Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Host Microbe ; 19(4): 515-528, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-27078069

ABSTRACT

Understanding the negative regulators of antiviral immune responses will be critical for advancing immune-modulated antiviral strategies. NLRX1, an NLR protein that negatively regulates innate immunity, was previously identified in an unbiased siRNA screen as required for HIV infection. We find that NLRX1 depletion results in impaired nuclear import of HIV-1 DNA in human monocytic cells. Additionally, NLRX1 was observed to reduce type-I interferon (IFN-I) and cytokines in response to HIV-1 reverse-transcribed DNA. NLRX1 sequesters the DNA-sensing adaptor STING from interaction with TANK-binding kinase 1 (TBK1), which is a requisite for IFN-1 induction in response to DNA. NLRX1-deficient cells generate an amplified STING-dependent host response to cytosolic DNA, c-di-GMP, cGAMP, HIV-1, and DNA viruses. Accordingly, Nlrx1(-/-) mice infected with DNA viruses exhibit enhanced innate immunity and reduced viral load. Thus, NLRX1 is a negative regulator of the host innate immune response to HIV-1 and DNA viruses.


Subject(s)
DNA Virus Infections/virology , DNA Viruses/physiology , HIV Infections/immunology , HIV-1/physiology , Interferon-beta/immunology , Membrane Proteins/immunology , Mitochondrial Proteins/metabolism , Virus Replication , Animals , DNA Virus Infections/immunology , Down-Regulation , Female , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , Humans , Immunity, Innate , Interferon-beta/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
2.
Peptides ; 48: 96-105, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23891650

ABSTRACT

Broad-spectrum antiviral drugs are urgently needed to treat individuals infected with new and re-emerging viruses, or with viruses that have developed resistance to antiviral therapies. Mammalian natural host defense peptides (mNHP) are short, usually cationic, peptides that have direct antimicrobial activity, and which in some instances activate cell-mediated antiviral immune responses. Although mNHP have potent activity in vitro, efficacy trials in vivo of exogenously provided mNHP have been largely disappointing, and no mNHP are currently licensed for human use. Mastoparan is an invertebrate host defense peptide that penetrates lipid bilayers, and we reasoned that a mastoparan analog might interact with the lipid component of virus membranes and thereby reduce infectivity of enveloped viruses. Our objective was to determine whether mastoparan-derived peptide MP7-NH2 could inactivate viruses of multiple types, and whether it could stimulate cell-mediated antiviral activity. We found that MP7-NH2 potently inactivated a range of enveloped viruses. Consistent with our proposed mechanism of action, MP7-NH2 was not efficacious against a non-enveloped virus. Pre-treatment of cells with MP7-NH2 did not reduce the amount of virus recovered after infection, which suggested that the primary mechanism of action in vitro was direct inactivation of virus by MP7-NH2. These results demonstrate for the first time that a mastoparan derivative has broad-spectrum antiviral activity in vitro and suggest that further investigation of the antiviral properties of mastoparan peptides in vivo is warranted.


Subject(s)
Peptides/pharmacology , Viruses/drug effects , Wasp Venoms/chemistry , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Invertebrates/chemistry , Mice , Microscopy, Electron , Peptides/chemistry , Viruses/ultrastructure
3.
PLoS One ; 7(10): e46516, 2012.
Article in English | MEDLINE | ID: mdl-23056330

ABSTRACT

Vaccines based on live viruses are attractive because they are immunogenic, cost-effective, and can be delivered by multiple routes. However, live virus vaccines also cause reactogenic side effects such as fever, myalgia, and injection site pain that have reduced their acceptance in the clinic. Several recent studies have linked vaccine-induced reactogenic side effects to production of the pro-inflammatory cytokine interleukin-1ß (IL-1ß) in humans. Our objective was therefore to determine whether IL-1ß contributed to pathology after immunization with recombinant vesicular stomatitis virus (rVSV) vaccine vectors, and if so, to identify strategies by which IL-1ß mediated pathology might be reduced without compromising immunogenicity. We found that an rVSV vaccine induced local and systemic production of IL-1ß in vivo, and that accumulation of IL-1ß correlated with acute pathology after rVSV immunization. rVSV-induced pathology was reduced in mice deficient in the IL-1 receptor Type I, but the IL-1R-/- mice were fully protected from lethal rechallenge with a high dose of VSV. This result demonstrated that IL-1 contributed to reactogenicity of the rVSV, but was dispensable for induction of protective immunity. The amount of IL-1ß detected in mice deficient in either caspase-1 or the inflammasome adaptor molecule ASC after rVSV immunization was not significantly different than that produced by wild type animals, and caspase-1-/- and ASC-/- mice were only partially protected from rVSV-induced pathology. Those data support the idea that some of the IL-1ß expressed in vivo in response to VSV may be activated by a caspase-1 and ASC-independent mechanism. Together these results suggest that rVSV vectors engineered to suppress the induction of IL-1ß, or signaling through the IL-1R would be less reactogenic in vivo, but would retain their immunogenicity and protective capacity. Such rVSV would be highly desirable as either vaccine vectors or oncolytic therapies, and would likely be better tolerated in human vaccinees.


