Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
PLoS Negl Trop Dis ; 9(10): e0004104, 2015.
Article in English | MEDLINE | ID: mdl-26496497

ABSTRACT

Mayaro virus (MAYV), an alphavirus similar to chikungunya virus (CHIKV), causes an acute debilitating disease which results in the development of long-term arthralgia in more than 50% of infected individuals. Currently, the immune response and its role in the development of MAYV-induced persistent arthralgia remain unknown. In this study, we evaluated the immune response of individuals with confirmed MAYV infection in a one-year longitudinal study carried out in Loreto, Peru. We report that MAYV infection elicits robust immune responses that result in the development of a strong neutralizing antibody response and the secretion of pro-inflammatory immune mediators. The composition of these inflammatory mediators, in some cases, differed to those previously observed for CHIKV. Key mediators such as IL-13, IL-7 and VEGF were strongly induced following MAYV infection and were significantly increased in subjects that eventually developed persistent arthralgia. Although a strong neutralizing antibody response was observed in all subjects, it was not sufficient to prevent the long-term outcomes of MAYV infection. This study provides initial immunologic insight that may eventually contribute to prognostic tools and therapeutic treatments against this emerging pathogen.


Subject(s)
Alphavirus Infections/complications , Alphavirus Infections/pathology , Alphavirus/immunology , Arthralgia/pathology , Cytokines/blood , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Arthralgia/immunology , Child , Female , Humans , Longitudinal Studies , Male , Middle Aged , Peru , Young Adult
2.
Vaccine ; 33(30): 3562-70, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26044496

ABSTRACT

Avian-derived influenza A zoonoses are closely monitored and may be an indication of virus strains with pandemic potential. Both successful vaccination and convalescence of influenza A virus in humans typically results in the induction of antibodies that can neutralize viral infection. To improve long-standing and new-generation methodologies for detection of neutralizing antibodies, we have employed a novel reporter-based approach that allows for multiple antigenic testing within a single sample. Central to this approach is a single-cycle infectious influenza A virus (sciIAV), where a functional hemagglutinin (HA) gene was changed to encode either the green or the monomeric red fluorescent protein (GFP and mRFP, respectively) and HA is complemented in trans by stable HA-expressing cell lines. By using fluorescent proteins with non-overlapping emission spectra, this novel bivalent fluorescence-based microneutralization assay (BiFMA) can be used to detect neutralizing antibodies against two distinct influenza isolates in a single reaction, doubling the speed of experimentation while halving the amount of sera required. Moreover, this approach can be used for the rapid identification of influenza broadly neutralizing antibodies. Importantly, this novel BiFMA can be used for any given influenza HA-pseudotyped virus under BSL-2 facilities, including highly pathogenic influenza HA isolates.


Subject(s)
Antibodies, Neutralizing/blood , Fluorometry/methods , Influenza A virus/immunology , Neutralization Tests/methods , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics
3.
Emerg Infect Dis ; 21(5): 781-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25898901

ABSTRACT

Our genetic analyses of uncharacterized bunyaviruses isolated in Peru identified a possible reassortant virus containing small and large gene segment sequences closely related to the Caraparu virus and a medium gene segment sequence potentially derived from an unidentified group C orthobunyavirus. Neutralization tests confirmed serologic distinction among the newly identified virus and the prototype and Caraparu strains. This virus, named Itaya, was isolated in 1999 and 2006 from febrile patients in the cities of Iquitos and Yurimaguas in Peru. The geographic distance between the 2 cases suggests that the Itaya virus could be widely distributed throughout the Amazon basin in northeastern Peru. Identification of a new Orthobunyavirus species that causes febrile disease in humans reinforces the need to expand viral disease surveillance in tropical regions of South America.


