Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29.120
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 41: 343-373, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36750314

ABSTRACT

A large body of evidence generated in the last two and a half years addresses the roles of T cells in SARS-CoV-2 infection and following vaccination. Infection or vaccination induces multi-epitope CD4 and CD8 T cell responses with polyfunctionality. Early T cell responses have been associated with mild COVID-19 outcomes. In concert with animal model data, these results suggest that while antibody responses are key to prevent infection, T cell responses may also play valuable roles in reducing disease severity and controlling infection. T cell memory after vaccination is sustained for at least six months. While neutralizing antibody responses are impacted by SARS-CoV-2 variants, most CD4 and CD8 T cell responses are preserved. This review highlights the extensive progress made, and the data and knowledge gaps that remain, in our understanding of T cell responses to SARS-CoV-2 and COVID-19 vaccines.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Humans , COVID-19 Vaccines , CD8-Positive T-Lymphocytes , Antibodies, Viral
2.
Cell ; 187(6): 1374-1386.e13, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38428425

ABSTRACT

The World Health Organization declared mpox a public health emergency of international concern in July 2022. To investigate global mpox transmission and population-level changes associated with controlling spread, we built phylogeographic and phylodynamic models to analyze MPXV genomes from five global regions together with air traffic and epidemiological data. Our models reveal community transmission prior to detection, changes in case reporting throughout the epidemic, and a large degree of transmission heterogeneity. We find that viral introductions played a limited role in prolonging spread after initial dissemination, suggesting that travel bans would have had only a minor impact. We find that mpox transmission in North America began declining before more than 10% of high-risk individuals in the USA had vaccine-induced immunity. Our findings highlight the importance of broader routine specimen screening surveillance for emerging infectious diseases and of joint integration of genomic and epidemiological information for early outbreak control.


Subject(s)
Communicable Diseases, Emerging , Epidemics , Mpox (monkeypox) , Humans , Disease Outbreaks , Mpox (monkeypox)/epidemiology , Mpox (monkeypox)/transmission , Mpox (monkeypox)/virology , Public Health , Monkeypox virus/physiology
3.
Cell ; 187(20): 5554-5571.e19, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39197450

ABSTRACT

Immunization with mosaic-8b (nanoparticles presenting 8 SARS-like betacoronavirus [sarbecovirus] receptor-binding domains [RBDs]) elicits more broadly cross-reactive antibodies than homotypic SARS-CoV-2 RBD-only nanoparticles and protects against sarbecoviruses. To investigate original antigenic sin (OAS) effects on mosaic-8b efficacy, we evaluated the effects of prior COVID-19 vaccinations in non-human primates and mice on anti-sarbecovirus responses elicited by mosaic-8b, admix-8b (8 homotypics), or homotypic SARS-CoV-2 immunizations, finding the greatest cross-reactivity for mosaic-8b. As demonstrated by molecular fate mapping, in which antibodies from specific cohorts of B cells are differentially detected, B cells primed by WA1 spike mRNA-LNP dominated antibody responses after RBD-nanoparticle boosting. While mosaic-8b- and homotypic-nanoparticles boosted cross-reactive antibodies, de novo antibodies were predominantly induced by mosaic-8b, and these were specific for variant RBDs with increased identity to RBDs on mosaic-8b. These results inform OAS mechanisms and support using mosaic-8b to protect COVID-19-vaccinated/infected humans against as-yet-unknown SARS-CoV-2 variants and animal sarbecoviruses with human spillover potential.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Cross Reactions , Nanoparticles , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Nanoparticles/chemistry , Cross Reactions/immunology , SARS-CoV-2/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , Mice , Spike Glycoprotein, Coronavirus/immunology , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Female , Antibodies, Neutralizing/immunology , Betacoronavirus/immunology , Vaccination , B-Lymphocytes/immunology , Mice, Inbred BALB C
4.
Annu Rev Immunol ; 34: 635-59, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27168247

