Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Gen Virol ; 97(7): 1489-1499, 2016 07.
Article in English | MEDLINE | ID: mdl-27031847

ABSTRACT

Multiple subtypes and many antigenic variants of influenza A virus (IAV) co-circulate in swine in the USA, complicating effective use of commercial vaccines to control disease and transmission. Whole inactivated virus (WIV) vaccines may provide partial protection against IAV with substantial antigenic drift, but have been shown to induce vaccine-associated enhanced respiratory disease (VAERD) when challenged with an antigenic variant of the same haemagglutinin (HA) subtype. This study investigated the role the immune response against HA, neuraminidase (NA) and nucleoprotein (NP) may play in VAERD by reverse engineering vaccine and challenge viruses on a common backbone and using them in a series of vaccination/challenge trials. Mismatched HA between vaccine and challenge virus was necessary to induce VAERD. However, vaccines containing a matched NA abrogated the VAERD phenomenon induced by the HA mismatch and this was correlated with NA-inhibiting (NI) antibodies. Divergence between the two circulating swine N2 lineages (92 % identity) resulted in a loss of NI cross-reactivity and also resulted in VAERD with the mismatched HA. The NP lineage selected for use in the WIV vaccine strains did not affect protection or pathology. Thus the combination of HA and NA in the vaccine virus strains played a substantial role in vaccine protection versus immunopathology, suggesting that vaccines that target the HA protein alone could be more prone to VAERD due to the absence of cross-protective NI antibodies.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Orthomyxoviridae Infections/virology , Swine Diseases/virology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Neuraminidase/immunology , Nucleoproteins/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Respiratory System/virology , Swine , Swine Diseases/immunology , Swine Diseases/prevention & control , Vaccination , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology
2.
Proc Natl Acad Sci U S A ; 108(51): 20748-53, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22143798

ABSTRACT

Seasonal epidemics caused by influenza virus are driven by antigenic changes (drift) in viral surface glycoproteins that allow evasion from preexisting humoral immunity. Antigenic drift is a feature of not only the hemagglutinin (HA), but also of neuraminidase (NA). We have evaluated the antigenic evolution of each protein in H1N1 and H3N2 viruses used in vaccine formulations during the last 15 y by analysis of HA and NA inhibition titers and antigenic cartography. As previously shown for HA, genetic changes in NA did not always lead to an antigenic change. The noncontinuous pattern of NA drift did not correspond closely with HA drift in either subtype. Although NA drift was demonstrated using ferret sera, we show that these changes also impact recognition by NA-inhibiting antibodies in human sera. Remarkably, a single point mutation in the NA of A/Brisbane/59/2007 was primarily responsible for the lack of inhibition by polyclonal antibodies specific for earlier strains. These data underscore the importance of NA inhibition testing to define antigenic drift when there are sequence changes in NA.


Subject(s)
Antigens/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/chemistry , Neuraminidase/chemistry , Animals , Evolution, Molecular , Ferrets , Humans , Influenza Vaccines/chemistry , Influenza, Human/prevention & control , Influenza, Human/virology , Models, Molecular , Molecular Conformation , Mutation , Reassortant Viruses/chemistry
3.
J Virol ; 86(19): 10597-605, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22811541

ABSTRACT

Control of swine influenza A virus (IAV) in the United States is hindered because inactivated vaccines do not provide robust cross-protection against the multiple antigenic variants cocirculating in the field. Vaccine efficacy can be limited further for vaccines administered to young pigs that possess maternally derived immunity. We previously demonstrated that a recombinant A/sw/Texas/4199-2/1998 (TX98) (H3N2) virus expressing a truncated NS1 protein is attenuated in swine and has potential for use as an intranasal live attenuated influenza virus (LAIV) vaccine. In the present study, we compared 1 dose of intranasal LAIV with 2 intramuscular doses of TX98 whole inactivated virus (WIV) with adjuvant in weanling pigs with and without TX98-specific maternally derived antibodies (MDA). Pigs were subsequently challenged with wild-type homologous TX98 H3N2 virus or with an antigenic variant, A/sw/Colorado/23619/1999 (CO99) (H3N2). In the absence of MDA, both vaccines protected against homologous TX98 and heterologous CO99 shedding, although the LAIV elicited lower hemagglutination inhibition (HI) antibody titers in serum. The efficacy of both vaccines was reduced by the presence of MDA; however, WIV vaccination of MDA-positive pigs led to dramatically enhanced pneumonia following heterologous challenge, a phenomenon known as vaccine-associated enhanced respiratory disease (VAERD). A single dose of LAIV administered to MDA-positive pigs still provided partial protection from CO99 and may be a safer vaccine for young pigs under field conditions, where dams are routinely vaccinated and diverse IAV strains are in circulation. These results have implications not only for pigs but also for other influenza virus host species.