Subject(s)
Bunyaviridae Infections/epidemiology , Bunyaviridae Infections/virology , Fever/epidemiology , Fever/virology , Orthobunyavirus/classification , Adult , Animals , Cell Line , Geography , Humans , Male , Neutralization Tests , Orthobunyavirus/genetics , Orthobunyavirus/isolation & purification , Peru/epidemiology , Phylogeny , Population Surveillance , RNA, Viral , Reassortant Viruses , Serotyping
4.
J Virol ; 88(8): 4572-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478431

ABSTRACT

UNLABELLED: Recognition of viral pathogens by the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) family results in the activation of type I interferon (IFN) responses. To avoid this response, most viruses have evolved strategies that target different essential steps in the activation of host innate immunity. In this study, we report that the nonstructural protein NSs of the newly described severe fever with thrombocytopenia syndrome virus (SFTSV) is a potent inhibitor of IFN responses. The SFTSV NSs protein was found to inhibit the activation of the beta interferon (IFN-ß) promoter induced by viral infection and by a RIG-I ligand. Astonishingly, we found that SFTSV NSs interacts with and relocalizes RIG-I, the E3 ubiquitin ligase TRIM25, and TANK-binding kinase 1 (TBK1) into SFTSV NSs-induced cytoplasmic structures. Interestingly, formation of these SFTSV NSs-induced structures occurred in the absence of the Atg7 gene, a gene essential for autophagy. Furthermore, confocal microscopy studies revealed that these SFTSV NSs-induced structures colocalize with Rab5 but not with Golgi apparatus or endoplasmic reticulum markers. Altogether, the data suggest that sequestration of RIG-I signaling molecules into endosome-like structures may be the mechanism used by SFTSV to inhibit IFN responses and point toward a novel mechanism for the suppression of IFN responses. IMPORTANCE: The mechanism by which the newly described SFTSV inhibits host antiviral responses has not yet been fully characterized. In this study, we describe the redistribution of RIG-I signaling components into virus-induced cytoplasmic structures in cells infected with SFTSV. This redistribution correlates with the inhibition of host antiviral responses. Further characterization of the interplay between the viral protein and components of the IFN responses could potentially provide targets for the rational development of therapeutic interventions.


Subject(s)
Bunyaviridae Infections/enzymology , DEAD-box RNA Helicases/metabolism , Endosomes/metabolism , Interferon Type I/immunology , Phlebovirus/metabolism , Viral Nonstructural Proteins/metabolism , Bunyaviridae Infections/genetics , Bunyaviridae Infections/immunology , Bunyaviridae Infections/virology , Cell Line , Cytoplasmic Structures , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Endosomes/genetics , Humans , Interferon Type I/genetics , Phlebovirus/genetics , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Receptors, Immunologic , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Viral Nonstructural Proteins/genetics
5.
Biomaterials ; 34(34): 8776-85, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23953841

ABSTRACT

Balancing immunogenicity with inflammation is a central tenet of vaccine design, especially for subunit vaccines that utilize traditional pro-inflammatory adjuvants. Here we report that by using a nanoparticulate peptide-based vaccine, immunogenicity and local inflammation could be decoupled. Self-assembled ß-sheet-rich peptide nanofibers, previously shown to elicit potent antibody responses in mice, were found to be non-cytotoxic in vitro and, remarkably, elicited no measurable inflammation in vivo-with none of the swelling at the injection site, accumulation of inflammatory cells or cytokines, or production of allergic IgE that were elicited by an alum-adjuvanted vaccine. Nanofibers were internalized by dendritic cells and macrophages at the injection site, and only dendritic cells that acquired the material increased their expression of the activation markers CD80 and CD86. Immunization with epitope-bearing nanofibers elicited antigen-specific differentiation of T cells into T follicular helper cells and B cells into germinal center cells, as well as high-titer, high-affinity IgG that cross-reacted with the native protein antigen and was neutralizing in an in vitro influenza hemagglutination inhibition assay. These responses were superior to those induced by alum and comparable to those induced by complete Freund's adjuvant. Thus, nanoparticulate assemblies may provide a new route to non-inflammatory immunotherapies and vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibody Formation/drug effects , B-Lymphocytes/immunology , Nanofibers/chemistry , Peptides/immunology , Vaccines, Subunit/chemistry , Alum Compounds/chemistry , Animals , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Biocompatible Materials/chemistry , Cytokines/metabolism , Dendritic Cells/immunology , Epitopes/immunology , Inflammation/immunology , Mice , Mice, Inbred C57BL , Peptides/chemistry , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptor 4/metabolism , Vaccines, Subunit/immunology
6.
Clin Vaccine Immunol ; 20(6): 867-76, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23576673

