Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(6): 966-978, 2023 06.
Article in English | MEDLINE | ID: mdl-37248417

ABSTRACT

High-risk groups, including Indigenous people, are at risk of severe COVID-19. Here we found that Australian First Nations peoples elicit effective immune responses to COVID-19 BNT162b2 vaccination, including neutralizing antibodies, receptor-binding domain (RBD) antibodies, SARS-CoV-2 spike-specific B cells, and CD4+ and CD8+ T cells. In First Nations participants, RBD IgG antibody titers were correlated with body mass index and negatively correlated with age. Reduced RBD antibodies, spike-specific B cells and follicular helper T cells were found in vaccinated participants with chronic conditions (diabetes, renal disease) and were strongly associated with altered glycosylation of IgG and increased interleukin-18 levels in the plasma. These immune perturbations were also found in non-Indigenous people with comorbidities, indicating that they were related to comorbidities rather than ethnicity. However, our study is of a great importance to First Nations peoples who have disproportionate rates of chronic comorbidities and provides evidence of robust immune responses after COVID-19 vaccination in Indigenous people.


Subject(s)
COVID-19 Vaccines , COVID-19 , Humans , BNT162 Vaccine , COVID-19/prevention & control , CD8-Positive T-Lymphocytes , Australia/epidemiology , SARS-CoV-2 , Immunoglobulin G , Antibodies, Neutralizing , Immunity , Antibodies, Viral , Vaccination
2.
Nat Immunol ; 23(1): 86-98, 2022 01.
Article in English | MEDLINE | ID: mdl-34845392

ABSTRACT

Ineffective antibody-mediated responses are a key characteristic of chronic viral infection. However, our understanding of the intrinsic mechanisms that drive this dysregulation are unclear. Here, we identify that targeting the epigenetic modifier BMI-1 in mice improves humoral responses to chronic lymphocytic choriomeningitis virus. BMI-1 was upregulated by germinal center B cells in chronic viral infection, correlating with changes to the accessible chromatin landscape, compared to acute infection. B cell-intrinsic deletion of Bmi1 accelerated viral clearance, reduced splenomegaly and restored splenic architecture. Deletion of Bmi1 restored c-Myc expression in B cells, concomitant with improved quality of antibody and coupled with reduced antibody-secreting cell numbers. Specifically, BMI-1-deficiency induced antibody with increased neutralizing capacity and enhanced antibody-dependent effector function. Using a small molecule inhibitor to murine BMI-1, we could deplete antibody-secreting cells and prohibit detrimental immune complex formation in vivo. This study defines BMI-1 as a crucial immune modifier that controls antibody-mediated responses in chronic infection.


Subject(s)
B-Lymphocytes/immunology , Immunity, Humoral/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Polycomb Repressive Complex 1/immunology , Proto-Oncogene Proteins/immunology , Adaptive Immunity/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody Formation/immunology , Female , Germinal Center/immunology , Male , Mice , Mice, Inbred C57BL
3.
Cell ; 167(2): 433-443.e14, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27667685

ABSTRACT

While a third of the world carries the burden of tuberculosis, disease control has been hindered by a lack of tools, including a rapid, point-of-care diagnostic and a protective vaccine. In many infectious diseases, antibodies (Abs) are powerful biomarkers and important immune mediators. However, in Mycobacterium tuberculosis (Mtb) infection, a discriminatory or protective role for humoral immunity remains unclear. Using an unbiased antibody profiling approach, we show that individuals with latent tuberculosis infection (Ltb) and active tuberculosis disease (Atb) have distinct Mtb-specific humoral responses, such that Ltb infection is associated with unique Ab Fc functional profiles, selective binding to FcγRIII, and distinct Ab glycosylation patterns. Moreover, compared to Abs from Atb, Abs from Ltb drove enhanced phagolysosomal maturation, inflammasome activation, and, most importantly, macrophage killing of intracellular Mtb. Combined, these data point to a potential role for Fc-mediated Ab effector functions, tuned via differential glycosylation, in Mtb control.


Subject(s)
Antibodies, Bacterial/immunology , Host-Pathogen Interactions/immunology , Immunity, Humoral , Latent Tuberculosis/immunology , Mycobacterium tuberculosis/immunology , Adult , Female , Glycosylation , Humans , Immunoglobulin Fc Fragments/immunology , Macrophage Activation , Male , Middle Aged , Polysaccharides/immunology , Protein Array Analysis , Receptors, IgG/immunology , Young Adult
4.
Cell ; 163(4): 988-98, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544943

