesis , Immunoglobulin A/immunology , Influenza Vaccines/administration & dosage , Mice , Orthomyxoviridae Infections/prevention & control
9.
Sci Rep ; 7: 40360, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28059146

ABSTRACT

Tunneling nanotubes (TNTs) represent a novel route of intercellular communication. While previous work has shown that TNTs facilitate the exchange of viral or prion proteins from infected to naïve cells, it is not clear whether the viral genome is also transferred via this mechanism and further, whether transfer via this route can result in productive replication of the infectious agents in the recipient cell. Here we present evidence that lung epithelial cells are connected by TNTs, and in spite of the presence of neutralizing antibodies and an antiviral agent, Oseltamivir, influenza virus can exploit these networks to transfer viral proteins and genome from the infected to naïve cell, resulting in productive viral replication in the naïve cells. These observations indicate that influenza viruses can spread using these intercellular networks that connect epithelial cells, evading immune and antiviral defenses and provide an explanation for the incidence of influenza infections even in influenza-immune individuals and vaccine failures.


Subject(s)
Movement , Nanotubes/chemistry , Orthomyxoviridae/physiology , Actins/metabolism , Animals , Cell Line , Coculture Techniques , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dogs , Epithelial Cells/cytology , Epithelial Cells/ultrastructure , Genome, Viral , Humans , Lung/cytology , Nanotubes/ultrastructure , Orthomyxoviridae/genetics , Polymerization , Viral Proteins/metabolism
10.
Sci Rep ; 6: 37341, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27849030

ABSTRACT

The association of seasonal trivalent influenza vaccine (TIV) with increased infection by 2009 pandemic H1N1 (A(H1N1)pdm09) virus, initially observed in Canada, has elicited numerous investigations on the possibility of vaccine-associated enhanced disease, but the potential mechanisms remain largely unresolved. Here, we investigated if prior immunization with TIV enhanced disease upon A(H1N1)pdm09 infection in mice. We found that A(H1N1)pdm09 infection in TIV-immunized mice did not enhance the disease, as measured by morbidity and mortality. Instead, TIV-immunized mice cleared A(H1N1)pdm09 virus and recovered at an accelerated rate compared to control mice. Prior TIV immunization was associated with potent inflammatory mediators and virus-specific CD8 T cell activation, but efficient immune regulation, partially mediated by IL-10R-signaling, prevented enhanced disease. Furthermore, in contrast to suggested pathological roles, pre-existing non-neutralizing antibodies (NNAbs) were not associated with enhanced virus replication, but rather with promoted antigen presentation through FcR-bearing cells that led to potent activation of virus-specific CD8 T cells. These findings provide new insights into interactions between pre-existing immunity and pandemic viruses.


Subject(s)
Antibodies, Viral/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Receptors, Interleukin-10/immunology , Seasons , Signal Transduction/immunology , Vaccination
11.
J Infect Dis ; 214(7): 1030-8, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27534687

ABSTRACT

BACKGROUND: Emergence of antigenically drifted influenza A(H3N2) viruses resulted in reduced vaccine effectiveness in all age groups during the 2014-2015 influenza season. In children, inactivated influenza vaccine (IIV) elicited neutralizing antibodies (Abs) against drifted strains at significantly lower levels than against the vaccine strain. Little is known about the cross-reactivity of cell-mediated immunity against drifted strains in children. METHODS: Children aged 3-17 years (n = 48) received IIV during the 2014-2015 influenza season. Peripheral blood mononuclear cells, collected before (on day 0) and after (on days 7 and 21) vaccination were evaluated for induction of cross-reactive plasmablasts, memory B cells, and cytokine-secreting CD4(+) and CD8(+) T cells against the vaccine and drifted A(H3N2) viruses by an enzyme-linked immunospot assay and flow cytometry. RESULTS: IIV increased frequencies of plasmablasts and memory B cells. The overall induction of the T-cell response was not significant. Both B-cell and T-cell responses showed significant cross-reactivity against A(H3N2) viruses. Age and preexisting immunity affected virus-specific plasmablast responses and fold-change of T-cell responses, respectively. The proportion of T-helper type 1-prone (ie, interferon γ- or tumor necrosis factor α-secreting) CD4(+) T cell responses also increased with age. CONCLUSIONS: In children aged 3-17 years, B- and T-cell responses following IIV receipt showed significant cross-reactivity against A(H3N2) viruses during a vaccine mismatch season.