ABSTRACT

HIV employs multiple means to evade the humoral immune response, particularly the elicitation of and recognition by broadly neutralizing antibodies (bnAbs). Such antibodies can act antivirally against a wide spectrum of viruses by targeting relatively conserved regions on the surface HIV envelope trimer spike. Elicitation of and recognition by bnAbs are hindered by the arrangement of spikes on virions and the relatively difficult access to bnAb epitopes on spikes, including the proximity of variable regions and a high density of glycans. Yet, in a small proportion of HIV-infected individuals, potent bnAb responses do develop, and isolation of the corresponding monoclonal antibodies has been facilitated by identification of favorable donors with potent bnAb sera and by development of improved methods for human antibody generation. Molecular studies of recombinant Env trimers, alone and in interaction with bnAbs, are providing new insights that are fueling the development and testing of promising immunogens aimed at the elicitation of bnAbs.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Neutralizing/metabolism , HIV Antibodies/metabolism , HIV Infections/immunology , HIV/immunology , Immunization, Passive/methods , Virion/immunology , Animals , Conserved Sequence , HIV Infections/prevention & control , Humans , Immune Evasion , Immunization, Passive/trends , Viral Envelope Proteins/immunology
5.
Cell ; 186(21): 4615-4631.e16, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37769658

ABSTRACT

SARS-CoV-2 primary strain-based vaccination exerts a protective effect against Omicron variants-initiated infection, symptom occurrence, and disease severity in a booster-dependent manner. Yet, the underlying mechanisms remain unclear. During the 2022 Omicron outbreak in Shanghai, we enrolled 122 infected adults and 50 uninfected controls who had been unvaccinated or vaccinated with two or three doses of COVID-19 inactive vaccines and performed integrative analysis of 41-plex CyTOF, RNA-seq, and Olink on their peripheral blood samples. The frequencies of HLA-DRhi classical monocytes, non-classical monocytes, and Th1-like Tem tended to increase, whereas the frequency of Treg was reduced by booster vaccine, and they influenced symptom occurrence in a vaccine dose-dependent manner. Intercorrelation and mechanistic analysis suggested that the booster vaccination induced monocytic training, which would prime monocytic activation and maturation rather than differentiating into myeloid-derived suppressive cells upon Omicron infections. Overall, our study provides insights into how booster vaccination elaborates protective immunity across SARS-CoV-2 variants.

6.
Cell ; 185(3): 485-492.e10, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35051367

ABSTRACT

An outbreak of over 1,000 COVID-19 cases in Provincetown, Massachusetts (MA), in July 2021-the first large outbreak mostly in vaccinated individuals in the US-prompted a comprehensive public health response, motivating changes to national masking recommendations and raising questions about infection and transmission among vaccinated individuals. To address these questions, we combined viral genomic and epidemiological data from 467 individuals, including 40% of outbreak-associated cases. The Delta variant accounted for 99% of cases in this dataset; it was introduced from at least 40 sources, but 83% of cases derived from a single source, likely through transmission across multiple settings over a short time rather than a single event. Genomic and epidemiological data supported multiple transmissions of Delta from and between fully vaccinated individuals. However, despite its magnitude, the outbreak had limited onward impact in MA and the US overall, likely due to high vaccination rates and a robust public health response.


Subject(s)
COVID-19/epidemiology , COVID-19/immunology , COVID-19/transmission , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/virology , Child , Child, Preschool , Contact Tracing/methods , Disease Outbreaks , Female , Genome, Viral , Humans , Infant , Infant, Newborn , Male , Massachusetts/epidemiology , Middle Aged , Molecular Epidemiology , Phylogeny , SARS-CoV-2/classification , Vaccination , Whole Genome Sequencing , Young Adult
7.
Cell ; 185(26): 4873-4886.e10, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36513064

ABSTRACT

Respiratory syncytial virus (RSV) infection is a major cause of severe lower respiratory tract infection and death in young infants and the elderly. With no effective prophylactic treatment available, current vaccine candidates aim to elicit neutralizing antibodies. However, binding and neutralization have poorly predicted protection in the past, and accumulating data across epidemiologic cohorts and animal models collectively point to a role for additional antibody Fc-effector functions. To begin to define the humoral correlates of immunity against RSV, here we profiled an adenovirus 26 RSV-preF vaccine-induced humoral immune response in a group of healthy adults that were ultimately challenged with RSV. Protection from infection was linked to opsonophagocytic functions, driven by IgA and differentially glycosylated RSV-specific IgG profiles, marking a functional humoral immune signature of protection against RSV. Furthermore, Fc-modified monoclonal antibodies able to selectively recruit effector functions demonstrated significant antiviral control in a murine model of RSV.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Mice , Animals , Respiratory Syncytial Virus Infections/prevention & control , Antibodies, Neutralizing , Antibodies, Viral , Immunoglobulin G , Immunoglobulin Fc Fragments , Viral Fusion Proteins
8.
Cell ; 185(6): 1041-1051.e6, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35202566

