Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.469
Filtrar
1.
Nat Commun ; 15(1): 7982, 2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39266537

RESUMO

Precise regulation of B cell differentiation is essential for an effective adaptive immune response. Here, we show that B cell development in mice with B cell-specific Maf deletion is unaffected, but marginal zone B cells, germinal centre B cells, and plasmablasts are significantly more frequent in the spleen of naive Maf-deficient mice compared to wild type controls. In the context of a T cell-dependent immunization, Maf deletion causes increased proliferation of germinal centre B cells and extrafollicular plasmablasts. This is accompanied by higher production of antigen-specific IgG1 antibodies with minimal modification of early memory B cells, but a reduction in plasma cell numbers. Single-cell RNA sequencing shows upregulation of genes associated with DNA replication and cell cycle progression, confirming the role of Maf in cell proliferation. Subsequent pathway analysis reveals that Maf influences cellular metabolism, transporter activity, and mitochondrial proteins, which have been implicated in controlling the germinal centre reaction. In summary, our findings demonstrate that Maf acts intrinsically in B cells as a negative regulator of late B cell differentiation, plasmablast proliferation and germinal centre B cell formation.


Assuntos
Linfócitos B , Diferenciação Celular , Proliferação de Células , Centro Germinativo , Plasmócitos , Proteínas Proto-Oncogênicas c-maf , Animais , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Centro Germinativo/citologia , Camundongos , Plasmócitos/imunologia , Plasmócitos/metabolismo , Plasmócitos/citologia , Diferenciação Celular/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proteínas Proto-Oncogênicas c-maf/metabolismo , Proteínas Proto-Oncogênicas c-maf/genética , Camundongos Knockout , Camundongos Endogâmicos C57BL , Baço/citologia , Baço/metabolismo , Baço/imunologia , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Feminino
2.
Transpl Int ; 37: 13196, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39228658

RESUMO

Following transplantation, human CD4+T cells can respond to alloantigen using three distinct pathways. Direct and semi-direct responses are considered potent, but brief, so contribute mostly to acute rejection. Indirect responses are persistent and prolonged, involve B cells as critical antigen presenting cells, and are an absolute requirement for development of donor specific antibody, so more often mediate chronic rejection. Novel in vitro techniques have furthered our understanding by mimicking in vivo germinal centre processes, including B cell antigen presentation to CD4+ T cells and effector cytokine responses following challenge with donor specific peptides. In this review we outline recent data detailing the contribution of CD4+ T follicular helper cells and antigen presenting B cells to donor specific antibody formation and antibody mediated rejection. Furthermore, multi-parametric flow cytometry analyses have revealed specific endogenous regulatory T and B subsets each capable of suppressing distinct aspects of the indirect response, including CD4+ T cell cytokine production, B cell maturation into plasmablasts and antibody production, and germinal centre maturation. These data underpin novel opportunities to control these aberrant processes either by targeting molecules critical to indirect alloresponses or potentiating suppression via exogenous regulatory cell therapy.


Assuntos
Linfócitos B , Rejeição de Enxerto , Isoanticorpos , Humanos , Rejeição de Enxerto/imunologia , Isoanticorpos/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Centro Germinativo/imunologia , Doença Crônica , Doadores de Tecidos , Isoantígenos/imunologia , Citocinas/metabolismo , Linfócitos T Reguladores/imunologia
3.
J Exp Med ; 221(9)2024 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-39093312

RESUMO

Adaptive immune cell function is regulated by a highly diverse receptor recombined from variable germline-encoded segments that can recognize an almost unlimited array of epitopes. While this diversity enables the recognition of any pathogen, it also poses a risk of self-recognition, leading to autoimmunity. Many layers of regulation are present during both the generation and activation of B cells to prevent this phenomenon, although they are evidently imperfect. In recent years, our ability to analyze immune repertoires at scale has drastically increased, both through advances in sequencing and single-cell analyses. Here, we review the current knowledge on B cell repertoire analyses, focusing on their implication for autoimmunity. These studies demonstrate that a failure of tolerance occurs at multiple independent checkpoints in different autoimmune contexts, particularly during B cell maturation, plasmablast differentiation, and within germinal centers. These failures are marked by distinct repertoire features that may be used to identify disease- or patient-specific therapeutic approaches.