Subject(s)
Antibodies/chemistry , Influenza Vaccines/metabolism , Respiratory Tract Infections/immunology , Vaccines, Attenuated/metabolism , Animals , Bronchoalveolar Lavage Fluid , Cell Line , Dogs , Hemagglutination Inhibition Tests , Influenza A Virus, H3N2 Subtype/metabolism , Lung/metabolism , Mucous Membrane/metabolism , Swine
4.
Rev Med Virol ; 22(4): 267-79, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22438243

ABSTRACT

Vaccines are instrumental in controlling the burden of influenza virus infection in humans and animals. Antibodies raised against both major viral surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA), can contribute to protective immunity. Vaccine-induced HA antibodies have been characterized extensively, and they generally confer protection by blocking the attachment and fusion of a homologous virus onto host cells. Although not as well characterized, some functions of NA antibodies in influenza vaccine-mediated immunity have been recognized for many years. In this review, we summarize the case for NA antibodies in influenza vaccine-mediated immunity. In the absence of well-matched HA antibodies, NA antibodies can provide varying degrees of protection against disease. NA proteins of seasonal influenza vaccines have been shown in some instances to elicit serum antibodies with cross-reactivity to avian-origin and swine-origin influenza strains, in addition to HA drift variants. NA-mediated immunity has been linked to (i) conserved NA epitopes amongst otherwise antigenically distinct strains, partly attributable to the segmented influenza viral genome; (ii) inhibition of NA enzymatic activity; and (iii) the NA content in vaccine formulations. There is a potential to enhance the effectiveness of existing and future influenza vaccines by focusing greater attention on the antigenic characteristics and potency of the NA protein.


Subject(s)
Antibodies, Viral/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Neuraminidase/immunology , Viral Proteins/immunology , Animals , Humans , Influenza A virus/enzymology , Influenza A virus/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/virology , Neuraminidase/genetics , Viral Proteins/genetics
5.
J Infect Dis ; 202(11): 1634-8, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20979454

ABSTRACT

Levels of preexisting antibodies to the hemagglutinin of pandemic influenza A(H1N1) 2009 (hereafter pandemic H1N1) virus positively correlate with age. The impact of contemporary seasonal influenza vaccines on establishing immunity to other pandemic H1N1 proteins is unknown. We measured serum antibodies to the neuraminidase (NA) of pandemic H1N1 in adults prior to and after vaccination with seasonal trivalent inactivated influenza vaccines. Serum antibodies to pandemic H1N1 NA were observed in all age groups; however, vaccination elevated levels of pandemic H1N1 NA antibodies predominately in elderly individuals (age, ⩾60 years). Therefore, contemporary seasonal vaccines likely contribute to reduction of pandemic H1N1-associated disease in older individuals.


Subject(s)
Antibodies, Viral/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Adult , Age Factors , Aged , Aged, 80 and over , British Columbia , Connecticut , Humans , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/immunology , Influenza, Human/prevention & control , Middle Aged , Neuraminidase/immunology , Pandemics/prevention & control , Vaccines, Inactivated/immunology , Young Adult
6.
Pediatr Infect Dis J ; 39(2): 134-136, 2020 02.
Article in English | MEDLINE | ID: mdl-31738324

ABSTRACT

We identified mortality predictors among HIV-exposed uninfected infants and infants living with HIV in Kenyan early infant diagnosis services between 2012 and 2017. Younger maternal age and absence of antenatal antiretroviral therapy among HIV-exposed uninfected infants (n = 2366) and travel time to hospital and delayed infant testing among infants living with HIV (n = 130) predicted mortality, highlighting the importance of supporting engagement in maternal/pediatric HIV services.


Subject(s)
HIV Infections/mortality , Infant Mortality , Anti-Retroviral Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Female , HIV Infections/drug therapy , HIV Infections/epidemiology , HIV Infections/transmission , Humans , Infant , Infant, Newborn , Infectious Disease Transmission, Vertical , Kenya/epidemiology , Male , Odds Ratio , Pregnancy , Public Health Surveillance , Retrospective Studies
7.
Article in English | MEDLINE | ID: mdl-30701079

ABSTRACT

BACKGROUND: Infant HIV diagnosis by HIV DNA polymerase chain reaction (PCR) testing at the standard 6 weeks of age is often late to mitigate the mortality peak that occurs in HIV positive infants' first 2-3 months of life. Kenya recently revised their early infant diagnosis (EID) guidelines to include HIV DNA PCR testing at birth (pilot only), 6 weeks, 6 months, and 12 months postnatal and a final 18-month antibody test. The World Health Organization (WHO) approved point-of-care (POC) diagnostic platforms for infant HIV testing in resource-limited countries that could simplify logistics and expedite infant diagnosis. Sustainable scale-up and optimal utility in Kenya and other high-prevalence countries depend on robust implementation studies in diverse clinical settings. METHODS: We will pilot the implementation of birth testing by HIV DNA PCR, as well as two POC testing systems (Xpert HIV-1 Qual [Xpert] and Alere q HIV-1/2 Detect [Alere q]), on specimens collected from Kenyan infants at birth (0 to 2 weeks) and 6 weeks (4 to < 24 weeks) postnatal. The formative phase will inform optimal implementation of birth testing and two POC testing technologies. Qualitative interviews with stakeholders (providers, parents of HIV-exposed infants, and community members) will assess attitudes, barriers, and recommendations to optimize implementation at their respective sites. A non-blinded pilot study at four Kenyan hospitals (n = 2 Xpert, n = 2 Alere q platforms) will evaluate infant HIV POC testing compared with standard of care HIV DNA PCR testing in both the birth and 6-week windows. Objectives of the pilot are to assess uptake, efficiency, quality, implementation variables, user experiences of birth testing with both POC testing systems or with HIV DNA PCR, and costs. DISCUSSION: This study will generate data on the clinical impact and feasibility of adding HIV testing at birth utilizing POC and traditional PCR HIV testing strategies in resource-limited settings. Data from this pilot will inform the optimal implementation of Kenya's birth testing guidelines and of POC testing systems for the improvement of EID outcomes. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03435887. Registered 26 February 2018.

8.
Vaccine ; 36(41): 6103-6110, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30181048

ABSTRACT

Influenza A virus (IAV) causes a disease burden in the swine industry in the US and is a challenge to prevent due to substantial genetic and antigenic diversity of IAV that circulate in pig populations. Whole inactivated virus (WIV) vaccines formulated with oil-in-water (OW) adjuvant are commonly used in swine. However, WIV-OW are associated with vaccine-associated enhanced respiratory disease (VAERD) when the hemagglutinin and neuraminidase of the vaccine strain are mismatched with the challenge virus. Here, we assessed if different types of adjuvant in WIV vaccine formulations impacted VAERD outcome. WIV vaccines with a swine δ1-H1N2 were formulated with different commercial adjuvants: OW1, OW2, nano-emulsion squalene-based (NE) and gel polymer (GP). Pigs were vaccinated twice by the intramuscular route, 3 weeks apart, then challenged with an H1N1pdm09 three weeks post-boost and necropsied at 5 days post infection. All WIV vaccines elicited antibodies detected using the hemagglutination inhibition (HI) assay against the homologous vaccine virus, but not against the heterologous challenge virus; in contrast, all vaccinated groups had cross-reactive IgG antibody and IFN-γ responses against H1N1pdm09, with a higher magnitude observed in OW groups. Both OW groups demonstrated robust homologous HI titers and cross-reactivity against heterologous H1 viruses in the same genetic lineage. However, both OW groups had severe immunopathology consistent with VAERD after challenge when compared to NE, GP, and non-vaccinated challenge controls. None of the WIV formulations protected pigs from heterologous virus replication in the lungs or nasal cavity. Thus, although the type of adjuvant in the WIV formulation played a significant role in the magnitude of immune response to homologous and antigenically similar H1, none tested here increased the breadth of protection against the antigenically-distinct challenge virus, and some impacted immunopathology after challenge.


Subject(s)
Influenza Vaccines/therapeutic use , Respiratory Tract Diseases/prevention & control , Vaccines, Inactivated/therapeutic use , Animals , Enzyme-Linked Immunosorbent Assay , Lung/metabolism , Lung/virology , Neuraminidase/antagonists & inhibitors , Respiratory Tract Diseases/immunology , Swine
9.
PLoS Med ; 4(2): e59, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17298168

ABSTRACT

BACKGROUND: A pandemic H5N1 influenza outbreak would be facilitated by an absence of immunity to the avian-derived virus in the human population. Although this condition is likely in regard to hemagglutinin-mediated immunity, the neuraminidase (NA) of H5N1 viruses (avN1) and of endemic human H1N1 viruses (huN1) are classified in the same serotype. We hypothesized that an immune response to huN1 could mediate cross-protection against H5N1 influenza virus infection. METHODS AND FINDINGS: Mice were immunized against the NA of a contemporary human H1N1 strain by DNA vaccination. They were challenged with recombinant A/Puerto Rico/8/34 (PR8) viruses bearing huN1 (PR8-huN1) or avN1 (PR8-avN1) or with H5N1 virus A/Vietnam/1203/04. Additional naïve mice were injected with sera from vaccinated mice prior to H5N1 challenge. Also, serum specimens from humans were analyzed for reactivity with avN1. Immunization elicited a serum IgG response to huN1 and robust protection against the homologous challenge virus. Immunized mice were partially protected from lethal challenge with H5N1 virus or recombinant PR8-avN1. Sera transferred from immunized mice to naïve animals conferred similar protection against H5N1 mortality. Analysis of human sera showed that antibodies able to inhibit the sialidase activity of avN1 exist in some individuals. CONCLUSIONS: These data reveal that humoral immunity elicited by huN1 can partially protect against H5N1 infection in a mammalian host. Our results suggest that a portion of the human population could have some degree of resistance to H5N1 influenza, with the possibility that this could be induced or enhanced through immunization with seasonal influenza vaccines.


Subject(s)
Antibodies, Viral/immunology , Influenza A Virus, H5N1 Subtype , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Female , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/immunology , Influenza, Human/virology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Treatment Outcome , Ultracentrifugation , Vaccines, DNA/therapeutic use
10.
Vet Immunol Immunopathol ; 185: 57-65, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28242003

ABSTRACT

While T cell contribution to IAV immunity is appreciated, data comparing methods to evaluate IFN-γ production by IAV-specific T cells elicited following vaccination is limited. To understand the differential immunogenicity between live-attenuated influenza virus (LAIV) and whole-inactivated virus (WIV) vaccines in relation to induction of peripheral T cell responses, ELISpot and ELISA were used to assess IFN-γ production by peripheral lymphocytes following antigen restimulation. Following restimulation, peripheral blood lymphocytes from WIV-vaccinated pigs had a greater quantity of IFN-γ secreting cells (SC) and IFN-γ secreted compared to LAIV vaccinated and non-vaccinated (NV) pigs. Pig age at time of WIV vaccination significantly impacted peripheral IAV-specific IFN-γ recall response, as did the inclusion of adjuvant in the WIV vaccine. Collectively, these data indicate that peripheral IAV-specific IFN-γ recall responses are not predictive of LAIV vaccination status, thus, are unlikely to be a useful surrogate for evaluating LAIV immunogenicity and predicting cross-protection. However, these data suggest that the evaluation of peripheral IFN-γ recall responses may be useful for identifying factors, such as animal age or vaccine formulation, that may impact parenterally-delivered WIV vaccine immunogenicity. Overall, results did not differ based upon the assay used to evaluate IFN-γ recall responses. Therefore, either ELISpot or ELISA could serve as a measure for evaluating IAV-specific IFN-γ cell-mediated immune responses in swine.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines/immunology , Interferon-gamma/metabolism , Animals , Dogs , Enzyme-Linked Immunosorbent Assay/veterinary , Immunity, Cellular , Immunogenicity, Vaccine , Madin Darby Canine Kidney Cells , Swine , Vaccines, Attenuated/immunology , Vaccines, Inactivated/immunology
11.
Virology ; 491: 79-88, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26874588

ABSTRACT

Control of influenza A virus (IAV) in pigs is done by vaccination of females to provide maternally-derived antibodies (MDA) through colostrum. Our aim was to evaluate if MDA interfere with IAV infection, clinical disease, and transmission in non-vaccinated piglets. In the first study, naïve sows were vaccinated with H1N2-δ1 whole inactivated virus (WIV) vaccine. In a follow-up study seropositive sows to 2009 pandemic H1N1 (H1N1pdm09) were boosted with H1N1pdm09 WIV or secondary experimental infection (EXP). MDA-positive pigs were challenged with homologous or heterologous virus, and MDA-negative control groups were included. WIV-MDA piglets were protected from homologous infection. However, piglets with WIV-derived MDA subsequently challenged with heterologous virus developed vaccine associated enhanced respiratory disease (VAERD), regardless of history of natural exposure in the sows. Our data indicates that although high titers of vaccine-derived MDA reduced homologous virus infection, transmission, and disease, MDA alone was sufficient to induce VAERD upon heterologous infection.


Subject(s)
Antibodies, Viral/immunology , Immunity, Maternally-Acquired , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H1N2 Subtype/physiology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Respiratory Tract Infections/veterinary , Swine Diseases/immunology , Animals , Female , Follow-Up Studies , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N2 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Swine , Swine Diseases/virology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
12.
Vaccine ; 34(33): 3773-9, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27325350

ABSTRACT

The neuraminidase (NA) protein of influenza A viruses (IAV) has important functional roles in the viral replication cycle. Antibodies specific to NA can reduce viral replication and limit disease severity, but are not routinely measured. We analyzed NA inhibiting (NI) antibody titers in serum and respiratory specimens of pigs vaccinated with intramuscular whole-inactivated virus (WIV), intranasal live-attenuated influenza virus (LAIV), and intranasal wild type (WT) IAV. NI titers were also analyzed in sera from an investigation of piglet vaccination in the presence of passive maternally-derived antibodies. Test antigens contained genetically divergent swine-lineage NA genes homologous or heterologous to the vaccines with mismatched hemagglutinin genes (HA). Naïve piglets responded to WIV and LAIV vaccines and WT infection with strong homologous serum NI titers. Cross-reactivity to heterologous NAs depended on the degree of genetic divergence between the NA genes. Bronchoalveolar lavage specimens of LAIV and WT-immunized groups also had significant NI titers against the homologous antigen whereas the WIV group did not. Piglets of vaccinated sows received high levels of passive NI antibody, but their NI responses to homologous LAIV vaccination were impeded. These data demonstrate the utility of the enzyme-linked lectin assay for efficient NI antibody titration of serum as well as respiratory tract secretions. Swine IAV vaccines that induce robust NI responses are likely to provide broader protection against the diverse and rapidly evolving IAV strains that circulate in pig populations. Mucosal antibodies to NA may be one of the protective immune mechanisms induced by LAIV vaccines.


Subject(s)
Antibodies, Viral/blood , Immunity, Maternally-Acquired , Influenza Vaccines/immunology , Neuraminidase/immunology , Orthomyxoviridae Infections/immunology , Swine Diseases/immunology , Animals , Antibody Formation , Cross Reactions , Female , Influenza A virus , Swine , Swine Diseases/virology , Vaccines, Attenuated/immunology
13.
Vaccines (Basel) ; 3(1): 22-73, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-26344946

ABSTRACT

Influenza A virus in swine (IAV-S) is one of the most important infectious disease agents of swine in North America. In addition to the economic burden of IAV-S to the swine industry, the zoonotic potential of IAV-S sometimes leads to serious public health concerns. Adjuvanted, inactivated vaccines have been licensed in the United States for over 20 years, and there is also widespread usage of autogenous/custom IAV-S vaccines. Vaccination induces neutralizing antibodies and protection against infection with very similar strains. However, IAV-S strains are so diverse and prone to mutation that these vaccines often have disappointing efficacy in the field. This scientific review was developed to help veterinarians and others to identify the best available IAV-S vaccine for a particular infected herd. We describe key principles of IAV-S structure and replication, protective immunity, currently available vaccines, and vaccine technologies that show promise for the future. We discuss strategies to optimize the use of available IAV-S vaccines, based on information gathered from modern diagnostics and surveillance programs. Improvements in IAV-S immunization strategies, in both the short term and long term, will benefit swine health and productivity and potentially reduce risks to public health.

14.
Viral Immunol ; 17(1): 39-49, 2004.
Article in English | MEDLINE | ID: mdl-15018661

ABSTRACT

Modified vaccinia virus Ankara (MVA) was used as a vector to express genes from bovine respiratory syncytial virus (BRSV). Using these recombinant viruses as recall antigens for cells from BRSV-immuned cattle proved to be problematic because non-recombinant MVA itself frequently stimulated high levels of T lymphocyte activation. This phenomenon was observed in a high percentage of cattle from multiple herds. Gamma delta TCR(+) T cells were more sensitive to activation by MVA than other classes of T cells. A serological assay for MVA neutralization detected low, fluctuating titers of serum virus neutralizing (SVN) activity toward MVA in some cattle, but these were lower titers than those observed in cattle that underwent MVA vaccination. T cell reactivity in non-vaccinated cattle did not correlate significantly (p > 0.05) with SVN activity, undermining the notion that any adaptive immune response was responsible for the observed T cell sensitivity. More probable explanations are that MVA has mitogenic or superantigenic properties, or that the virus induces gammadelta TCR(+) T cell activation through interactions with innate pattern recognition receptors.


Subject(s)
Lymphocyte Activation , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/immunology , Vaccinia virus/immunology , Animals , Antigens, Viral/immunology , Cattle , Cells, Cultured , Chick Embryo , Fibroblasts/virology , Genetic Vectors , Receptors, Interleukin-2/metabolism , Respiratory Syncytial Virus, Bovine/genetics , Respiratory Syncytial Virus, Bovine/immunology , Vaccinia virus/genetics , Vaccinia virus/physiology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
15.
Viral Immunol ; 17(1): 13-23, 2004.
Article in English | MEDLINE | ID: mdl-15018659

ABSTRACT

Passive antibody to bovine viral diarrhea virus (BVDV) acquired through colostrum intake may interfere with the development of a protective immune response by calves to this virus. The objective of this study was to determine if calves, with a high level of maternal antibody to bovine viral diarrhea virus (BVDV), develop CD4(+), CD8(+), or gammadelta T lymphocyte responses to BVDV in the absence of a measurable humoral immune response. Colostrum or milk replacer fed calves were challenged with virulent BVDV at 2-5 weeks of age and/or after maternal antibody had waned. Calves exposed to BVDV while passive antibody levels were high did not mount a measurable humoral immune response to BVDV. However, compared to nonexposed animals, these animals had CD4(+), CD8(+), and gammadelta T lymphocytes that were activated by BVDV after exposure to in vitro BVDV. The production of IFNgamma by lymphocytes after in vitro BVDV exposure was also much greater in lymphocytes from calves exposed to BVDV in the presence of maternal antibody compared to the nonexposed calves. These data indicate that calves exposed to BVDV while maternal antibody levels are high can develop antigen specific CD4(+), CD8(+), and gammadelta T lymphocytes in the absence of an active antibody response. A manuscript presented separately demonstrates that the calves with T lymphocytes specific for BVDV in this study were also protected from virulent BVDV genotype 2 challenge after maternal antibody became undetectable.


Subject(s)
Bovine Virus Diarrhea-Mucosal Disease/immunology , Diarrhea Viruses, Bovine Viral/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Bovine Virus Diarrhea-Mucosal Disease/prevention & control , Bovine Virus Diarrhea-Mucosal Disease/virology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cattle , Colostrum , Female , Immunity, Maternally-Acquired , Interferon-gamma/metabolism , Male , Receptors, Antigen, T-Cell, gamma-delta/metabolism
16.
Vet Immunol Immunopathol ; 95(3-4): 123-33, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12963273

ABSTRACT

T cell activity is a critical component of immunity to bovine respiratory syncytial virus (BRSV). We tested the effects of immunization by modified-live and inactivated BRSV vaccines on cell-mediated and humoral immunity in young calves. The two forms of vaccine stimulated similar serum neutralizing antibody production, although the early kinetics of those responses differed. CD4+, CD8+, and gammadelta T cells were analyzed before and after immunization for BRSV-specific in vitro recall responses, as evaluated by CD25 upregulation measured by flow cytometry. Modified-live virus (MLV) primed each of the three subsets for statistically significant in vitro responses to antigen. Inactivated vaccine also primed each T cell population for significant antigen-driven CD25 upregulation, including responses by CD4+ and gammadelta T cells that were stronger and longer-lasting than those primed by MLV. Monoclonal antibody was used in additional assays to block MHC class I during incubation of BRSV antigen with peripheral blood mononuclear cells from an animal in the inactivated vaccine group. The recall response by CD8+ T cells was more inhibited by this treatment than the other subsets, further suggesting that the inactivated vaccine had primed antigen-specific CD8+ T cells. In summary, the data indicate that balanced BRSV-specific T cell responses can be induced by inactivated, as well as modified-live, conventional vaccines, which may implicate an alternative pathway of MHC class I antigen presentation.


Subject(s)
Cattle Diseases/immunology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus, Bovine/immunology , T-Lymphocyte Subsets/immunology , Viral Vaccines/immunology , Animals , Cattle , Cattle Diseases/virology , Flow Cytometry/veterinary , Histocompatibility Antigens Class I/immunology , Immunization/veterinary , Lymphocyte Activation/immunology , Male , Neutralization Tests/veterinary , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , T-Lymphocyte Subsets/virology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/therapeutic use , Vaccines, Inactivated/immunology , Vaccines, Inactivated/therapeutic use , Viral Vaccines/therapeutic use
17.
Vet Immunol Immunopathol ; 84(1-2): 111-23, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11825602

ABSTRACT

Calves lacking detectable serum antibodies against bovine respiratory syncytial virus (BRSV) were screened for virus-specific T-cell memory. Peripheral blood mononuclear cells were cultured in vitro with live BRSV and analyzed by dual-color flow cytometry for surface expression of CD25 on CD4(+), CD8(+), and gammadeltaT-cells. Significant recall responses were detected in some of the seronegative calves. Modified live BRSV vaccine was administered to these and to a group of non-responding calves. Following vaccination, virus-specific IgG, virus neutralizing antibody, and T-cell recall responses were all elevated more rapidly in the group with BRSV-sensitive T-cells than in the T-cell-negative group, which suggested that calves in the first group were previously exposed to BRSV. This demonstrates that exposure to BRSV can induce T and B cell memory in young calves without causing seroconversion. The calves were presumably exposed to BRSV while they had maternal antibody, which inhibited the calves from developing an antibody response.


Subject(s)
Respiratory Syncytial Virus, Bovine/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Viral/blood , Cattle , Cells, Cultured , Immunization , Viral Vaccines/immunology
18.
Methods Mol Biol ; 1161: 337-45, 2014.
Article in English | MEDLINE | ID: mdl-24899442

ABSTRACT

Neuraminidase (NA) is an envelope glycoprotein of influenza viruses, including swine-lineage influenza A viruses. NA possesses sialidase activity, which is functionally important at multiple points in viral replication, counter-balancing the sialic acid receptor binding activity of the hemagglutinin (HA), the other major envelope glycoprotein. The NA proteins of influenza A viruses have been classified into nine serological subtypes, and they undergo antigenic drift variation similar to that of HA. Antibodies to NA are analyzed much less often than antibodies to HA. The conventional assay for NA inhibition (NI) antibody titration, established decades ago, is widely considered unwieldy and inefficient for routine use. In recent years, a few new formats have been developed which still measure inhibition of NA enzymatic function, but more efficiently and with less chemical waste produced. Described here is the enzyme-linked lectin assay (ELLA), which is performed in 96-well plates and analyzed on a spectrophotometric plate reader. An important factor in adoption of the ELLA technique for animal studies, such as swine, is the choice of NA antigen, which may be purified protein or whole virus containing an antigenically irrelevant HA protein. This NI assay, in conjunction with the hemagglutination inhibiting (HI) antibody assay, offers a practical way to characterize viral isolates more fully and to quantify antibodies induced by infection or vaccination.


