Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51.556
Filter
Add more filters

Publication year range
1.
Cell ; 186(23): 5098-5113.e19, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37918395

ABSTRACT

Drug-resistant Pseudomonas aeruginosa (PA) poses an emerging threat to human health with urgent need for alternative therapeutic approaches. Here, we deciphered the B cell and antibody response to the virulence-associated type III secretion system (T3SS) in a cohort of patients chronically infected with PA. Single-cell analytics revealed a diverse B cell receptor repertoire directed against the T3SS needle-tip protein PcrV, enabling the production of monoclonal antibodies (mAbs) abrogating T3SS-mediated cytotoxicity. Mechanistic studies involving cryoelectron microscopy identified a surface-exposed C-terminal PcrV epitope as the target of highly neutralizing mAbs with broad activity against drug-resistant PA isolates. These anti-PcrV mAbs were as effective as treatment with conventional antibiotics in vivo. Our study reveals that chronically infected patients represent a source of neutralizing antibodies, which can be exploited as therapeutics against PA.


Subject(s)
Antibodies, Bacterial , Antibodies, Neutralizing , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Antibodies, Bacterial/pharmacology , Cryoelectron Microscopy , Immunoglobulins/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas Infections/drug therapy
2.
Nat Immunol ; 25(2): 316-329, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38182669

ABSTRACT

Pneumococcal infections cause serious illness and death among older adults. The capsular polysaccharide vaccine PPSV23 and conjugated alternative PCV13 can prevent these infections; yet, underlying immunological responses and baseline predictors remain unknown. We vaccinated 39 older adults (>60 years) with PPSV23 or PCV13 and observed comparable antibody responses (day 28) and plasmablast transcriptional responses (day 10); however, the baseline predictors were distinct. Analyses of baseline flow cytometry and bulk and single-cell RNA-sequencing data revealed a baseline phenotype specifically associated with weaker PCV13 responses, which was characterized by increased expression of cytotoxicity-associated genes, increased frequencies of CD16+ natural killer cells and interleukin-17-producing helper T cells and a decreased frequency of type 1 helper T cells. Men displayed this phenotype more robustly and mounted weaker PCV13 responses than women. Baseline expression levels of a distinct gene set predicted PPSV23 responses. This pneumococcal precision vaccinology study in older adults uncovered distinct baseline predictors that might transform vaccination strategies and initiate novel interventions.


Subject(s)
Antibodies, Bacterial , Streptococcus pneumoniae , Male , Humans , Female , Aged , Vaccines, Conjugate , Double-Blind Method , Vaccination , Pneumococcal Vaccines , Polysaccharides
3.
Nat Immunol ; 22(12): 1515-1523, 2021 12.
Article in English | MEDLINE | ID: mdl-34811542

ABSTRACT

Development of an effective tuberculosis (TB) vaccine has suffered from an incomplete understanding of the correlates of protection against Mycobacterium tuberculosis (Mtb). Intravenous (i.v.) vaccination with Bacille Calmette-Guérin (BCG) provides nearly complete protection against TB in rhesus macaques, but the antibody response it elicits remains incompletely defined. Here we show that i.v. BCG drives superior antibody responses in the plasma and the lungs of rhesus macaques compared to traditional intradermal BCG administration. While i.v. BCG broadly expands antibody titers and functions, IgM titers in the plasma and lungs of immunized macaques are among the strongest markers of reduced bacterial burden. IgM was also enriched in macaques that received protective vaccination with an attenuated strain of Mtb. Finally, an Mtb-specific IgM monoclonal antibody reduced Mtb survival in vitro. Collectively, these data highlight the potential importance of IgM responses as a marker and mediator of protection against TB.


