Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.380
Filtrar
1.
J Clin Invest ; 134(7)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38557496

RESUMO

Programmed cell death protein 1 (PD-1) is an immune checkpoint marker commonly expressed on memory T cells and enriched in latently HIV-infected CD4+ T cells. We engineered an anti-PD-1 chimeric antigen receptor (CAR) to assess the impact of PD-1 depletion on viral reservoirs and rebound dynamics in SIVmac239-infected rhesus macaques (RMs). Adoptive transfer of anti-PD-1 CAR T cells was done in 2 SIV-naive and 4 SIV-infected RMs on antiretroviral therapy (ART). In 3 of 6 RMs, anti-PD-1 CAR T cells expanded and persisted for up to 100 days concomitant with the depletion of PD-1+ memory T cells in blood and tissues, including lymph node CD4+ follicular helper T (TFH) cells. Loss of TFH cells was associated with depletion of detectable SIV RNA from the germinal center (GC). However, following CAR T infusion and ART interruption, there was a marked increase in SIV replication in extrafollicular portions of lymph nodes, a 2-log higher plasma viremia relative to controls, and accelerated disease progression associated with the depletion of CD8+ memory T cells. These data indicate anti-PD-1 CAR T cells depleted PD-1+ T cells, including GC TFH cells, and eradicated SIV from this immunological sanctuary.


Assuntos
Linfócitos T CD4-Positivos , Receptores de Antígenos Quiméricos , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Linfócitos T CD4-Positivos/imunologia , Centro Germinativo/imunologia , Infecções por HIV/terapia , Macaca mulatta/metabolismo , Receptor de Morte Celular Programada 1 , Receptores de Antígenos Quiméricos/genética , Síndrome de Imunodeficiência Adquirida dos Símios/terapia
2.
Nature ; 628(8008): 612-619, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38509366

RESUMO

There is increasing interest in how immune cells in the meninges-the membranes that surround the brain and spinal cord-contribute to homeostasis and disease in the central nervous system1,2. The outer layer of the meninges, the dura mater, has recently been described to contain both innate and adaptive immune cells, and functions as a site for B cell development3-6. Here we identify organized lymphoid structures that protect fenestrated vasculature in the dura mater. The most elaborate of these dural-associated lymphoid tissues (DALT) surrounded the rostral-rhinal confluence of the sinuses and included lymphatic vessels. We termed this structure, which interfaces with the skull bone marrow and a comparable venous plexus at the skull base, the rostral-rhinal venolymphatic hub. Immune aggregates were present in DALT during homeostasis and expanded with age or after challenge with systemic or nasal antigens. DALT contain germinal centre B cells and support the generation of somatically mutated, antibody-producing cells in response to a nasal pathogen challenge. Inhibition of lymphocyte entry into the rostral-rhinal hub at the time of nasal viral challenge abrogated the generation of germinal centre B cells and class-switched plasma cells, as did perturbation of B-T cell interactions. These data demonstrate a lymphoid structure around vasculature in the dura mater that can sample antigens and rapidly support humoral immune responses after local pathogen challenge.


Assuntos
Dura-Máter , Imunidade Humoral , Tecido Linfoide , Veias , Administração Intranasal , Antígenos/administração & dosagem , Antígenos/imunologia , Medula Óssea/imunologia , Sistema Nervoso Central/irrigação sanguínea , Sistema Nervoso Central/imunologia , Dura-Máter/irrigação sanguínea , Dura-Máter/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Vasos Linfáticos/imunologia , Tecido Linfoide/irrigação sanguínea , Tecido Linfoide/imunologia , Plasmócitos/imunologia , Crânio/irrigação sanguínea , Linfócitos T/imunologia , Veias/fisiologia , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Animais , Camundongos , Idoso de 80 Anos ou mais
3.
Nature ; 627(8003): 407-415, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38383779