Subject(s)
Interleukin-1beta/biosynthesis , Vaccines, Synthetic/immunology , Vesiculovirus/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Immunity, Cellular , Injections, Intramuscular , Mice , Mice, Knockout , Receptors, Interleukin-1/genetics , Vesiculovirus/physiology , Virus Replication
4.
Blood ; 119(26): 6344-53, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22596261

ABSTRACT

A major challenge in allogeneic hematopoietic cell transplantation is how to transfer T-cell immunity without causing graft-versus-host disease (GVHD). Effector memory T cells (CD62L(-)) are a cell subset that can potentially address this challenge because they do not induce GVHD. Here, we investigated how CD62L(-) T cells contributed to phenotypic and functional T-cell reconstitution after transplantation. On transfer into allogeneic recipients, CD62L(-) T cells were activated and expressed multiple cytokines and cytotoxic molecules. CD62L(-) T cells were able to deplete host radioresistant T cells and facilitate hematopoietic engraftment, resulting in enhanced de novo T-cell regeneration. Enhanced functional immune reconstitution was demonstrated in CD62L(-) T-cell recipients using a tumor and an influenza virus challenge model. Even though CD62L(-) T cells are able to respond to alloantigens and deplete host radioresistant immune cells in GVHD recipients, alloreactive CD62L(-) T cells lost the reactivity over time and were eventually tolerant to alloantigens as a result of prolonged antigen exposure, suggesting a mechanism by which CD62L(-) T cells were able to eliminate host resistance without causing GVHD. These data further highlight the unique characteristics of CD62L(-) T cells and their potential applications in clinical hematopoietic cell transplantation.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cell Transplantation , Immunologic Memory/immunology , L-Selectin/metabolism , T-Lymphocytes/physiology , Transplantation Tolerance/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Female , Immunity, Innate/immunology , Immunocompromised Host/immunology , Immunocompromised Host/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Homologous
5.
PLoS One ; 6(10): e25797, 2011.
Article in English | MEDLINE | ID: mdl-22039424

ABSTRACT

BACKGROUND: During the recent H1N1 influenza pandemic, excess morbidity and mortality was seen in young but not older adults suggesting that prior infection with influenza strains may have protected older subjects. In contrast, a history of recent seasonal trivalent vaccine in younger adults was not associated with protection. METHODS AND FINDINGS: To study hemagglutinin (HA) antibody responses in influenza immunization and infection, we have studied the day 7 plasma cell repertoires of subjects immunized with seasonal trivalent inactivated influenza vaccine (TIV) and compared them to the plasma cell repertoires of subjects experimentally infected (EI) with influenza H3N2 A/Wisconsin/67/2005. The majority of circulating plasma cells after TIV produced influenza-specific antibodies, while most plasma cells after EI produced antibodies that did not react with influenza HA. While anti-HA antibodies from TIV subjects were primarily reactive with single or few HA strains, anti-HA antibodies from EI subjects were isolated that reacted with multiple HA strains. Plasma cell-derived anti-HA antibodies from TIV subjects showed more evidence of clonal expansion compared with antibodies from EI subjects. From an H3N2-infected subject, we isolated a 4-member clonal lineage of broadly cross-reactive antibodies that bound to multiple HA subtypes and neutralized both H1N1 and H3N2 viruses. This broad reactivity was not detected in post-infection plasma suggesting this broadly reactive clonal lineage was not immunodominant in this subject. CONCLUSION: The presence of broadly reactive subdominant antibody responses in some EI subjects suggests that improved vaccine designs that make broadly reactive antibody responses immunodominant could protect against novel influenza strains.


Subject(s)
Antibodies, Viral/biosynthesis , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza Vaccines/immunology , Influenza, Human/immunology , Antibodies, Viral/immunology , Antibody Specificity , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique, Indirect , Humans , Influenza, Human/virology , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance
6.
Virology ; 417(1): 87-97, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21658738

ABSTRACT

Cowpox virus infection induces interleukin-10 (IL-10) production from mouse bone marrow-derived dendritic cells (BMDCs) or cells of the mouse macrophage line (RAW264.7) at about 1800 pg/ml, whereas infections with vaccinia virus (strains WR or MVA) induced much less IL-10. Similarly, in vivo, IL-10 levels in bronchoalveolar lavage fluids of mice infected with cowpox virus were significantly higher than those after vaccinia virus infection. However, after intranasal cowpox virus infection, although dendritic and T-cell accumulations in the lungs of IL-10 deficient mice were greater than those in wild-type mice, weight-loss and viral burdens were not significantly different. IL-10 deficient mice were more susceptible than wild-type mice to re-infection with cowpox virus even though titers of neutralizing antibodies and virus-specific CD8 T cells were similar between IL-10 deficient and wild-type mice. Greater bronchopneumonia in IL-10 deficient mice than wild-type mice suggests that IL-10 contributes to the suppression of immunopathology in the lungs.