ABSTRACT

The 2009 pandemic H1N1 (pH1N1) influenza virus carried a swine-origin hemagglutinin (HA) that was closely related to the HAs of pre-1947 H1N1 viruses but highly divergent from the HAs of recently circulating H1N1 strains. Consequently, prior exposure to pH1N1-like viruses was mostly limited to individuals over the age of about 60 years. We related age and associated differences in immune history to the B cell response to an inactivated monovalent pH1N1 vaccine given intramuscularly to subjects in three age cohorts: 18 to 32 years, 60 to 69 years, and ≥70 years. The day 0 pH1N1-specific hemagglutination inhibition (HAI) and microneutralization (MN) titers were generally higher in the older cohorts, consistent with greater prevaccination exposure to pH1N1-like viruses. Most subjects in each cohort responded well to vaccination, with early formation of circulating virus-specific antibody (Ab)-secreting cells and ≥4-fold increases in HAI and MN titers. However, the response was strongest in the 18- to 32-year cohort. Circulating levels of HA stalk-reactive Abs were increased after vaccination, especially in the 18- to 32-year cohort, raising the possibility of elevated levels of cross-reactive neutralizing Abs. In the young cohort, an increase in MN activity against the seasonal influenza virus A/Brisbane/59/07 after vaccination was generally associated with an increase in the anti-Brisbane/59/07 HAI titer, suggesting an effect mediated primarily by HA head-reactive rather than stalk-reactive Abs. Our findings support recent proposals that immunization with a relatively novel HA favors the induction of Abs against conserved epitopes. They also emphasize the need to clarify how the level of circulating stalk-reactive Abs relates to resistance to influenza.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Animals , Cohort Studies , Female , Hemagglutination Inhibition Tests , Humans , Influenza Vaccines/administration & dosage , Injections, Intramuscular , Male , Middle Aged , Neutralization Tests , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Young Adult
7.
Vaccine ; 30(31): 4606-16, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22609035

ABSTRACT

Antibodies directed against the influenza hemagglutinin (HA) protein largely mediate virus neutralization and confer protection against infection. Consequently, many studies and assays of influenza vaccines are focused on HA-specific immune responses. Recombinant HA (rHA) proteins can be produced in a number of protein expression and cell culture systems. These range from baculovirus infection of insect cell cultures, to transient transfection of plants, to stably transfected human cell lines. Furthermore, the rHA proteins may contain genetic modifications, such as histidine tags or trimerization domains, intended to ease purification or enhance protein stability. However, no systematic study of these different forms of the HA protein have been conducted. It is not clear which, if any, of these different protein expression systems or structural modifications improve or diminish the biological behavior of the proteins as immunogens or antigens in immune assays. Therefore we set out to perform systematic evaluation of rHA produced in different proteins expression systems and with varied modifications. Five rHA proteins based on recent strains of seasonal influenza A and five based on influenza B HA were kindly provided by the Biodefense and Emerging Infections Reagent Repository (BEIR). These proteins were evaluated in a combination of biochemical and structural assays, in vitro humoral and cellular immune assays, and in an animal vaccination model. Marked differences in the behavior of the individual proteins was evident suggesting that they are not equal when being used to detect an immune response. They were, nevertheless, similar at eliciting neutralizing antibody responses.