RESUMO

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Assuntos
Aquaporina 4 , Autoanticorpos , Autoantígenos , Linfócitos B , Tolerância Imunológica , Neuromielite Óptica , Animais , Humanos , Camundongos , 60533 , Aquaporina 4/deficiência , Aquaporina 4/genética , Aquaporina 4/imunologia , Aquaporina 4/metabolismo , Autoanticorpos/imunologia , Autoantígenos/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Antígenos CD40/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Neuromielite Óptica/imunologia , Neuromielite Óptica/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Células Epiteliais da Tireoide/imunologia , Células Epiteliais da Tireoide/metabolismo , Transcriptoma
4.
Science ; 383(6684): eadg0564, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38359115

RESUMO

Influenza viruses escape immunity owing to rapid antigenic evolution, which requires vaccination strategies that allow for broadly protective antibody responses. We found that the lipid globotriaosylceramide (Gb3) expressed on germinal center (GC) B cells is essential for the production of high-affinity antibodies. Mechanistically, Gb3 bound and disengaged CD19 from its chaperone CD81, permitting CD19 to translocate to the B cell receptor complex to trigger signaling. Moreover, Gb3 regulated major histocompatibility complex class II expression to increase diversity of T follicular helper and GC B cells reactive with subdominant epitopes. In influenza infection, elevating Gb3, either endogenously or exogenously, promoted broadly reactive antibody responses and cross-protection. These data demonstrate that Gb3 determines the affinity and breadth of B cell immunity and has potential as a vaccine adjuvant.


Assuntos
Anticorpos Antivirais , Linfócitos B , Centro Germinativo , Infecções por Orthomyxoviridae , Orthomyxoviridae , Triexosilceramidas , Formação de Anticorpos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Centro Germinativo/efeitos dos fármacos , Centro Germinativo/imunologia , Triexosilceramidas/metabolismo , Triexosilceramidas/farmacologia , Animais , Camundongos , Camundongos Knockout , Humanos , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/imunologia , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia
5.
Nature ; 617(7961): 592-598, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37011668

RESUMO

The primary two-dose SARS-CoV-2 mRNA vaccine series are strongly immunogenic in humans, but the emergence of highly infectious variants necessitated additional doses and the development of vaccines aimed at the new variants1-4. SARS-CoV-2 booster immunizations in humans primarily recruit pre-existing memory B cells5-9. However, it remains unclear whether the additional doses induce germinal centre reactions whereby re-engaged B cells can further mature, and whether variant-derived vaccines can elicit responses to variant-specific epitopes. Here we show that boosting with an mRNA vaccine against the original monovalent SARS-CoV-2 mRNA vaccine or the bivalent B.1.351 and B.1.617.2 (Beta/Delta) mRNA vaccine induced robust spike-specific germinal centre B cell responses in humans. The germinal centre response persisted for at least eight weeks, leading to significantly more mutated antigen-specific bone marrow plasma cell and memory B cell compartments. Spike-binding monoclonal antibodies derived from memory B cells isolated from individuals boosted with either the original SARS-CoV-2 spike protein, bivalent Beta/Delta vaccine or a monovalent Omicron BA.1-based vaccine predominantly recognized the original SARS-CoV-2 spike protein. Nonetheless, using a more targeted sorting approach, we isolated monoclonal antibodies that recognized the BA.1 spike protein but not the original SARS-CoV-2 spike protein from individuals who received the mRNA-1273.529 booster; these antibodies were less mutated and recognized novel epitopes within the spike protein, suggesting that they originated from naive B cells. Thus, SARS-CoV-2 booster immunizations in humans induce robust germinal centre B cell responses and can generate de novo B cell responses targeting variant-specific epitopes.