Subject(s)
Antibodies, Viral/blood , Antibodies, Viral/immunology , Immunoassay/methods , Influenza A virus/enzymology , Lectins/immunology , Neuraminidase/immunology , Swine , Animals , Lectins/metabolism
19.
Virology ; 464-465: 45-54, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25043588

ABSTRACT

Live-attenuated influenza virus (LAIV) prime-boost vaccination previously conferred protection against heterologous H3N2 swine influenza challenge, including in piglets with maternally derived antibodies (MDA). Conversely, a whole-inactivated virus (WIV) vaccine was associated with enhanced disease. This study was aimed at identifying immune correlates of cross-protection. Piglets with and without MDA received intramuscular adjuvanted WIV or intranasal LAIV, and were challenged with heterologous H3N2. WIV induced cross-reactive IgG, inhibited by MDA, and a moderate T cell response. LAIV elicited mucosal antibodies and T cells cross-reactive to the heterologous challenge strain. The presence of MDA at LAIV vaccination blocked lung and nasal antibody production, but did not interfere with T cell priming. Even without mucosal antibodies, MDA-positive LAIV vaccinates were protected, indicating a likely role for T cells. Based on the data, one LAIV dose can induce cell-mediated immunity against antigenically divergent H3N2 influenza virus despite passive antibody interference with humoral immune responses.


Subject(s)
Immunity, Maternally-Acquired , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Swine Diseases/immunology , Animals , Antibodies, Viral/immunology , Cross Protection , Female , Immunization , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Swine , Swine Diseases/prevention & control , Swine Diseases/virology , T-Lymphocytes/immunology
20.
Vaccine ; 30(2): 280-8, 2012 Jan 05.
Article in English | MEDLINE | ID: mdl-22067263

ABSTRACT

The diversity of contemporary swine influenza virus (SIV) strains impedes effective immunization of swine herds. Mucosally delivered, attenuated virus vaccines are one approach with potential to provide broad cross-protection. Reverse genetics-derived H3N2 SIV virus with truncated NS1 (NS1Δ126 TX98) is attenuated and immunogenic when delivered intranasally in young pigs. We analyzed T-cell priming and cross-protective efficacy in weanling piglets after intranasal inoculation with NS1Δ126 TX98 versus wild type TX98. In vivo replication of the truncation mutant was minimal compared to the wild type virus. T-cell responses were greater in magnitude in pigs infected with the wild type virus in in vitro restimulation assays. According to the expression of activation marker CD25, peripheral T cell recall responses in NS1Δ126 TX98 infected pigs were minimal. However, intracellular IFN-γ data indicate that the attenuated virus induced virus-specific CD4(+)CD8(-), CD4(+)CD8(+), CD4(-)CD8(+), and γδ T cells within 28 days. The IFN-γ response appeared to contract, as responses were reduced at later time points prior to challenge. CD4(+)CD8(+) cells isolated 5 days after heterosubtypic H1N1 challenge (day 70 overall) showed an elevated CD25 response to virus restimulation. Pigs previously infected with wild type TX98 were protected from replication of the H1N1 challenge virus. Vaccination with NS1Δ126 TX98 was associated with significantly lower levels of Th1-associated cytokines in infected lungs but provided partial cross-protection against the H1N1 challenge. These results demonstrate that NS1Δ SIV vaccines can elicit cell-mediated cross-protection against antigenically divergent strains.


Subject(s)
Cross Protection , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , T-Lymphocytes/immunology , Viral Nonstructural Proteins/genetics , Animals , Animals, Newborn , Disease Models, Animal , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Orthomyxoviridae Infections/immunology , Swine , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology
SELECTION OF CITATIONS
SEARCH DETAIL