Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 220(2): 219-227, 2019 06 19.
Article in English | MEDLINE | ID: mdl-30895307

ABSTRACT

BACKGROUND: Dengue virus is an emerging mosquito-borne flavivirus responsible for considerable morbidity and mortality worldwide. The Division of Intramural Research, National Institute of Allergy and Infectious Diseases of the US National Institutes of Health (NIH) has developed live attenuated vaccines to each of the 4 serotypes of dengue virus (DENV1-4). While overall levels of DENV neutralizing antibodies (nAbs) in humans have been correlated with protection, these correlations vary depending on DENV serotype, prevaccination immunostatus, age, and study site. By combining both the level and molecular specificity of nAbs to each serotype, it may be possible to develop more robust correlates that predict long-term outcome. METHODS: Using depletions and recombinant chimeric epitope transplant DENVs, we evaluate the molecular specificity and mapped specific epitopes and antigenic regions targeted by vaccine-induced nAbs in volunteers who received the NIH monovalent vaccines against each DENV serotype. RESULTS: After monovalent vaccination, subjects developed high levels of nAbs that mainly targeted epitopes that are unique (type-specific) to each DENV serotype. The DENV1, 2, and 4 monovalent vaccines induced type-specific nAbs directed to quaternary structure envelope epitopes known to be targets of strongly neutralizing antibodies induced by wild-type DENV infections. CONCLUSIONS: Our results reported here on the molecular specificity of NIH vaccine-induced antibodies enable new strategies, beyond the absolute levels of nAbs, for determining correlates and mechanisms of protective immunity.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Vaccines/immunology , Dengue Virus/immunology , Dengue/immunology , Epitopes/immunology , Amino Acid Sequence , Dengue/virology , Epitope Mapping/methods , Humans , National Institutes of Health (U.S.) , Serogroup , United States , Vaccination/methods , Vaccines, Attenuated/immunology , Viral Envelope Proteins/immunology
2.
Cell Rep ; 25(5): 1214-1224, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30380413

ABSTRACT

There are four distinct DENV serotypes, and within DENV4, there are five distinct genotypes. The impact of genotypic diversity is not known, nor is it clear whether infection with one DENV4 genotype results in protective immunity against the other genotypes. To measure the impact of DENV4 genetic diversity, we generated an isogenic panel of viruses containing the envelope protein from the different genotypes. We characterized many properties of these viruses and find that a small number of amino acids changes within the envelope have disproportionate impacts on virus biology. Additionally, we observe large differences in the ability of DENV4 antibodies, immune sera, and vaccine sera to neutralize the panel, suggesting that DENV4 immunity might not be equally protective against all DENV4s. Our results support the monitoring of changing or emerging DENV genotypes and their role in escaping pre-existing neutralizing antibodies in people who have been vaccinated or exposed to natural DENV4 infections.


Subject(s)
Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Dengue Virus/genetics , Genetic Variation , Animals , Antibodies, Monoclonal/metabolism , Cell Line , Chlorocebus aethiops , Cross Reactions/immunology , Genotype , Glycosylation , Humans , Immune Sera/metabolism , Kinetics , Neutralization Tests , Phylogeny , Serogroup , Temperature , Vaccination , Vero Cells
3.
PLoS One ; 13(10): e0200043, 2018.
Article in English | MEDLINE | ID: mdl-30332435

ABSTRACT

Infection of immunocompromised individuals with normally benign opportunistic viruses is a major health burden globally. Infections with viruses such as Epstein-Barr virus (EBV), human cytomegalovirus (HCMV), Kaposi's sarcoma virus (KSHV), adenoviruses (AdV), BK virus (BKPyV), John Cunningham virus (JCPyV), and human papillomavirus (HPV) are significant concerns for the immunocompromised, including when these viruses exist as a co-infection with human immunodeficiency virus (HIV). These viral infections are more complicated in patients with a weakened immune system, and often manifest as malignancies resulting in significant morbidity and mortality. Vaccination is not an attractive option for these immune compromised individuals due to defects in their adaptive immune response. Verdinexor is part of a novel class of small molecules known as SINE (Selective Inhibitor of Nuclear Export) compounds. These small molecules demonstrate specificity for the nuclear export protein XPO1, to which they bind and block function, resulting in sequestration of XPO1-dependent proteins in the nucleus of the cell. In antiviral screening, verdinexor demonstrated varying levels of efficacy against all of the aforementioned viruses including previously with HIV. Studies by other labs have discussed likely mechanisms of action for verdinexor (ie. XPO1-dependence) against each virus. GLP toxicology studies suggest that anti-viral activity can be achieved at a tolerable dose range, based on the safety profile of a previous phase 1 clinical trial of verdinexor in healthy human volunteers. Taken together, these results indicate verdinexor has the potential to be a broad spectrum antiviral for immunocompromised subjects for which vaccination is a poor option.