Assuntos
Linfócitos B , Vacinas contra COVID-19 , COVID-19 , Centro Germinativo , Imunização Secundária , Humanos , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , COVID-19/virologia , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/imunologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Plasmócitos/citologia , Plasmócitos/imunologia , Células B de Memória/citologia , Células B de Memória/imunologia , Epitopos de Linfócito B/genética , Epitopos de Linfócito B/imunologia
6.
J Immunol ; 210(7): 905-915, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36779803

RESUMO

High-affinity allergen-specific IgE is essential for the severe allergic anaphylaxis response. High-affinity Abs are formed by successive rounds of selection of Ag-specific B cells in the germinal center (GC); however, several studies have shown that IgE+ GC B cells are impaired in their ability to undergo selection in the GC. A pathway, known as the "indirect switching pathway" for IgE, has been described whereby Ag-specific B cells initially switch to the IgG1 isotype and undergo affinity selection in the GC, with a secondary switch to the IgE isotype after affinity selection. In previous work, using a food allergy model in mice, we investigated how high-affinity IgE develops in the GC, but we did not test the indirect switching model. In this study, we analyzed the importance of the indirect switching pathway by constructing IgG1-cre Bcl6-fl/fl mice. In these mice, once B cells switch to IgG1, they delete Bcl6 and thus cannot enter or persist in the GC. When we tested IgG1-cre Bcl6-fl/fl mice with our food allergy model, we found that, as expected, IgG1 Abs had decreased affinity, but unexpectedly, the affinity of IgE for allergen was unchanged. IgG1-cre Bcl6-fl/fl mice underwent anaphylaxis in response to allergen, consistent with the formation of high-affinity IgE. Thus, in a food allergy response, high-affinity IgE can be efficiently formed in the absence of indirect switching to IgG1, either by direct selection of IgE+ GC B cells or indirect selection of IgM+ GC B cells that later switch to IgE.


Assuntos
Hipersensibilidade Alimentar , Centro Germinativo , Imunoglobulina E , Animais , Camundongos , Imunoglobulina E/biossíntese , Imunoglobulina E/imunologia , Imunoglobulina G , Centro Germinativo/imunologia , Hipersensibilidade Alimentar/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Switching de Imunoglobulina
7.
Cell ; 186(1): 131-146.e13, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36565697

RESUMO

Germinal centers (GCs) form in secondary lymphoid organs in response to infection and immunization and are the source of affinity-matured B cells. The duration of GC reactions spans a wide range, and long-lasting GCs (LLGCs) are potentially a source of highly mutated B cells. We show that rather than consisting of continuously evolving B cell clones, LLGCs elicited by influenza virus or SARS-CoV-2 infection in mice are sustained by progressive replacement of founder clones by naive-derived invader B cells that do not detectably bind viral antigens. Rare founder clones that resist replacement for long periods are enriched in clones with heavily mutated immunoglobulins, including some with very high affinity for antigen, that can be recalled by boosting. Our findings reveal underappreciated aspects of the biology of LLGCs generated by respiratory virus infection and identify clonal replacement as a potential constraint on the development of highly mutated antibodies within these structures.


Assuntos
Linfócitos B , Centro Germinativo , Infecções por Vírus de RNA , Animais , Camundongos , Linfócitos B/citologia , Linfócitos B/imunologia , Células Clonais , COVID-19 , Centro Germinativo/citologia , Centro Germinativo/imunologia , SARS-CoV-2 , Influenza Humana , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/patologia , Infecções por Vírus de RNA/virologia
8.
Nature ; 613(7945): 735-742, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36473496

RESUMO

Feedback inhibition of humoral immunity by antibodies was first documented in 19091. Subsequent studies showed that, depending on the context, antibodies can enhance or inhibit immune responses2,3. However, little is known about how pre-existing antibodies influence the development of memory B cells. Here we examined the memory B cell response in individuals who received two high-affinity anti-SARS-CoV-2 monoclonal antibodies and subsequently two doses of an mRNA vaccine4-8. We found that the recipients of the monoclonal antibodies produced antigen-binding and neutralizing titres that were only fractionally lower compared than in control individuals. However, the memory B cells of the individuals who received the monoclonal antibodies differed from those of control individuals in that they predominantly expressed low-affinity IgM antibodies that carried small numbers of somatic mutations and showed altered receptor binding domain (RBD) target specificity, consistent with epitope masking. Moreover, only 1 out of 77 anti-RBD memory antibodies tested neutralized the virus. The mechanism underlying these findings was examined in experiments in mice that showed that germinal centres formed in the presence of the same antibodies were dominated by low-affinity B cells. Our results indicate that pre-existing high-affinity antibodies bias germinal centre and memory B cell selection through two distinct mechanisms: (1) by lowering the activation threshold for B cells, thereby permitting abundant lower-affinity clones to participate in the immune response; and (2) through direct masking of their cognate epitopes. This may in part explain the shifting target profile of memory antibodies elicited by booster vaccinations9.


