Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
EMBO Mol Med ; 16(3): 641-663, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38332201

ABSTRACT

Communications between immune cells are essential to ensure appropriate coordination of their activities. Here, we observed the infiltration of activated macrophages into the joint-footpads of chikungunya virus (CHIKV)-infected animals. Large numbers of CD64+MHCII+ and CD64+MHCII- macrophages were present in the joint-footpad, preceded by the recruitment of their CD11b+Ly6C+ inflammatory monocyte precursors. Recruitment and differentiation of these myeloid subsets were dependent on CD4+ T cells and GM-CSF. Transcriptomic and gene ontology analyses of CD64+MHCII+ and CD64+MHCII- macrophages revealed 89 differentially expressed genes, including genes involved in T cell proliferation and differentiation pathways. Depletion of phagocytes, including CD64+MHCII+ macrophages, from CHIKV-infected mice reduced disease pathology, demonstrating that these cells play a pro-inflammatory role in CHIKV infection. Together, these results highlight the synergistic dynamics of immune cell crosstalk in driving CHIKV immunopathogenesis. This study provides new insights in the disease mechanism and offers opportunities for development of novel anti-CHIKV therapeutics.


Subject(s)
Chikungunya Fever , Chikungunya virus , Animals , Mice , T-Lymphocytes/metabolism , Chikungunya virus/genetics , Macrophages , CD4-Positive T-Lymphocytes
2.
Nat Rev Immunol ; 22(10): 597-613, 2022 10.
Article in English | MEDLINE | ID: mdl-36064780

ABSTRACT

Monkeypox virus (MPXV), which causes disease in humans, has for many years been restricted to the African continent, with only a handful of sporadic cases in other parts of the world. However, unprecedented outbreaks of monkeypox in non-endemic regions have recently taken the world by surprise. In less than 4 months, the number of detected MPXV infections has soared to more than 48,000 cases, recording a total of 13 deaths. In this Review, we discuss the clinical, epidemiological and immunological features of MPXV infections. We also highlight important research questions and new opportunities to tackle the ongoing monkeypox outbreak.


Subject(s)
Humans , Monkeypox virus
3.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-35039441

ABSTRACT

O'nyongnyong virus (ONNV) is a re-emerging alphavirus previously known to be transmitted by main malaria vectors, thus suggesting the possibility of coinfections with arboviruses in co-endemic areas. However, the pathological outcomes of such infections remain unknown. Using murine coinfection models, we demonstrated that a preexisting blood-stage Plasmodium infection suppresses ONNV-induced pathologies. We further showed that suppression of viremia and virus dissemination are dependent on Plasmodium-induced IFNγ and are associated with reduced infection of CD45- cells at the site of virus inoculation. We further proved that treatment with IFNγ or plasma samples from Plasmodium vivax-infected patients containing IFNγ are able to restrict ONNV infection in human fibroblast, synoviocyte, skeletal muscle, and endothelial cell lines. Mechanistically, the role of IFNγ in restricting ONNV infection was confirmed in in vitro infection assays through the generation of an IFNγ receptor 1 α chain (IFNγR1)-deficient cell line.


Subject(s)
Alphavirus Infections , Coinfection , Malaria , O'nyong-nyong Virus/pathogenicity , Animals , Cell Line , Coinfection/parasitology , Coinfection/virology , Disease Models, Animal , Host-Pathogen Interactions , Mice , Microbial Interactions
4.
Proc Natl Acad Sci U S A ; 117(40): 24620-24626, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32883878

ABSTRACT

The reduced development of COVID-19 for children compared to adults provides some tantalizing clues on the pathogenesis and transmissibility of this pandemic virus. First, ACE2, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor, is reduced in the respiratory tract in children. Second, coronavirus associated with common colds in children may offer some protection, due to cross-reactive humoral immunity and T cell immunity between common coronaviruses and SARS-CoV-2. Third, T helper 2 immune responses are protective in children. Fourth, surprisingly, eosinophilia, associated with T helper 2, may be protective. Fifth, children generally produce lower levels of inflammatory cytokines. Finally, the influence of the downturn in the global economy, the impact of living in quarters among families who are the most at risk, and factors including the openings of some schools, are considered. Those most disadvantaged socioeconomically may suffer disproportionately with COVID-19.