Subject(s)
Antibodies, Bacterial/blood , BCG Vaccine/administration & dosage , Immunogenicity, Vaccine , Immunoglobulin M/blood , Mycobacterium tuberculosis/immunology , Tuberculosis/prevention & control , Vaccination , Administration, Intravenous , Animals , Biomarkers/blood , Disease Models, Animal , Host-Pathogen Interactions , Macaca mulatta , Mycobacterium tuberculosis/pathogenicity , Time Factors , Tuberculosis/immunology , Tuberculosis/microbiology
4.
Cell ; 167(2): 306-307, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27716502

ABSTRACT

In this issue of Cell, Lu et al. provide important insights on the efficacy of human antibodies to Mycobacterium tuberculosis and on how functional heterogeneity of the antibody response may explain a century of contradictory evidence for the role of humoral immunity in defense against tuberculosis.


Subject(s)
Antibodies, Bacterial/immunology , Mycobacterium tuberculosis/immunology , Humans , Immunity, Humoral , Immunoglobulins , Tuberculosis/immunology
5.
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
6.
Nat Immunol ; 19(6): 617-624, 2018 06.
Article in English | MEDLINE | ID: mdl-29760533

ABSTRACT

Humoral immune responses to microbial polysaccharide surface antigens can prevent bacterial infection but are typically strain specific and fail to mediate broad protection against different serotypes. Here we describe a panel of affinity-matured monoclonal human antibodies from peripheral blood immunoglobulin M-positive (IgM+) and IgA+ memory B cells and clonally related intestinal plasmablasts, directed against the lipopolysaccharide (LPS) O-antigen of Klebsiella pneumoniae, an opportunistic pathogen and major cause of antibiotic-resistant nosocomial infections. The antibodies showed distinct patterns of in vivo cross-specificity and protection against different clinically relevant K. pneumoniae serotypes. However, cross-specificity was not limited to K. pneumoniae, as K. pneumoniae-specific antibodies recognized diverse intestinal microbes and neutralized not only K. pneumoniae LPS but also non-K. pneumoniae LPS. Our data suggest that the recognition of minimal glycan epitopes abundantly expressed on microbial surfaces might serve as an efficient humoral immunological mechanism to control invading pathogens and the large diversity of the human microbiota with a limited set of cross-specific antibodies.


Subject(s)
Antibodies, Bacterial/immunology , Antibody Specificity/immunology , Klebsiella pneumoniae/immunology , O Antigens/immunology , Antibodies, Monoclonal/immunology , Cross Reactions/immunology , Humans
7.
Nat Immunol ; 19(10): 1100-1111, 2018 10.
Article in English | MEDLINE | ID: mdl-30250184

ABSTRACT

Females have an overall advantage over males in resisting Gram-negative bacteremias, thus hinting at sexual dimorphism of immunity during infections. Here, through intravital microscopy, we observed a sex-biased difference in the capture of blood-borne bacteria by liver macrophages, a process that is critical for the clearance of systemic infections. Complement opsonization was indispensable for the capture of enteropathogenic Escherichia coli (EPEC) in male mice; however, a faster complement component 3-independent process involving abundant preexisting antibodies to EPEC was detected in female mice. These antibodies were elicited predominantly in female mice at puberty in response to estrogen regardless of microbiota-colonization conditions. Estrogen-driven antibodies were maternally transferrable to offspring and conferred protection during infancy. These antibodies were conserved in humans and recognized specialized oligosaccharides integrated into the bacterial lipopolysaccharide and capsule. Thus, an estrogen-driven, innate antibody-mediated immunological strategy conferred protection to females and their offspring.


Subject(s)
Antibodies, Bacterial/immunology , Escherichia coli Infections/immunology , Immunity, Innate/immunology , Sex Characteristics , Animals , Enteropathogenic Escherichia coli , Estrogens/immunology , Female , Humans , Infant , Kupffer Cells/immunology , Male , Maternal-Fetal Exchange/immunology , Mice , Pregnancy
8.
Immunity ; 53(1): 172-186.e6, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32610078