Assuntos
Autoimunidade , Linfócitos B , Tolerância Imunológica , Humanos , Autoimunidade/imunologia , Linfócitos B/imunologia , Animais , Tolerância Imunológica/imunologia , Centro Germinativo/imunologia , Diferenciação Celular/imunologia
4.
Nat Immunol ; 25(9): 1704-1717, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39143398

RESUMO

The mammalian Brg1/Brm-associated factor (BAF) complexes are major regulators of nucleosomal remodeling that are commonly mutated in several cancers, including germinal center (GC)-derived B cell lymphomas. However, the specific roles of different BAF complexes in GC B cell biology are not well understood. Here we show that the AT-rich interaction domain 1a (Arid1a) containing canonical BAF (cBAF) complex is required for maintenance of GCs and high-affinity antibody responses. While Arid1a-deficient B cells undergo initial activation, they fail to sustain the GC program. Arid1a establishes permissive chromatin landscapes for B cell activation and is concomitantly required to suppress inflammatory gene programs. The inflammatory signatures instigated by Arid1a deficiency promoted the recruitment of neutrophils and inflammatory monocytes. Dampening of inflammatory cues through interleukin-1ß blockade or glucocorticoid receptor agonist partially rescued Arid1a-deficient GCs, highlighting a critical role for inflammation in impeding GCs. Our work reveals essential functions of Arid1a-dependent cBAF in promoting efficient GC responses.


Assuntos
Linfócitos B , Proteínas de Ligação a DNA , Centro Germinativo , Inflamação , Camundongos Knockout , Fatores de Transcrição , Animais , Centro Germinativo/imunologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Camundongos Endogâmicos C57BL , Ativação Linfocitária/imunologia , Interleucina-1beta/metabolismo , Cromatina/metabolismo
5.
Immunity ; 57(9): 2191-2201.e5, 2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39168129

RESUMO

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.


Assuntos
Anticorpos Antivirais , Células Produtoras de Anticorpos , Vacinas contra COVID-19 , COVID-19 , Células B de Memória , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , SARS-CoV-2/imunologia , Células B de Memória/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , Anticorpos Antivirais/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Células Produtoras de Anticorpos/imunologia , Vacinas contra COVID-19/imunologia , Vacinação , Afinidade de Anticorpos/imunologia , Diferenciação Celular/imunologia , Centro Germinativo/imunologia , Memória Imunológica/imunologia , Anticorpos Monoclonais/imunologia , Adulto , Feminino
6.
Trends Immunol ; 45(9): 693-704, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39168721

RESUMO

When mature B cells are activated by antigens, the selection of these activated B cells takes place particularly during T cell-dependent immune responses in which an improved antibody repertoire is generated through somatic hypermutation in germinal centers (GCs). In this process the importance of antigen presentation by GC B cells, and subsequent T follicular helper (Tfh) cell help in positive selection of GC B cells, has been well appreciated. By contrast, the role of B cell receptor (BCR) signaling per se remains unclear. Strong experimental support for the involvement of BCR signaling in GC B cell selection has now been provided. Interestingly, these studies suggest that several checkpoints operating through the BCR ensure affinity maturation.