Subject(s)
Acrylamides/pharmacology , Hydrazines/pharmacology , Immunocompromised Host/drug effects , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Virus Diseases/drug therapy , Adenoviridae Infections/drug therapy , Animals , Cell Line, Tumor , Cytomegalovirus Infections/drug therapy , Drug Evaluation, Preclinical , Epstein-Barr Virus Infections/drug therapy , Fibroblasts/virology , Guinea Pigs , HEK293 Cells , HIV Infections/complications , HeLa Cells , Humans , Mice , Papillomavirus Infections/drug therapy , Polyomavirus Infections/drug therapy , Reproducibility of Results , Sarcoma, Kaposi/drug therapy , Tumor Virus Infections/drug therapy , Virus Diseases/complications , Exportin 1 Protein
4.
J Infect Dis ; 217(12): 1932-1941, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29800370

ABSTRACT

Background: Dengue virus serotypes 1-4 (DENV-1-4) are the most common vector-borne viral pathogens of humans and the etiological agents of dengue fever and dengue hemorrhagic syndrome. A live-attenuated tetravalent dengue vaccine (TDV) developed by Takeda Vaccines has recently progressed to phase 3 safety and efficacy evaluation. Methods: We analyzed the qualitative features of the neutralizing antibody (nAb) response induced in naive and DENV-immune individuals after TDV administration. Using DENV-specific human monoclonal antibodies (mAbs) and recombinant DENV displaying different serotype-specific Ab epitopes, we mapped the specificity of TDV-induced nAbs against DENV-1-3. Results: Nearly all subjects had high levels of DENV-2-specific nAbs directed to epitopes centered on domain III of the envelope protein. In some individuals, the vaccine induced nAbs that tracked with a DENV-1-specific neutralizing epitope centered on domain I of the envelope protein. The vaccine induced binding Abs directed to a DENV-3 type-specific neutralizing epitope, but findings of mapping of DENV-3 type-specific nAbs were inconclusive. Conclusion: Here we provide qualitative measures of the magnitude and epitope specificity of the nAb responses to TDV. This information will be useful for understanding the performance of TDV in clinical trials and for identifying correlates of protective immunity.


Subject(s)
Antibodies, Viral/blood , Antibody Formation/immunology , Severe Dengue/blood , Severe Dengue/immunology , Vaccines, Attenuated/immunology , Adolescent , Adult , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line, Tumor , Dengue Vaccines/immunology , Dengue Virus/immunology , Double-Blind Method , Female , Humans , Immunity/immunology , Male , Middle Aged , U937 Cells , Vaccination/methods , Young Adult
5.
Nat Med ; 24(3): 368-374, 2018 03.
Article in English | MEDLINE | ID: mdl-29400709

ABSTRACT

Zika virus (ZIKV) is a flavivirus with teratogenic effects on fetal brain, but the spectrum of ZIKV-induced brain injury is unknown, particularly when ultrasound imaging is normal. In a pregnant pigtail macaque (Macaca nemestrina) model of ZIKV infection, we demonstrate that ZIKV-induced injury to fetal brain is substantial, even in the absence of microcephaly, and may be challenging to detect in a clinical setting. A common and subtle injury pattern was identified, including (i) periventricular T2-hyperintense foci and loss of fetal noncortical brain volume, (ii) injury to the ependymal epithelium with underlying gliosis and (iii) loss of late fetal neuronal progenitor cells in the subventricular zone (temporal cortex) and subgranular zone (dentate gyrus, hippocampus) with dysmorphic granule neuron patterning. Attenuation of fetal neurogenic output demonstrates potentially considerable teratogenic effects of congenital ZIKV infection even without microcephaly. Our findings suggest that all children exposed to ZIKV in utero should receive long-term monitoring for neurocognitive deficits, regardless of head size at birth.