ABSTRACT

B-1 B cells derive from a developmental program distinct from that of conventional B cells, through B cell receptor (BCR)-dependent positive selection of fetally derived precursors. Here, we used direct labeling of B cells reactive with the N-acetyl-D-glucosamine (GlcNAc)-containing Lancefield group A carbohydrate of Streptococcus pyogenes to study the effects of bacterial antigens on the emergent B-1 B cell clonal repertoire. The number, phenotype, and BCR clonotypes of GlcNAc-reactive B-1 B cells were modulated by neonatal exposure to heat-killed S. pyogenes bacteria. GlcNAc-reactive B-1 clonotypes and serum antibodies were reduced in germ-free mice compared with conventionally raised mice. Colonization of germ-free mice with a conventional microbiota promoted GlcNAc-reactive B-1 B cell development and concomitantly elicited clonally related IgA+ plasma cells in the small intestine. Thus, exposure to microbial antigens in early life determines the clonality of the mature B-1 B cell repertoire and ensuing antibody responses, with implications for vaccination approaches and schedules.


Subject(s)
Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , B-Lymphocyte Subsets/immunology , Polysaccharides, Bacterial/immunology , Streptococcus pyogenes/immunology , Acetylglucosamine/metabolism , Animals , Animals, Newborn/immunology , Germ-Free Life/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology
9.
Cell ; 159(6): 1277-89, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25480293

ABSTRACT

Glycosylation processes are under high natural selection pressure, presumably because these can modulate resistance to infection. Here, we asked whether inactivation of the UDP-galactose:ß-galactoside-α1-3-galactosyltransferase (α1,3GT) gene, which ablated the expression of the Galα1-3Galß1-4GlcNAc-R (α-gal) glycan and allowed for the production of anti-α-gal antibodies (Abs) in humans, confers protection against Plasmodium spp. infection, the causative agent of malaria and a major driving force in human evolution. We demonstrate that both Plasmodium spp. and the human gut pathobiont E. coli O86:B7 express α-gal and that anti-α-gal Abs are associated with protection against malaria transmission in humans as well as in α1,3GT-deficient mice, which produce protective anti-α-gal Abs when colonized by E. coli O86:B7. Anti-α-gal Abs target Plasmodium sporozoites for complement-mediated cytotoxicity in the skin, immediately after inoculation by Anopheles mosquitoes. Vaccination against α-gal confers sterile protection against malaria in mice, suggesting that a similar approach may reduce malaria transmission in humans.


Subject(s)
Escherichia coli/physiology , Immunoglobulin M/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/transmission , Plasmodium/physiology , Polysaccharides/immunology , Adult , Animals , Anopheles/parasitology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Autoantigens/immunology , Cell Line, Tumor , Child , Escherichia coli/classification , Escherichia coli/immunology , Female , Galactosyltransferases/genetics , Galactosyltransferases/metabolism , Gastrointestinal Tract/microbiology , Germ-Free Life , Humans , Immunoglobulin M/blood , Malaria, Falciparum/microbiology , Malaria, Falciparum/parasitology , Mice , Plasmodium/classification , Plasmodium/growth & development , Plasmodium/immunology , Plasmodium falciparum/immunology , Plasmodium falciparum/physiology , Sporozoites/immunology , Toll-Like Receptor 9/agonists
10.
Immunity ; 50(4): 1099-1114.e10, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30876876

ABSTRACT

Inflammatory bowel disease is a chronic, relapsing condition with two subtypes, Crohn's disease (CD) and ulcerative colitis (UC). Genome-wide association studies (GWASs) in UC implicate a FCGR2A variant that alters the binding affinity of the antibody receptor it encodes, FcγRIIA, for immunoglobulin G (IgG). Here, we aimed to understand the mechanisms whereby changes in FcγRIIA affinity would affect inflammation in an IgA-dominated organ. We found a profound induction of anti-commensal IgG and a concomitant increase in activating FcγR signaling in the colonic mucosa of UC patients. Commensal-IgG immune complexes engaged gut-resident FcγR-expressing macrophages, inducing NLRP3- and reactive-oxygen-species-dependent production of interleukin-1ß (IL-1ß) and neutrophil-recruiting chemokines. These responses were modulated by the FCGR2A genotype. In vivo manipulation of macrophage FcγR signal strength in a mouse model of UC determined the magnitude of intestinal inflammation and IL-1ß-dependent type 17 immunity. The identification of an important contribution of IgG-FcγR-dependent inflammation to UC has therapeutic implications.