ABSTRACT

The SARS-CoV-2 Omicron variant (B.1.1.529) contains mutations that mediate escape from antibody responses, although the extent to which these substitutions in spike and non-spike proteins affect T cell recognition is unknown. In this study, we show that T cell responses in individuals with prior infection, vaccination, both prior infection and vaccination, and boosted vaccination are largely preserved to Omicron spike and non-spike proteins. However, we also identify a subset of individuals (∼21%) with a >50% reduction in T cell reactivity to the Omicron spike. Evaluation of functional CD4+ and CD8+ memory T cell responses confirmed these findings and revealed that reduced recognition to Omicron spike is primarily observed within the CD8+ T cell compartment potentially due to escape from HLA binding. Booster vaccination enhanced T cell responses to Omicron spike. In contrast to neutralizing immunity, these findings suggest preservation of T cell responses to the Omicron variant, although with reduced reactivity in some individuals.

9.
Cell ; 185(23): 4333-4346.e14, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36257313

ABSTRACT

SARS-CoV-2 mRNA booster vaccines provide protection from severe disease, eliciting strong immunity that is further boosted by previous infection. However, it is unclear whether these immune responses are affected by the interval between infection and vaccination. Over a 2-month period, we evaluated antibody and B cell responses to a third-dose mRNA vaccine in 66 individuals with different infection histories. Uninfected and post-boost but not previously infected individuals mounted robust ancestral and variant spike-binding and neutralizing antibodies and memory B cells. Spike-specific B cell responses from recent infection (<180 days) were elevated at pre-boost but comparatively less so at 60 days post-boost compared with uninfected individuals, and these differences were linked to baseline frequencies of CD27lo B cells. Day 60 to baseline ratio of BCR signaling measured by phosphorylation of Syk was inversely correlated to days between infection and vaccination. Thus, B cell responses to booster vaccines are impeded by recent infection.


Subject(s)
B-Lymphocytes , COVID-19 , Viral Vaccines , Humans , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , SARS-CoV-2 , Vaccination , B-Lymphocytes/immunology , mRNA Vaccines
10.
Cell ; 185(3): 457-466.e4, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34995482

ABSTRACT

Recent surveillance has revealed the emergence of the SARS-CoV-2 Omicron variant (BA.1/B.1.1.529) harboring up to 36 mutations in spike protein, the target of neutralizing antibodies. Given its potential to escape vaccine-induced humoral immunity, we measured the neutralization potency of sera from 88 mRNA-1273, 111 BNT162b, and 40 Ad26.COV2.S vaccine recipients against wild-type, Delta, and Omicron SARS-CoV-2 pseudoviruses. We included individuals that received their primary series recently (<3 months), distantly (6-12 months), or an additional "booster" dose, while accounting for prior SARS-CoV-2 infection. Remarkably, neutralization of Omicron was undetectable in most vaccinees. However, individuals boosted with mRNA vaccines exhibited potent neutralization of Omicron, only 4-6-fold lower than wild type, suggesting enhanced cross-reactivity of neutralizing antibody responses. In addition, we find that Omicron pseudovirus infects more efficiently than other variants tested. Overall, this study highlights the importance of additional mRNA doses to broaden neutralizing antibody responses against highly divergent SARS-CoV-2 variants.

11.
Cell ; 185(4): 603-613.e15, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35026152

ABSTRACT

SARS-CoV-2 mRNA vaccines induce robust anti-spike (S) antibody and CD4+ T cell responses. It is not yet clear whether vaccine-induced follicular helper CD4+ T (TFH) cell responses contribute to this outstanding immunogenicity. Using fine-needle aspiration of draining axillary lymph nodes from individuals who received the BNT162b2 mRNA vaccine, we evaluated the T cell receptor sequences and phenotype of lymph node TFH. Mining of the responding TFH T cell receptor repertoire revealed a strikingly immunodominant HLA-DPB1∗04-restricted response to S167-180 in individuals with this allele, which is among the most common HLA alleles in humans. Paired blood and lymph node specimens show that while circulating S-specific TFH cells peak one week after the second immunization, S-specific TFH persist at nearly constant frequencies for at least six months. Collectively, our results underscore the key role that robust TFH cell responses play in establishing long-term immunity by this efficacious human vaccine.