Assuntos
Linfócitos B , Centro Germinativo , Receptores de Antígenos de Linfócitos B , Transdução de Sinais , Centro Germinativo/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Humanos , Transdução de Sinais/imunologia , Linfócitos B/imunologia , Seleção Clonal Mediada por Antígeno , Ativação Linfocitária/imunologia
7.
Sci Rep ; 14(1): 17767, 2024 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-39090233

RESUMO

The germinal centers (GCs) are structure found within secondary lymphoid organs and are important for the antibody-producing response against foreign antigens. In GCs, antigen-specific B cells proliferate intensely, inducing immunoglobulin class switching. Recent studies have shown that GCs are also an important site for class switching to IgE, which is implicated in allergy. However, the mechanisms by which IgE production is regulated in GCs remain unclear. Here, we found impairment in IgE-specific production and a reduction of GC B cells after immunization in mice deficient in the Aps/Sh2b2 gene encoding the Lnk/Sh2b family adaptor protein Aps. GC B cells express higher levels of the Aps gene than non-GC B cells, and cell death of Aps-/- GC B cells is enhanced compared to wild-type GC B cells. An in vitro culture system with purified Aps-/- B cells induced the same level of IgE production and frequencies of IgE+ B cells as wild-type B cells. We found that Aps deficiency in B cells resulted in augmented depletion of IgE+ blasts by B cell receptor crosslinking with anti-CD79b antibodies compared to wild-type IgE+ cells. These results suggest that Aps regulates IgE production by controlling the survival of GC B cells and IgE+ plasma cells and may serve as a potential therapeutic target to control IgE production.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Linfócitos B , Sobrevivência Celular , Centro Germinativo , Imunoglobulina E , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linfócitos B/imunologia , Linfócitos B/metabolismo , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Domínios de Homologia de src
8.
Artigo em Chinês | MEDLINE | ID: mdl-39118513

RESUMO

Objective:To investigate the clinical features, imaging findings, pathological phenotype, treatment and prognosis of unicentric Castleman disease in the children's neck, in order to improve the understanding of CD among Otolaryngology Head and Neck Surgery. Methods:Retrospective cross-sectional, observational study was undertaken in Kunming Children's Hospital, from the archival data between January July 2015 and June 2020. Only 6 cases of CD were identified after studying the histomorphological characteristics and neck mass diagnosed. The imaging and pathological features were summarized and the pathogenesis was discussed. Results:Among the 6 cases of Castleman disease, five were male and one was female. Histopathology: Five cases were hyaline vascular subtype, one was mixed type. The uniform clinicopathologic features seen in all hyaline vascular subtype of CD included atrophic germinal centre with lymphocyte depletion, concentric rings of small lymphocytes, increased vascularity and predominance of high endothelial vessels in interfollicular region. Twinning, in which two or more germinal centers are combined and surrounded by lymphocytes in the mantle zone was observed in two cases with lollipop pattern at the same time. All the cases underwent complete surgical resection, the median follow-up time was 48 months(26, 84), both of them had good prognosis. Conclusion:Most cases of unicentric type CD in children are diagnosed late, which is clinical showed by painless lymphadenopathy. The most common pathological type is hyaline vascular. The overall prognosis of surgical treatment was good.


Assuntos
Hiperplasia do Linfonodo Gigante , Pescoço , Humanos , Hiperplasia do Linfonodo Gigante/patologia , Masculino , Feminino , Criança , Estudos Retrospectivos , Estudos Transversais , Prognóstico , Adolescente , Centro Germinativo/patologia , Pré-Escolar
9.
J Med Virol ; 96(8): e29851, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39132689

RESUMO

Here, we performed single-cell RNA sequencing of S1 and receptor binding domain protein-specific B cells from convalescent COVID-19 patients with different clinical manifestations. This study aimed to evaluate the role and developmental pathway of atypical memory B cells (MBCs) in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The results revealed a proinflammatory signature across B cell subsets associated with disease severity, as evidenced by the upregulation of genes such as GADD45B, MAP3K8, and NFKBIA in critical and severe individuals. Furthermore, the analysis of atypical MBCs suggested a developmental pathway similar to that of conventional MBCs through germinal centers, as indicated by the expression of several genes involved in germinal center processes, including CXCR4, CXCR5, BCL2, and MYC. Additionally, the upregulation of genes characteristic of the immune response in COVID-19, such as ZFP36 and DUSP1, suggested that the differentiation and activation of atypical MBCs may be influenced by exposure to SARS-CoV-2 and that these genes may contribute to the immune response for COVID-19 recovery. Our study contributes to a better understanding of atypical MBCs in COVID-19 and the role of other B cell subsets across different clinical manifestations.