Subject(s)
Antibodies, Bacterial/immunology , Colitis, Ulcerative/immunology , Gastrointestinal Microbiome/immunology , Immunoglobulin G/immunology , Interleukin-1beta/immunology , Th17 Cells/immunology , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Gene Expression Regulation , Genotype , Humans , Inflammation , Interleukin-8/biosynthesis , Interleukin-8/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Macrophages/immunology , Mice , Phagocytes/immunology , RNA, Messenger/biosynthesis , Reactive Oxygen Species , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , Receptors, IgG/immunology
11.
N Engl J Med ; 389(3): 215-227, 2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37467497

ABSTRACT

BACKGROUND: Natural history studies have correlated serotype-specific anti-capsular polysaccharide (CPS) IgG in newborns with a reduced risk of group B streptococcal disease. A hexavalent CPS-cross-reactive material 197 glycoconjugate vaccine (GBS6) is being developed as a maternal vaccine to prevent invasive group B streptococcus in young infants. METHODS: In an ongoing phase 2, placebo-controlled trial involving pregnant women, we assessed the safety and immunogenicity of a single dose of various GBS6 formulations and analyzed maternally transferred anti-CPS antibodies. In a parallel seroepidemiologic study that was conducted in the same population, we assessed serotype-specific anti-CPS IgG concentrations that were associated with a reduced risk of invasive disease among newborns through 89 days of age to define putative protective thresholds. RESULTS: Naturally acquired anti-CPS IgG concentrations were associated with a reduced risk of disease among infants in the seroepidemiologic study. IgG thresholds that were determined to be associated with 75 to 95% reductions in the risk of disease were 0.184 to 0.827 µg per milliliter. No GBS6-associated safety signals were observed among the mothers or infants. The incidence of adverse events and of serious adverse events were similar across the trial groups for both mothers and infants; more local reactions were observed in the groups that received GBS6 containing aluminum phosphate. Among the infants, the most common serious adverse events were minor congenital anomalies (umbilical hernia and congenital dermal melanocytosis). GBS6 induced maternal antibody responses to all serotypes, with maternal-to-infant antibody ratios of approximately 0.4 to 1.3, depending on the dose. The percentage of infants with anti-CPS IgG concentrations above 0.184 µg per milliliter varied according to serotype and formulation, with 57 to 97% of the infants having a seroresponse to the most immunogenic formulation. CONCLUSIONS: GBS6 elicited anti-CPS antibodies against group B streptococcus in pregnant women that were transferred to infants at levels associated with a reduced risk of invasive group B streptococcal disease. (Funded by Pfizer and the Bill and Melinda Gates Foundation; C1091002 ClinicalTrials.gov number, NCT03765073.).


Subject(s)
Streptococcal Infections , Streptococcal Vaccines , Streptococcus agalactiae , Female , Humans , Infant , Infant, Newborn , Pregnancy , Antibodies, Bacterial , Immunoglobulin G , Seroepidemiologic Studies , Streptococcal Infections/epidemiology , Streptococcal Infections/immunology , Streptococcal Infections/prevention & control , Vaccines, Combined/administration & dosage , Vaccines, Combined/adverse effects , Vaccines, Combined/immunology , Vaccines, Combined/therapeutic use , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/adverse effects , Vaccines, Conjugate/immunology , Vaccines, Conjugate/therapeutic use , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/adverse effects , Streptococcal Vaccines/immunology , Streptococcal Vaccines/therapeutic use , Immunity, Maternally-Acquired/immunology
12.
PLoS Pathog ; 20(1): e1011957, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38241393