Subject(s)
Betacoronavirus/metabolism , Coronavirus Infections/immunology , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/immunology , Respiratory Mucosa/metabolism , Respiratory Tract Infections/immunology , Angiotensin-Converting Enzyme 2 , COVID-19 , Child , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Host-Pathogen Interactions/immunology , Humans , Pandemics , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Respiratory Tract Infections/virology , SARS-CoV-2 , Th2 Cells/physiology
5.
Front Immunol ; 11: 894, 2020.
Article in English | MEDLINE | ID: mdl-32477364

ABSTRACT

O'nyong-nyong virus (ONNV) is an arthritogenic alphavirus that caused two large epidemics in 1959 and 1996, affecting millions of people in Africa. More recently, sero-surveillance of healthy blood donors conducted in 2019 revealed high rates of unreported ONNV infection in Uganda. Due to similar clinical symptoms with other endemic mosquito-borne pathogens in the region, including chikungunya virus, dengue virus and malaria, ONNV infections are often un- or misdiagnosed. Elucidating the immunopathogenic factors of this re-emerging arbovirus is critical with the expanding geographic distribution of competent vectors. This study reports the establishment of an immune competent C57BL6/J mouse model to mechanistically characterize ONNV infection and assess potential treatment efficacy. This mouse model successfully recapitulated arthralgia and viremia profiles seen in ONNV patients. Furthermore, longitudinal in-vivo PET imaging with [18F]FB-IL-2 (CD25+CD4+ binding probe) and histopathological assessment in this model demonstrated the pathogenic role of CD4+ T cells in driving joint pathology. Concordantly, in vivo CD4+ T cell depletion, or suppression with fingolimod, an FDA-approved immunomodulating drug, abrogated CD4+ T cell-mediated disease. This study demonstrates the importance of this immune competent ONNV model for future studies on factors influencing disease pathogenesis, which could shape the discovery of novel therapeutic strategies for arthritogenic alphaviruses.


Subject(s)
Alphavirus Infections/immunology , Alphavirus Infections/pathology , O'nyong-nyong Virus/immunology , O'nyong-nyong Virus/pathogenicity , Alphavirus Infections/virology , Animals , CD4-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Drug Repositioning , Female , Fingolimod Hydrochloride/administration & dosage , Humans , Immunocompetence , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Positron-Emission Tomography , Viremia
6.
Clin Transl Immunology ; 9(4): e1126, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32346479

ABSTRACT

OBJECTIVES: Zika virus (ZIKV) is a mosquito-borne flavivirus that re-emerged in 2015. The association between ZIKV and neurological complications initiated the development of relevant animal models to understand the mechanisms underlying ZIKV-induced pathologies. Transient inhibition of the type I interferon (IFN) pathway through the use of an IFNAR1-blocking antibody, MAR1-5A3, could efficiently permit active virus replication in immunocompetent animals. Type I IFN signalling is involved in the regulation of humoral responses, and thus, it is crucial to investigate the potential effects of type I IFN blockade towards B-cell responses. METHODS: In this study, comparative analysis was conducted using serum samples collected from ZIKV-infected wild-type (WT) animals either administered with or without MAR1-5A3. RESULTS: Serological assays revealed a more robust ZIKV-specific IgG response and subtype switching upon inhibition of type I IFN due to the abundance of antigen availability. This observation was corroborated by an increase in germinal centres, plasma cells and germinal centre B cells. Interestingly, although both groups of animals recognised different B-cell linear epitopes in the E and NS1 regions, there was no difference in neutralising capacity. Further characterisation of these epitopes in the E protein revealed a detrimental role of antibodies that were generated in the absence of type I IFN. CONCLUSION: This study highlights the role of type I IFN in shaping the anti-ZIKV antibody response to generate beneficial antibodies and will help guide development of better vaccine candidates triggering efficient neutralising antibodies and avoiding detrimental ones.