Assuntos
COVID-19 , Células B de Memória , SARS-CoV-2 , Análise de Célula Única , Humanos , COVID-19/imunologia , COVID-19/virologia , COVID-19/genética , SARS-CoV-2/imunologia , SARS-CoV-2/genética , Células B de Memória/imunologia , Masculino , Adulto , Feminino , Pessoa de Meia-Idade , Perfilação da Expressão Gênica , Transcriptoma , Centro Germinativo/imunologia , Linfócitos B/imunologia , Idoso
10.
Front Immunol ; 15: 1396808, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39136032

RESUMO

Introduction: Tertiary lymphoid structures (TLSs) are analogues of secondary lymphoid organs that contain various immune cells. The spatial distribution, maturation and composition of TLSs have differential effects on prognosis, and the roles of TLSs in gastric adenocarcinoma (GA) have not been revealed. Methods: Thus, we evaluated the prognostic value of TLSs in GA through analysis of bulk RNA sequencing(RNA-seq) data from public databases and validated our findings in tumour samples from the Fudan University Shanghai Cancer Center (FUSCC) cohort. The spatial distribution,maturation, and composition of TLSs in GA were analysed by reviewing H&E-stained sections and by multiplex immunofluorescence (mIF) staining. Results: We found that TLSs, especially TLSs with germinal centres (GCs) and TLSs located in the invasive margin (IM), were correlated with prolonged overall survival (OS). Second, analysis of public RNA-seq data showed that high dendritic cell (DC) scores were a favourable prognostic factor in GA patients with high scores for both TLSs and GCs. In the FUSCC cohort, DC-LAMP+ DCs weresignificantly enriched in IM-TLSs with GCs, suggesting a potential correlation between the tumour immune activation milieu and the DC abundance. Third, compared to that in TLSs without GCs, the proportion of FOXP3+CD8+ Treg cells was significantly decreased in IM-TLSs with GCs, and the percentage of PD1+CD20+ B cells was significantly increased in TLSs with GCs. Discussion: Our results demonstrate that the spatial arrangement and maturation of TLSs significantly affect prognosis and indicate that TLSs could be a new additional factor for histopathological evaluation.


Assuntos
Adenocarcinoma , Neoplasias Gástricas , Estruturas Linfoides Terciárias , Humanos , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/genética , Estruturas Linfoides Terciárias/imunologia , Estruturas Linfoides Terciárias/patologia , Adenocarcinoma/imunologia , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Prognóstico , Masculino , Feminino , Pessoa de Meia-Idade , Células Dendríticas/imunologia , Idoso , Centro Germinativo/imunologia , Centro Germinativo/patologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Microambiente Tumoral/imunologia
11.
Front Immunol ; 15: 1427509, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39188715