Subject(s)
Fetus/virology , Pregnancy Complications, Infectious/physiopathology , Zika Virus Infection/virology , Zika Virus/pathogenicity , Animals , Disease Models, Animal , Female , Fetus/physiopathology , Humans , Macaca nemestrina/virology , Microcephaly/diagnostic imaging , Microcephaly/physiopathology , Microcephaly/virology , Neurogenesis/genetics , Pregnancy , Pregnancy Complications, Infectious/diagnostic imaging , Pregnancy Complications, Infectious/virology , Zika Virus/genetics , Zika Virus Infection/genetics , Zika Virus Infection/physiopathology
6.
PLoS Pathog ; 14(2): e1006934, 2018 02.
Article in English | MEDLINE | ID: mdl-29481552

ABSTRACT

Dengue virus (DENV) infection causes dengue fever, dengue hemorrhagic fever and dengue shock syndrome. It is estimated that a third of the world's population is at risk for infection, with an estimated 390 million infections annually. Dengue virus serotype 2 (DENV2) causes severe epidemics, and the leading tetravalent dengue vaccine has lower efficacy against DENV2 compared to the other 3 serotypes. In natural DENV2 infections, strongly neutralizing type-specific antibodies provide protection against subsequent DENV2 infection. While the epitopes of some human DENV2 type-specific antibodies have been mapped, it is not known if these are representative of the polyclonal antibody response. Using structure-guided immunogen design and reverse genetics, we generated a panel of recombinant viruses containing amino acid alterations and epitope transplants between different serotypes. Using this panel of recombinant viruses in binding, competition, and neutralization assays, we have finely mapped the epitopes of three human DENV2 type-specific monoclonal antibodies, finding shared and distinct epitope regions. Additionally, we used these recombinant viruses and polyclonal sera to dissect the epitope-specific responses following primary DENV2 natural infection and monovalent vaccination. Our results demonstrate that antibodies raised following DENV2 infection or vaccination circulate as separate populations that neutralize by occupying domain III and domain I quaternary epitopes. The fraction of neutralizing antibodies directed to different epitopes differs between individuals. The identification of these epitopes could potentially be harnessed to evaluate epitope-specific antibody responses as correlates of protective immunity, potentially improving vaccine design.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Dengue Virus/immunology , Epitopes/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antibody Formation , Antibody Specificity , Cells, Cultured , Chlorocebus aethiops , Cross Reactions , Dengue/immunology , Dengue/virology , Dengue Vaccines/immunology , Epitopes/chemistry , Epitopes/immunology , Humans , Protein Structure, Quaternary , Serogroup , Vaccination , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
7.
Sci Rep ; 7(1): 17169, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215033

ABSTRACT

Dengue vaccine trials have revealed deficits in our understanding of the mechanisms of protective immunity, demonstrating a need to measure epitope-specific antibody responses against each DENV serotype. HmAb 5J7 binds to a complex, 3-monomer spanning quaternary epitope in the DENV3 envelope (E) protein, but it is unclear whether all interactions are needed for neutralization. Structure guided design and reverse genetics were used to sequentially transplant larger portions of the DENV3-specific 5J7 mAb epitope into dengue virus serotype 4 (DENV4). We observed complete binding and neutralization only when the entire 3 monomer spanning epitope was transplanted into DENV4, providing empirical proof that cooperative monomer-hmAb 5J7 interactions maximize activity. The rDENV4/3 virus containing the most expanded 5J7 epitope was also significantly more sensitive than WT DENV4 to neutralization by DENV3 primary immune sera. We conclude that the hinge-spanning region of the 5J7 quaternary epitope is a target for serotype-specific neutralizing antibodies after DENV3 infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Dengue/immunology , Epitopes/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing/genetics , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Chlorocebus aethiops , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Humans , Neutralization Tests , Protein Structure, Quaternary , Sequence Homology , Serogroup , U937 Cells , Vero Cells
8.
mBio ; 8(2)2017 03 07.
Article in English | MEDLINE | ID: mdl-28270583