8.
Clin Transl Immunology ; 8(11): e01082, 2019.
Article in English | MEDLINE | ID: mdl-31709049

ABSTRACT

OBJECTIVES: Effects of Zika virus (ZIKV) infection on placental development during pregnancy are unclear. METHODS: Full-term placentas from three women, each infected with ZIKV during specific pregnancy trimesters, were harvested for anatomic, immunologic and transcriptomic analysis. RESULTS: In this study, each woman exhibited a unique immune response with raised IL-1RA, IP-10, EGF and RANTES expression and neutrophil numbers during the acute infection phase. Although ZIKV NS3 antigens co-localised to placental Hofbauer cells, the placentas showed no anatomic defects. Transcriptomic analysis of samples from the placentas revealed that infection during trimester 1 caused a disparate cellular response centred on differential eIF2 signalling, mitochondrial dysfunction and oxidative phosphorylation. Despite these, the babies were delivered without any congenital anomalies. CONCLUSION: These findings should translate to improve clinical prenatal screening procedures for virus-infected pregnant patients.

9.
Clin Transl Immunology ; 8(7): e1066, 2019.
Article in English | MEDLINE | ID: mdl-31372218

ABSTRACT

OBJECTIVES: Recent Zika virus (ZIKV) outbreaks challenged existing laboratory diagnostic standards, especially for serology-based methods. Because of the genetic and structural similarity of ZIKV with other flaviviruses, this results in cross-reactive antibodies, which confounds serological interpretations. METHODS: Plasma from Singapore ZIKV patients was screened longitudinally for antibody responses and neutralising capacities against ZIKV. Samples from healthy controls, ZIKV patients and DENV patients were further assessed using ZIKV and DENV peptides of precursor membrane (prM), envelope (E) or non-structural 1 (NS1) viral proteins in a peptide-based ELISA for epitope identification. Identified epitopes were re-validated and diagnostically evaluated using sera of patients with DENV, bacteria or unknown infections from Thailand. RESULTS: Long-lasting ZIKV-neutralising antibodies were elicited during ZIKV infection. Thirteen potential linear B-cell epitopes were identified, and of these, four common flavivirus, three ZIKV-specific and one DENV-specific differential epitopes had more than 50% sensitivity and specificity. Notably, ZIKV-specific peptide 26 on domain I/II of E protein (amino acid residues 271-288) presented 80% sensitivity and 85.7% specificity. Importantly, the differential epitopes also showed significance in differentiating non-flavivirus patient samples. CONCLUSION: Linear B-cell epitope candidates to differentiate between ZIKV and DENV infections were identified, providing the first step towards the design of a much-needed serology-based assay.

10.
EMBO Mol Med ; 11(6)2019 06.
Article in English | MEDLINE | ID: mdl-31015278

ABSTRACT

Currently, there are no commercially available live-attenuated vaccines against chikungunya virus (CHIKV). Here, CHIKVs with mutations in non-structural proteins (nsPs) were investigated for their suitability as attenuated CHIKV vaccines. R532H mutation in nsP1 caused reduced infectivity in mouse tail fibroblasts but an enhanced type-I IFN response compared to WT-CHIKV Adult mice infected with this nsP-mutant exhibited a mild joint phenotype with low-level viremia that rapidly cleared. Mechanistically, ingenuity pathway analyses revealed a tilt in the anti-inflammatory IL-10 versus pro-inflammatory IL-1ß and IL-18 balance during CHIKV nsP-mutant infection that modified acute antiviral and cell signaling canonical pathways. Challenging CHIKV nsP-mutant-infected mice with WT-CHIKV or the closely related O'nyong-nyong virus resulted in no detectable viremia, observable joint inflammation, or damage. Challenged mice showed high antibody titers with efficient neutralizing capacity, indicative of immunological memory. Manipulating molecular processes that govern CHIKV replication could lead to plausible vaccine candidates against alphavirus infection.