RESUMO

The successful development of germinal centers (GC) relies heavily on innate mechanisms to amplify the initial inflammatory cascade. In addition to their role in antigen presentation, innate cells are essential for the redirection of circulating lymphocytes toward the draining lymph node (dLN) to maximize antigen surveillance. Sphingosine-1-Phosphate (S1P) and its receptors (S1PR1-5) affect various aspects of immunity; however, the role of S1PR4 in regulating an immune response is not well understood. Here we use a footpad model of localized TH1 inflammation to carefully monitor changes in leukocyte populations within the blood, the immunized tissue, and the dLN. Within hours of immunization, neutrophils failed to adequately mobilize and infiltrate into the footpad tissue of S1PR4-/- mice, thereby diminishing the local vascular changes thought to be necessary for redirecting circulating cells toward the inflamed region. Neutrophil depletion with anti-Ly6G antibodies significantly reduced early tissue edema as well as the redirection and initial accumulation of naïve lymphocytes in dLN of WT mice, while the effects were less prominent or absent in S1PR4-/- dLN. Adoptive transfer experiments further demonstrated that the lymphocyte homing deficiencies in vivo were not intrinsic to the donor S1PR4-/- lymphocytes, but were instead attributed to differences within the S1PR4-deficient host. Reduced cell recruitment in S1PR4-/- mice would seed the dLN with fewer antigen-respondent lymphocytes and indeed, dLN hypertrophy at the peak of the immune response was severely diminished, with attenuated GC and activation pathways in these mice. Histological examination of the S1PR4-/- dLN also revealed an underdeveloped vascular network with reduced expression of the leukocyte tethering ligand, PNAd, within high endothelial venule regions, suggesting inadequate growth of the dLN meant to support a robust GC response. Thus, our study reveals that S1PR4 may link early immune modulation by neutrophils to the initial recruitment of circulating lymphocytes and downstream expansion and maturation of the dLN, thereby contributing to optimal GC development during an adaptive response.


Assuntos
Centro Germinativo , Inflamação , Linfonodos , Camundongos Knockout , Neutrófilos , Receptores de Esfingosina-1-Fosfato , Animais , Centro Germinativo/imunologia , Neutrófilos/imunologia , Camundongos , Linfonodos/imunologia , Receptores de Esfingosina-1-Fosfato/genética , Receptores de Esfingosina-1-Fosfato/metabolismo , Inflamação/imunologia , Camundongos Endogâmicos C57BL , Linfócitos/imunologia , Células Th1/imunologia
12.
Nat Commun ; 15(1): 7195, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39179580

RESUMO

The myocyte enhancer factor 2B (MEF2B) transcription factor is frequently mutated in germinal center (GC)-derived B-cell lymphomas. Its ammino (N)-terminal mutations drive lymphomagenesis by escaping interaction with transcriptional repressors, while the function of carboxy (C)-terminal mutations remains to be elucidated. Here, we show that MEF2B C-tail is physiologically phosphorylated at specific residues and phosphorylation at serine (S)324 is impaired by lymphoma-associated mutations. Lack of phosphorylation at S324 enhances the interaction of MEF2B with the SWI/SNF chromatin remodeling complex, leading to higher transcriptional activity. In addition, these mutants show an increased protein stability due to impaired interaction with the CUL3/KLHL12 ubiquitin complex. Mice expressing a phosphorylation-deficient lymphoma-associated MEF2B mutant display GC enlargement and develop GC-derived lymphomas, when crossed with Bcl2 transgenic mice. These results unveil converging mechanisms of action for a diverse spectrum of MEF2B mutations, all leading to its dysregulation and GC B-cell lymphomagenesis.


Assuntos
Linfoma de Células B , Fatores de Transcrição MEF2 , Mutação , Animais , Humanos , Camundongos , Linfócitos B/metabolismo , Linhagem Celular Tumoral , Centro Germinativo/metabolismo , Células HEK293 , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/genética , Camundongos Transgênicos , Fosforilação , Estabilidade Proteica , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética
13.
Sci Immunol ; 9(98): eadd4874, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39121196

RESUMO

Dedicator of cytokinesis 8 (DOCK8) immunodeficiency syndrome is characterized by a failure of the germinal center response, a process involving the proliferation and positive selection of antigen-specific B cells. Here, we describe how DOCK8-deficient B cells are blocked at a light-zone checkpoint in the germinal centers of immunized mice, where they are unable to respond to T cell-dependent survival and selection signals and consequently differentiate into plasma cells or memory B cells. Although DOCK8-deficient B cells can acquire and present antigen to initiate activation of cognate T cells, integrin up-regulation, B cell-T cell conjugate formation, and costimulation are insufficient for sustained B cell and T cell activation when antigen availability is limited. Our findings provide an explanation for the failure of the humoral response in DOCK8 immunodeficiency syndrome and insight into how the level of available antigen modulates B cell-T cell cross-talk to fine-tune humoral immune responses and immunological memory.