ABSTRACT

Zika virus (ZIKV), a mosquito-borne flavivirus discovered in 1947, has only recently caused large outbreaks and emerged as a significant human pathogen. In 2015, ZIKV was detected in Brazil, and the resulting epidemic has spread throughout the Western Hemisphere. Severe complications from ZIKV infection include neurological disorders such as Guillain-Barré syndrome in adults and a variety of fetal abnormalities, including microcephaly, blindness, placental insufficiency, and fetal demise. There is an urgent need for tools and reagents to study the pathogenesis of epidemic ZIKV and for testing vaccines and antivirals. Using a reverse genetics platform, we generated six ZIKV infectious clones and derivative viruses representing diverse temporal and geographic origins. These include three versions of MR766, the prototype 1947 strain (with and without a glycosylation site in the envelope protein), and H/PF/2013, a 2013 human isolate from French Polynesia representative of the virus introduced to Brazil. In the course of synthesizing a clone of a circulating Brazilian strain, phylogenetic studies identified two distinct ZIKV clades in Brazil. We reconstructed viable clones of strains SPH2015 and BeH819015, representing ancestral members of each clade. We assessed recombinant virus replication, binding to monoclonal antibodies, and virulence in mice. This panel of molecular clones and recombinant virus isolates will enable targeted studies of viral determinants of pathogenesis, adaptation, and evolution, as well as the rational attenuation of contemporary outbreak strains to facilitate the design of vaccines and therapeutics.IMPORTANCE Viral emergence is a poorly understood process as evidenced by the sudden emergence of Zika virus in Latin America and the Caribbean. Malleable reagents that both predate and span an expanding epidemic are key to understanding the virologic determinants that regulate pathogenesis and transmission. We have generated representative cDNA molecular clones and recombinant viruses that span the known ZIKV family tree, including early Brazilian isolates. Recombinant viruses replicated efficiently in cell culture and were pathogenic in immunodeficient mice, providing a genetic platform for rational vaccine and therapeutic design.


Subject(s)
Molecular Biology/methods , Reverse Genetics/methods , Virology/methods , Zika Virus/genetics , Zika Virus/physiology , Humans , Zika Virus/immunology , Zika Virus/pathogenicity
9.
mSphere ; 2(1)2017.
Article in English | MEDLINE | ID: mdl-28251184

ABSTRACT

Despite the clinical relevance, dengue virus (DENV) research has been hampered by the absence of robust reverse genetic systems to manipulate the viral serotypes for propagation and generation of mutant viruses. In this article, we describe application of an infectious clone system for DENV serotype 1 (DENV1). Similar to previous clones in both flaviviruses and coronaviruses, the approach constructs a panel of contiguous cDNAs that span the DENV genome and can be systematically and directionally assembled to produce viable, full-length viruses. Comparison of the virus derived from the infectious clone with the original viral isolate reveals identical sequence, comparable endpoint titers, and similar focus staining. Both focus-forming assays and percent infection by flow cytometry revealed overlapping replication levels in two different cell types. Moreover, serotype-specific monoclonal antibodies (MAbs) bound similarly to infectious clone and the natural isolate. Using the clone, we were able to insert a DENV4 type-specific epitope recognized by primate MAb 5H2 into envelope (E) protein domain I (EDI) of DENV1 and recover a viable chimeric recombinant virus. The recombinant DENV1 virus was recognized and neutralized by the DENV4 type-specific 5H2 MAb. The introduction of the 5H2 epitope ablated two epitopes on DENV1 EDI recognized by human MAbs (1F4 and 14C10) that strongly neutralize DENV1. Together, the work demonstrates the utility of the infectious clone and provides a resource to rapidly manipulate the DENV1 serotype for generation of recombinant and mutant viruses. IMPORTANCE Dengue viruses (DENVs) are significant mosquito-transmitted pathogens that cause widespread infection and can lead to severe infection and complications. Here we further characterize a novel and robust DENV serotype 1 (DENV1) infectious clone system that can be used to support basic and applied research. We demonstrate how the system can be used to probe the antigenic relationships between strains by creating viable recombinant viruses that display or lack major antibody epitopes. The DENV1 clone system and recombinant viruses can be used to analyze existing vaccine immune responses and inform second-generation bivalent vaccine designs.