Subject(s)
Chikungunya Fever/prevention & control , Chikungunya virus , Mutation , Viral Nonstructural Proteins , Viral Vaccines , Animals , Chikungunya Fever/genetics , Chikungunya Fever/immunology , Chikungunya virus/genetics , Chikungunya virus/immunology , Chlorocebus aethiops , Mice , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
11.
J Infect Dis ; 220(2): 203-212, 2019 06 19.
Article in English | MEDLINE | ID: mdl-30901054

ABSTRACT

BACKGROUND: Zika virus (ZIKV) infections have reemerged as a global health issue due to serious clinical complications. Development of specific serological assays to detect and differentiate ZIKV from other cocirculating flaviviruses for accurate diagnosis remains a challenge. METHODS: We investigated antibody responses in 51 acute ZIKV-infected adult patients from Campinas, Brazil, including 7 pregnant women who later delivered during the study. Using enzyme-linked immunosorbent assays, levels of antibody response were measured and specific epitopes identified. RESULTS: Several antibody-binding hot spots were identified in ZIKV immunogenic antigens, including membrane, envelope (E) and nonstructural protein 1 (NS1). Interestingly, specific epitopes (2 from E and 2 from NS1) strongly recognized by ZIKV-infected patients' antibodies were identified and were not cross-recognized by dengue virus (DENV)-infected patients' antibodies. Corresponding DENV peptides were not strongly recognized by ZIKV-infected patients' antibodies. Notably, ZIKV-infected pregnant women had specific epitope recognition for ZIKV NS1 (amino acid residues 17-34), which could be a potential serological marker for early ZIKV detection. CONCLUSIONS: This study identified 6 linear ZIKV-specific epitopes for early detection of ZIKV infections. We observed differential epitope recognition between ZIKV-infected and DENV-infected patients. This information will be useful for developing diagnostic methods that differentiate between closely related flaviviruses.


Subject(s)
Epitopes/immunology , Viral Nonstructural Proteins/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Acute Disease , Adolescent , Adult , Aged , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibody Formation/immunology , Brazil , Cross Reactions/immunology , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Male , Middle Aged , Serologic Tests , Young Adult , Zika Virus Infection/virology
12.
Life Sci Alliance ; 2(1)2019 02.
Article in English | MEDLINE | ID: mdl-30665948

ABSTRACT

Chikungunya virus (CHIKV) has been a worldwide threat since its reemergence in La Reunion Island in 2004. Expression of the interferon-stimulated protein Viperin correlates with viral load burden in patients, and studies in mice have demonstrated its role to limit disease severity against CHIKV infection. Using Viperin -/- mice, we aimed to understand the contribution of Viperin to the T-cell immune response against CHIKV. CD4 T-cell depletion in Viperin -/- mice showed that increased late acute joint inflammation (5-8 d postinfection) was exclusively mediated by T cells. Specifically, CHIKV-infected Viperin -/- mice showed an increased INFγ Th1 profile of CD4 T cells, enhanced INFγ stimulation by APCs, an increased INFγ secretion profile in the joint microenvironment, and increased numbers of inflammatory monocytes in virus-infected joints compared with WT mice. Bone marrow grafting experiments showed that Viperin expression in both hematopoietic and non-hematopoietic cells is instrumental in reducing disease severity associated with a CD4 T-cell response.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chikungunya Fever/virology , Chikungunya virus/pathogenicity , Interferon-gamma/metabolism , Proteins/metabolism , Animals , Antigen-Presenting Cells/metabolism , Arthritis/metabolism , Arthritis/virology , Bone Marrow Transplantation , Chikungunya virus/isolation & purification , Female , Gene Knockout Techniques , HEK293 Cells , Humans , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Proteins/genetics , Viral Load
13.
Sci Rep ; 8(1): 17125, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30459473

