Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
mBio ; 12(5): e0198721, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34579572

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has raised concerns about the detrimental effects of antibodies. Antibody-dependent enhancement (ADE) of infection is one of the biggest concerns in terms of not only the antibody reaction to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) upon reinfection with the virus but also the reaction to COVID-19 vaccines. In this study, we evaluated ADE of infection by using COVID-19 convalescent-phase plasma and BHK cells expressing human Fcγ receptors (FcγRs). We found that FcγRIIA and FcγRIIIA mediated modest ADE of infection against SARS-CoV-2. Although ADE of infection was observed in monocyte-derived macrophages infected with SARS-CoV-2, including its variants, proinflammatory cytokine/chemokine expression was not upregulated in macrophages. SARS-CoV-2 infection thus produces antibodies that elicit ADE of infection, but these antibodies do not contribute to excess cytokine production by macrophages. IMPORTANCE Viruses infect cells mainly via specific receptors at the cell surface. Antibody-dependent enhancement (ADE) of infection is an alternative mechanism of infection for viruses to infect immune cells that is mediated by antibodies and IgG receptors (FcγRs). Because ADE of infection contributes to the pathogenesis of some viruses, such as dengue virus and feline coronavirus, it is important to evaluate the precise mechanism of ADE and its contribution to the pathogenesis of SARS-CoV-2. Here, using convalescent-phase plasma from COVID-19 patients, we found that two types of FcγRs, FcγRIIA and FcγRIIIA, mediate ADE of SARS-CoV-2 infection. Although ADE of infection was observed for SARS-CoV-2 and its recent variants, proinflammatory cytokine production in monocyte-derived macrophages was not upregulated. These observations suggest that SARS-CoV-2 infection produces antibodies that elicit ADE of infection, but these antibodies may not be involved in aberrant cytokine release by macrophages during SARS-CoV-2 infection.


Subject(s)
Cytokines/metabolism , Macrophages/metabolism , Receptors, IgG/metabolism , SARS-CoV-2/pathogenicity , Animals , Antibody-Dependent Enhancement/physiology , Cell Line , Cricetinae , Humans , Real-Time Polymerase Chain Reaction , Receptors, IgG/genetics
2.
J Pharmacol Exp Ther ; 368(3): 475-489, 2019 03.
Article in English | MEDLINE | ID: mdl-30606761

ABSTRACT

Acinetobacter baumannii has become a pathogen of increasing medical importance because of the emergence of multidrug-resistant strains and the high rate of mortality of infected patients. Promising animal study results have been reported recently with active and passive immunization against A. baumannii virulence factors. In the present study, a monoclonal IgG3 antibody, 8E3, was developed with specificity for the K2 capsular polysaccharide of A. baumannii, and its therapeutic potential was assessed. 8E3 enhanced macrophage-mediated bactericidal activity against the A. baumannii clinical strain AB899. However, 8E3 treatment (passive immunization) of AB899-infected mice led to a substantial increase in mortality and to substantial increases in bacterial load in blood, lung, and in splenic samples. In vitro investigations showed a large binding capacity in the supernatant of bacterial cultures, suggesting that shed capsule components act as a binding sink for 8E3. Investigations of 8E3 pharmacokinetics in mice demonstrated that unbound concentrations of the antibody dropped below detection limits within 24 hours after a 200 mg/kg dose. However, total concentrations of antibody declined slowly, with an apparent terminal half-life (t 1/2) of 6.7-8.0 days, suggesting that the vast majority of 8E3 in blood is bound (e.g., with soluble capsule components in blood). We hypothesize that high concentrations of 8E3-capsule immune complexes act to inhibit bacterial clearance in vivo. To the best of our knowledge, this is the first demonstration of antibody-dependent enhancement of A. baumannii infection, and these observations highlight the complexity of antibody-based therapy for A. baumannii infections.


Subject(s)
Acinetobacter baumannii/isolation & purification , Acinetobacter baumannii/metabolism , Antibody-Dependent Enhancement/physiology , Disease Models, Animal , Pneumonia, Bacterial/metabolism , Acinetobacter baumannii/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/metabolism , Female , Humans , Male , Mice , Mice, Inbred BALB C , Pneumonia, Bacterial/drug therapy , Protein Binding/physiology
3.
BMC Infect Dis ; 18(1): 641, 2018 Dec 10.
Article in English | MEDLINE | ID: mdl-30526531

ABSTRACT

BACKGROUND: Dengue Virus (DENV) and Zika Virus (ZIKV) are closely related flaviviruses, circulating in overlapping geographical regions. The recent ZIKV epidemic has been linked to an explosion in reports of microcephaly and neurological defects. It is conceivable that our knowledge of DENV might potentiate the development of a ZIKV vaccine due to the close phylogenetic relationship between these flaviviruses and cross-reactive antibodies, principally to the envelope protein (E protein). Alternatively, cross-reactive antibodies that are generated following vaccination or infection, might become damaging during subsequent infections. MAIN BODY: The aims of this review are to collate and analyse data from a recent series of DENV-derived monoclonal antibody (mAb) panels from different research groups. These panels measured DENV-mAb activity against ZIKV in terms of antibody-dependent enhancement (ADE) and neutralisation. Methodology used across groups was compared and critiqued. Furthermore, the specific antibody targets on E protein were considered and their therapeutic potential evaluated. Shortcomings of hmAb panels suggest ADE may be over-estimated and neutralisation underestimated, as compared to clinical situations. It remains unknown whether preference of enhancement or neutralisation by antibodies to ZIKV E protein is dictated by quantitative aspects of antibody titre or epitope specific variation. Additionally, little is known about how duration between flavivirus reinfections affect secondary antibody response. CONCLUSION: This review concludes that our current knowledge of cross-reactive antibodies to E protein is inadequate to anticipate the outcome of deploying an E protein based vaccine to regions co-infected by DENV and ZIKV.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Dengue Virus/immunology , Dengue/prevention & control , Viral Envelope Proteins/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody-Dependent Enhancement/physiology , Cross Reactions/immunology , Dengue/epidemiology , Dengue Virus/pathogenicity , Humans , Zika Virus/pathogenicity , Zika Virus Infection/epidemiology
4.
Theor Biol Med Model ; 15(1): 13, 2018 09 03.
Article in English | MEDLINE | ID: mdl-30173664

ABSTRACT

BACKGROUND: Many vector-borne diseases co-circulate, as the viruses from the same family are also transmitted by the same vector species. For example, Zika and dengue viruses belong to the same Flavivirus family and are primarily transmitted by a common mosquito species Aedes aegypti. Zika outbreaks have also commonly occurred in dengue-endemic areas, and co-circulation and co-infection of both viruses have been reported. As recent immunological cross-reactivity studies have confirmed that convalescent plasma following dengue infection can enhance Zika infection, and as global efforts of developing dengue and Zika vaccines are intensified, it is important to examine whether and how vaccination against one disease in a large population may affect infection dynamics of another disease due to antibody-dependent enhancement. METHODS: Through a conceptual co-infection dynamics model parametrized by reported dengue and Zika epidemic and immunological cross-reactivity characteristics, we evaluate impact of a hypothetical dengue vaccination program on Zika infection dynamics in a single season when only one particular dengue serotype is involved. RESULTS: We show that an appropriately designed and optimized dengue vaccination program can not only help control the dengue spread but also, counter-intuitively, reduce Zika infections. We identify optimal dengue vaccination coverages for controlling dengue and simultaneously reducing Zika infections, as well as the critical coverages exceeding which dengue vaccination will increase Zika infections. CONCLUSION: This study based on a conceptual model shows the promise of an integrative vector-borne disease control strategy involving optimal vaccination programs, in regions where different viruses or different serotypes of the same virus co-circulate, and convalescent plasma following infection from one virus (serotype) can enhance infection against another virus (serotype). The conceptual model provides a first step towards well-designed regional and global vector-borne disease immunization programs.


Subject(s)
Antibody-Dependent Enhancement/physiology , Dengue/prevention & control , Models, Theoretical , Vaccination/standards , Zika Virus Infection/prevention & control , Animals , Antibody-Dependent Enhancement/drug effects , Dengue/epidemiology , Dengue Vaccines/therapeutic use , Dengue Virus/drug effects , Dengue Virus/physiology , Humans , Vaccination/methods , Vaccination/statistics & numerical data , Vaccines/therapeutic use , Zika Virus/drug effects , Zika Virus/physiology , Zika Virus Infection/epidemiology
5.
Proc Natl Acad Sci U S A ; 111(7): 2722-7, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550301

ABSTRACT

Viruses must evade the host innate defenses for replication and dengue is no exception. During secondary infection with a heterologous dengue virus (DENV) serotype, DENV is opsonized with sub- or nonneutralizing antibodies that enhance infection of monocytes, macrophages, and dendritic cells via the Fc-gamma receptor (FcγR), a process termed antibody-dependent enhancement of DENV infection. However, this enhancement of DENV infection is curious as cross-linking of activating FcγRs signals an early antiviral response by inducing the type-I IFN-stimulated genes (ISGs). Entry through activating FcγR would thus place DENV in an intracellular environment unfavorable for enhanced replication. Here we demonstrate that, to escape this antiviral response, antibody-opsonized DENV coligates leukocyte Ig-like receptor-B1 (LILRB1) to inhibit FcγR signaling for ISG expression. This immunoreceptor tyrosine-based inhibition motif-bearing receptor recruits Src homology phosphatase-1 to dephosphorylate spleen tyrosine kinase (Syk). As Syk is a key intermediate of FcγR signaling, LILRB1 coligation resulted in reduced ISG expression for enhanced DENV replication. Our findings suggest a unique mechanism for DENV to evade an early antiviral response for enhanced infection.


Subject(s)
Antibody-Dependent Enhancement/physiology , Antigens, CD/metabolism , Dengue Virus/metabolism , Dengue/physiopathology , Receptors, Immunologic/metabolism , Antibody-Dependent Enhancement/immunology , Blotting, Western , Cell Line , Dengue/immunology , Dengue Virus/physiology , Humans , Leukocyte Immunoglobulin-like Receptor B1 , Microarray Analysis , RNA, Small Interfering/genetics , Receptors, IgG/metabolism
7.
Vopr Virusol ; 56(4): 4-8, 2011.
Article in Russian | MEDLINE | ID: mdl-21899061

ABSTRACT

The overview analyzes an update on and current concepts of the initial stage of viral infection of sensitive cells. It considers the nature of virus receptors, the mechanisms of virus-receptor interaction, methodical approaches to identifying the receptor role of cell molecules for various viruses, and the association of the initial stage of viral infection with its subsequent ones.


Subject(s)
Nucleocapsid/metabolism , Receptors, Virus , Virus Attachment , Antibody-Dependent Enhancement/physiology , Antigens, Surface/metabolism , Glycolipids/metabolism , Humans , Membrane Glycoproteins/metabolism , Receptors, Virus/classification , Receptors, Virus/genetics , Receptors, Virus/metabolism , Toll-Like Receptors/metabolism , Viral Interference/physiology , Virus Diseases/microbiology
8.
J Virol ; 74(17): 7814-23, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933688

ABSTRACT

Although prior studies have investigated cellular infection by dengue virus (DV), many have used highly passaged strains. We have reassessed cellular infection by DV type 2 (DV2) using prototype and low-passage isolates representing genotypes from different geographic areas. We observed marked variation in the susceptibility to infection among cell types by different DV2 strains. HepG2 hepatoma cells were susceptible to infection by all DV2 strains assayed. Although the prototype strain generated higher titers of secreted virus than the low-passage isolates, this difference did not correspond to positive- or negative-strand viral RNA levels and thus may reflect variation in efficiency among DV2 isolates to translate viral proteins or package and/or secrete virus. In contrast, human foreskin fibroblasts were susceptible to the prototype and low-passage Thai isolates but not to five Nicaraguan strains tested, as reflected by the absence of accumulation of negative-strand viral RNA, viral antigen, and infectious virus. A similar pattern was observed with the antibody-dependent pathway of infection. U937 and THP-1 myeloid cells and peripheral blood monocytes were infected in the presence of enhancing antibodies by the prototype strain but not by low-passage Nicaraguan isolates. Again, the barrier appeared to be prior to negative-strand accumulation. Thus, depending on the cell type and viral isolate, blocks that limit the production of infectious virus in vitro may occur at distinct steps in the pathway of cellular infection.


Subject(s)
Antibody-Dependent Enhancement/physiology , Dengue Virus/pathogenicity , Animals , Cells, Cultured , Cricetinae , Dengue Virus/isolation & purification , Flow Cytometry , Humans , RNA, Viral/analysis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...