Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Immunol ; 189(6): 3150-8, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22896629

ABSTRACT

Semaphorin 7A (Sema7A) is a membrane-associated/secreted protein that plays an essential role in connecting the vertebrate neuronal and immune systems. However, the role of Sema7A has not been elucidated in viral pathogenesis. In this study, we show that abrogation of Sema7A protects mice from lethal West Nile virus (WNV) infection. Mice lacking Sema7A showed increased survival, reduced viral burden, and less blood-brain barrier permeability upon WNV infection. Increased Sema7A levels were evident in murine tissues, as well as in murine cortical neurons and primary human macrophages upon WNV infection. Treatment with Sema7A Ab blocked WNV infection in both of these cell types. Furthermore, Sema7A positively regulates the production of TGF-ß1 and Smad6 to facilitate WNV pathogenesis in mice. Collectively, these data elucidate the role of Sema7A in shared signaling pathways used by the immune and nervous systems during viral pathogenesis that may lead to the development of Sema7A-blocking therapies for WNV and possibly other flaviviral infections.


Subject(s)
Antigens, CD/physiology , Semaphorins/physiology , Signal Transduction/immunology , Smad6 Protein/physiology , Transforming Growth Factor beta1/physiology , West Nile virus/immunology , West Nile virus/pathogenicity , Animals , Cell Line , Cells, Cultured , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/virology , Disease Models, Animal , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Virus Replication/immunology
2.
J Immunol ; 183(1): 650-60, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19535627

ABSTRACT

West Nile virus is an emerging pathogen that can cause fatal neurological disease. A recombinant human mAb, mAb11, has been described as a candidate for the prevention and treatment of West Nile disease. Using a yeast surface display epitope mapping assay and neutralization escape mutant, we show that mAb11 recognizes the fusion loop, at the distal end of domain II of the West Nile virus envelope protein. Ab mAb11 cross-reacts with all four dengue viruses and provides protection against dengue (serotypes 2 and 4) viruses. In contrast to the parental West Nile virus, a neutralization escape variant failed to cause lethal encephalitis (at higher infectious doses) or induce the inflammatory responses associated with blood-brain barrier permeability in mice, suggesting an important role for the fusion loop in viral pathogenesis. Our data demonstrate that an intact West Nile virus fusion loop is critical for virulence, and that human mAb11 targeting this region is efficacious against West Nile virus infection. These experiments define the molecular determinant on the envelope protein recognized by mAb11 and demonstrate the importance of this region in causing West Nile encephalitis.


Subject(s)
Antibodies, Monoclonal/metabolism , Binding Sites, Antibody , Peptides/immunology , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , West Nile Fever/immunology , West Nile virus/pathogenicity , Animals , Antibodies, Monoclonal/therapeutic use , Cell Line , Cross Reactions , Dengue Virus/immunology , Dengue Virus/pathogenicity , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/metabolism , West Nile Fever/therapy , West Nile Fever/virology , West Nile virus/immunology
3.
J Clin Invest ; 117(10): 3059-66, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17909632

ABSTRACT

The flavivirus West Nile virus (WNV) is an emerging pathogen that causes life-threatening encephalitis in susceptible individuals. We investigated the role of the proinflammatory cytokine macrophage migration inhibitory factor (MIF), which is an upstream mediator of innate immunity, in WNV immunopathogenesis. We found that patients suffering from acute WNV infection presented with increased MIF levels in plasma and in cerebrospinal fluid. MIF expression also was induced in WNV-infected mice. Remarkably, abrogation of MIF action by 3 distinct approaches (antibody blockade, small molecule pharmacologic inhibition, and genetic deletion) rendered mice more resistant to WNV lethality. Mif(-/-) mice showed a reduced viral load and inflammatory response in the brain when compared with wild-type mice. Our results also indicate that MIF favors viral neuroinvasion by compromising the integrity of the blood-brain barrier. In conclusion, the data obtained from this study provide direct evidence for the involvement of MIF in viral pathogenesis and suggest that pharmacotherapeutic approaches targeting MIF may hold promise for the treatment of WNV encephalitis.