10.
J Virol ; 91(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-28031369

ABSTRACT

The four dengue virus (DENV) serotypes are mosquito-borne flaviviruses responsible for dengue fever and dengue hemorrhagic fever. People exposed to DENV develop antibodies (Abs) that strongly neutralize the serotype responsible for infection. Historically, infection with DENV serotype 4 (DENV4) has been less common and less studied than infections with the other three serotypes. However, DENV4 has been responsible for recent large and sustained epidemics in Asia and Latin America. The neutralizing antibody responses and the epitopes targeted against DENV4 have not been characterized in human infection. In this study, we mapped and characterized epitopes on DENV4 recognized by neutralizing antibodies in people previously exposed to DENV4 infections or to a live attenuated DENV4 vaccine. To study the fine specificity of DENV4 neutralizing human antibodies, B cells from two people exposed to DENV4 were immortalized and screened to identify DENV-specific clones. Two human monoclonal antibodies (MAbs) that neutralized DENV4 were isolated, and their epitopes were finely mapped using recombinant viruses and alanine scan mutation array techniques. Both antibodies bound to quaternary structure epitopes near the hinge region between envelope protein domain I (EDI) and EDII. In parallel, to characterize the serum neutralizing antibody responses, convalescence-phase serum samples from people previously exposed to primary DENV4 natural infections or a monovalent DENV4 vaccine were analyzed. Natural infection and vaccination also induced serum-neutralizing antibodies that targeted similar epitope domains at the EDI/II hinge region. These studies defined a target of neutralizing antigenic site on DENV4 targeted by human antibodies following natural infection or vaccination.IMPORTANCE The four serotypes of dengue virus are the causative agents of dengue fever and dengue hemorrhagic fever. People exposed to primary DENV infections develop long-term neutralizing antibody responses, but these principally recognize only the infecting serotype. An effective vaccine against dengue should elicit long-lasting protective antibody responses to all four serotypes simultaneously. We and others have defined antigenic sites on the envelope (E) protein of viruses of dengue virus serotypes 1, 2, and 3 targeted by human neutralizing antibodies. The epitopes on DENV4 E protein targeted by the human neutralizing antibodies and the mechanisms of serotype 4 neutralization are poorly understood. Here, we report the properties of human antibodies that neutralize dengue virus serotype 4. People exposed to serotype 4 infections or a live attenuated serotype 4 vaccine developed neutralizing antibodies that bound to similar sites on the viral E protein. These studies have provided a foundation for developing and evaluating DENV4 vaccines.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , Dengue Virus/immunology , Dengue/prevention & control , Adaptive Immunity , Aedes , Animals , Antibodies, Viral/drug effects , Cell Line , Dengue/immunology , Dengue/virology , Epitope Mapping , Humans , Immunologic Memory , Protein Binding , Protein Domains , Vaccination , Vaccines, Attenuated/immunology , Viral Vaccines/immunology
11.
Nat Commun ; 7: 13679, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27882950

ABSTRACT

The rapid spread of Zika virus (ZIKV), which causes microcephaly and Guillain-Barré syndrome, signals an urgency to identify therapeutics. Recent efforts to rescreen dengue virus human antibodies for ZIKV cross-neutralization activity showed antibody C10 as one of the most potent. To investigate the ability of the antibody to block fusion, we determined the cryoEM structures of the C10-ZIKV complex at pH levels mimicking the extracellular (pH8.0), early (pH6.5) and late endosomal (pH5.0) environments. The 4.0 Å resolution pH8.0 complex structure shows that the antibody binds to E proteins residues at the intra-dimer interface, and the virus quaternary structure-dependent inter-dimer and inter-raft interfaces. At pH6.5, antibody C10 locks all virus surface E proteins, and at pH5.0, it locks the E protein raft structure, suggesting that it prevents the structural rearrangement of the E proteins during the fusion event-a vital step for infection. This suggests antibody C10 could be a good therapeutic candidate.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Viral Envelope Proteins/immunology , Zika Virus/immunology , Antibodies, Neutralizing/ultrastructure , Antibodies, Viral/ultrastructure , Cross Reactions/immunology , Cryoelectron Microscopy , Dengue Virus/immunology , Hydrogen-Ion Concentration , Zika Virus/ultrastructure
12.
J Virol ; 90(10): 5090-5097, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26962223