Assuntos
Anticorpos Antivirais , Linfócitos B , Vacinas contra COVID-19 , COVID-19 , Retroalimentação Fisiológica , Memória Imunológica , Vacinação , Vacinas de mRNA , Animais , Camundongos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , COVID-19/terapia , COVID-19/virologia , SARS-CoV-2/imunologia , Vacinas de mRNA/imunologia , Vacinas contra COVID-19/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Imunoglobulina M/imunologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Imunização Secundária , Hipermutação Somática de Imunoglobulina
9.
Nature ; 609(7929): 998-1004, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36131022

RESUMO

Germinal centres are the engines of antibody evolution. Here, using human immunodeficiency virus (HIV) Env protein immunogen priming in rhesus monkeys followed by a long period without further immunization, we demonstrate germinal centre B (BGC) cells that last for at least 6 months. A 186-fold increase in BGC cells was present by week 10 compared with conventional immunization. Single-cell transcriptional profiling showed that both light- and dark-zone germinal centre states were sustained. Antibody somatic hypermutation of BGC cells continued to accumulate throughout the 29-week priming period, with evidence of selective pressure. Env-binding BGC cells were still 49-fold above baseline at 29 weeks, which suggests that they could remain active for even longer periods of time. High titres of HIV-neutralizing antibodies were generated after a single booster immunization. Fully glycosylated HIV trimer protein is a complex antigen, posing considerable immunodominance challenges for B cells1,2. Memory B cells generated under these long priming conditions had higher levels of antibody somatic hypermutation, and both memory B cells and antibodies were more likely to recognize non-immunodominant epitopes. Numerous BGC cell lineage phylogenies spanning more than the 6-month germinal centre period were identified, demonstrating continuous germinal centre activity and selection for at least 191 days with no further antigen exposure. A long-prime, slow-delivery (12 days) immunization approach holds promise for difficult vaccine targets and suggests that patience can have great value for tuning of germinal centres to maximize antibody responses.


Assuntos
Afinidade de Anticorpos , Linfócitos B , Movimento Celular , Células Clonais , Centro Germinativo , Anticorpos Anti-HIV , Imunização , Animais , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/imunologia , Afinidade de Anticorpos/genética , Afinidade de Anticorpos/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Células Clonais/citologia , Células Clonais/imunologia , Epitopos de Linfócito B/imunologia , Perfilação da Expressão Gênica , Centro Germinativo/citologia , Centro Germinativo/imunologia , Anticorpos Anti-HIV/genética , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Imunização Secundária , Macaca mulatta/imunologia , Macaca mulatta/virologia , Células B de Memória/citologia , Células B de Memória/imunologia , Análise de Célula Única , Hipermutação Somática de Imunoglobulina/genética , Hipermutação Somática de Imunoglobulina/imunologia , Fatores de Tempo , Produtos do Gene env do Vírus da Imunodeficiência Humana/administração & dosagem , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia
10.
Proc Natl Acad Sci U S A ; 119(32): e2123362119, 2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35921433