ABSTRACT

Animal models that recapitulate the human pathophysiology have been developed as useful research tools. Although laboratory mice are widely used, they are phylogenetically "distant" to humans. New world monkeys, such as the common marmoset (Callithrix jacchus) have steadily gained prominence. In this report, marmosets are explored as an alternate in vivo model to investigate infection and immunity of Zika virus (ZIKV). Multimodal platforms, including ultrasound and magnetic resonance imaging (MRI), flow cytometry, and multiplex microbead immunoassays were established to comprehensively decipher immune responses and pathophysiological outcomes. While ZIKV-infected marmosets had detectable ZIKV RNA load in various body fluids, animals did not develop any observable lesions in their testes and brains as shown by ultrasound and MRI. Immune-phenotyping detected differences in the numbers of B cells, CD8+ T cells and HLADR+ NK cells during the first two weeks of infection. Neutralizing ZIKV-specific antibodies were elicited to high levels and targeted epitopes in the E protein. This study presents a one-stop-shop platform to study infection and pathophysiology in marmosets. While marmoset-specific research tools are being refined, the research values of these animals present them as a good model for immune-based therapies.


Subject(s)
Callithrix/immunology , Callithrix/virology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , HEK293 Cells , Humans , Killer Cells, Natural/immunology , RNA, Viral/immunology , Zika Virus Infection/virology
14.
Nat Commun ; 9(1): 3905, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254309

ABSTRACT

Co-infection with Plasmodium and chikungunya virus (CHIKV) has been reported in humans, but the impact of co-infection on pathogenesis remains unclear. Here, we show that prior exposure to Plasmodium suppresses CHIKV-associated pathologies in mice. Mechanistically, Plasmodium infection induces IFNγ, which reduces viraemia of a subsequent CHIKV infection and suppresses tissue viral load and joint inflammation. Conversely, concomitant infection with both pathogens limits the peak of joint inflammation with no effect on CHIKV viraemia. Reduced peak joint inflammation is regulated by elevated apoptosis of CD4+ T-cells in the lymph nodes and disrupted CXCR3-mediated CD4+ T-cell migration that abolishes their infiltration into the joints. Virus clearance from tissues is delayed in both infection scenarios, and is associated with a disruption of B cell affinity-maturation in the spleen that reduces CHIKV-neutralizing antibody production.


Subject(s)
Chikungunya Fever/immunology , Chikungunya virus/immunology , Coinfection/immunology , Malaria/immunology , Plasmodium/immunology , Animals , Apoptosis/immunology , Arthritis/genetics , Arthritis/immunology , Arthritis/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chikungunya Fever/virology , Chikungunya virus/physiology , Coinfection/parasitology , Coinfection/virology , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Malaria/metabolism , Malaria/parasitology , Male , Mice, Inbred C57BL , Mice, Knockout , Plasmodium/physiology , Viral Load/immunology , Viremia/immunology , Viremia/virology
15.
Viruses ; 10(5)2018 05 17.
Article in English | MEDLINE | ID: mdl-29772762