Subject(s)
Cowpox virus/physiology , Cowpox/immunology , Dendritic Cells/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , Animals , Bone Marrow Cells/metabolism , Cell Line , Female , Gene Expression Regulation/physiology , Interleukin-10/genetics , Lung/cytology , Mice , Mice, Inbred C57BL , Mutation , T-Lymphocytes , Weight Loss
7.
Vaccine ; 28(1): 79-89, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19819211

ABSTRACT

Vaccines currently licensed for the prevention of seasonal influenza induce antibodies against the influenza hemagglutinin (HA) and neuraminidase (NA) contained in the vaccine preparation but require at least 2 weeks after immunization for the development of protective immunity. These vaccines do not induce protective responses quickly enough to blunt the effects of infection when administered after exposure. We have developed a novel vaccine based on recombinant vesicular stomatitis virus which expresses the influenza hemagglutinin (rVSV HA) and protects mice from lethal influenza challenge when the vaccine is administered intramuscularly at least 24h after delivery of the influenza challenge virus. To our knowledge this is the first vaccine that effectively protects animals from lethal influenza challenge when delivered by a systemic route after influenza exposure has occurred. The induction of HA-specific immune responses by the vaccine is necessary for full protection from challenge, because animals immunized with an empty rVSV vector were not protected equally. Our results are consistent with a model in which vaccination induces an immediate antiviral cytokine response, followed by development of humoral and cellular immune responses which act to reduce pulmonary viral loads and accelerate recovery. Consistent with this model, mice vaccinated with the specific vaccine rVSV HA had high levels of IFN-alpha in the serum by 24h after challenge/vaccination, developed serum neutralizing Ab to influenza 2 days prior to control animals, and had detectable anti-HA CD8 T cells present in the peripheral blood 3 days prior to control mice.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cricetinae , Female , Influenza A virus/immunology , Injections, Intramuscular , Interferon-alpha/blood , Mice , Mice, Inbred BALB C , Neutralization Tests , Orthomyxoviridae Infections/immunology , Vesicular stomatitis Indiana virus/immunology , Viral Load , Viral Plaque Assay
8.
Clin Vaccine Immunol ; 16(4): 488-98, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19244472

ABSTRACT

Live attenuated vaccine vectors based on recombinant vesicular stomatitis viruses (rVSVs) expressing foreign antigens are highly effective vaccines in animal models. In this study, we report that an rVSV expressing influenza nucleoprotein (VSV NP) from the first position of the VSV genome induces robust anti-NP CD8 T cells in immunized mice. These CD8 T cells are phenotypically similar to those induced by natural influenza infection and are cytotoxic in vivo. Animals immunized with an rVSV expressing the influenza hemagglutinin (rVSV HA) were protected but still exhibited considerable morbidity after challenge. Animals receiving a cocktail vaccine of rVSV NP and rVSV HA had reduced pulmonary viral loads, less weight loss, and reduced clinical signs of illness after influenza virus challenge, relative to those vaccinated with rVSV HA alone. Influenza NP is a highly conserved antigen, and induction of protective anti-NP responses may be a productive strategy for generating heterologous protection against divergent influenza strains.


Subject(s)
Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , RNA-Binding Proteins/immunology , Vesiculovirus/genetics , Viral Core Proteins/immunology , Animals , Body Weight , Female , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/genetics , Lung/virology , Mice , Nucleocapsid Proteins , Orthomyxoviridae Infections/pathology , RNA-Binding Proteins/genetics , Survival Analysis , Viral Core Proteins/genetics
9.
Vaccine ; 26(48): 6108-18, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18809447

ABSTRACT

The prevention of infectious disease via prophylactic immunization is a mainstay of global public health efforts. Vaccine design would be facilitated by a better understanding of the type and durability of immune responses generated by different vaccine vectors. We report here the results of a comparative immunogenicity trial of six different vaccine vectors expressing the same insert antigen, cowpox virus B5 (CPXV-B5). Of those vectors tested, recombinant adenovirus (rAd5) was the most immunogenic, inducing the highest titer anti-B5 antibodies and conferring protection from sublethal vaccinia virus challenge in mice after a single immunization. We tested select heterologous prime-boost combinations and identified recombinant vesicular stomatitis virus (rVSV) and recombinant Venezuelan equine encephalitis virus replicons (VRP) as the most synergistic regimen. Comparative data such as those presented here are critical to efforts to generate protective vaccines for emerging infectious diseases as well as for biothreat agents.


Subject(s)
Genetic Vectors/immunology , Vaccines/genetics , Vaccines/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Administration, Intranasal , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/genetics , Bioterrorism , Comet Assay , Cowpox virus/genetics , Cowpox virus/immunology , Dose-Response Relationship, Immunologic , Encephalitis Virus, Venezuelan Equine/immunology , Immunization, Secondary , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Mycobacterium smegmatis/immunology , Plasmids/genetics , Vaccines/administration & dosage , Vaccines, Synthetic/immunology , Vaccinia virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...