RESUMO

The germinal center (GC) plays a central role in the generation of antigen-specific B cells and antibodies. Tight regulation of the GC is essential due to the inherent risks of tumorigenesis and autoimmunity posed by inappropriate GC B cell processes. Gammaherpesviruses such as Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) utilize numerous armaments to drive infected naïve B cells, independent of antigen, through GC reactions to expand the latently infected B cell population and establish a stable latency reservoir. We previously demonstrated that the MHV68 microRNA (miRNA) mghv-miR-M1-7-5p represses host EWSR1 (Ewing sarcoma breakpoint region 1) to promote B cell infection. EWSR1 is a transcription and splicing regulator that is recognized for its involvement as a fusion protein in Ewing sarcoma. A function for EWSR1 in B cell responses has not been previously reported. Here, we demonstrate that 1) B cell-specific deletion of EWSR1 had no effect on generation of mature B cell subsets or basal immunoglobulin levels in naïve mice, 2) repression or ablation of EWSR1 in B cells promoted expansion of MHV68 latently infected GC B cells, and 3) B cell-specific deletion of EWSR1 during a normal immune response to nonviral antigen resulted in significantly elevated numbers of antigen-specific GC B cells, plasma cells, and circulating antibodies. Notably, EWSR1 deficiency did not affect the proliferation or survival of GC B cells but instead resulted in the generation of increased numbers of precursor GC B cells. Cumulatively, these findings demonstrate that EWSR1 is a negative regulator of B cell responses.


Assuntos
Linfócitos B , Gammaherpesvirinae , Centro Germinativo , Infecções por Herpesviridae , MicroRNAs , Proteína EWS de Ligação a RNA , Infecções Tumorais por Vírus , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Gammaherpesvirinae/genética , Gammaherpesvirinae/fisiologia , Deleção de Genes , Centro Germinativo/imunologia , Centro Germinativo/virologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína EWS de Ligação a RNA/genética , Proteína EWS de Ligação a RNA/metabolismo , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia , Latência Viral
11.
Cancer Immunol Res ; 10(9): 1040, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35917325

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy has achieved remarkable milestones in the treatment of B-cell malignancies. However, cancer cells frequently survive CAR T-cell killing in a large cohort of patients. Relapse oftentimes is associated with antigen loss. In this issue, Im and colleagues report a new mechanism of leukemic-cell resistance to anti-CD19 CAR T cells: Leukemic cells can enable a B-cell activation and germinal center reaction signature, which causes CD19 transcriptional downregulation and survival from CAR exposure. See related article by Im et al., p. 1055 (5).


Assuntos
Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Antígenos CD19/imunologia , Centro Germinativo/imunologia , Humanos , Imunoterapia Adotiva , Linfoma de Células B/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia
12.
Nature ; 608(7924): 724-732, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35948631

RESUMO

The lymphocyte genome is prone to many threats, including programmed mutation during differentiation1, antigen-driven proliferation and residency in diverse microenvironments. Here, after developing protocols for expansion of single-cell lymphocyte cultures, we sequenced whole genomes from 717 normal naive and memory B and T cells and haematopoietic stem cells. All lymphocyte subsets carried more point mutations and structural variants than haematopoietic stem cells, with higher burdens in memory cells than in naive cells, and with T cells accumulating mutations at a higher rate throughout life. Off-target effects of immunological diversification accounted for approximately half of the additional differentiation-associated mutations in lymphocytes. Memory B cells acquired, on average, 18 off-target mutations genome-wide for every on-target IGHV mutation during the germinal centre reaction. Structural variation was 16-fold higher in lymphocytes than in stem cells, with around 15% of deletions being attributable to off-target recombinase-activating gene activity. DNA damage from ultraviolet light exposure and other sporadic mutational processes generated hundreds to thousands of mutations in some memory cells. The mutation burden and signatures of normal B cells were broadly similar to those seen in many B-cell cancers, suggesting that malignant transformation of lymphocytes arises from the same mutational processes that are active across normal ontogeny. The mutational landscape of normal lymphocytes chronicles the off-target effects of programmed genome engineering during immunological diversification and the consequences of differentiation, proliferation and residency in diverse microenvironments.