ABSTRACT

Since 2005, Chikungunya virus (CHIKV) re-emerged and caused numerous outbreaks in the world, and finally, was introduced into the Americas in 2013. The lack of CHIKV-specific therapies has led to the use of non-specific drugs. Chloroquine, which is commonly used to treat febrile illnesses in the tropics, has been shown to inhibit CHIKV replication in vitro. To assess the in vivo effect of chloroquine, two complementary studies were performed: (i) a prophylactic study in a non-human primate model (NHP); and (ii) a curative study "CuraChik", which was performed during the Reunion Island outbreak in 2006 in a human cohort. Clinical, biological, and immunological data were compared between treated and placebo groups. Acute CHIKV infection was exacerbated in NHPs treated with prophylactic administration of chloroquine. These NHPs displayed a higher viremia and slower viral clearance (p < 0.003). Magnitude of viremia was correlated to the type I IFN response (Rho = 0.8, p < 0.001) and severe lymphopenia (Rho = 0.8, p < 0.0001), while treatment led to a delay in both CHIKV-specific cellular and IgM responses (p < 0.02 and p = 0.04, respectively). In humans, chloroquine treatment did not affect viremia or clinical parameters during the acute stage of the disease (D1 to D14), but affected the levels of C-reactive Protein (CRP), IFNα, IL-6, and MCP1 over time (D1 to D16). Importantly, no positive effect could be detected on prevalence of persistent arthralgia at Day 300. Although inhibitory in vitro, chloroquine as a prophylactic treatment in NHPs enhances CHIKV replication and delays cellular and humoral response. In patients, curative chloroquine treatment during the acute phase decreases the levels of key cytokines, and thus may delay adaptive immune responses, as observed in NHPs, without any suppressive effect on peripheral viral load.


Subject(s)
Chikungunya Fever/immunology , Chikungunya virus/immunology , Chloroquine/pharmacology , Disease Outbreaks , Immunity/drug effects , Macaca fascicularis/virology , Viral Load/drug effects , Animals , Cells, Cultured , Chikungunya Fever/drug therapy , Chikungunya Fever/epidemiology , Chikungunya Fever/virology , Chloroquine/therapeutic use , Cohort Studies , Disease Models, Animal , Fibroblasts/virology , Humans , Macaca fascicularis/immunology , Macrophages/virology , Male , Reunion/epidemiology , Virus Replication/drug effects
16.
J Infect Dis ; 218(5): 814-824, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29672707

ABSTRACT

Background: Since its unexpected reemergence, Zika virus (ZIKV) has caused numerous outbreaks globally. This study characterized the host immune responses during ZIKV infection. Methods: Patient samples were collected longitudinally during the acute, convalescence and recovery phases of ZIKV infection over 6 months during the Singapore outbreak in late 2016. Plasma immune mediators were profiled via multiplex microbead assay, while changes in blood cell numbers were determined with immunophenotyping. Results: Data showed the involvement of various immune mediators during acute ZIKV infection accompanied by a general reduction in blood cell numbers for all immune subsets except CD14+ monocytes. Importantly, viremic patients experiencing moderate symptoms had significantly higher quantities of interferon γ-induced protein 10, monocyte chemotactic protein 1, interleukin 1 receptor antagonist, interleukin 8, and placental growth factor 1, accompanied by reduced numbers of peripheral CD8+ T cells, CD4+ T cells, and double-negative T cells. Levels of T-cell associated mediators, including interferon γ-induced protein 10, interferon γ, and interleukin 10, were high in recovery phases of ZIKV infection, suggesting a functional role for T cells. The identification of different markers at specific disease phases emphasizes the dynamics of a balanced cytokine environment in disease progression. Conclusions: This is the first comprehensive study that highlights specific cellular changes and immune signatures during ZIKV disease progression, and it provides valuable insights into ZIKV immunopathogenesis.


Subject(s)
Cytokines/blood , Zika Virus Infection/immunology , Zika Virus Infection/pathology , Zika Virus/immunology , Adolescent , Adult , Aged , Disease Outbreaks , Female , Humans , Immunoassay , Longitudinal Studies , Male , Middle Aged , Plasma/chemistry , Singapore/epidemiology , T-Lymphocyte Subsets/immunology , Young Adult , Zika Virus Infection/epidemiology
17.
mSphere ; 3(2)2018.
Article in English | MEDLINE | ID: mdl-29600283