Subject(s)
COVID-19/immunology , COVID-19/virology , Immunity/immunology , SARS-CoV-2/immunology , T Follicular Helper Cells/immunology , Vaccination , Vaccines, Synthetic/immunology , mRNA Vaccines/immunology , Adult , B-Lymphocytes/immunology , BNT162 Vaccine/immunology , COVID-19/blood , Clone Cells , Cohort Studies , Cytokines/metabolism , Female , Germinal Center/immunology , HLA-DP beta-Chains/immunology , Humans , Immunodominant Epitopes/immunology , Jurkat Cells , Lymph Nodes/metabolism , Male , Middle Aged , Peptides/chemistry , Peptides/metabolism , Protein Multimerization , Receptors, Antigen, T-Cell/metabolism
12.
Cell ; 184(15): 3936-3948.e10, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34192529

ABSTRACT

In this study we profiled vaccine-induced polyclonal antibodies as well as plasmablast-derived mAbs from individuals who received SARS-CoV-2 spike mRNA vaccine. Polyclonal antibody responses in vaccinees were robust and comparable to or exceeded those seen after natural infection. However, the ratio of binding to neutralizing antibodies after vaccination was greater than that after natural infection and, at the monoclonal level, we found that the majority of vaccine-induced antibodies did not have neutralizing activity. We also found a co-dominance of mAbs targeting the NTD and RBD of SARS-CoV-2 spike and an original antigenic-sin like backboost to spikes of seasonal human coronaviruses OC43 and HKU1. Neutralizing activity of NTD mAbs but not RBD mAbs against a clinical viral isolate carrying E484K as well as extensive changes in the NTD was abolished, suggesting that a proportion of vaccine-induced RBD binding antibodies may provide substantial protection against viral variants carrying single E484K RBD mutations.


Subject(s)
Antibodies, Viral/immunology , COVID-19 Vaccines/immunology , RNA, Messenger/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Amino Acid Substitution , Angiotensin-Converting Enzyme 2/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Binding, Competitive , Humans , Immunoglobulin G/metabolism , Mutation/genetics , Protein Domains , Somatic Hypermutation, Immunoglobulin/genetics
13.
Cell ; 184(26): 6229-6242.e18, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34910927

ABSTRACT

SARS-CoV-2 variants of concern exhibit varying degrees of transmissibility and, in some cases, escape from acquired immunity. Much effort has been devoted to measuring these phenotypes, but understanding their impact on the course of the pandemic-especially that of immune escape-has remained a challenge. Here, we use a mathematical model to simulate the dynamics of wild-type and variant strains of SARS-CoV-2 in the context of vaccine rollout and nonpharmaceutical interventions. We show that variants with enhanced transmissibility frequently increase epidemic severity, whereas those with partial immune escape either fail to spread widely or primarily cause reinfections and breakthrough infections. However, when these phenotypes are combined, a variant can continue spreading even as immunity builds up in the population, limiting the impact of vaccination and exacerbating the epidemic. These findings help explain the trajectories of past and present SARS-CoV-2 variants and may inform variant assessment and response in the future.


Subject(s)
COVID-19/immunology , COVID-19/transmission , Immune Evasion , SARS-CoV-2/immunology , COVID-19/epidemiology , COVID-19/virology , Computer Simulation , Humans , Immunity , Models, Biological , Reinfection , Vaccination
14.
Cell ; 184(13): 3467-3473.e11, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34133941

ABSTRACT

We previously reported that a single immunization with an adenovirus serotype 26 (Ad26)-vector-based vaccine expressing an optimized SARS-CoV-2 spike (Ad26.COV2.S) protected rhesus macaques against SARS-CoV-2 challenge. To evaluate reduced doses of Ad26.COV2.S, 30 rhesus macaques were immunized once with 1 × 1011, 5 × 1010, 1.125 × 1010, or 2 × 109 viral particles (vp) Ad26.COV2.S or sham and were challenged with SARS-CoV-2. Vaccine doses as low as 2 × 109 vp provided robust protection in bronchoalveolar lavage, whereas doses of 1.125 × 1010 vp were required for protection in nasal swabs. Activated memory B cells and binding or neutralizing antibody titers following vaccination correlated with protective efficacy. At suboptimal vaccine doses, viral breakthrough was observed but did not show enhancement of disease. These data demonstrate that a single immunization with relatively low dose of Ad26.COV2.S effectively protected against SARS-CoV-2 challenge in rhesus macaques, although a higher vaccine dose may be required for protection in the upper respiratory tract.