Subject(s)
B-Lymphocytes/immunology , Genetic Drift , Immunity, Heterologous , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , T-Lymphocytes/immunology , Adolescent , Child , Child, Preschool , Cross Reactions , Female , Humans , Influenza A Virus, H3N2 Subtype/genetics , Male
12.
J Infect Dis ; 214(10): 1477-1486, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27571905

ABSTRACT

BACKGROUND: Influenza viruses gradually accumulate point mutations, reducing the effectiveness of prior immune protection. METHODS: Children aged 9-14 years received 2010-2011 trivalent inactivated influenza vaccine (TIV). Vaccination history, hemagglutination-inhibition (HI) titers, and cell-mediated immune responses were assessed to investigate the cross-reactivity with past and future influenza virus strains. RESULTS: 2010-2011 TIV induced significant T-cell responses and HI titers of ≥160, with a fold-rise of ≥4 and titers of ≥100 maintained for >7 months in the majority of children. Pre-existing memory B cells in these children differentiated quickly to antibody-secreting cells to the new vaccine antigens. Children vaccinated in the previous year maintained high HI titers well into 2010, demonstrating elevated HI titers against A/Perth/16/2009, the future (in 2010-2011) H3N2 component. Prior vaccination enhanced CD8+ T-cell responses to A/Perth/16/2009. Children vaccinated with the prior 2009-2010 seasonal vaccine also demonstrated higher preexisting levels of interferon γ-secreting CD4+CD69+ T cells to 2009 pandemic influenza A(H1N1). Children previously vaccinated with 2009-2010 seasonal influenza vaccine also showed greater expansion of tumor necrosis factor α-secreting CD8+CD69+ T cells to 2009 pandemic influenza A(H1N1) upon vaccination in the 2010-2011 season than those who were not previously vaccinated. CONCLUSIONS: Seasonal influenza viruses continuously drift, which allows them to circumvent protective immunity, but conserved epitopes provide immunological cross-reactivity in children through either vaccination directly or through prime/boost in the prior influenza season.


Subject(s)
Immunity, Cellular , Immunity, Humoral , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Orthomyxoviridae/immunology , Adolescent , Antibodies, Viral/blood , Child , Cross Reactions , Female , Hemagglutination Inhibition Tests , Humans , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Male , T-Lymphocytes/immunology , Time Factors , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
13.
Vaccine ; 34(38): 4594-4601, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27473306

ABSTRACT

UNLABELLED: High-dose (HD) influenza vaccine shows improved relative efficacy against influenza disease compared to standard-dose (SD) vaccine in individuals ⩾65years. This has been partially credited to superior serological responses, but a comprehensive understanding of cell-mediated immunity (CMI) of HD vaccine remains lacking. In the current study, a total of 105 participants were randomly administered HD or SD vaccine and were evaluated for serological responses. Subsets of the group (n=12-26 per group) were evaluated for B and T cell responses at days 0, 7, 14 and 28 post-vaccination by flow cytometry or ELISPOT assay. HD vaccine elicited significantly higher hemagglutination inhibition (HI) titers than SD vaccine at d28, but comparable titers at d365 post-vaccination. HD vaccine also elicited higher vaccine-specific plasmablast responses at d7 post-vaccination than SD vaccine. However, long-lived memory B cell induction, cytokine-secreting T cell responses and persistence of serological memory were comparable regardless of vaccine dose. More strategies other than increased Ag amount may be needed to improve CMI in older adults. TRIAL REGISTRATION: ClinicalTrials.gov NCT 01189123.