ABSTRACT

UNLABELLED: The four dengue virus (DENV) serotypes, DENV1 through 4, are endemic throughout tropical and subtropical regions of the world. While first infection confers long-term protective immunity against viruses of the infecting serotype, a second infection with virus of a different serotype carries a greater risk of severe dengue disease, including dengue hemorrhagic fever and dengue shock syndrome. Recent studies demonstrate that humans exposed to DENV infections develop neutralizing antibodies that bind to quaternary epitopes formed by the viral envelope (E) protein dimers or higher-order assemblies required for the formation of the icosahedral viral envelope. Here we show that the quaternary epitope target of the human DENV3-specific neutralizing monoclonal antibody (MAb) 5J7 can be partially transplanted into a DENV1 strain by changing the core residues of the epitope contained within a single monomeric E molecule. MAb 5J7 neutralized the recombinant DENV1/3 strain in cell culture and was protective in a mouse model of infection with the DENV1/3 strain. However, the 5J7 epitope was only partially recreated by transplantation of the core residues because MAb 5J7 bound and neutralized wild-type (WT) DENV3 better than the DENV1/3 recombinant. Our studies demonstrate that it is possible to transplant a large number of discontinuous residues between DENV serotypes and partially recreate a complex antibody epitope, while retaining virus viability. Further refinement of this approach may lead to new tools for measuring epitope-specific antibody responses and new vaccine platforms. IMPORTANCE: Dengue virus is the most important mosquito-borne pathogen of humans worldwide, with approximately one-half the world's population living in regions where dengue is endemic. Dengue immunity following infection is robust and thought to be conferred by antibodies raised against the infecting virus. However, the specific viral components that these antibodies recognize and how they neutralize the virus have been incompletely described. Here we map a region on dengue virus serotype 3 recognized by the human neutralizing antibody 5J7 and then test the functional significance of this region by transplanting it into a serotype 1 virus. Our studies demonstrate a region on dengue virus necessary for 5J7 binding and neutralization. Our work also demonstrates the technical feasibility of engineering dengue viruses to display targets of protective antibodies. This technology can be used to develop new dengue vaccines and diagnostic assays.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Epitopes , Animals , Antibodies, Neutralizing/genetics , Antibodies, Viral/chemistry , Antibodies, Viral/genetics , Cross Reactions , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Disease Models, Animal , Epitopes/genetics , Epitopes/immunology , Genetic Engineering , Humans , Mice , Neutralization Tests , Serogroup
13.
PLoS One ; 10(8): e0136708, 2015.
Article in English | MEDLINE | ID: mdl-26301593

ABSTRACT

Dengue virus is a major global health threat and can lead to life-threatening hemorrhagic complications due to immune activation and cytokine production. Cross-reactive antibodies to an earlier dengue virus infection are a recognized risk factor for severe disease. These antibodies bind heterologous dengue serotypes and enhance infection into Fc-receptor-bearing cells, a process known as antibody-dependent enhancement of infection. One crucial cytokine seen elevated in severe dengue patients is IL-1ß, a potent inflammatory cytokine matured by the inflammasome. We used a highly-physiologic system by studying antibody-dependent enhancement of IL-1ß in primary human monocytes with anti-dengue human monoclonal antibodies isolated from patients. Antibody-enhancement increased viral replication in primary human monocytes inoculated with supernatant harvested from Vero cells infected with dengue virus serotype 2 (DENV-2) 16681. Surprisingly, IL-1ß secretion induced by infectious supernatant harvested from two independent Vero cell lines was not enhanced by antibody. Secretion of multiple other inflammatory cytokines was also independent of antibody signaling. However, IL-1ß secretion did require NLRP3 and caspase-1 activity. Immunodepletion of dengue virions from the infectious supernatant confirmed that virus was not the main IL-1ß-inducing agent, suggesting that a supernatant component(s) not associated with the virion induced IL-1ß production. We excluded RNA, DNA, contaminating LPS, viral NS1 protein, complement, and cytokines. In contrast, purified Vero-derived DENV-2 16681 exhibited antibody-enhancement of both infection and IL-1ß induction. Furthermore, C6/36 mosquito cells did not produce such an inflammatory component, as crude supernatant harvested from insect cells infected with DENV-2 16681 induced antibody-dependent IL-1ß secretion. This study indicates that Vero cells infected with DENV-2 16681 may produce inflammatory components during dengue virus propagation that mask the virus-specific immune response. Thus, the choice of host cell and viral purity should be carefully considered, while insect-derived virus represents a system that elicits antibody-dependent cytokine responses to dengue virus with fewer confounding issues.


Subject(s)
Antibodies, Viral/immunology , Dengue Virus/immunology , Interleukin-1beta/metabolism , Severe Dengue/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Chlorocebus aethiops , Dengue Virus/pathogenicity , Humans , Interleukin-1beta/immunology , Monocytes/immunology , Monocytes/virology , Severe Dengue/virology , Vero Cells , Virus Replication/immunology
14.
Expert Rev Vaccines ; 14(1): 5-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25249440

ABSTRACT

Dengue virus is the most significant arboviral pathogen worldwide with nearly 400 million infections annually and half the global population at risk of disease. Despite this tremendous public health burden, there are no licensed treatments or vaccines to prevent dengue in humans. Results from clinical trials of leading vaccine candidates have demonstrated that our current understanding of the correlates of protection from dengue is incomplete, and as such vaccine performance has been moderate, but with considerable room for improvement. Here we highlight new findings revealing key neutralizing epitopes that regulate serotype-specific immunity, and discuss their implications for design and evaluation of future vaccine candidates.