ABSTRACT

Streptococcus suis serotype 2 is an important encapsulated bacterial swine pathogen and zoonotic agent for which no effective vaccine exists. The interaction with B cells and the humoral response against S. suis are poorly understood despite their likely relevance for a potential vaccine. We evaluated germinal center (GC) B cell kinetics, as well as the production and role of S. suis-specific antibodies following infections in a mouse model. We found that mice infected with S. suis developed GC that peaked 13-21 days post-infection. GC further increased and persisted upon periodic reinfection that mimics real life conditions in swine farms. Anti-S. suis IgM and several IgG subclasses were produced, but antibodies against the S. suis capsular polysaccharide (CPS) were largely IgM. Interestingly, depletion of total IgG from the wild-type mice sera had no effect on bacterial killing by opsonophagocytosis in vitro. Somatic hypermutation and isotype switching were dispensable for controlling the infection or anti-CPS IgM production. However, T cell-deficient (Tcrb-/-) mice were unable to control bacteremia, produce optimal anti-CPS IgM titers, or elicit antibodies with opsonophagocytic activity. SAP deficiency, which prevents GC formation but not extrafollicular B cell responses, ablated anti S. suis-IgG production but maintained IgM production and eliminated the infection. In contrast, B cell deficient mice were unable to control bacteremia. Collectively, our results indicate that the antibody response plays a large role in immunity against S. suis, with GC-independent but T cell-dependent germline IgM being the major effective antibody specificities. Our results further highlight the importance IgM, and potentially anti-CPS antibodies, in clearing S. suis infections and provide insight for future development of S. suis vaccines.


Subject(s)
Bacteremia , Streptococcal Infections , Streptococcus suis , Vaccines , Animals , Mice , Swine , Streptococcus suis/genetics , Antibodies, Bacterial , Immunoglobulin G , Immunoglobulin M , T-Lymphocytes , Streptococcal Infections/microbiology
13.
Mol Cell Proteomics ; 23(5): 100753, 2024 May.
Article in English | MEDLINE | ID: mdl-38527648

ABSTRACT

Bacterial or viral antigens can contain subdominant protein regions that elicit weak antibody responses upon vaccination or infection although there is accumulating evidence that antibody responses against subdominant regions can enhance the protective immune response. One proposed mechanism for subdominant protein regions is the binding of host proteins that prevent antibody production against epitopes hidden within the protein binding interfaces. Here, we used affinity purification combined with quantitative mass spectrometry (AP-MS) to examine the level of competition between antigen-specific antibodies and host-pathogen protein interaction networks using the M1 protein from Streptococcus pyogenes as a model system. As most humans have circulating antibodies against the M1 protein, we first used AP-MS to show that the M1 protein interspecies protein network formed with human plasma proteins is largely conserved in naïve mice. Immunizing mice with the M1 protein generated a time-dependent increase of anti-M1 antibodies. AP-MS analysis comparing the composition of the M1-plasma protein network from naïve and immunized mice showed significant enrichment of 292 IgG peptides associated with 56 IgG chains in the immune mice. Despite the significant increase of bound IgGs, the levels of interacting plasma proteins were not significantly reduced in the immune mice. The results indicate that the antigen-specific polyclonal IgG against the M1 protein primarily targets epitopes outside the other plasma protein binding interfaces. In conclusion, this study demonstrates that AP-MS is a promising strategy to determine the relationship between antigen-specific antibodies and host-pathogen interaction networks that could be used to define subdominant protein regions of relevance for vaccine development.


Subject(s)
Antigens, Bacterial , Immunoglobulin G , Protein Binding , Streptococcus pyogenes , Animals , Streptococcus pyogenes/immunology , Streptococcus pyogenes/metabolism , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Mice , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Adaptive Immunity , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/metabolism , Antibodies, Bacterial/immunology , Protein Interaction Maps , Mass Spectrometry , Carrier Proteins/metabolism , Carrier Proteins/immunology , Female , Host-Pathogen Interactions/immunology
14.
Blood ; 142(2): 202-209, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37172200