Subject(s)
Dose-Response Relationship, Immunologic , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Plasma Cells/immunology , Aged , Antibodies, Viral/blood , B-Lymphocytes/immunology , Female , Hemagglutination Inhibition Tests , Humans , Immunity, Cellular , Immunity, Humoral , Immunologic Memory , Male
14.
PLoS One ; 11(6): e0157903, 2016.
Article in English | MEDLINE | ID: mdl-27315117

ABSTRACT

In order to better understand inflammation associated with influenza virus infection, we measured cell trafficking, via flow cytometry, to various tissues in the ferret model following infection with an A(H3N2) human seasonal influenza virus (A/Perth/16/2009). Changes in immune cells were observed in the blood, bronchoalveolar lavage fluid, and spleen, as well as lymph nodes associated with the site of infection or distant from the respiratory system. Nevertheless clinical symptoms were mild, with circulating leukocytes exhibiting rapid, dynamic, and profound changes in response to infection. Each of the biological compartments examined responded differently to influenza infection. Two days after infection, when infected ferrets showed peak fever, a marked, transient lymphopenia and granulocytosis were apparent in all infected animals. Both draining and distal lymph nodes demonstrated significant accumulation of T cells, B cells, and granulocytes at days 2 and 5 post-infection. CD8+ T cells significantly increased in spleen at days 2 and 5 post-infection; CD4+ T cells, B cells and granulocytes significantly increased at day 5. We interpret our findings as showing that lymphocytes exit the peripheral blood and differentially home to lymph nodes and tissues based on cell type and proximity to the site of infection. Monitoring leukocyte homing and trafficking will aid in providing a more detailed view of the inflammatory impact of influenza virus infection.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Ferrets/virology , Inflammation/virology , Influenza, Human/virology , Animals , Antibodies, Viral/isolation & purification , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Ferrets/blood , Humans , Inflammation/blood , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza, Human/blood , Leukocyte Count , Lymph Nodes/pathology , Lymph Nodes/virology , Seasons
15.
Cell Immunol ; 304-305: 55-8, 2016.
Article in English | MEDLINE | ID: mdl-27106062

ABSTRACT

Avian H7N9 influenza virus infection with fatal outcomes continues to pose a pandemic threat and highly immunogenic vaccines are urgently needed. In this report we show that baculovirus-derived recombinant H7 hemagglutinin protein, when delivered with RIG-I ligand, induced enhanced antibody and T cell responses and conferred protection against lethal challenge with a homologous H7N9 virus. These findings indicate the potential utility of RIG-I ligands as vaccine adjuvants to increase the immunogenicity of recombinant H7 hemagglutinin.


Subject(s)
DEAD Box Protein 58/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Orthomyxoviridae Infections/prevention & control , T-Lymphocytes/immunology , Adjuvants, Immunologic , Animals , Cells, Cultured , Female , Humans , Immunity, Humoral , Influenza A Virus, H7N9 Subtype/metabolism , Influenza, Human/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Receptors, Immunologic , Receptors, Pattern Recognition/metabolism , T-Lymphocytes/virology , Vaccines, Synthetic
16.
Nanomedicine ; 12(7): 1909-1917, 2016 10.
Article in English | MEDLINE | ID: mdl-27112307

ABSTRACT

To enhance the immunogenicity of the Influenza H5N1 vaccine, we developed an oil-in-water nanoemulsion (NE) adjuvant. NE displayed good temperature stability and maintained particle size. More importantly, it significantly enhanced IL-6 and MCP-1 production to recruit innate cells, including neutrophils, monocytes/macrophages and dendritic cells to the local environment. Furthermore, NE enhanced dendritic cell function to induce robust antigen-specific T and B cell immune responses. NE-adjuvanted H5N1 vaccine not only elicited significantly higher and long-lasting antibody responses, but also conferred enhanced protection against homologous clade 1 as well as heterologous clade 2 H5N1 virus challenge in young as well as in aged mice. The pre-existing immunity to seasonal influenza did not affect the immunogenicity of NE-adjuvanted H5N1 vaccine.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza Vaccines/administration & dosage , Nanoparticles/chemistry , Adjuvants, Immunologic , Animals , Antibodies, Viral , Emulsions , Humans , Influenza, Human/prevention & control , Mice
17.
Vaccine ; 34(6): 744-9, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26765287