Assuntos
Linfócitos , Mutação , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Diferenciação Celular , Proliferação de Células , Microambiente Celular , Dano ao DNA/genética , Dano ao DNA/efeitos da radiação , Centro Germinativo/citologia , Centro Germinativo/imunologia , Humanos , Memória Imunológica/genética , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Linfócitos/patologia , Neoplasias/genética , Neoplasias/patologia
13.
Proc Natl Acad Sci U S A ; 119(37): e2205598119, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36006981

RESUMO

The humoral immune response, a key arm of adaptive immunity, consists of B cells and their products. Upon infection or vaccination, B cells undergo a Darwinian evolutionary process in germinal centers (GCs), resulting in the production of antibodies and memory B cells. We developed a computational model to study how humoral memory is recalled upon reinfection or booster vaccination. We find that upon reexposure to the same antigen, affinity-dependent selective expansion of available memory B cells outside GCs (extragerminal center compartments [EGCs]) results in a rapid response made up of the best available antibodies. Memory B cells that enter secondary GCs can undergo mutation and selection to generate even more potent responses over time, enabling greater protection upon subsequent exposure to the same antigen. GCs also generate a diverse pool of B cells, some with low antigen affinity. These results are consistent with our analyses of data from humans vaccinated with two doses of a COVID-19 vaccine. Our results further show that the diversity of memory B cells generated in GCs is critically important upon exposure to a variant antigen. Clones drawn from this diverse pool that cross-react with the variant are rapidly expanded in EGCs to provide the best protection possible while new secondary GCs generate a tailored response for the new variant. Based on a simple evolutionary model, we suggest that the complementary roles of EGC and GC processes we describe may have evolved in response to complex organisms being exposed to evolving pathogen families for millennia.


Assuntos
Antígenos , Linfócitos B , Imunidade Humoral , Memória Imunológica , Antígenos/imunologia , Linfócitos B/imunologia , COVID-19/prevenção & controle , Vacinas contra COVID-19/imunologia , Simulação por Computador , Centro Germinativo/imunologia , Humanos , Modelos Biológicos
14.
Cancer Immunol Res ; 10(9): 1055-1068, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35759797

RESUMO

Chimeric antigen receptor (CAR) T-cell therapy has led to tremendous successes in the treatment of B-cell malignancies. However, a large fraction of treated patients relapse, often with disease expressing reduced levels of the target antigen. Here, we report that exposing CD19+ B-cell acute lymphoblastic leukemia (B-ALL) cells to CD19 CAR T cells reduced CD19 expression within hours. Initially, CD19 CAR T cells caused clustering of CD19 at the T cell-leukemia cell interface followed by CD19 internalization and decreased CD19 surface expression on the B-ALL cells. CD19 expression was then repressed by transcriptional rewiring. Using single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin using sequencing, we demonstrated that a subset of refractory CD19low cells sustained decreased CD19 expression through transcriptional programs of physiologic B-cell activation and germinal center reaction. Inhibiting B-cell activation programs with the Bruton's tyrosine kinase inhibitor ibrutinib increased the cytotoxicity of CD19 CAR T cells without affecting CAR T-cell viability. These results demonstrate transcriptional plasticity as an underlying mechanism of escape from CAR T cells and highlight the importance of combining CAR T-cell therapy with targeted therapies that aim to overcome this plasticity. See related Spotlight by Zhao and Melenhorst, p. 1040.


Assuntos
Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Antígenos CD19/imunologia , Centro Germinativo/imunologia , Humanos , Imunoterapia Adotiva/métodos , Linfoma de Células B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia
15.
Biochem Biophys Res Commun ; 618: 86-92, 2022 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-35716600

RESUMO

The transcription factor Bach2 serves as a crucial regulator of the germinal center (GC) reaction, which is required for production of high-affinity antibodies and establishment of long-lived B cell memory. However, the stage at which Bach2 controls the GC programs and the precise mechanism underlying these processes remain poorly understood. In this study, we show that genetic ablation of Bach2 in GC B cells of mice impairs their survival and maintenance, and memory B cell formation. These defects can be rescued by enforced expression of anti-apoptotic gene Bcl2. As expected, Bach2-deficient GC B cells are defective in antibody affinity maturation, but have normal somatic hyper mutation and class switch recombination of immunoglobulin genes. Mechanistically, Bach2 controls the GC programs by directly repressing pro-apoptotic gene Bim and a set of genes involved in cell stress response and metabolic processes. Thus, our work reveals the precise roles of Bach2 in the GC biology, and demonstrates that Bach2 acts as a crucial survival regulator of GC B cells, providing a key mechanism underlying GC B maintenance and B cell memory formation.