Subject(s)
Brain/virology , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Virus Internalization , West Nile Fever/virology , West Nile virus/physiology , Animals , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Mutant Strains , Viral Load , West Nile Fever/genetics
4.
Vaccine ; 27(2): 213-22, 2009 Jan 07.
Article in English | MEDLINE | ID: mdl-18996430

ABSTRACT

In this study, a recombinant truncated West Nile virus envelope protein antigen (rWNV-E) was produced in serum-free cultures of the expresSF+ insect cell line via baculovirus infection. This production system was selected based on its use in the production of candidate human and animal vaccine antigens. A defined fermentation and purification process for the rWNV-E antigen was established to control for purity and immunogenicity of each protein batch. The material formulated with aluminum hydroxide was stable for greater than 8months at 4 degrees C. The recombinant vaccine candidate was evaluated for immunogenicity and protective efficacy in several animal models. In mouse and hamster WNV challenge models, the vaccine candidate induced viral protection that correlated with anti-rWNV-E immunogenicity and WNV neutralizing antibody titers. The rWNV-E vaccine candidate was used to boost horses previously immunized with the Fort Dodge inactivated WNV vaccine and also to induce WNV neutralizing titers in naïve foals that were at least 14weeks of age. Furthermore, the vaccine candidate was found safe when high doses were injected into rats, with no detectable treatment-related clinical adverse effects. These observations demonstrate that baculovirus-produced rWNV-E can be formulated with aluminum hydroxide to produce a stable and safe vaccine which induces humoral immunity that can protect against WNV infection.


Subject(s)
Recombinant Proteins/metabolism , Spodoptera/metabolism , Viral Envelope Proteins/metabolism , West Nile Fever/prevention & control , West Nile Virus Vaccines/metabolism , West Nile virus/immunology , Animals , Antibodies, Viral/blood , Baculoviridae/genetics , Baculoviridae/metabolism , Cells, Cultured , Cricetinae , Disease Models, Animal , Horse Diseases/immunology , Horse Diseases/prevention & control , Horse Diseases/virology , Horses , Humans , Mice , Rats , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spodoptera/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/immunology , West Nile virus/genetics
5.
Vaccine ; 27(23): 3013-21, 2009 May 18.
Article in English | MEDLINE | ID: mdl-19428913

ABSTRACT

Innate immune system activation is a critical step in the initiation of an effective adaptive immune response; therefore, activation of a class of innate pathogen receptors called pattern recognition receptors (PRR) is a central feature of many adjuvant systems. It has recently been shown that one member of an intracellular PRR, the NLRP3 inflammasome, is activated by a number of classical adjuvants including aluminum hydroxide and saponins [Eisenbarth SC, Colegio OR, O'Connor W, Sutterwala FS, Flavell RA. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature 2008;453(June (7198)):1122-6; Li H, Willingham SB, Ting JP, Re F. Cutting edge: inflammasome activation by alum and alum's adjuvant effect are mediated by NLRP3. J Immunol 2008;181(July (1)):17-21]. Inflammasome activation in vitro requires signaling of both the Toll-like receptor (TLR) and NLRP3 in antigen-presenting cells. Here we present a class of nanomaterials endowed with these two signals for rapid optimization of vaccine design. We constructed this system using a simple approach that incorporates lipopolysaccharides (LPS) onto the surface of nanoparticles constructed from a biocompatible polyester, poly(lactic-co-glycolic acid) (PLGA), loaded with antigen. We demonstrate that LPS-modified particles are preferentially internalized by dendritic cells compared to uncoated nanoparticles and the system, when administered to mice, elicits potent humoral and cellular immunity against a model antigen, ovalbumin. Wild-type macrophages pulsed with LPS-modified nanoparticles resulted in production of the proinflammatory cytokine IL-1beta consistent with inflammasome activation. In comparison, NLRP3-deficient and caspase-1-deficient macrophages showed negligible production of IL-1beta. Furthermore, when endocytosis and lysosomal destabilization were inhibited, inflammasome activity was diminished, supporting the notion that nanoparticles rupture lysosomal compartments and behave as 'danger signals' [Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol 2008;9(August (8)):847-56]. The generality of this vaccination approach is tested by encapsulation of a recombinant West Nile envelope protein and demonstrated by protection against a murine model of West Nile encephalitis. The design of such an antigen delivery mechanism with the ability to stimulate two potent innate immune pathways represents a potent new approach to simultaneous antigen and adjuvant delivery.