Subject(s)
Antigens, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza B virus/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antigens, Viral/biosynthesis , Antigens, Viral/genetics , Female , Hemagglutination Inhibition Tests , Hemagglutination Tests , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza Vaccines/immunology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology
8.
Vaccine ; 29(48): 8888-97, 2011 Nov 08.
Article in English | MEDLINE | ID: mdl-21963871

ABSTRACT

Substantial H5 influenza HA directed immunity is elicited after vaccination of human subjects who had been previously immunized with a drifted H5 HA variant. We sought to investigate the characteristics of H5 HA specific immune responses in more depth by developing an animal model of H5 HA vaccination using drift variants of recombinant H5 HA proteins. HA proteins derived from influenzas A/Vietnam/1203/04 (Clade 1) and A/Indonesia/05/05 (Clade 2.1) were chosen. The sequence of vaccination consisted of two doses of homologous protein, followed by one additional dose of the homologous or heterologous, drifted HA protein. Each dose of HA was combined with CpG as an adjuvant and was injected subcutaneously. All the animals exhibited a serum IgG antibody response that cross-reacted with both HAs in an ELISA. However, those animals that received the drifted variant exhibited higher reactivity to the heterologous HA. Competitive ELISA of serum from drift-variant recipients showed evidence of antibody focusing towards the drifted HA, suggesting modification of the response towards improved cross-reactivity, though development of neutralizing antibodies was limited. Nevertheless, animals were protected against live-virus challenge, and passive transfer of serum was sufficient to confer protection to otherwise naïve mice, indicating that both neutralizing and non-neutralizing antibodies offer some degree of protection. These findings suggest that pre-vaccination against H5 influenza has the potential to prime immunity against emerging drifted H5 strains, and could also lower the dose requirements of vaccination in the event of a pandemic.


Subject(s)
Cross Protection , Genetic Drift , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody Formation , Cross Reactions , Disease Models, Animal , Immunoglobulin G/blood , Immunoglobulin G/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Male , Mice , Mice, Inbred C57BL , Neutralization Tests , Orthomyxoviridae Infections/immunology
9.
J Virol ; 85(1): 448-55, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980523

ABSTRACT

Genetic mutation and reassortment of influenza virus gene segments, in particular those of hemagglutinin (HA) and neuraminidase (NA), that lead to antigenic drift and shift are the major strategies for influenza virus to escape preexisting immunity. The most recent example of such phenomena is the first pandemic of H1N1 influenza of the 21st century, which started in 2009. Cross-reactive antibodies raised against H1N1 viruses circulating before 1930 show protective activity against the 2009 pandemic virus. Cross-reactive T-cell responses can also contribute to protection, but in vivo support of this view is lacking. To explore the protection mechanisms in vivo, we primed mice with H1 and H3 influenza virus isolates and rechallenged them with a virus derived from the 2009 H1N1 A/CA/04/09 virus, named CA/E3/09. We found that priming with influenza viruses of both H1 and H3 homo- and heterosubtypes protected against lethal CA/E3/09 virus challenge. Convalescent-phase sera from these primed mice conferred no neutralization activity in vitro and no protection in vivo. However, T-cell depletion studies suggested that both CD4 and CD8 T cells contributed to the protection. Taken together, these results indicate that cross-reactive T cells established after initial priming with distally related viruses can be a vital component for prevention of disease and control of pandemic H1N1 influenza virus infection. Our results highlight the importance of establishing cross-reactive T-cell responses for protecting against existing or newly emerging pandemic influenza viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype/classification , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , T-Lymphocytes/immunology , Animals , Antibodies, Viral/blood , Cross Reactions , Disease Models, Animal , Female , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/virology , Mice , Mice, Inbred C57BL , Pandemics , Virulence
10.
Vaccine ; 28(4): 907-15, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19932673

ABSTRACT

B cell responses after immunization with a drifted H5 influenza/A/Vietnam/1203/04 vaccine were characterized in the peripheral blood of human subjects primed with experimental recombinant H5 influenza A/Hong Kong/156/97 vaccine. Antibody secreting cells were assayed by ELISPOT against a panel of recombinant hemagglutinin and control proteins. Increased frequencies of H5 HA-specific antibody secreting and memory B cells could be observed within 7 days of re-vaccination. Furthermore, these responses were cross-reactive to both H5 HA variants, but not H3 or avian H6 HA strains. These observations suggest prior vaccination against H5 influenza HA induces cellular immune responses that cross-react among drifted variants, without precluding a response to new, or existing HA strains.