ABSTRACT

Frequent plateletpheresis is associated with severe lymphopenia of uncertain clinical significance. We assessed the functional impact of frequent platelet donations and associated lymphopenia on the response to neoantigens. We conducted a prospective study of 102 platelet donors (HIV uninfected) who were naive to meningococcal vaccination recruited at Brigham and Women's Hospital. One dose of quadrivalent meningococcal conjugate vaccine was administered. Seroresponse was defined as a fourfold increase of serum bactericidal antibody titers and seroprotection was defined as postvaccination titers of ≥1:8, for each of the 4 vaccine antigens (A, C, W, and Y). Mean age of participants was 61 years, 69% were male, and medial number of platelet donations in prior year was 14 (interquartile range, 4-20). Frequent platelet donors had a low CD4 count (14% with ≤200/µL and 34% with ≤350/µL). Seroresponse rates varied from 68% for serogroup Y to 86% for serogroup A and were higher for participants with baseline titers of <1:8. Postvaccination seroprotection rates varied from 76% for serogroup Y to 96% for serogroup A. After adjustments for age, sex, and frequent donations, lower total lymphocyte or lower CD4 counts were not associated with lower responses. These data suggest no impairment by plateletpheresis-associated lymphopenia on response to these neoantigens. This trial was registered at www.clinicaltrials.gov as #NCT04224311.


Subject(s)
Lymphopenia , Meningococcal Infections , Meningococcal Vaccines , Female , Humans , Male , Middle Aged , Antibodies, Bacterial , Meningococcal Infections/prevention & control , Prospective Studies , Vaccines, Conjugate
15.
Int Immunol ; 36(8): 393-404, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-38536954

ABSTRACT

Invasive meningococcal disease (IMD) is caused by Neisseria meningitidis, with the main serogroups responsible for the disease being A, B, C, W, X, and Y. To date, several vaccines targeting N. meningitidis have been developed albeit with a short-lived protection. Given that MenW and MenB are the most common causes of IMD in Europe, Turkey, and the Middle East, we aimed to develop an outer membrane vesicle (OMV) based bivalent vaccine as the heterologous antigen source. Herein, we compared the immunogenicity, and breadth of serum bactericidal activity (SBA) assay-based protective coverage of OMV vaccine to the X serotype with existing commercial meningococcal conjugate and polysaccharide (PS) vaccines in a murine model. BALB/c mice were immunized with preclinical batches of the W + B OMV vaccine, either adjuvanted with Alum, CpG ODN, or their combinations, and compared with a MenACYW conjugate vaccine (NimenrixTM, Pfizer), and a MenB OMV-based vaccine (Bexsero®, GSK), The immune responses were assessed through enzyme-linked immunosorbent assay (ELISA) and SBA assay. Antibody responses and SBA titers were significantly higher in the W + B OMV vaccine when adjuvanted with Alum or CpG ODN, as compared to the control groups. Moreover, the SBA titers were not only significantly higher than those achieved with available conjugated ACYW vaccines but also on par with the 4CMenB vaccines. In conclusion, the W + B OMV vaccine demonstrated the capacity to elicit robust antibody responses, surpassing or matching the levels induced by licensed meningococcal vaccines. Consequently, the W + B OMV vaccine could potentially serve as a viable alternative or supplement to existing meningococcal vaccines.


Subject(s)
Alum Compounds , Meningococcal Infections , Meningococcal Vaccines , Mice, Inbred BALB C , Neisseria meningitidis , Oligodeoxyribonucleotides , Animals , Meningococcal Vaccines/immunology , Meningococcal Vaccines/administration & dosage , Mice , Neisseria meningitidis/immunology , Alum Compounds/administration & dosage , Oligodeoxyribonucleotides/immunology , Oligodeoxyribonucleotides/administration & dosage , Female , Meningococcal Infections/prevention & control , Meningococcal Infections/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Immunogenicity, Vaccine , Bacterial Outer Membrane/immunology
16.
J Immunol ; 210(1): 50-60, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36351696