Assuntos
Linfócitos B , Fatores de Troca do Nucleotídeo Guanina , Camundongos Endogâmicos C57BL , Linfócitos T , Animais , Fatores de Troca do Nucleotídeo Guanina/imunologia , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Linfócitos B/imunologia , Camundongos , Linfócitos T/imunologia , Ativação Linfocitária/imunologia , Camundongos Knockout , Antígenos/imunologia , Centro Germinativo/imunologia
14.
Nat Immunol ; 25(9): 1742-1753, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39164477

RESUMO

The differentiation and specificity of human CD4+ T follicular helper cells (TFH cells) after influenza vaccination have been poorly defined. Here we profiled blood and draining lymph node (LN) samples from human volunteers for over 2 years after two influenza vaccines were administered 1 year apart to define the evolution of the CD4+ TFH cell response. The first vaccination induced an increase in the frequency of circulating TFH (cTFH) and LN TFH cells at week 1 postvaccination. This increase was transient for cTFH cells, whereas the LN TFH cells further expanded during week 2 and remained elevated in frequency for at least 3 months. We observed several distinct subsets of TFH cells in the LN, including pre-TFH cells, memory TFH cells, germinal center (GC) TFH cells and interleukin-10+ TFH cell subsets beginning at baseline and at all time points postvaccination. The shift toward a GC TFH cell phenotype occurred with faster kinetics after the second vaccine compared to the first vaccine. We identified several influenza-specific TFH cell clonal lineages, including multiple responses targeting internal influenza virus proteins, and found that each TFH cell state was attainable within a clonal lineage. Thus, human TFH cells form a durable and dynamic multitissue network.


Assuntos
Diferenciação Celular , Centro Germinativo , Vacinas contra Influenza , Influenza Humana , Células T Auxiliares Foliculares , Vacinação , Humanos , Vacinas contra Influenza/imunologia , Células T Auxiliares Foliculares/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Centro Germinativo/imunologia , Diferenciação Celular/imunologia , Linfonodos/imunologia , Adulto , Feminino , Masculino , Pessoa de Meia-Idade , Interleucina-10/imunologia , Interleucina-10/metabolismo , Memória Imunológica/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Adulto Jovem
15.
Nat Commun ; 15(1): 6971, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-39138218

RESUMO

Ligation of the B cell antigen receptor (BCR) initiates humoral immunity. However, BCR signaling without appropriate co-stimulation commits B cells to death rather than to differentiation into immune effector cells. How BCR activation depletes potentially autoreactive B cells while simultaneously primes for receiving rescue and differentiation signals from cognate T lymphocytes remains unknown. Here, we use a mass spectrometry-based proteomic approach to identify cytosolic/nuclear shuttling elements and uncover transcription factor EB (TFEB) as a central BCR-controlled rheostat that drives activation-induced apoptosis, and concurrently promotes the reception of co-stimulatory rescue signals by supporting B cell migration and antigen presentation. CD40 co-stimulation prevents TFEB-driven cell death, while enhancing and prolonging TFEB's nuclear residency, which hallmarks antigenic experience also of memory B cells. In mice, TFEB shapes the transcriptional landscape of germinal center B cells. Within the germinal center, TFEB facilitates the dark zone entry of light-zone-residing centrocytes through regulation of chemokine receptors and, by balancing the expression of Bcl-2/BH3-only family members, integrates antigen-induced apoptosis with T cell-provided CD40 survival signals. Thus, TFEB reprograms antigen-primed germinal center B cells for cell fate decisions.