Subject(s)
Adenoviridae/immunology , COVID-19 Vaccines/immunology , COVID-19/immunology , SARS-CoV-2/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Female , Immunogenicity, Vaccine/immunology , Immunologic Memory/immunology , Macaca mulatta , Male , Spike Glycoprotein, Coronavirus/immunology , Vaccination/methods
15.
Cell ; 183(2): 315-323.e9, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32941801

ABSTRACT

BCG vaccination in children protects against heterologous infections and improves survival independently of tuberculosis prevention. The phase III ACTIVATE trial assessed whether BCG has similar effects in the elderly. In this double-blind, randomized trial, elderly patients (n = 198) received BCG or placebo vaccine at hospital discharge and were followed for 12 months for new infections. At interim analysis, BCG vaccination significantly increased the time to first infection (median 16 weeks compared to 11 weeks after placebo). The incidence of new infections was 42.3% (95% CIs 31.9%-53.4%) after placebo vaccination and 25.0% (95% CIs 16.4%-36.1%) after BCG vaccination; most of the protection was against respiratory tract infections of probable viral origin (hazard ratio 0.21, p = 0.013). No difference in the frequency of adverse effects was found. Data show that BCG vaccination is safe and can protect the elderly against infections. Larger studies are needed to assess protection against respiratory infections, including COVID-19 (ClinicalTrials.gov NCT03296423).


Subject(s)
BCG Vaccine/adverse effects , BCG Vaccine/immunology , Respiratory Tract Infections/prevention & control , Aged , Aged, 80 and over , BCG Vaccine/administration & dosage , Double-Blind Method , Female , Hospitalization , Humans , Male , Middle Aged , Respiratory Tract Infections/immunology , Virus Diseases/immunology , Virus Diseases/prevention & control
16.
Cell ; 178(1): 202-215.e14, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31204102

ABSTRACT

Despite the worldwide success of vaccination, newborns remain vulnerable to infections. While neonatal vaccination has been hampered by maternal antibody-mediated dampening of immune responses, enhanced regulatory and tolerogenic mechanisms, and immune system immaturity, maternal pre-natal immunization aims to boost neonatal immunity via antibody transfer to the fetus. However, emerging data suggest that antibodies are not transferred equally across the placenta. To understand this, we used systems serology to define Fc features associated with antibody transfer. The Fc-profile of neonatal and maternal antibodies differed, skewed toward natural killer (NK) cell-activating antibodies. This selective transfer was linked to digalactosylated Fc-glycans that selectively bind FcRn and FCGR3A, resulting in transfer of antibodies able to efficiently leverage innate immune cells present at birth. Given emerging data that vaccination may direct antibody glycosylation, our study provides insights for the development of next-generation maternal vaccines designed to elicit antibodies that will most effectively aid neonates.


Subject(s)
Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Placenta/metabolism , Polysaccharides/metabolism , Receptors, Fc/immunology , Receptors, Fc/metabolism , Adolescent , Adult , Belgium , Cell Degranulation , Cohort Studies , Female , Glycosylation , Humans , Infant, Newborn , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Male , Pregnancy , Receptors, IgG/metabolism , THP-1 Cells , United States , Vaccination , Young Adult
17.
Immunity ; 57(7): 1618-1628.e4, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38838672

ABSTRACT

Re-exposure to an antigen generates abundant antibody responses and drives the formation of secondary germinal centers (GCs). Recall GCs in mice consist almost entirely of naïve B cells, whereas recall antibodies derive overwhelmingly from memory B cells. Here, we examine this division between cellular and serum compartments. After repeated immunization with the same antigen, tetramer analyses of recall GCs revealed a marked decrease in the ability of B cells in these structures to bind the antigen. Boosting with viral variant proteins restored antigen binding in recall GCs, as did genetic ablation of primary-derived antibody-secreting cells through conditional deletion of Prdm1, demonstrating suppression of GC recall responses by pre-existing antibodies. In hapten-carrier experiments in which B and T cell specificities were uncoupled, memory T cell help allowed B cells with undetectable antigen binding to access GCs. Thus, antibody-mediated feedback steers recall GC B cells away from previously targeted epitopes and enables specific targeting of variant epitopes, with implications for vaccination protocols.