Subject(s)
Dengue Virus/immunology , Protein Structure, Tertiary , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Clinical Trials as Topic , Epitopes, B-Lymphocyte/immunology , Humans , Viral Vaccines/isolation & purification
15.
Nat Med ; 20(8): 927-35, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25064127

ABSTRACT

Oxidative tissue injury often accompanies viral infection, yet there is little understanding of how it influences virus replication. We show that multiple hepatitis C virus (HCV) genotypes are exquisitely sensitive to oxidative membrane damage, a property distinguishing them from other pathogenic RNA viruses. Lipid peroxidation, regulated in part through sphingosine kinase-2, severely restricts HCV replication in Huh-7 cells and primary human hepatoblasts. Endogenous oxidative membrane damage lowers the 50% effective concentration of direct-acting antivirals in vitro, suggesting critical regulation of the conformation of the NS3-4A protease and the NS5B polymerase, membrane-bound HCV replicase components. Resistance to lipid peroxidation maps genetically to transmembrane and membrane-proximal residues within these proteins and is essential for robust replication in cell culture, as exemplified by the atypical JFH1 strain of HCV. Thus, the typical, wild-type HCV replicase is uniquely regulated by lipid peroxidation, providing a mechanism for attenuating replication in stressed tissue and possibly facilitating long-term viral persistence.


Subject(s)
Hepacivirus/enzymology , Lipid Peroxidation , Oxidative Stress , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects , Adaptor Proteins, Signal Transducing/genetics , Antiviral Agents/pharmacology , Cell Line , Cell Membrane/pathology , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C/drug therapy , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Interference , RNA, Small Interfering/genetics , Viral Nonstructural Proteins/genetics
16.
Virology ; 450-451: 278-89, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24503091

ABSTRACT

Macrophages encounter flaviviruses early after injection by arthropod vectors. Using in vivo imaging of mice inoculated with firefly luciferase-expressing single-cycle flavivirus particles (FLUC-SCFV), we examined the initial dissemination of virus particles in the presence or absence of lymph node (LN)-resident macrophages. Higher luciferase activity, indicating higher SCFV gene expression, was detected in the footpad of macrophage-depleted mice after 24h post infection (hpi). Moreover, FLUC-SCFV particles disseminated to the spleen within 14 hpi in macrophage-depleted, but not control mice. Although macrophages presented SCFV to naïve T cells in vitro, depletion of subcapsular sinus (SCS) macrophages did not alter the magnitude or effector function of the WNV-specific CD8(+) T cell response. Together, these results indicate that SCS macrophages play a role in limiting the dissemination of SCFV early in infection but are not required for the generation of a polyfunctional WNV-specific CD8(+) T cell response in the draining LN.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Macrophages/virology , West Nile Fever/immunology , West Nile virus/physiology , Animals , Chemokine CCL2/immunology , Interleukin-6/immunology , Lymph Nodes/immunology , Lymph Nodes/virology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Species Specificity , Spleen/immunology , Spleen/virology , West Nile Fever/virology , West Nile virus/genetics
17.
Methods Mol Biol ; 1031: 91-6, 2013.
Article in English | MEDLINE | ID: mdl-23824891

ABSTRACT

Type I interferons are critical cytokines produced by the host innate immune response to viral infection. They act collectively to initiate expression of a multitude of antiviral genes that serve to inhibit viral replication and spread. Despite the great importance of interferons to the host response to viral infection, assays to measure their presence can be costly and require a great deal of optimization for success. Here, we describe an inexpensive approach for the determination of murine type I interferon activity in a given set of samples, which is based on using 50 % protection of a cell monolayer from virus-induced cytopathic effects as an endpoint measurement. The following protocol allows for the accurate and sensitive measurement of interferon activity without the use of highly specialized equipment or reagents.


Subject(s)
Immunity, Innate , Infections/metabolism , Interferon Type I/isolation & purification , Animals , Infections/virology , Interferon Type I/metabolism , Mice
18.
Immunity ; 36(6): 933-46, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22749352

ABSTRACT

The mitochondrial protein MAVS (also known as IPS-1, VISA, and CARDIF) interacts with RIG-I-like receptors (RLRs) to induce type I interferon (IFN-I). NLRX1 is a mitochondrial nucleotide-binding, leucine-rich repeats (NLR)-containing protein that attenuates MAVS-RLR signaling. Using Nlrx1(-/-) cells, we confirmed that NLRX1 attenuated IFN-I production, but additionally promoted autophagy during viral infection. This dual function of NLRX1 paralleled the previously described functions of the autophagy-related proteins Atg5-Atg12, but NLRX1 did not associate with Atg5-Atg12. High-throughput quantitative mass spectrometry and endogenous protein-protein interaction revealed an NLRX1-interacting partner, mitochondrial Tu translation elongation factor (TUFM). TUFM interacted with Atg5-Atg12 and Atg16L1 and has similar functions as NLRX1 by inhibiting RLR-induced IFN-I but promoting autophagy. In the absence of NLRX1, increased IFN-I and decreased autophagy provide an advantage for host defense against vesicular stomatitis virus. This study establishes a link between an NLR protein and the viral-induced autophagic machinery via an intermediary partner, TUFM.