ABSTRACT

While antibody titers and neutralization are considered the gold standard for the selection of successful vaccines, these parameters are often inadequate predictors of protective immunity. As antibodies mediate an array of extra-neutralizing Fc functions, when neutralization fails to predict protection, investigating Fc-mediated activity may help identify immunological correlates and mechanism(s) of humoral protection. Here, we used an integrative approach termed Systems Serology to analyze relationships among humoral responses elicited in four HIV vaccine trials. Each vaccine regimen induced a unique humoral "Fc fingerprint." Moreover, analysis of case:control data from the first moderately protective HIV vaccine trial, RV144, pointed to mechanistic insights into immune complex composition that may underlie protective immunity to HIV. Thus, multi-dimensional relational comparisons of vaccine humoral fingerprints offer a unique approach for the evaluation and design of novel vaccines against pathogens for which correlates of protection remain elusive.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Viral/immunology , Immunoglobulin G/immunology , Animals , Antibodies, Viral/blood , Antibody-Dependent Cell Cytotoxicity , Antigen-Antibody Complex/immunology , Clinical Trials as Topic , Drug Design , HIV Infections/immunology , Humans , Immunoglobulin G/blood , Receptors, Fc/immunology
5.
Nat Immunol ; 17(9): 1067-74, 2016 09.
Article in English | MEDLINE | ID: mdl-27455421

ABSTRACT

The activating natural killer (NK)-cell receptor KIR3DS1 has been linked to the outcome of various human diseases, including delayed progression of disease caused by human immunodeficiency virus type 1 (HIV-1), yet a ligand that would account for its biological effects has remained unknown. We screened 100 HLA class I proteins and found that KIR3DS1 bound to HLA-F, a result we confirmed biochemically and functionally. Primary human KIR3DS1(+) NK cells degranulated and produced antiviral cytokines after encountering HLA-F and inhibited HIV-1 replication in vitro. Activation of CD4(+) T cells triggered the transcription and surface expression of HLA-F mRNA and HLA-F protein, respectively, and induced binding of KIR3DS1. HIV-1 infection further increased the transcription of HLA-F mRNA but decreased the binding of KIR3DS1, indicative of a mechanism for evading recognition by KIR3DS1(+) NK cells. Thus, we have established HLA-F as a ligand of KIR3DS1 and have demonstrated cell-context-dependent expression of HLA-F that might explain the widespread influence of KIR3DS1 in human disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/physiology , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Receptors, KIR3DS1/metabolism , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Progression , Histocompatibility Antigens Class I/genetics , Humans , Immune Evasion , Jurkat Cells , Ligands , Lymphocyte Activation , Primary Cell Culture , Receptors, KIR3DS1/agonists , Receptors, KIR3DS1/genetics , Virus Latency , Virus Replication
6.
Trends Immunol ; 43(6): 417-419, 2022 06.
Article in English | MEDLINE | ID: mdl-35537983

ABSTRACT

Severe coronavirus disease 2019 (COVID-19) has been associated with cytokine storms and hyperinflammation. In a recent study, Junqueira et al. provide evidence that antibody-mediated uptake of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus by monocytes and macrophages may contribute to this inflammation by activating inflammasomes which trigger pyroptosis.


Subject(s)
COVID-19 , Cytokine Release Syndrome , Humans , Inflammasomes , RNA, Viral , SARS-CoV-2
7.
Trends Immunol ; 43(10): 815-825, 2022 10.
Article in English | MEDLINE | ID: mdl-35995705

ABSTRACT

A single dose of human papillomavirus (HPV) vaccine against HPV infection (prerequisite for cervical cancer) appears to be as efficacious as two or three doses, despite inducing lower antibody titers. Neutralizing antibodies are thought to be the primary mediator of protection, but the threshold for protection is unknown. Antibody functions beyond neutralization have not been explored for HPV vaccines. Here, we discuss the immune mechanisms of HPV vaccines, with a focus on non-neutralizing antibody effector functions. In the context of single-dose HPV vaccination where antibody is limiting, we propose that non-neutralizing antibody functions may contribute to preventing HPV infection. Understanding the immunological basis of protection for single-dose HPV vaccination will provide a rationale for implementing single-dose HPV vaccine regimens.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Antibodies, Neutralizing , Antibodies, Viral , Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18 , Humans , Papillomavirus Infections/prevention & control
8.
BMC Med ; 22(1): 388, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267089

ABSTRACT

BACKGROUND: Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) proteins are expressed on the surface of infected erythrocytes, mediating parasite sequestration in the vasculature. PfEMP1 is a major target of protective antibodies, but the features of the antibody response are poorly defined. METHODS: In Malawian children with cerebral or uncomplicated malaria, we characterized the antibody response to 39 recombinant PfEMP1 Duffy binding like (DBL) domains or cysteine-rich interdomain regions (CIDRs) in detail, including measures of antibody classes, subclasses, and engagement with Fcγ receptors and complement. Using elastic net regularized logistic regression, we identified a combination of seven antibody targets and Fc features that best distinguished between children with cerebral and uncomplicated malaria. To confirm the role of the selected targets and Fc features, we measured antibody-dependent neutrophil and THP-1 cell phagocytosis of intercellular adhesion molecule-1 (ICAM-1) and endothelial protein C (EPCR) co-binding infected erythrocytes. RESULTS: The selected features distinguished between children with cerebral and uncomplicated malaria with 87% accuracy (median, 80-96% interquartile range) and included antibody to well-characterized DBLß3 domains and a less well-characterized CIDRγ12 domain. The abilities of antibodies to engage C1q and FcγRIIIb, rather than levels of IgG, correlated with protection. In line with a role of FcγRIIIb binding antibodies to DBLß3 domains, antibody-dependent neutrophil phagocytosis of ICAM-1 and EPCR co-binding IE was higher in uncomplicated malaria (15% median, 8-38% interquartile range) compared to cerebral malaria (7%, 30-15%, p < 0.001). CONCLUSIONS: Antibodies associated with protection from cerebral malaria target a subset of PfEMP1 domains. The Fc features of protective antibody response include engagement of FcγRIIIb and C1q, and ability to induce antibody-dependent neutrophil phagocytosis of infected erythrocytes. Identifying the targets and Fc features of protective immunity could facilitate the development of PfEMP1-based therapeutics for cerebral malaria.