Assuntos
Linfócitos B , Fatores de Transcrição de Zíper de Leucina Básica , Centro Germinativo , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Sobrevivência Celular/fisiologia , Centro Germinativo/citologia , Centro Germinativo/imunologia , Memória Imunológica , Camundongos , Fatores de Transcrição/metabolismo
16.
Immunity ; 55(3): 385-387, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35263564

RESUMO

Some bacteria and parasites, such as Salmonella, actively disrupt germinal centers and elicit only low affinity antibodies, but the mechanisms by which microbes alter these responses is poorly understood. In this issue of Immunity, Biram et al. (2022) uncover a mechanism by which Salmonella recruits Sca-1+ monocytes to germinal centers and impairs metabolic adaptation.


Assuntos
Infecções Bacterianas , Monócitos , Centro Germinativo/imunologia , Humanos , Salmonella
17.
Front Immunol ; 13: 844837, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35296091

RESUMO

In this work, we evaluated recombinant receptor binding domain (RBD)-based vaccine formulation prototypes with potential for further clinical development. We assessed different formulations containing RBD plus alum, AddaS03, AddaVax, or the combination of alum and U-Omp19: a novel Brucella spp. protease inhibitor vaccine adjuvant. Results show that the vaccine formulation composed of U-Omp19 and alum as adjuvants has a better performance: it significantly increased mucosal and systemic neutralizing antibodies in comparison to antigen plus alum, AddaVax, or AddaS03. Antibodies induced with the formulation containing U-Omp19 and alum not only increased their neutralization capacity against the ancestral virus but also cross-neutralized alpha, lambda, and gamma variants with similar potency. Furthermore, the addition of U-Omp19 to alum vaccine formulation increased the frequency of RBD-specific geminal center B cells and plasmablasts. Additionally, U-Omp19+alum formulation induced RBD-specific Th1 and CD8+ T-cell responses in spleens and lungs. Finally, this vaccine formulation conferred protection against an intranasal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge of K18-hACE2 mice.


Assuntos
Adjuvantes Imunológicos/metabolismo , Linfócitos B/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella/metabolismo , Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Centro Germinativo/imunologia , SARS-CoV-2/fisiologia , Compostos de Alúmen/metabolismo , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais , Formação de Anticorpos , Proteínas da Membrana Bacteriana Externa/imunologia , Brucella/imunologia , Resistência à Doença , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Glicoproteína da Espícula de Coronavírus/imunologia
18.
Front Immunol ; 13: 838328, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251032

RESUMO

Confirmed SARS-coronavirus-2 infection with gastrointestinal symptoms and changes in microbiota associated with coronavirus disease 2019 (COVID-19) severity have been previously reported, but the disease impact on the architecture and cellularity of ileal Peyer's patches (PP) remains unknown. Here we analysed post-mortem tissues from throughout the gastrointestinal (GI) tract of patients who died with COVID-19. When virus was detected by PCR in the GI tract, immunohistochemistry identified virus in epithelium and lamina propria macrophages, but not in lymphoid tissues. Immunohistochemistry and imaging mass cytometry (IMC) analysis of ileal PP revealed depletion of germinal centres (GC), disruption of B cell/T cell zonation and decreased potential B and T cell interaction and lower nuclear density in COVID-19 patients. This occurred independent of the local viral levels. The changes in PP demonstrate that the ability to mount an intestinal immune response is compromised in severe COVID-19, which could contribute to observed dysbiosis.