Subject(s)
B-Lymphocytes/immunology , Blood/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Lymphocyte Subsets/immunology , Adult , Cross Reactions , Enzyme-Linked Immunosorbent Assay/methods , Female , Hong Kong , Humans , Immunologic Memory , Male , Middle Aged
11.
Brain Behav Immun ; 21(7): 872-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17681745

ABSTRACT

During the period from 1987 to 2007, our understanding of nervous system-T lymphocyte bi-directional communication advanced exponentially. Progress in exploring these relationships was aided by the constant development of new, cutting-edge technologies, and by a steady growth in interest, and number, of scientists who recognized the need to conduct cross-disciplinary research. In this brief review of 20 years of Brain, Behavior, and Immunity (BBI), we highlight just a small number of the important studies published in the journal that collectively have provided the foundation for our current understanding of brain, behavior, and, specifically, T cells.


Subject(s)
Antibody Formation/immunology , Brain/immunology , Neuroimmunomodulation/immunology , T-Lymphocytes/immunology , Animals , Humans
12.
Proc Natl Acad Sci U S A ; 100(26): 15953-8, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14671329

ABSTRACT

Neurological symptoms are common manifestations of Lyme disease; however, the paucibacillary nature of the spirochete in this environment has precluded a molecular analysis of the spirochete in the CNS. We have now adapted differential expression analysis by using a custom-amplified library (DECAL) in conjunction with Borrelia burgdorferi whole-genome and subgenome arrays to examine in vivo gene expression by B. burgdorferi in a non-human primate (NHP) model of neuroborreliosis. The expression profile of B. burgdorferi was examined in the CNS and heart of steroid-treated and immunocompetent NHPs. Eighty-six chromosomal genes and 80 plasmid-encoded genes were expressed at similar levels in the CNS and heart tissue of both immunocompetent and steroid-treated NHPs. The expression of 66 chromosomal genes and 32 plasmid-encoded genes was increased in the CNS of both immunocompetent and steroid-treated NHPs. It is likely that the expression of these genes is governed by physiological factors specific to the CNS milieu. However, 83 chromosomal and 114 plasmid-encoded genes showed contrasting expression profiles in steroid-treated and immunocompetent NHPs. The effect of dexamethasone on the immune status of the host as well as on the host metabolic pathways could contribute to these differences in the B. burgdorferi transcriptome. Results obtained herein underscore the complex interplay of host factors on B. burgdorferi gene expression in vivo. The results provide a global snapshot of the spirochetal transcriptome in the CNS and should spur the design of experiments aimed at understanding the molecular basis of neuroborreliosis.


Subject(s)
Borrelia burgdorferi/genetics , Brain/microbiology , Gene Expression Regulation, Bacterial/genetics , Genome, Bacterial , Medulla Oblongata/microbiology , Transcription, Genetic , Animals , Borrelia burgdorferi/isolation & purification , Gene Library , Heart/microbiology , Macaca mulatta , Oligonucleotide Array Sequence Analysis , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
13.
J Bacteriol ; 184(11): 3122-5, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12003955

ABSTRACT

Borrelia burgdorferi gene expression within the guts of engorging Ixodes scapularis ticks was examined by use of differential immunoscreening and differential expression with a customized amplified library. Fourteen chromosomal genes involved in energy metabolism, substrate transport, and signal transduction and 10 (4 chromosomal and 6 plasmid) genes encoding putative lipoproteins and periplasmic proteins were preferentially expressed in engorging ticks. These data demonstrate a new approach to the global analysis of B. burgdorferi genes that are preferentially expressed within the vector during feeding.


Subject(s)
Arachnid Vectors/microbiology , Bacterial Proteins/genetics , Borrelia burgdorferi/genetics , Ixodes/microbiology , Animals , Bacterial Proteins/analysis , Blotting, Southern , Borrelia burgdorferi/metabolism , Feeding Behavior , Gene Amplification , Gene Expression , Genomic Library , Ixodes/physiology , Mice , Mice, Inbred C3H , RNA, Bacterial/analysis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...