Subject(s)
Antibodies, Protozoan , Malaria, Cerebral , Plasmodium falciparum , Protozoan Proteins , Humans , Malaria, Cerebral/immunology , Malawi , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Protozoan Proteins/immunology , Child, Preschool , Plasmodium falciparum/immunology , Male , Female , Child , Infant , Intercellular Adhesion Molecule-1/immunology , Endothelial Protein C Receptor/immunology , Phagocytosis , Erythrocytes/parasitology , Erythrocytes/immunology , Malaria, Falciparum/immunology , Antigens, Protozoan/immunology
9.
PLoS Pathog ; 18(10): e1010891, 2022 10.
Article in English | MEDLINE | ID: mdl-36206307

ABSTRACT

Although antibody-inducing split virus vaccines (SV) are currently the most effective way to combat seasonal influenza, their efficacy can be modest, especially in immunologically-naïve individuals. We investigated immune responses towards inactivated whole influenza virus particle vaccine (WPV) formulations, predicated to be more immunogenic, in a non-human primate model, as an important step towards clinical testing in humans. Comprehensive analyses were used to capture 46 immune parameters to profile how WPV-induced responses differed to those elicited by antigenically-similar SV formulations. Naïve cynomolgus macaques vaccinated with either monovalent or quadrivalent WPV consistently induced stronger antibody responses and hemagglutination inhibition (HI) antibody titres against vaccine-matched viruses compared to SV formulations, while acute reactogenic effects were similar. Responses in WPV-primed animals were further increased by boosting with the same formulation, conversely to modest responses after priming and boosting with SV. 28-parameter multiplex bead array defined key antibody features and showed that while both WPV and SV induced elevated IgG responses against A/H1N1 nucleoprotein, only WPV increased IgG responses against A/H1N1 hemagglutinin (HA) and HA-Stem, and higher IgA responses to A/H1N1-HA after each vaccine dose. Antibodies to A/H1N1-HA and HA-Stem that could engage FcγR2a and FcγR3a were also present at higher levels after one dose of WPV compared to SV and remained elevated after the second dose. Furthermore, WPV-enhanced antibody responses were associated with higher frequencies of HA-specific B-cells and IFN-γ-producing CD4+ T-cell responses. Our data additionally demonstrate stronger boosting of HI titres by WPV following prior infection and support WPV administered as a priming dose irrespective of the follow up vaccine for the second dose. Our findings thus show that compared to SV vaccination, WPV-induced humoral responses are significantly increased in scope and magnitude, advocating WPV vaccination regimens for priming immunologically-naïve individuals and also in the event of a pandemic outbreak.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Animals , Humans , Hemagglutinins , Antibodies, Viral , Vaccination , Hemagglutination Inhibition Tests , Vaccines, Inactivated , Macaca fascicularis , Virion , Immunoglobulin A , Immunoglobulin G , Nucleoproteins
10.
J Med Virol ; 96(6): e29732, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38874202

ABSTRACT

Neutralizing antibodies (NAbs) are considered the primary mechanism of vaccine-mediated protection against human papillomaviruses (HPV), the causative agent of cervical cancer. However, the minimum level of NAb needed for protection is currently unknown. The HPV pseudovirion-based neutralization assay (PBNA) is the gold standard method for assessing HPV antibody responses but is time-consuming and labor-intensive. With the development of higher valency HPV vaccines, alternative serological assays with the capacity for multiplexing would improve efficiency and output. Here we describe a multiplex bead-based immunoassay to characterize the antibody responses to the seven oncogenic HPV types (HPV16/18/31/33/45/52/58) contained in the current licensed nonavalent HPV vaccine. This assay can measure antibody isotypes and subclasses (total IgG, IgM, IgA1-2, IgG1-4), and can be adapted to measure other antibody features (e.g., Fc receptors) that contribute to vaccine immunity. When tested with serum samples from unvaccinated and vaccinated individuals, we found high concordance between HPV-specific IgG using this multiplex assay and NAbs measured with PBNA. Overall, this assay is high-throughput, sample-sparing, and time-saving, providing an alternative to existing assays for the measurement and characterization of HPV antibody responses.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Immunoglobulin G , Papillomavirus Infections , Papillomavirus Vaccines , Humans , Antibodies, Viral/blood , Immunoassay/methods , Female , Papillomavirus Infections/diagnosis , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Immunoglobulin G/blood , Papillomaviridae/immunology , Human Papillomavirus Viruses
SELECTION OF CITATIONS
SEARCH DETAIL