ABSTRACT

Streptococcus pneumoniae persists as a leading cause of bacterial pneumonia despite the widespread use of polysaccharide-based vaccines. The limited serotype coverage of current vaccines has led to increased incidence of nonvaccine serotypes, as well as an increase in antibiotic resistance among these serotypes. Pneumococcal infection often follows a primary viral infection such as influenza virus, which hinders host defense and results in bacterial spread to the lungs. We previously isolated human monoclonal Abs (mAbs) against the conserved surface Ag pneumococcal histidine triad protein D (PhtD), and we demonstrated that mAbs to this Ag are protective against lethal pneumococcal challenge prophylactically and therapeutically. In this study, we elucidated the mechanism of protection of a protective anti-pneumococcal human mAb, PhtD3, which is mediated by the presence of complement and macrophages in a mouse model of pneumococcal infection. Treatment with mAb PhtD3 reduced blood and lung bacterial burden in mice, and mAb PhtD3 is able to bind to bacteria in the presence of the capsular polysaccharide, indicating exposure of surface PhtD on encapsulated bacteria. In a mouse model of secondary pneumococcal infection, protection mediated by mAb PhtD3 and another mAb targeting a different epitope, PhtD7, was reduced; however, robust protection was restored by combining mAb PhtD3 with mAb PhtD7, indicating a synergistic effect. Overall, these studies provide new insights into anti-pneumococcal mAb protection and demonstrate, to our knowledge, for the first time, that mAbs to pneumococcal surface proteins can protect against secondary pneumococcal infection in the mouse model.


Subject(s)
Pneumococcal Infections , Streptococcus pneumoniae , Humans , Animals , Mice , Antibodies, Monoclonal , Epitopes , Lung , Pneumococcal Vaccines , Antibodies, Bacterial , Bacterial Proteins
17.
J Immunol ; 211(10): 1540-1549, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37782044

ABSTRACT

Infection with Borrelia burgdorferi causes Lyme disease in humans. In small rodents, the natural reservoir species of this spirochete, infections lead to only modest disease manifestations, despite causing persistence infection. Although B cell responses are central for controlling bacterial tissue burden and disease manifestations, they lack classical aspects of T-dependent responses, such as sustained IgG affinity maturation and longevity, corresponding with a rapid collapse of germinal centers. Instead, the Ab response is characterized by strong and ongoing secretion of IgM, whose origins and impact on protective immunity to B. burgdorferi remain unknown. In this article, we demonstrate that B. burgdorferi infection-induced IgM in mice was produced continuously, mainly by conventional B, not B-1 cells, in a T-independent manner. Although IgM was passively protective and restricted early bacteremia, its production had no effects on bacterial dissemination into solid tissues, nor did it affect Borrelia tissue burden. The latter was controlled by the induction of bactericidal IgG, as shown comparing infections in wild type mice with those of mice lacking exclusively secreted IgM-/-, all class-switched Abs via deletion of aicda (AID-/-), and all secreted Abs (secreted IgM-/- × AID-/-). Consistent with the notion that B. burgdorferi infection drives production of IgM over more tissue-penetrable IgG, we demonstrated increased short- and long-term IgM Ab responses also to a coadministered, unrelated Ag. Thus, the continued production of IgM may explain the absence of B. burgdorferi in the blood.


Subject(s)
Bacteremia , Borrelia burgdorferi , Lyme Disease , Humans , Mice , Animals , Antibodies, Bacterial , Immunoglobulin M , Immunoglobulin G
18.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35058363

ABSTRACT

Gram-positive organisms with their thick envelope cannot be lysed by complement alone. Nonetheless, antibody-binding on the surface can recruit complement and mark these invaders for uptake and killing by phagocytes, a process known as opsonophagocytosis. The crystallizable fragment of immunoglobulins (Fcγ) is key for complement recruitment. The cell surface of S. aureus is coated with Staphylococcal protein A (SpA). SpA captures the Fcγ domain of IgG and interferes with opsonization by anti-S. aureus antibodies. In principle, the Fcγ domain of therapeutic antibodies could be engineered to avoid the inhibitory activity of SpA. However, the SpA-binding site on Fcγ overlaps with that of the neonatal Fc receptor (FcRn), an interaction that is critical for prolonging the half-life of serum IgG. This evolutionary adaptation poses a challenge for the exploration of Fcγ mutants that can both weaken SpA-IgG interactions and retain stability. Here, we use both wild-type and transgenic human FcRn mice to identify antibodies with enhanced half-life and increased opsonophagocytic killing in models of S. aureus infection and demonstrate that antibody-based immunotherapy can be improved by modifying Fcγ. Our experiments also show that by competing for FcRn-binding, staphylococci effectively reduce the half-life of antibodies during infection. These observations may have profound impact in treating cancer, autoimmune, and asthma patients colonized or infected with S. aureus and undergoing monoclonal antibody treatment.


Subject(s)
Antibodies, Bacterial/genetics , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Opsonization/immunology , Protein Engineering , Amino Acid Sequence , Antibody-Dependent Cell Cytotoxicity/immunology , Complement Activation , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Humans , Phagocytosis/immunology , Protein Binding , Protein Engineering/methods , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/immunology , Receptors, Fc/genetics , Staphylococcal Protein A/immunology , Staphylococcus aureus/immunology
19.
Proc Natl Acad Sci U S A ; 119(11): e2109667119, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35275791

ABSTRACT

SignificanceYersinia pestis, the etiologic agent of plague, has been responsible for high mortality in several epidemics throughout human history. This plague bacillus has been used as a biological weapon during human history and is currently one of the deadliest biological threats. Currently, no licensed plague vaccines are available in the Western world. Since an array of immunogens are enclosed in outer membrane vesicles (OMVs), immune responses elicited by OMVs against a diverse range of antigens may reduce the likelihood of antigen circumvention. Therefore, self-adjuvanting OMVs from a remodeled Yersinia pseudotuberculosis strain as a type of plague vaccine could diversify prophylactic choices and solve current vaccine limitations.


Subject(s)
Antigens, Bacterial , Lipid A , Plague Vaccine , Plague , Pore Forming Cytotoxic Proteins , Yersinia pseudotuberculosis , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Lethal Dose 50 , Lipid A/genetics , Lipid A/immunology , Mice , Plague/prevention & control , Plague Vaccine/administration & dosage , Plague Vaccine/genetics , Plague Vaccine/immunology , Plasmids/genetics , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis/immunology
20.
J Infect Dis ; 229(2): 376-383, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-37565807

ABSTRACT

BACKGROUND: The United States has experienced a resurgence of pertussis following the introduction of acellular pertussis (aP) vaccines. This is likely due to the failure of aP vaccines to induce durable immunity and prevent infection, carriage, and transmission. METHODS: To evaluate the impact of aP vaccination on the immune response to infection and test the ability of infection to reprogram aP-imprinted immune responses, we challenged unvaccinated and aP-vaccinated baboons with Bordetella pertussis multiple times and accessed the immune responses and outcomes of infections after each exposure. RESULTS: Multiple infections were required to elicit T-helper 17 responses and protection in aP-vaccinated animals comparable to responses seen in unvaccinated animals after a single challenge. Even after 3 challenges, T-helper 1 responses were not observed in aP-vaccinated animals. Immunoglobulin G responses to vaccine and nonvaccine antigens were not negatively affected in aP-vaccinated animals. CONCLUSIONS: Our results indicate that it is possible to retrain aP-primed immune responses, but it will likely require an optimal booster and multiple doses. Our results in the baboon model suggest that circulation of B. pertussis in aP-vaccinated populations is concentrated in the younger age bands of the population, providing information that can guide improved modeling of B. pertussis epidemiology in aP-vaccinated populations.


Subject(s)
Whooping Cough , Animals , Whooping Cough/prevention & control , Bordetella pertussis , Papio , Antibodies, Bacterial , Pertussis Vaccine , Vaccines, Acellular
SELECTION OF CITATIONS
SEARCH DETAIL