Assuntos
Apoptose , Linfócitos B , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Antígenos CD40 , Centro Germinativo , Receptores de Antígenos de Linfócitos B , Animais , Centro Germinativo/imunologia , Centro Germinativo/citologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Camundongos , Antígenos CD40/metabolismo , Antígenos CD40/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Antígenos de Linfócitos B/imunologia , Camundongos Endogâmicos C57BL , Ativação Linfocitária/imunologia , Diferenciação Celular/imunologia , Transdução de Sinais , Apresentação de Antígeno/imunologia
16.
Hum Immunol ; 85(5): 110838, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38970880

RESUMO

BACKGROUND: T follicular helper (Tfh) cells are a subdivision of T helper cells involved in antigen-specific B cell immunity. Tfh cells play an essential role in the interaction of T cells/B cells in the germinal centers (GC), and dysregulation of Tfh actions can offer pathogenic autoantibody formation and lead to the development of autoimmune diseases. This study seeks to evaluate changes in Tfh frequency and its related cytokines in autoimmune disease, its association with disease phase, severity, prognosis, and the effect of immunosuppressive treatment on the Tfh population. METHOD: The study adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 Statement. Electronic databases, including PubMed, Scopus, Web of Science, and Embase, were systematically searched for potentially eligible studies up to January 1, 2024. RESULTS: We identified 4998 articles in the initial search, from which 1686 similar titles were removed. A total of 3312 articles were initially screened, and 3051 articles were excluded by title/abstract screening. A total of 261 studies were considered for full-text assessment, and 205 articles were excluded by reason. Finally, a total of 56 studies were included in our review. CONCLUSION: The population of Tfh cells is generally higher in autoimmune diseases versus Health control. Moreover, the number of Tfh cells is associated with the disease severity and can be considered for determining the prognosis of studies. Also, peripheral blood circulating Tfh (cTfh) cells are an available sample that can be used as an indicator for diagnosing diseases.


Assuntos
Doenças Autoimunes , Doenças Reumáticas , Células T Auxiliares Foliculares , Humanos , Células T Auxiliares Foliculares/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/diagnóstico , Doenças Reumáticas/imunologia , Doenças Reumáticas/diagnóstico , Doenças Reumáticas/tratamento farmacológico , Centro Germinativo/imunologia , Imunossupressores/uso terapêutico , Linfócitos B/imunologia , Prognóstico , Citocinas/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia
17.
Immunity ; 57(7): 1454-1456, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38986440

RESUMO

The association of tertiary lymphoid structures (TLSs) with survival and immunotherapy response brought B cells to center stage. In a pan-cancer B cells atlas in Science, Ma et al. show that germinal center reaction generating anti-tumor antibody-secreting cells (ASCs) from B memory cells in mature TLSs co-exist in tumors with extra-follicular reaction generating auto-reactive ASCs from memory B cells in immature TLSs.


Assuntos
Linfócitos B , Centro Germinativo , Neoplasias , Humanos , Linfócitos B/imunologia , Centro Germinativo/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Animais , Estruturas Linfoides Terciárias/imunologia , Células B de Memória/imunologia , Imunoterapia/métodos
18.
Trends Immunol ; 45(8): 574-576, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39060142

RESUMO

The immune and sensory nervous systems communicate to maintain homeostasis. Wu et al. recently demonstrated that sensory neurons innervate the mouse spleen. These neurons promote calcitonin gene-related peptide (CGRP)-dependent responses in splenic B cell germinal centers (GCs) and antigen-specific antibody production. Dietary capsaicin activates these neurons to enhance humoral immunity against influenza virus infection.