ABSTRACT

Zika virus (ZIKV) has reemerged in the population and caused unprecedented global outbreaks. Here, the transcriptomic consequences of ZIKV infection were studied systematically first in human peripheral blood CD14+ monocytes and monocyte-derived macrophages with high-density RNA sequencing. Analyses of the ZIKV genome revealed that the virus underwent genetic diversification, and differential mRNA abundance was found in host cells during infection. Notably, there was a significant change in the cellular response, with cross talk between monocytes and natural killer (NK) cells as one of the highly identified pathways. Immunophenotyping of peripheral blood from ZIKV-infected patients further confirmed the activation of NK cells during acute infection. ZIKV infection in peripheral blood cells isolated from healthy donors led to the induction of gamma interferon (IFN-γ) and CD107a-two key markers of NK cell function. Depletion of CD14+ monocytes from peripheral blood resulted in a reduction of these markers and reduced priming of NK cells during infection. This was complemented by the immunoproteomic changes observed. Mechanistically, ZIKV infection preferentially counterbalances monocyte and/or NK cell activity, with implications for targeted cytokine immunotherapies. IMPORTANCE ZIKV reemerged in recent years, causing outbreaks in many parts of the world. Alarmingly, ZIKV infection has been associated with neurological complications such as Guillain-Barré syndrome (GBS) in adults and congenital fetal growth-associated anomalies in newborns. Host peripheral immune cells are one of the first to interact with the virus upon successful transmission from an infected female Aedes mosquito. However, little is known about the role of these immune cells during infection. In this work, the immune responses of monocytes, known target cells of ZIKV infection, were investigated by high-density transcriptomics. The analysis saw a robust immune response being elicited. Importantly, it also divulged that monocytes prime NK cell activities during virus infection. Removal of monocytes during the infection changed the immune milieu, which in turn reduced NK cell stimulation. This study provides valuable insights into the pathobiology of the virus and allows for the possibility of designing novel targeted therapeutics.


Subject(s)
Killer Cells, Natural/immunology , Macrophages/immunology , Monocytes/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Cells, Cultured , Genome, Viral , Host-Pathogen Interactions , Humans , Interferon-gamma/immunology , Killer Cells, Natural/virology , Lymphocyte Activation , Lymphocyte Depletion , Lysosomal-Associated Membrane Protein 1/immunology , Macrophages/virology , Monocytes/virology , Proteomics , RNA, Viral/genetics , Sequence Analysis, RNA , Transcriptome , Virus Replication , Zika Virus/genetics
18.
Sci Rep ; 8(1): 1860, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29382880

ABSTRACT

The arthropod-transmitted chikungunya virus (CHIKV) causes a flu-like disease that is characterized by incapacitating arthralgia. The re-emergence of CHIKV and the continual risk of new epidemics have reignited research in CHIKV pathogenesis. Virus-specific antibodies have been shown to control virus clearance, but antibodies present at sub-neutralizing concentrations can also augment virus infection that exacerbates disease severity. To explore this occurrence, CHIKV infection was investigated in the presence of CHIKV-specific antibodies in both primary human cells and a murine macrophage cell line, RAW264.7. Enhanced attachment of CHIKV to the primary human monocytes and B cells was observed while increased viral replication was detected in RAW264.7 cells. Blocking of specific Fc receptors (FcγRs) led to the abrogation of these observations. Furthermore, experimental infection in adult mice showed that animals had higher viral RNA loads and endured more severe joint inflammation in the presence of sub-neutralizing concentrations of CHIKV-specific antibodies. In addition, CHIKV infection in 11 days old mice under enhancing condition resulted in higher muscles viral RNA load detected and death. These observations provide the first evidence of antibody-mediated enhancement in CHIKV infection and pathogenesis and could also be relevant for other important arboviruses such as Zika virus.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Arthralgia/virology , Chikungunya Fever/virology , Chikungunya virus/immunology , Severity of Illness Index , Animals , Arthralgia/immunology , Arthralgia/pathology , Cells, Cultured , Chikungunya Fever/immunology , Chikungunya Fever/pathology , Humans , Interferon-gamma/physiology , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgG/metabolism , Viral Load , Virus Replication
19.
EMBO Mol Med ; 10(1): 121-138, 2018 01.
Article in English | MEDLINE | ID: mdl-29113976

ABSTRACT

Arboviral diseases have risen significantly over the last 40 years, increasing the risk of co-infection with other endemic disease such as malaria. However, nothing is known about the impact arboviruses have on the host response toward heterologous pathogens during co-infection. Here, we investigate the effects of Chikungunya virus (CHIKV) co-infection on the susceptibility and severity of malaria infection. Using the Plasmodium berghei ANKA (PbA) experimental cerebral malaria (ECM) model, we show that concurrent co-infection induced the most prominent changes in ECM manifestation. Concurrent co-infection protected mice from ECM mortality without affecting parasite development in the blood. This protection was mediated by the alteration of parasite-specific CD8+ T-cell trafficking through an IFNγ-mediated mechanism. Co-infection with CHIKV induced higher splenic IFNγ levels that lead to high local levels of CXCL9 and CXCL10. This induced retention of CXCR3-expressing pathogenic CD8+ T cells in the spleen and prevented their migration to the brain. This then averts all downstream pathogenic events such as parasite sequestration in the brain and disruption of blood-brain barrier that prevents ECM-induced mortality in co-infected mice.


Subject(s)
Brain/pathology , CD8-Positive T-Lymphocytes/pathology , Chikungunya Fever/pathology , Chikungunya virus/physiology , Coinfection/pathology , Malaria, Cerebral/pathology , Plasmodium berghei/physiology , Animals , Brain/parasitology , Brain/virology , CD8-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/virology , Cell Movement , Chikungunya Fever/parasitology , Chikungunya Fever/virology , Coinfection/parasitology , Coinfection/virology , Female , Malaria, Cerebral/parasitology , Malaria, Cerebral/virology , Male , Mice , Mice, Inbred C57BL , Neuropathology , Protective Factors
20.
J Infect Dis ; 216(2): 172-181, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28838147

ABSTRACT

Background: Zika virus (ZIKV) infections have been linked to different levels of clinical outcomes, ranging from mild rash and fever to severe neurological complications and congenital malformations. Methods: We investigated the clinical and immunological response, focusing on the immune mediators profile in 95 acute ZIKV-infected adult patients from Campinas, Brazil. These patients included 6 pregnant women who later delivered during the course of this study. Clinical observations were recorded during hospitalization. Levels of 45 immune mediators were quantified using multiplex microbead-based immunoassays. Results: Whereas 11.6% of patients had neurological complications, 88.4% displayed mild disease of rash and fever. Several immune mediators were specifically higher in ZIKV-infected patients, and levels of interleukin 10, interferon gamma-induced protein 10 (IP-10), and hepatocyte growth factor differentiated between patients with or without neurological complications. Interestingly, higher levels of interleukin 22, monocyte chemoattractant protein 1, TNF-α, and IP-10 were observed in ZIKV-infected pregnant women carrying fetuses with fetal growth-associated malformations. Notably, infants with congenital central nervous system deformities had significantly higher levels of interleukin 18 and IP-10 but lower levels of hepatocyte growth factor than those without such abnormalities born to ZIKV-infected mothers. Conclusions: This study identified several key markers for the control of ZIKV pathogenesis. This will allow a better understanding of the molecular mechanisms of ZIKV infection in patients.


Subject(s)
Cytokines/blood , Nervous System Malformations/epidemiology , Pregnancy Complications, Infectious/epidemiology , Zika Virus Infection/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Brazil/epidemiology , Child , Female , Fetal Growth Retardation/virology , Humans , Infant, Newborn , Male , Middle Aged , Nervous System Malformations/virology , Pregnancy , Pregnancy Complications, Infectious/virology , Pregnancy Outcome , Viral Load , Young Adult , Zika Virus , Zika Virus Infection/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...