ABSTRACT

Since the first case of human infection in March 2013, continued reports of H7N9 cases highlight a potential pandemic threat. Highly immunogenic vaccines to this virus are urgently needed to protect vulnerable populations who lack protective immunity. In this study, an egg- and adjuvant-independent adenoviral vector-based, hemagglutinin H7 subtype influenza vaccine (HAd-H7HA) demonstrated enhanced cell-mediated immunity as well as serum antibody responses in a mouse model. Most importantly, this vaccine provided complete protection against homologous A/H7N9 viral challenge suggesting its potential utility as a pandemic vaccine.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Cellular , Influenza A Virus, H7N9 Subtype , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Adenoviridae , Animals , Antibodies, Viral/blood , Immunity, Humoral , Mice , Mice, Inbred BALB C , Neutralization Tests
18.
Vaccine ; 34(4): 495-502, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26706277

ABSTRACT

The role of pre-existing immunity for influenza vaccine responses is of great importance for public health, and thus has been studied in various contexts, yet the impact of differential priming on vaccine responses in the midst of antigenic drift remains to be elucidated. To address this with antigenically related viruses, mice were first primed by either infection or immunization with A/Puerto Rico/8/34 (PR8) virus, then immunized with whole-inactivated A/Fort Monmouth/1/47 (FM1) virus. The ensuing vaccine responses and the protective efficacy of FM1 were superior in PR8 infection-primed mice compared to PR8 immunization-primed or unprimed mice. Increased FM1-specific Ab responses of PR8 infection-primed mice also broadened cross-reactivity against contemporary as well as antigenically more drifted strains. Further, prior infection heightened the protective efficacy of antigenically distant strains, such as A/Brisbane/59/2006 infection followed by immunization with split pandemic H1N1 vaccine (A/California/07/2009). Therefore, influenza infection is a significant priming event that intensifies future vaccine responses against drift strains.


Subject(s)
Antigenic Variation , Cross Reactions , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Animals , Antibodies, Viral/immunology , Dose-Response Relationship, Immunologic , Immunologic Memory , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Neutralization Tests , Orthomyxoviridae Infections/prevention & control , Vaccination
19.
Open Forum Infect Dis ; 2(2): ofv052, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26380344

ABSTRACT

Background. Influenza disproportionately impacts older adults while current vaccines have reduced effectiveness in the older population. Methods. We conducted a comprehensive evaluation of cellular and humoral immune responses of adults aged 50 years and older to the 2008-2009 seasonal trivalent inactivated influenza vaccine and assessed factors influencing vaccine response. Results. Vaccination increased hemagglutination inhibition and neutralizing antibody; however, 66.3% of subjects did not reach hemagglutination inhibition titers ≥ 40 for H1N1, compared with 22.5% for H3N2. Increasing age had a minor negative impact on antibody responses, whereas prevaccination titers were the best predictors of postvaccination antibody levels. Preexisting memory B cells declined with age, especially for H3N2. However, older adults still demonstrated a significant increase in antigen-specific IgG(+) and IgA(+) memory B cells postvaccination. Despite reduced frequency of preexisting memory B cells associated with advanced age, fold-rise in memory B cell frequency in subjects 60+ was comparable to subjects age 50-59. Conclusions. Older adults mounted statistically significant humoral and cell-mediated immune responses, but many failed to reach hemagglutination inhibition titers ≥40, especially for H1N1. Although age had a modest negative effect on vaccine responses, prevaccination titers were the best predictor of postvaccination antibody levels, irrespective of age.

20.
J Infect Dis ; 212(12): 1923-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26068782

ABSTRACT

We compared the innate immune response to a newly emerged swine-origin influenza A(H3N2) variant containing the M gene from 2009 pandemic influenza A(H1N1), termed "A(H3N2)vpM," to the immune responses to the 2010 swine-origin influenza A(H3N2) variant and seasonal influenza A(H3N2). Our results demonstrated that A(H3N2)vpM-induced myeloid dendritic cells secreted significantly lower levels of type I interferon (IFN) but produced significantly higher levels of proinflammatory cytokines and induced potent inflammasome activation. The reduction in antiviral immunity with increased inflammatory responses upon A(H3N2)vpM infection suggest that these viruses have the potential for increased disease severity in susceptible hosts.


Subject(s)
Inflammasomes/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Leukocytes, Mononuclear/immunology , Animals , Cell Line , Cytokines/metabolism , Dendritic Cells/immunology , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/isolation & purification , Orthomyxoviridae Infections/virology , Swine , Swine Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...