Subject(s)
Autophagy/physiology , Interferon Type I/biosynthesis , Mitochondrial Proteins/physiology , Peptide Elongation Factor Tu/physiology , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Animals , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Carrier Proteins/physiology , Cytokines/biosynthesis , Cytokines/genetics , DEAD Box Protein 58 , DEAD-box RNA Helicases/physiology , Fibroblasts/metabolism , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Interferon Type I/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Molecular Sequence Data , Multiprotein Complexes/physiology , Peptide Elongation Factor Tu/chemistry , Protein Interaction Mapping , Proteins/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Specific Pathogen-Free Organisms , Vesiculovirus/physiology
19.
Vaccine ; 30(8): 1465-75, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22226862

ABSTRACT

Type I interferons (IFNs) are critical for controlling pathogenic virus infections and can enhance immune responses. Hence their impact on the effectiveness of live-attenuated vaccines involves a balance between limiting viral antigen expression and enhancing the development of adaptive immune responses. We examined the influence of type I IFNs on these parameters following immunization with RepliVAX WN, a single-cycle flavivirus vaccine (SCFV) against West Nile virus (WNV) disease. RepliVAX WN-immunized mice produced IFN-α and displayed increased IFN-stimulated gene transcription in draining lymph nodes (LN). SCFV gene expression was over 100 fold-higher on days 1-3 post-infection in type I IFN receptor knockout mice (IFNAR(-/-)) compared to wild-type (wt) mice indicating a profound IFN-mediated suppression of SCFV gene expression in the wt animals. IFNAR(-/-) mice produced nearly equivalent levels of WNV-specific serum IgG and WNV-specific CD4(+) T cell responses compared to wt mice. However, significantly higher numbers of WNV-specific CD8(+) T cells were produced by IFNAR(-/-) mice and a significantly greater percentage of these T cells from IFNAR(-/-) mice produced only IFN-γ following antigen-specific re-stimulation. This altered cytokine expression was not associated with increased antigen load suggesting the loss of type I IFN receptor signaling was responsible for the altered quality of the CD8(+) effector T cell response. Together, these results indicate that although type I IFN is not essential for the intrinsic adjuvanting of RepliVAX WN, it plays a role in shaping the cytokine secretion profiles of CD8(+) effector T cells elicited by this SCFV.


Subject(s)
Adjuvants, Immunologic/pharmacology , Receptor, Interferon alpha-beta/immunology , Signal Transduction , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunoglobulin G/blood , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , West Nile Fever/immunology
20.
Virology ; 421(2): 96-104, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-21999990

ABSTRACT

We previously described a single-cycle dengue vaccine (RepliVAX D2) engineered from a capsid (C) gene-deleted West Nile virus (WNV) expressing dengue virus serotype 2 (DENV2) prM/E genes in place of the corresponding WNV genes. That work demonstrated that adaptation of RepliVAX D2 to grow in WNV C-expressing cells resulted in acquisition of non-synonymous mutations in the DENV2 prM/E and WNV NS2A/NS3 genes. Here we demonstrate that the prM/E mutations increase the specific infectivity of chimeric virions and the NS2A/NS3 mutations independently enhance packaging. Studies with the NS2A mutant demonstrated that it was unable to produce a larger form of NS1 (NS1'), suggesting that the mutation had been selected to eliminate a ribosomal frame-shift "slippage site" in NS2A. Evaluation of a synonymous mutation at this slippage site confirmed that genomes that failed to make NS1' were packaged more efficiently than WT genomes supporting a role for NS1/NS1' in orchestrating virion assembly.


Subject(s)
Dengue Virus/genetics , Dengue Virus/physiology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Structural Proteins/metabolism , Virus Assembly/genetics , Animals , Cell Line , Cricetinae , Dengue Virus/pathogenicity , Frameshifting, Ribosomal , Mutation , RNA Helicases/genetics , RNA Helicases/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...