Subject(s)
Carrier Proteins/metabolism , Nanoparticles/therapeutic use , Vaccination/methods , West Nile Fever/prevention & control , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/therapeutic use , Animals , Antibody Formation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carrier Proteins/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lactic Acid/immunology , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile Fever/metabolism , West Nile Virus Vaccines/immunology , West Nile Virus Vaccines/therapeutic use
6.
J Infect Dis ; 196(12): 1741-8, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18190253

ABSTRACT

The flavivirus envelope (E) protein mediates cellular attachment and fusion with host cell membranes and is recognized by virus-neutralizing antibodies. We raised antibodies against a broad range of epitopes by immunizing a horse with recombinant West Nile virus (WNV) E protein. To define epitopes recognized by protective antibodies, we selected, by affinity chromatography, immunoglobulins against immobilized linear peptides derived from parts of the E protein. Immunoglobulins binding 9 different peptides from domains I, II, and III of the E protein neutralized WNV in vitro. This indicates that multiple protective epitopes can be found in the E protein. Immunoglobulins recognizing 3 peptides derived from domains I and II of E protein protected mice against a lethal challenge with WNV. These immunoglobulins recognized the E proteins of related flaviviruses, demonstrating that antibodies targeting specific E protein epitopes could be developed for prevention and treatment of multiple flavivirus infections.


Subject(s)
Antibodies, Viral/immunology , Immunization, Passive/methods , Immunoglobulins/immunology , Viral Envelope Proteins/immunology , West Nile Fever/prevention & control , West Nile virus/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/pharmacology , Antibody Affinity/immunology , Chlorocebus aethiops , Drosophila , Epitopes/immunology , Female , Fluorescent Antibody Technique/methods , Horses , Mice , Mice, Inbred C3H , Molecular Sequence Data , Peptide Fragments/immunology , Vero Cells , West Nile Fever/immunology , West Nile Fever/virology
7.
J Infect Dis ; 195(11): 1607-17, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17471430

ABSTRACT

A chimeric protein West Nile virus (WNV) vaccine capable of delivering both innate and adaptive immune signals was designed by fusing a modified version of bacterial flagellin (STF2 Delta ) to the EIII domain of the WNV envelope protein. This fusion protein stimulated interleukin-8 production in a Toll-like receptor (TLR)-5-dependent fashion, confirming appropriate in vitro TLR5 bioactivity, and also retained critical WNV-E-specific conformation-dependent neutralizing epitopes as measured by enzyme-linked immunosorbent assay. When administered without adjuvant to C3H/HeN mice, the fusion protein elicited a strong WNV-E-specific immunoglobulin G antibody response that neutralized viral infectivity and conferred protection against a lethal WNV challenge. This potent EIII-specific immune response requires a direct linkage of EIII to STF2 Delta , given that a simple mixture of the 2 components failed to induce an antibody response or to provide protection against virus challenge. The presence of a functional TLR5 gene in vivo is also required--TLR5-deficient mice elicited only a minimal antigen-specific response. These results confirm that vaccines designed to coordinately regulate the innate and adaptive immune responses can induce protective immune responses without the need for potentially toxic adjuvants. They also support the further development of an effective WNV vaccine and novel monovalent and multivalent vaccines for related flaviviruses.


Subject(s)
Antibodies, Viral/blood , Flagellin/immunology , Recombinant Fusion Proteins/immunology , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , Antibody Specificity , Cell Line , Flagellin/genetics , Flagellin/metabolism , Immunity, Cellular , Immunity, Innate , Mice , Mice, Inbred C3H , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Plaque Assay , West Nile Fever/virology , West Nile Virus Vaccines/administration & dosage
8.
Vaccine ; 23(30): 3915-24, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15917113

ABSTRACT

West Nile (WN) virus is a flavivirus that first appeared in North America in 1999. Since then, more than 600 human deaths and 22,000 equine infections have been attributed to the virus in the United States. We expressed a truncated form of WN virus envelope (E) protein in Drosophila S2 cells. This soluble recombinant E protein was recognized by antibodies from naturally infected horses, indicating that it contains native epitopes. Mice and horses produced high-titer antibodies when immunized with recombinant E protein combined with aluminum hydroxide. Immunized mice were resistant to challenge with a lethal viral dose. Sera from immunized horses, administered to naive mice, conferred resistance against a lethal WN viral challenge. In addition, sera of immunized horses neutralized West Nile virus in vitro, as demonstrated by plaque reduction assays. This recombinant form of E protein, combined with aluminum hydroxide, is a candidate vaccine that may protect humans and horses against WN virus infections.


Subject(s)
Viral Envelope Proteins/immunology , Viral Vaccines/immunology , West Nile Fever/prevention & control , West Nile virus/immunology , Adjuvants, Immunologic , Aluminum Hydroxide/pharmacology , Animals , Cells, Cultured , Drosophila , Female , Horses , Immunization, Passive , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Male , Mice , Mice, Inbred C57BL , Protein Conformation , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , Viral Plaque Assay
SELECTION OF CITATIONS
SEARCH DETAIL