Assuntos
Atrofia/imunologia , COVID-19/imunologia , Centro Germinativo/imunologia , Mucosa Intestinal/imunologia , Nódulos Linfáticos Agregados/imunologia , Linfócitos B/imunologia , Humanos , Tecido Linfoide/imunologia , Macrófagos/imunologia , SARS-CoV-2/imunologia , Linfócitos T/imunologia
19.
Cell Rep ; 38(8): 110399, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35139367

RESUMO

Follicular helper T (Tfh) cells promote, whereas follicular regulatory T (Tfr) cells restrain, germinal center (GC) reactions. However, the precise roles of these cells in the complex GC reaction remain poorly understood. Here, we perturb Tfh or Tfr cells after SARS-CoV-2 spike protein vaccination in mice. We find that Tfh cells promote the frequency and somatic hypermutation (SHM) of Spike-specific GC B cells and regulate clonal diversity. Tfr cells similarly control SHM and clonal diversity in the GC but do so by limiting clonal competition. In addition, deletion of Tfh or Tfr cells during primary vaccination results in changes in SHM after vaccine boosting. Aged mice, which have altered Tfh and Tfr cells, have lower GC responses, presenting a bimodal distribution of SHM. Together, these data demonstrate that GC responses to SARS-CoV-2 spike protein vaccines require a fine balance of positive and negative follicular T cell help to optimize humoral immunity.


Assuntos
COVID-19/prevenção & controle , Centro Germinativo/imunologia , Glicoproteína da Espícula de Coronavírus/administração & dosagem , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Envelhecimento , Animais , Anticorpos Antivirais/sangue , Linfócitos B/imunologia , Linfócitos B/metabolismo , COVID-19/virologia , Centro Germinativo/citologia , Centro Germinativo/metabolismo , Imunidade Humoral , Camundongos , Camundongos Endogâmicos C57BL , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Vacinação , Vacinas de Subunidades/imunologia
20.
PLoS Pathog ; 18(2): e1009831, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35130312

RESUMO

During chronic human immunodeficiency virus (HIV) or simian immunodeficiency virus (SIV) infection prior to AIDS progression, the vast majority of viral replication is concentrated within B cell follicles of secondary lymphoid tissues. We investigated whether infusion of T cells expressing an SIV-specific chimeric antigen receptor (CAR) and the follicular homing receptor, CXCR5, could successfully kill viral-RNA+ cells in targeted lymphoid follicles in SIV-infected rhesus macaques. In this study, CD4 and CD8 T cells from rhesus macaques were genetically modified to express antiviral CAR and CXCR5 moieties (generating CAR/CXCR5-T cells) and autologously infused into a chronically infected animal. At 2 days post-treatment, the CAR/CXCR5-T cells were located primarily in spleen and lymph nodes both inside and outside of lymphoid follicles. Few CAR/CXCR5-T cells were detected in the ileum, rectum, and lung, and no cells were detected in the bone marrow, liver, or brain. Within follicles, CAR/CXCR5-T cells were found in direct contact with SIV-viral RNA+ cells. We next infused CAR/CXCR5-T cells into ART-suppressed SIV-infected rhesus macaques, in which the animals were released from ART at the time of infusion. These CAR/CXCR5-T cells replicated in vivo within both the extrafollicular and follicular regions of lymph nodes and accumulated within lymphoid follicles. CAR/CXR5-T cell concentrations in follicles peaked during the first week post-infusion but declined to undetectable levels after 2 to 4 weeks. Overall, CAR/CXCR5-T cell-treated animals maintained lower viral loads and follicular viral RNA levels than untreated control animals, and no outstanding adverse reactions were noted. These findings indicate that CAR/CXCR5-T cell treatment is safe and holds promise as a future treatment for the durable remission of HIV.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Receptores CXCR5/imunologia , Receptores de Antígenos Quiméricos/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/terapia , Vírus da Imunodeficiência Símia/imunologia , Animais , Linfócitos B/imunologia , Centro Germinativo/imunologia , Humanos , Imunoterapia , Linfonodos/imunologia , Macaca mulatta , RNA Viral , Receptores CXCR5/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...