Assuntos
Linfócitos B , Centro Germinativo , Imunidade Humoral , Baço , Animais , Baço/imunologia , Baço/inervação , Humanos , Centro Germinativo/imunologia , Camundongos , Linfócitos B/imunologia , Células Receptoras Sensoriais/imunologia , Células Receptoras Sensoriais/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/imunologia , Capsaicina/farmacologia
19.
Sci Rep ; 14(1): 16970, 2024 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-39043800

RESUMO

B cells and the antibodies they produce are critical in host defense against pathogens and contribute to various immune-mediated diseases. B cells responding to activating signals in vitro release extracellular vesicles (EV) that carry surface antibodies, yet B cell production of EVs that express antibodies and their function in vivo is incompletely understood. Using transgenic mice expressing the Cre recombinase in B cells switching to IgG1 to induce expression of fusion proteins between emerald green fluorescent protein (emGFP) and the EV tetraspanin CD63 as a model, we identify emGFP expression in B cells responding to foreign antigen in vivo and characterize the emGFP+ EVs they release. Our data suggests that emGFP+ germinal center B cells undergoing immunoglobulin class switching to express IgG and their progeny memory B cells and plasma cells, also emGFP+, are sources of circulating antigen-specific IgG+ EVs. Furthermore, using a mouse model of influenza virus infection, we find that IgG+ EVs specific for the influenza hemagglutinin antigen protect against virus infection. In addition, crossing the B cell Cre driver EV reporter mice onto the Nba2 lupus-prone strain revealed increased circulating emGFP+ EVs that expressed surface IgG against nuclear antigens linked to autoimmunity. These data identify EVs loaded with antibodies as a novel route for antibody secretion in B cells that contribute to adaptive immune responses, with important implications for different functions of IgG+ EVs in infection and autoimmunity.


Assuntos
Linfócitos B , Vesículas Extracelulares , Imunoglobulina G , Camundongos Transgênicos , Animais , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Camundongos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Infecções por Orthomyxoviridae/imunologia , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Fluorescência Verde/genética , Antígenos/imunologia , Switching de Imunoglobulina , Camundongos Endogâmicos C57BL , Centro Germinativo/imunologia , Centro Germinativo/metabolismo
20.
Nat Commun ; 15(1): 6421, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39080316

RESUMO

The rodent-borne Andes virus (ANDV) causes a severe disease in humans. We developed an ANDV mRNA vaccine based on the M segment of the viral genome, either with regular uridine (U-mRNA) or N1-methylpseudouridine (m1Ψ-mRNA). Female mice immunized by m1Ψ-mRNA developed slightly greater germinal center (GC) responses than U-mRNA-immunized mice. Single cell RNA and BCR sequencing of the GC B cells revealed similar levels of activation, except an additional cluster of cells exhibiting interferon response in animals vaccinated with U-mRNA but not m1Ψ-mRNA. Similar immunoglobulin class-switching and somatic hypermutations were observed in response to the vaccines. Female Syrian hamsters were immunized via a prime-boost regimen with two doses of each vaccine. The titers of glycoprotein-binding antibodies were greater for U-mRNA construct than for m1Ψ-mRNA construct; however, the titers of ANDV-neutralizing antibodies were similar. Vaccinated animals were challenged with a lethal dose of ANDV, along with a naïve control group. All control animals and two animals vaccinated with a lower dose of m1Ψ-mRNA succumbed to infection whereas other vaccinated animals survived without evidence of virus replication. The data demonstrate the development of a protective vaccine against ANDV and the lack of a substantial effect of m1Ψ modification on immunogenicity and protection in rodents.


Assuntos
Mesocricetus , Uridina , Vacinas Virais , Animais , Feminino , Camundongos , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/imunologia , Anticorpos Antivirais/imunologia , Orthohantavírus/imunologia , Orthohantavírus/genética , Anticorpos Neutralizantes/imunologia , Centro Germinativo/imunologia , Pseudouridina/imunologia , Cricetinae , Vacinas de mRNA , Febre Hemorrágica Americana/prevenção & controle , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/virologia , RNA Viral/genética , RNA Viral/imunologia , Linfócitos B/imunologia , Humanos , Desenvolvimento de Vacinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA