Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 695
Filter
1.
Sci Transl Med ; 16(749): eadn2199, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809964

ABSTRACT

Infection with any of the four dengue virus serotypes (DENV1-4) can protect against or enhance subsequent dengue depending on preexisting antibodies and infecting serotype. Additionally, primary infection with the related flavivirus Zika virus (ZIKV) is associated with increased risk of DENV2 disease. Here, we measured how prior DENV and ZIKV immunity influenced risk of disease caused by DENV1-4 in a pediatric Nicaraguan cohort. Of 3412 participants in 2022, 10.6% experienced dengue cases caused by DENV1 (n = 139), DENV4 (n = 133), DENV3 (n = 54), DENV2 (n = 9), or an undetermined serotype (n = 39). Longitudinal clinical and serological data were used to define infection histories, and generalized linear and additive models adjusted for age, sex, time since last infection, and year, and repeat measurements were used to predict disease risk. Compared with flavivirus-naïve participants, primary ZIKV infection was associated with increased risk of disease caused by DENV4 (relative risk = 2.62, 95% confidence interval: 1.48 to 4.63) and DENV3 (2.90, 1.34 to 6.27), but not DENV1 infection. Primary DENV infection or DENV followed by ZIKV infection was also associated with increased risk of DENV4 disease. We reanalyzed 19 years of cohort data and demonstrated that prior flavivirus immunity and antibody titer had distinct associations with disease risk depending on incoming serotype. We thus find that prior ZIKV infection, like prior DENV infection, is associated with increased risk of disease with certain DENV serotypes. Cross-reactivity among flaviviruses should be considered when assessing vaccine safety and efficacy.


Subject(s)
Dengue Virus , Dengue , Serogroup , Zika Virus Infection , Zika Virus , Humans , Zika Virus/immunology , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Child , Female , Male , Nicaragua/epidemiology , Child, Preschool , Risk Factors , Adolescent , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cohort Studies
2.
Vaccine ; 42(17): 3674-3683, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38749821

ABSTRACT

The Zika virus (ZIKV) is considered a public health problem worldwide due to its association with the development of microcephaly and the Guillain-Barré syndrome. Currently, there is no specific treatment or vaccine approved to combat this disease, and thus, developing safe and effective vaccines is a relevant goal. In this study, a multi-epitope protein called rpZDIII was designed based on a series of ZIKV antigenic sequences, a bacterial carrier, and linkers. The analysis of the predicted 3D structure of the rpZDIII chimeric antigen was performed on the AlphaFold 2 server, and it was produced in E. coli and purified from inclusion bodies, followed by solubilization and refolding processes. The yield achieved for rpZDIII was 11 mg/L in terms of pure soluble recombinant protein per liter of fermentation. rpZDIII was deemed immunogenic since it induced serum IgG and IgM responses in mice upon subcutaneous immunization in a three-dose scheme. Moreover, sera from mice immunized with rpZDIII showed neutralizing activity against ZIKV. Therefore, this study reveals rpZDIII as a promising immunogen for the development of a rationally designed multi-epitope vaccine against ZIKV, and completion of its preclinical evaluation is guaranteed.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Antigens, Viral , Zika Virus Infection , Zika Virus , Animals , Zika Virus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Mice , Antibodies, Viral/immunology , Antibodies, Viral/blood , Zika Virus Infection/prevention & control , Zika Virus Infection/immunology , Antigens, Viral/immunology , Antigens, Viral/genetics , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Epitopes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Female , Escherichia coli/genetics , Escherichia coli/metabolism , Immunoglobulin M/immunology , Immunoglobulin M/blood , Mice, Inbred BALB C
3.
PLoS One ; 19(5): e0302684, 2024.
Article in English | MEDLINE | ID: mdl-38722858

ABSTRACT

BACKGROUND: In most cases, Zika virus (ZIKV) causes a self-limited acute illness in adults, characterized by mild clinical symptoms that resolve within a few days. Immune responses, both innate and adaptive, play a central role in controlling and eliminating virus-infected cells during the early stages of infection. AIM: To test the hypothesis that circulating T cells exhibit phenotypic and functional activation characteristics during the viremic phase of ZIKV infection. METHODS: A comprehensive analysis using mass cytometry was performed on peripheral blood mononuclear cells obtained from patients with acute ZIKV infection (as confirmed by RT-PCR) and compared with that from healthy donors (HD). The frequency of IFN-γ-producing T cells in response to peptide pools covering immunogenic regions of structural and nonstructural ZIKV proteins was quantified using an ELISpot assay. RESULTS: Circulating CD4+ and CD8+ T lymphocytes from ZIKV-infected patients expressed higher levels of IFN-γ and pSTAT-5, as well as cell surface markers associated with proliferation (Ki-67), activation ((HLA-DR, CD38) or exhaustion (PD1 and CTLA-4), compared to those from HD. Activation of CD4+ and CD8+ memory T cell subsets, including Transitional Memory T Cells (TTM), Effector Memory T cells (TEM), and Effector Memory T cells Re-expressing CD45RA (TEMRA), was prominent among CD4+ T cell subset of ZIKV-infected patients and was associated with increased levels of IFN-γ, pSTAT-5, Ki-67, CTLA-4, and PD1, as compared to HD. Additionally, approximately 30% of ZIKV-infected patients exhibited a T cell response primarily directed against the ZIKV NS5 protein. CONCLUSION: Circulating T lymphocytes spontaneously produce IFN-γ and express elevated levels of pSTAT-5 during the early phase of ZIKV infection whereas recognition of ZIKV antigen results in the generation of virus-specific IFN-γ-producing T cells.


Subject(s)
CD8-Positive T-Lymphocytes , Interferon-gamma , Zika Virus Infection , Zika Virus , Humans , Zika Virus Infection/immunology , Zika Virus Infection/epidemiology , Adult , Zika Virus/immunology , Female , Male , Interferon-gamma/metabolism , Interferon-gamma/immunology , Brazil/epidemiology , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Middle Aged , Young Adult , Epidemics , Lymphocyte Activation/immunology , T-Lymphocytes/immunology
4.
Viruses ; 16(5)2024 05 19.
Article in English | MEDLINE | ID: mdl-38793688

ABSTRACT

Arboviral diseases are serious threats to global health with increasing prevalence and potentially severe complications. Significant arthropod-borne viruses are the dengue viruses (DENV 1-4), the Zika virus (ZIKV), and the chikungunya virus (CHIKV). Among the areas most affected is the South Pacific Region (SPR). Here, arboviruses not only cause a high local burden of disease, but the region has also proven to contribute to their global spread. Outpatient serum samples collected between 08/2016 and 04/2017 on three islands of the island states of Vanuatu and the Cook Islands were tested for anti-DENV- and anti-ZIKV-specific antibodies (IgG) using enzyme-linked immunosorbent assays (ELISA). ELISA test results showed 89% of all test sera from the Cook Islands and 85% of the Vanuatu samples to be positive for anti-DENV-specific antibodies. Anti-ZIKV antibodies were identified in 66% and 52%, respectively, of the test populations. Statistically significant differences in standardized immunity levels were found only at the intranational level. Our results show that in both the Cook Islands and Vanuatu, residents were exposed to significant Flavivirus transmission. Compared to other seroprevalence studies, the marked difference between ZIKV immunity levels and previously published CHIKV seroprevalence rates in our study populations is surprising. We propose the timing of ZIKV and CHIKV emergence in relation to recurrent DENV outbreaks and the impact of seasonality as explanatory external factors for this observation. Our data add to the knowledge of arboviral epidemics in the SPR and contribute to a better understanding of virus spread, including external conditions with potential influence on outbreak dynamics. These data may support preventive and rapid response measures in the affected areas, travel-related risk assessment, and infection identification in locals and returning travelers.


Subject(s)
Antibodies, Viral , Dengue Virus , Dengue , Zika Virus Infection , Zika Virus , Humans , Zika Virus Infection/epidemiology , Zika Virus Infection/blood , Zika Virus Infection/immunology , Zika Virus Infection/virology , Seroepidemiologic Studies , Dengue Virus/immunology , Zika Virus/immunology , Vanuatu/epidemiology , Dengue/epidemiology , Dengue/immunology , Dengue/blood , Dengue/virology , Polynesia/epidemiology , Antibodies, Viral/blood , Adult , Female , Adolescent , Young Adult , Male , Middle Aged , Aged , Child , Enzyme-Linked Immunosorbent Assay , Child, Preschool , Immunoglobulin G/blood , Infant
5.
Proc Natl Acad Sci U S A ; 121(21): e2312755121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38743628

ABSTRACT

Antigenic similarities between Zika virus (ZIKV) and other flaviviruses pose challenges to the development of virus-specific diagnostic tools and effective vaccines. Starting with a DNA-encoded one-bead-one-compound combinatorial library of 508,032 synthetic, non-natural oligomers, we selected and characterized small molecules that mimic ZIKV epitopes. High-throughput fluorescence-activated cell sorter-based bead screening was used to select molecules that bound IgG from ZIKV-immune but not from dengue-immune sera. Deep sequencing of the DNA from the "Zika-only" beads identified 40 candidate molecular structures. A lead candidate small molecule "CZV1-1" was selected that correctly identifies serum specimens from Zika-experienced patients with good sensitivity and specificity (85.3% and 98.4%, respectively). Binding competition studies of purified anti-CZV1-1 IgG against known ZIKV-specific monoclonal antibodies (mAbs) showed that CZV1-1 mimics a nonlinear, neutralizing conformational epitope in the domain III of the ZIKV envelope. Purified anti-CZV1-1 IgG neutralized infection of ZIKV in cell cultures with potencies comparable to highly specific ZIKV-neutralizing mAbs. This study demonstrates an innovative approach for identification of synthetic non-natural molecular mimics of conformational virus epitopes. Such molecular mimics may have value in the development of accurate diagnostic assays for Zika, as well as for other viruses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Epitopes , Zika Virus Infection , Zika Virus , Zika Virus/immunology , Epitopes/immunology , Humans , Zika Virus Infection/immunology , Zika Virus Infection/virology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunoglobulin G/immunology , Antibodies, Monoclonal/immunology , Molecular Mimicry/immunology
6.
Emerg Microbes Infect ; 13(1): 2348528, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38662785

ABSTRACT

Zika is a systemic inflammatory disease caused by infection with Zika virus (ZIKV). ZIKV infection in adults is associated with encephalitis marked by elevated expression of pro-inflammatory cytokines and chemokines, as well as increased brain infiltration of immune cells. In this study, we demonstrate that ZIKV encephalitis in a mouse infection model exhibits increased brain TSPO expression. TSPO expression on brain-resident and infiltrating immune cells in ZIKV infection correlates with disease and inflammation status in the brain. Brain TSPO expression can also be sensitively detected ex vivo and in vitro using radioactive small molecule probes that specifically bind to TSPO, such as [3H]PK11195. TSPO expression on brain-resident and infiltrating immune cells is a biomarker of ZIKV neuroinflammation, which can also be a general biomarker of acute viral neuroinflammatory disease.


Subject(s)
Biomarkers , Brain , Neuroinflammatory Diseases , Receptors, GABA , Zika Virus Infection , Zika Virus , Animals , Zika Virus Infection/virology , Zika Virus Infection/immunology , Zika Virus Infection/metabolism , Mice , Receptors, GABA/metabolism , Receptors, GABA/genetics , Zika Virus/immunology , Brain/virology , Brain/metabolism , Brain/pathology , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/metabolism , Disease Models, Animal , Humans , Mice, Inbred C57BL , Female , Cytokines/metabolism
7.
J Virol ; 98(5): e0019424, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38567950

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne flavivirus that caused an epidemic in the Americas in 2016 and is linked to severe neonatal birth defects, including microcephaly and spontaneous abortion. To better understand the host response to ZIKV infection, we adapted the 10× Genomics Chromium single-cell RNA sequencing (scRNA-seq) assay to simultaneously capture viral RNA and host mRNA. Using this assay, we profiled the antiviral landscape in a population of human monocyte-derived dendritic cells infected with ZIKV at the single-cell level. The bystander cells, which lacked detectable viral RNA, expressed an antiviral state that was enriched for genes coinciding predominantly with a type I interferon (IFN) response. Within the infected cells, viral RNA negatively correlated with type I IFN-dependent and -independent genes (the antiviral module). We modeled the ZIKV-specific antiviral state at the protein level, leveraging experimentally derived protein interaction data. We identified a highly interconnected network between the antiviral module and other host proteins. In this work, we propose a new paradigm for evaluating the antiviral response to a specific virus, combining an unbiased list of genes that highly correlate with viral RNA on a per-cell basis with experimental protein interaction data. IMPORTANCE: Zika virus (ZIKV) remains a public health threat given its potential for re-emergence and the detrimental fetal outcomes associated with infection during pregnancy. Understanding the dynamics between ZIKV and its host is critical to understanding ZIKV pathogenesis. Through ZIKV-inclusive single-cell RNA sequencing (scRNA-seq), we demonstrate on the single-cell level the dynamic interplay between ZIKV and the host: the transcriptional program that restricts viral infection and ZIKV-mediated inhibition of that response. Our ZIKV-inclusive scRNA-seq assay will serve as a useful tool for gaining greater insight into the host response to ZIKV and can be applied more broadly to the flavivirus field.


Subject(s)
Dendritic Cells , Single-Cell Analysis , Zika Virus Infection , Zika Virus , Humans , Zika Virus/physiology , Zika Virus Infection/virology , Zika Virus Infection/immunology , Dendritic Cells/virology , Dendritic Cells/immunology , RNA, Viral/metabolism , RNA, Viral/genetics , Interferon Type I/metabolism , Host-Pathogen Interactions , Sequence Analysis, RNA
8.
Microbiol Spectr ; 12(6): e0075824, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38687079

ABSTRACT

Human immunoglobulin preparations contain a diverse range of polyclonal antibodies that reflect past immune responses against pathogens encountered by the blood donor population. In this study, we examined a panel of intravenous immunoglobulins (IGIVs) manufactured over the past two decades (1998-2020) for their capacity to neutralize or enhance Zika virus (ZIKV) infection in vitro. These IGIVs were selected specifically based on their production dates in relation to the occurrences of two flavivirus outbreaks in the U.S.: the West Nile virus (WNV) outbreak in 1999 and the ZIKV outbreak in 2015. As demonstrated by enzyme-linked immunosorbent assay (ELISA) experiments, IGIVs made before the ZIKV outbreak already harbored antibodies that bind to various peptides across the envelope protein of ZIKV because of the WNV outbreak. Using phage display, the most dominant binding site was mapped precisely to the P2 peptide between residues 211 and 230 within domain II, where BF1176-56, an anti-ZIKV monoclonal antibody, also binds. When tested in permissive Vero E6 cells for ZIKV neutralization, the IGIVs, even after undergoing rigorous enrichment for P2 binding specificity, failed, as did BF1176-56. Meanwhile, BF1176-56 enhanced ZIKV infection in both FcγRII-expressing K562 cells and human peripheral blood mononuclear cells. However, for enhancement by the IGIVs to be detected in these cells, a substantial increase in their P2 binding specificity was required, thus linking the P2 site with ZIKV enhancement in vitro. Our findings warrant further study of the significance of elevated levels of anti-WNV antibodies in IGIVs, considering that various mechanisms operating in vivo may modulate ZIKV infection outcomes.IMPORTANCEWe investigated the capacity of intravenous immunoglobulins manufactured previously over two decades (1998-2020) to neutralize or enhance Zika virus infection in vitro. West Nile virus antibodies in IGIVs could not neutralize Zika virus initially; however, once the IGIVs were concentrated further, they enhanced its infection. These findings lay the groundwork for exploring how preexisting WNV antibodies in IGIVs could impact Zika infection, both in vitro and in vivo. Our observations are historically significant, since we tested a panel of IGIV lots that were carefully selected based on their production dates which covered two major flavivirus outbreaks in the U.S.: the WNV outbreak in 1999 and the ZIKV outbreak in 2015. These findings will facilitate our understanding of the interplay among closely related viral pathogens, particularly from a historical perspective regarding large blood donor populations. They should remain relevant for future outbreaks of emerging flaviviruses that may potentially affect vulnerable populations.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , West Nile Fever , West Nile virus , Zika Virus Infection , Zika Virus , Humans , Zika Virus/immunology , West Nile virus/immunology , Antibodies, Viral/immunology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Animals , Chlorocebus aethiops , Vero Cells , West Nile Fever/immunology , West Nile Fever/virology , Antibodies, Neutralizing/immunology , Binding Sites , Immunoglobulins, Intravenous/immunology , Viral Envelope Proteins/immunology , Enzyme-Linked Immunosorbent Assay
9.
Am J Trop Med Hyg ; 110(6): 1178-1179, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38653230

ABSTRACT

We report strong Zika virus (ZIKV) neutralizing antibody responses in African green monkeys (Chlorocebus sabaeus) up to 1,427 days after ZIKV exposure via the subcutaneous, intravaginal, or intrarectal routes. Our results suggest that immunocompetent African green monkeys previously infected with ZIKV are likely protected from reinfection for years, possibly life, and would not contribute to virus amplification during ZIKV epizootics.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Zika Virus Infection , Zika Virus , Animals , Chlorocebus aethiops , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Zika Virus/immunology , Zika Virus Infection/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Female
10.
Viruses ; 16(4)2024 04 07.
Article in English | MEDLINE | ID: mdl-38675911

ABSTRACT

Zika virus (ZIKV) remains a public health concern, with epidemics in endemic regions and sporadic outbreaks in new areas posing significant threats. Several mosquito-borne flaviviruses that can cause human illness, including West Nile, Usutu, and St. Louis encephalitis, have associations with birds. However, the susceptibility of chickens to ZIKV and their role in viral epidemiology is not currently known. We investigated the susceptibility of chickens to experimental ZIKV infection using chickens ranging from 1-day-old chicks to 6-week-old birds. ZIKV caused no clinical signs in chickens of all age groups tested. Viral RNA was detected in the blood and tissues during the first 5 days post-inoculation in 1-day and 4-day-old chicks inoculated with a high viral dose, but ZIKV was undetectable in 6-week-old birds at all timepoints. Minimal antibody responses were observed in 6-week-old birds, and while present in younger chicks, they waned by 28 days post-infection. Innate immune responses varied significantly between age groups. Robust type I interferon and inflammasome responses were measured in older chickens, while limited innate immune activation was observed in younger chicks. Signal transducer and activator of transcription 2 (STAT2) is a major driver of host restriction to ZIKV, and chicken STAT2 is distinct from human STAT2, potentially contributing to the observed resistance to ZIKV infection. The rapid clearance of the virus in older chickens coincided with an effective innate immune response, highlighting age-dependent susceptibility. Our study indicates that chickens are not susceptible to productive ZIKV infection and are unlikely to play a role in the ZIKV epidemiology.