Subject(s)
B-Lymphocytes , Germinal Center , Immunologic Memory , Germinal Center/immunology , Animals , Mice , Immunologic Memory/immunology , B-Lymphocytes/immunology , Memory T Cells/immunology , Mice, Inbred C57BL , Positive Regulatory Domain I-Binding Factor 1/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Antibody Formation/immunology , Memory B Cells/immunology , Mice, Knockout
18.
Immunity ; 57(9): 2095-2107.e8, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39153479

ABSTRACT

Although the Bacille-Calmette-Guérin (BCG) vaccine is used to prevent tuberculosis, it also offers protection against a diverse range of non-mycobacterial infections. However, the underlying protective mechanisms in humans are not yet fully understood. Here, we surveyed at single-cell resolution the gene expression and chromatin landscape of human bone marrow, aspirated before and 90 days after BCG vaccination or placebo. We showed that BCG alters both the gene expression and epigenetic profiles of human hematopoietic stem and progenitor cells (HSPCs). Changes in gene expression occurred primarily within uncommitted stem cells. By contrast, changes in chromatin accessibility were most prevalent within differentiated progenitor cells at sites influenced by Kruppel-like factor (KLF) and early growth response (EGR) transcription factors and were highly correlated (r > 0.8) with the interleukin (IL)-1ß secretion capacity of paired peripheral blood mononuclear cells (PBMCs). Our findings shed light on BCG vaccination's profound and lasting effects on HSPCs and its influence on innate immune responses and trained immunity.


Subject(s)
BCG Vaccine , Epigenesis, Genetic , Immunity, Innate , Vaccination , Humans , BCG Vaccine/immunology , Epigenesis, Genetic/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Interleukin-1beta/metabolism , Bone Marrow/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Adult , Leukocytes, Mononuclear/immunology , Chromatin/metabolism , Female , Male , Cell Differentiation/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/immunology
19.
Immunity ; 57(5): 1160-1176.e7, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38697118

ABSTRACT

Multimodal single-cell profiling methods can capture immune cell variations unfolding over time at the molecular, cellular, and population levels. Transforming these data into biological insights remains challenging. Here, we introduce a framework to integrate variations at the human population and single-cell levels in vaccination responses. Comparing responses following AS03-adjuvanted versus unadjuvanted influenza vaccines with CITE-seq revealed AS03-specific early (day 1) response phenotypes, including a B cell signature of elevated germinal center competition. A correlated network of cell-type-specific transcriptional states defined the baseline immune status associated with high antibody responders to the unadjuvanted vaccine. Certain innate subsets in the network appeared "naturally adjuvanted," with transcriptional states resembling those induced uniquely by AS03-adjuvanted vaccination. Consistently, CD14+ monocytes from high responders at baseline had elevated phospho-signaling responses to lipopolysaccharide stimulation. Our findings link baseline immune setpoints to early vaccine responses, with positive implications for adjuvant development and immune response engineering.


Subject(s)
B-Lymphocytes , Influenza Vaccines , Single-Cell Analysis , Humans , Influenza Vaccines/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Vaccination , Antibodies, Viral/immunology , Adjuvants, Immunologic , Adjuvants, Vaccine , Monocytes/immunology , Polysorbates , Squalene/immunology , Immunity, Innate/immunology
20.
Immunity ; 57(9): 2191-2201.e5, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39168129

ABSTRACT

Memory B cells (MBCs) formed over the individual's lifetime constitute nearly half of the circulating B cell repertoire in humans. These pre-existing MBCs dominate recall responses to their cognate antigens, but how they respond to recognition of novel antigens is not well understood. Here, we tracked the origin and followed the differentiation paths of MBCs in the early anti-spike (S) response to mRNA vaccination in SARS-CoV-2-naive individuals on single-cell and monoclonal antibody levels. Pre-existing, highly mutated MBCs showed no signs of germinal center re-entry and rapidly developed into mature antibody-secreting cells (ASCs). By contrast, and despite similar levels of S reactivity, naive B cells showed strong signs of antibody affinity maturation before differentiating into MBCs and ASCs. Thus, pre-existing human MBCs differentiate into ASCs in response to novel antigens, but the quality of the humoral and cellular anti-S response improved through the clonal selection and affinity maturation of naive precursors.


Subject(s)
Antibodies, Viral , Antibody-Producing Cells , COVID-19 Vaccines , COVID-19 , Memory B Cells , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , Memory B Cells/immunology , COVID-19/immunology , COVID-19/prevention & control , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibody-Producing Cells/immunology , COVID-19 Vaccines/immunology , Vaccination , Antibody Affinity/immunology , Cell Differentiation/immunology , Germinal Center/immunology , Immunologic Memory/immunology , Antibodies, Monoclonal/immunology , Adult , Female
SELECTION OF CITATIONS
SEARCH DETAIL