Subject(s)
Chickens , Immunity, Innate , Poultry Diseases , Zika Virus Infection , Zika Virus , Animals , Chickens/virology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Disease Susceptibility , Poultry Diseases/virology , Poultry Diseases/immunology , Age Factors , Antibodies, Viral/blood , RNA, Viral/genetics
11.
Virus Res ; 345: 199376, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38643856

ABSTRACT

Zika virus (ZIKV) and Japanese encephalitis virus (JEV) are antigenically related flaviviruses that co-circulate in many countries/territories. The interaction between the two viruses needs to be determined. Recent findings by ourselves and other labs showed that JEV-elicited antibodies (Abs) and CD8+T cells exacerbate and protect against subsequent ZIKV infection, respectively. However, the impact of JEV envelope (E) protein domain III (EDIII)-induced immune responses on ZIKV infection is unclear. We show here that sera from JEV-EDIII-vaccinated mice cross-react with ZIKV-EDIII in vitro, and transfer of the same sera to mice significantly decreases death upon lethal ZIKV infection at a dose-dependent manner. Maternally acquired anti-JEV-EDIII Abs also significantly reduce the mortality of neonatal mice born to JEV-EDIII-immune mothers post ZIKV challenge. Similarly, transfer of ZIKV-EDIII-reactive IgG purified from JEV-vaccinated humans increases the survival of ZIKV-infected mice. Notably, transfer of an extremely low volume of JEV-EDIII-immune sera or ZIKV-EDIII-reactive IgG does not mediate the Ab-mediated enhancement (ADE) of ZIKV infection. Similarly, transfer of JEV-EDIII-elicited CD8+T cells protects recipient mice against ZIKV challenge. These results demonstrate that JEV-EDIII-induced immune components including Abs and T cells have protective roles in ZIKV infection, suggesting EDIII is a promising immunogen for developing effective and safety JEV vaccine.


Subject(s)
Antibodies, Viral , CD8-Positive T-Lymphocytes , Cross Protection , Encephalitis Virus, Japanese , Viral Envelope Proteins , Zika Virus Infection , Zika Virus , Animals , Zika Virus Infection/prevention & control , Zika Virus Infection/immunology , CD8-Positive T-Lymphocytes/immunology , Zika Virus/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Envelope Proteins/immunology , Mice , Encephalitis Virus, Japanese/immunology , Cross Protection/immunology , Female , Cross Reactions , Encephalitis, Japanese/prevention & control , Encephalitis, Japanese/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin G/blood , Disease Models, Animal , Immunization
12.
Diagn Microbiol Infect Dis ; 109(2): 116238, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38554539

ABSTRACT

The interpretation for Zika virus serology results is challenging due to high antibody cross reactivity with other flaviviruses. This limits availability of reliable and accurate methods for serosurveillance studies to understand the disease burden. Therefore, we conducted study to harmonize anti-Zika IgG antibody detection assays with 1st WHO International Standard (16/352) and working standard (16/320) for anti-Zika virus antibody.Additionally, evaluated NuGenTMZIKA-IgG and NovaLisa®ZIKA virus IgG-Capture ELISA using a panel of 278 seraFurther, 106 samples positive for other-flavi viruses were taken for assessing cross-reactivity of the assay, all serums were further tested by Zika-PRNT. The results of this study indicates satisfactory performance of both the assays. Serological and neutralization assays were calibrated according to the international standards. This will help in understanding antibody dynamics in serosurveillance and vaccine studies. However the performance of the kits with possibilities of cross-reactivity will have to be verified by coupling ZIKV and DENV specific ELISA.


Subject(s)
Antibodies, Viral , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G , Zika Virus Infection , Zika Virus , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/standards , Zika Virus/immunology , Immunoglobulin G/blood , Humans , Antibodies, Viral/blood , Zika Virus Infection/diagnosis , Zika Virus Infection/immunology , Zika Virus Infection/blood , Reagent Kits, Diagnostic/standards , Serologic Tests/standards , Serologic Tests/methods , Sensitivity and Specificity , Female , Adult , Adolescent , Young Adult
13.
J Virol ; 98(1): e0078923, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38168677

ABSTRACT

Zika virus (ZIKV) infection caused neurological complications and male infertility, leading to the accumulation of antigen-specific immune cells in immune-privileged organs (IPOs). Thus, it is important to understand the immunological responses to ZIKV in IPOs. We extensively investigated the ZIKV-specific T cell immunity in IPOs in Ifnar1-/- mice, based on an immunodominant epitope E294-302 tetramer. The distinct kinetics and functions of virus-specific CD8+ T cells infiltrated into different IPOs were characterized, with late elevation in the brain and spinal cord. Single epitope E294-302-specific T cells can account for 20-60% of the total CD8+ T cells in the brain, spinal cord, and testicle and persist for at least 90 days in the brain and spinal cord. The E294-302-specific TCRαßs within the IPOs are featured with the majority of clonotypes utilizing TRAV9N-3 paired with diverse TRBV chains, but with distinct αß paired clonotypes in 7 and 30 days post-infection. Specific chemokine receptors, Ccr2 and Ccr5, were selectively expressed in the E294-302-specific CD8+ T cells within the brain and testicle, indicating an IPO-oriented migration of virus-specific CD8+ T cells after infection. Overall, this study adds to the understanding of virus-specific CD8+ T cell responses for controlling and clearing ZIKV infection in IPOs.IMPORTANCEThe immune-privileged organs (IPOs), such as the central nervous system and testicles, presented pathogenicity and inflammation after Zika virus (ZIKV) infection with infiltrated CD8+ T cells. Our data show that CD8+ T cells keep up with virus increases and decreases in immune-privileged organs. Furthermore, our study provides the first ex vivo comparative analyses of the composition and diversity related to TCRα/ß clonotypes across anatomical sites and ZIKV infection phases. We show that the vast majority of TCRα/ß clonotypes in tissues utilize TRAV9N-3 with conservation. Specific chemokine expression, including Ccr2 and Ccr5, was found to be selectively expressed in the E294-302-specific CD8+ T cells within the brain and testicle, indicating an IPO-oriented migration of the virus-specific CD8+ T cells after the infection. Our study adds insights into the anti-viral immunological characterization and chemotaxis mechanism of virus-specific CD8+ T cells after ZIKV infection in different IPOs.


Subject(s)
CD8-Positive T-Lymphocytes , Immune Privilege , Zika Virus Infection , Animals , Male , Mice , Brain/immunology , Brain/virology , CD8-Positive T-Lymphocytes/immunology , Receptor, Interferon alpha-beta/genetics , Zika Virus , Zika Virus Infection/immunology , Mice, Knockout , Testis/immunology , Testis/virology
14.
J Virol ; 98(2): e0195423, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38289102

ABSTRACT

During the life cycle of mosquito-borne flaviviruses, substantial subgenomic flaviviral RNA (sfRNA) is produced via incomplete degradation of viral genomic RNA by host XRN1. Zika virus (ZIKV) sfRNA has been detected in mosquito and mammalian somatic cells. Human neural progenitor cells (hNPCs) in the developing brain are the major target cells of ZIKV, and antiviral RNA interference (RNAi) plays a critical role in hNPCs. However, whether ZIKV sfRNA was produced in ZIKV-infected hNPCs as well as its function remains not known. In this study, we demonstrate that abundant sfRNA was produced in ZIKV-infected hNPCs. RNA pulldown and mass spectrum assays showed ZIKV sfRNA interacted with host proteins RHA and PACT, both of which are RNA-induced silencing complex (RISC) components. Functionally, ZIKV sfRNA can antagonize RNAi by outcompeting small interfering RNAs (siRNAs) in binding to RHA and PACT. Furthermore, the 3' stem loop (3'SL) of sfRNA was responsible for RISC components binding and RNAi inhibition, and 3'SL can enhance the replication of a viral suppressor of RNAi (VSR)-deficient virus in a RHA- and PACT-dependent manner. More importantly, the ability of binding to RISC components is conversed among multiple flaviviral 3'SLs. Together, our results identified flavivirus 3'SL as a potent VSR in RNA format, highlighting the complexity in virus-host interaction during flavivirus infection.IMPORTANCEZika virus (ZIKV) infection mainly targets human neural progenitor cells (hNPCs) and induces cell death and dysregulated cell-cycle progression, leading to microcephaly and other central nervous system abnormalities. RNA interference (RNAi) plays critical roles during ZIKV infections in hNPCs, and ZIKV has evolved to encode specific viral proteins to antagonize RNAi. Herein, we first show that abundant sfRNA was produced in ZIKV-infected hNPCs in a similar pattern to that in other cells. Importantly, ZIKV sfRNA acts as a potent viral suppressor of RNAi (VSR) by competing with siRNAs for binding RISC components, RHA and PACT. The 3'SL of sfRNA is responsible for binding RISC components, which is a conserved feature among mosquito-borne flaviviruses. As most known VSRs are viral proteins, our findings highlight the importance of viral non-coding RNAs during the antagonism of host RNAi-based antiviral innate immunity.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Humans , Mammals/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Induced Silencing Complex/metabolism , Subgenomic RNA , Viral Proteins/metabolism , Virus Replication , Zika Virus/physiology , Zika Virus Infection/immunology , Zika Virus Infection/virology
15.
J Virol ; 97(11): e0141423, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37943046

ABSTRACT

IMPORTANCE: The wide endemic range of mosquito-vectored flaviviruses-such as Zika virus and dengue virus serotypes 1-4-places hundreds of millions of people at risk of infection every year. Despite this, there are no widely available vaccines, and treatment of severe cases is limited to supportive care. An avenue toward development of more widely applicable vaccines and targeted therapies is the characterization of monoclonal antibodies that broadly neutralize all these viruses. Here, we measure how single amino acid mutations in viral envelope protein affect neutralizing antibodies with both broad and narrow specificities. We find that broadly neutralizing antibodies with potential as vaccine prototypes or biological therapeutics are quantifiably more difficult to escape than narrow, virus-specific neutralizing antibodies.


Subject(s)
Antibodies, Viral , Broadly Neutralizing Antibodies , Viral Envelope Proteins , Zika Virus Infection , Zika Virus , Animals , Humans , Cross Reactions , Mutation , Vaccines , Viral Envelope , Viral Envelope Proteins/genetics , Zika Virus/genetics , Zika Virus Infection/immunology , Zika Virus Infection/therapy
16.
mBio ; 14(5): e0093423, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37732809

ABSTRACT

IMPORTANCE: One of the fundamental features that make viruses intracellular parasites is the necessity to use cellular translational machinery. Hence, this is a crucial checkpoint for controlling infections. Here, we show that dengue and Zika viruses, responsible for nearly 400 million infections every year worldwide, explore such control for optimal replication. Using immunocompetent cells, we demonstrate that arrest of protein translations happens after sensing of dsRNA and that the information required to avoid this blocking is contained in viral 5'-UTR. Our work, therefore, suggests that the non-canonical translation described for these viruses is engaged when the intracellular stress response is activated.


Subject(s)
Dengue Virus , Stress, Physiological , Virus Replication , Zika Virus , eIF-2 Kinase , Animals , Humans , A549 Cells , Chlorocebus aethiops , Dengue/immunology , Dengue/virology , Dengue Virus/physiology , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Eukaryotic Initiation Factor-2/metabolism , Gene Deletion , Protein Biosynthesis/genetics , Protein Biosynthesis/immunology , Stress, Physiological/genetics , Stress, Physiological/immunology , Vero Cells , Virus Replication/genetics , Virus Replication/immunology , Zika Virus/physiology , Zika Virus Infection/immunology , Zika Virus Infection/virology , RNA, Double-Stranded/metabolism
17.
J Virol ; 97(6): e0055623, 2023 06 29.
Article in English | MEDLINE | ID: mdl-37191498

ABSTRACT

During the 2015-2016 Zika virus (ZIKV) epidemic, ZIKV-associated neurological diseases were reported in adults, including microcephaly, Guillain-Barre syndrome, myelitis, meningoencephalitis, and fatal encephalitis. However, the mechanisms underlying the neuropathogenesis of ZIKV infection are not yet fully understood. In this study, we used an adult ZIKV infection mouse model (Ifnar1-/-) to investigate the mechanisms underlying neuroinflammation and neuropathogenesis. ZIKV infection induced the expression of proinflammatory cytokines, including interleukin-1ß (IL-1ß), IL-6, gamma interferon, and tumor necrosis factor alpha, in the brains of Ifnar1-/- mice. RNA-seq analysis of the infected mouse brain also revealed that genes involved in innate immune responses and cytokine-mediated signaling pathways were significantly upregulated at 6 days postinfection. Furthermore, ZIKV infection induced macrophage infiltration and activation and augmented IL-1ß expression, whereas microgliosis was not observed in the brain. Using human monocyte THP-1 cells, we confirmed that ZIKV infection promotes inflammatory cell death and increases IL-1ß secretion. In addition, expression of the complement component C3, which is associated with neurodegenerative diseases and known to be upregulated by proinflammatory cytokines, was induced by ZIKV infection through the IL-1ß-mediated pathway. An increase in C5a produced by complement activation in the brains of ZIKV-infected mice was also verified. Taken together, our results suggest that ZIKV infection in the brain of this animal model augments IL-1ß expression in infiltrating macrophages and elicits IL-1ß-mediated inflammation, which can lead to the destructive consequences of neuroinflammation. IMPORTANCE Zika virus (ZIKV) associated neurological impairments are an important global health problem. Our results suggest that ZIKV infection in the mouse brain can induce IL-1ß-mediated inflammation and complement activation, thereby contributing to the development of neurological disorders. Thus, our findings reveal a mechanism by which ZIKV induces neuroinflammation in the mouse brain. Although we used adult type I interferon receptor IFNAR knockout (Ifnar1-/-) mice owing to the limited mouse models of ZIKV pathogenesis, our conclusions contributed to the understanding ZIKV-associated neurological diseases to develop treatment strategies for patients with ZIKV infection based on these findings.


Subject(s)
Brain , Interleukin-1beta , Macrophages , Zika Virus Infection , Animals , Humans , Mice , Brain/immunology , Cytokines/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Macrophages/immunology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/virology , Zika Virus , Zika Virus Infection/immunology , Transcriptome/immunology , Disease Models, Animal , Neurons/immunology , Neurons/virology
18.
Arch Virol ; 168(2): 70, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36658439

ABSTRACT

Zika virus (ZIKV) is an arbovirus that was responsible for multiple outbreaks from 2007 to 2015. It has been linked to cases of microcephaly in Brazil in 2015, among other neurological disorders. Differences among strains might be the reason for different clinical outcomes of infection. To evaluate this hypothesis, we performed a comparative proteomic analysis of Vero cells infected with the African strain MR766 (ZIKVAFR) and the Brazilian strain 17 SM (ZIKVBR). A total of 550 proteins were identified as differentially expressed in ZIKVAFR- or ZIKVBR-infected cells compared to the control. The main findings included upregulation of immune system pathways (neutrophil degranulation and adaptive/innate immune system) and potential activation of immune-system-related pathways by ZIKVAFR (mTOR, JAK-STAT, NF-κB, and others) compared with the ZIKVBR/control. In addition, phagocytosis by macrophages and engulfment of leukocytes were activated in ZIKVAFR infection. An in vivo analysis using an immunocompetent C57BL/6N mouse model identified interstitial pneumonia with neutrophil infiltration in the lungs only in mice infected with ZIKVBR at 48 hours postinfection, with a significant amount of virus detected. Likewise, only animals infected with ZIKVBR had viral material in the cytoplasm of lung macrophages. These results suggest that activation of the immune system by ZIKVAFR infection may lead to faster viral clearance by immune cells.


Subject(s)
Immune Evasion , Zika Virus Infection , Zika Virus , Animals , Mice , Brazil , Chlorocebus aethiops , Mice, Inbred C57BL , Proteomics , Vero Cells , Zika Virus/physiology , Zika Virus Infection/immunology
19.
Virology ; 579: 101-110, 2023 02.
Article in English | MEDLINE | ID: mdl-36623351

ABSTRACT

Zika virus (ZIKV), a mosquito-borne pathogen, is associated with neurological complications in adults and congenital abnormalities in newborns. There are no vaccines or treatments for ZIKV infection. Understanding the specificity of natural antibody responses to ZIKV could help inform vaccine efforts. Here, we used a technology called Deep Sequence-Coupled Biopanning to map the targets of the human antibody responses to ZIKV infection. A bacteriophage virus-like particle (VLP) library displaying overlapping linear peptides derived from the ZIKV polyprotein was generated. The library was panned using IgG from 23 ZIKV-infected patients from Panama and deep sequencing identified common targets of anti-ZIKV antibodies within the ZIKV envelope glycoprotein. These included epitopes within the fusion loop within domain II and four epitopes within domain III. Additionally, we showed that VLPs displaying selected epitopes elicited antibodies that bound to native ZIKV envelope protein but failed to prevent infection in a mouse challenge model.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Humans , Mice , Antibodies, Neutralizing , Antibodies, Viral , Antibody Formation , Epitopes , Viral Envelope Proteins/chemistry , Zika Virus Infection/immunology
20.
J Biomol Struct Dyn ; 41(9): 3762-3771, 2023 06.
Article in English | MEDLINE | ID: mdl-35318896

ABSTRACT

Zika virus (ZIKV), an RNA virus, rapidly spreads Aedes mosquito-borne sickness. Currently, there are neither effective vaccines nor therapeutics available to prevent or treat ZIKV infection. In this study, to address these unmet medical needs, we aimed to design B- and T-cell candidate multi-epitope-based subunit against ZIKV using an in silico approach. In this study we applied immunoinformatics, molecular docking, and dynamic simulation assessments targeting the most immunogenic proteins; the capsid (C), envelope (E) proteins and the non-stuctural protein (NS1), described in our previous study, and which predicted immunodominant B and T cell epitopes. The final non-allergenic and highly antigenic multi-epitope was constituted of immunogenic screened-epitopes (3 CTL and 3 HTL) and the ß-defensin as an adjuvant that have been linked using EAAAK, AAY, and GPGPG linkers, respectively. The final construct containing 143 amino acids was characterized for its allergenicity, antigenicity, and physiochemical properties; and found to be safe and immunogenic with a good prediction of solubility. The existence of IFN-γ epitopes asserts the capacity to trigger strong immune responses. Subsequently, the molecular docking among vaccine and immune receptors (TLR2/TLR4) was revealed with a good binding affinity with and stable molecular interactions. Molecular dynamics simulation confirmed the stability of the complexes. Finally, the construct was subjected to in silico cloning demonstrating the efficiently of its expression in E.coli. However, this study needs the experimental validation to demonstrate vaccine safety and efficacy.Communicated by Ramaswamy H. Sarma.


Subject(s)
Computer Simulation , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Viral Vaccines , Zika Virus Infection , Zika Virus , Cloning, Molecular , Codon/genetics , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Molecular Docking Simulation , Solubility , Toll-Like Receptors/immunology , Viral Vaccines/adverse effects , Viral Vaccines/chemistry , Viral Vaccines/immunology , Zika Virus/chemistry , Zika Virus/immunology , Zika Virus Infection/immunology , Zika Virus Infection/prevention & control , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...