Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Front Immunol ; 12: 685718, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34234784

RESUMO

Antigen-specific B cells (ASBCs) can drive autoimmune disease by presenting autoantigen to cognate T cells to drive their activation, proliferation, and effector cell differentiation and/or by differentiating into autoantibody-secreting cells. Autoantibodies are frequently used to predict risk and diagnose several autoimmune diseases. ASBCs can drive type 1 diabetes even when immune tolerance mechanisms block their differentiation into antibody-secreting cells. Furthermore, anti-histidyl tRNA synthetase syndrome patients have expanded IgM+ Jo-1-binding B cells, which clinically diagnostic IgG Jo-1 autoantibodies may not fully reflect. Given the potential disconnect between the pathologic function of ASBCs and autoantibody secretion, direct study of ASBCs is a necessary step towards developing better therapies for autoimmune diseases, which often have no available cure. We therefore developed a high-throughput screening pipeline to 1) phenotypically identify specific B cell subsets, 2) expand them in vitro, 3) drive them to secrete BCRs as antibody, and 4) identify wells enriched for ASBCs through ELISA detection of antibody. We tested the capacity of several B cell subset(s) to differentiate into antibody-secreting cells following this robust stimulation. IgM+ and/or IgD+, CD27- memory, memory, switched memory, and BND B cells secreted B cell receptor (BCR) as antibody following in vitro stimulation, whereas few plasmablasts responded. Bimodal responses were observed across autoimmune donors for IgM+ CD21lo and IgM- CD21lo B cells, consistent with documented heterogeneity within the CD21lo subset. Using this approach, we detected insulin-binding B cell bias towards CD27- memory and CD27+ memory subsets in pre-symptomatic type 1 diabetes donors. We took advantage of routine detection of Jo-1-binding B cells in Jo-1+ anti-histidyl tRNA synthetase syndrome patients to show that Jo-1-binding B cells and total B cells expanded 20-30-fold using this culture system. Overall, these studies highlight technology that is amenable to small numbers of cryopreserved peripheral blood mononuclear cells that enables interrogation of phenotypic and repertoire attributes of ASBCs derived from autoimmune patients.


Assuntos
Doenças Autoimunes/imunologia , Subpopulações de Linfócitos B/fisiologia , Linfócitos B/imunologia , Imunoglobulina D/metabolismo , Imunoglobulina M/metabolismo , Adulto , Idoso , Autoanticorpos/imunologia , Autoantígenos/imunologia , Feminino , Humanos , Imunoglobulina D/genética , Imunoglobulina M/genética , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
2.
Science ; 373(6553)2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-34083450

RESUMO

The meninges contain adaptive immune cells that provide immunosurveillance of the central nervous system (CNS). These cells are thought to derive from the systemic circulation. Through single-cell analyses, confocal imaging, bone marrow chimeras, and parabiosis experiments, we show that meningeal B cells derive locally from the calvaria, which harbors a bone marrow niche for hematopoiesis. B cells reach the meninges from the calvaria through specialized vascular connections. This calvarial-meningeal path of B cell development may provide the CNS with a constant supply of B cells educated by CNS antigens. Conversely, we show that a subset of antigen-experienced B cells that populate the meninges in aging mice are blood-borne. These results identify a private source for meningeal B cells, which may help maintain immune privilege within the CNS.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B/fisiologia , Células da Medula Óssea/fisiologia , Sistema Nervoso Central/imunologia , Dura-Máter/citologia , Linfopoese , Meninges/citologia , Meninges/imunologia , Crânio/anatomia & histologia , Envelhecimento , Animais , Subpopulações de Linfócitos B/imunologia , Movimento Celular , Sistema Nervoso Central/fisiologia , Dura-Máter/imunologia , Fibroblastos/fisiologia , Homeostase , Privilégio Imunológico , Camundongos , Plasmócitos/fisiologia , Análise de Célula Única
3.
Front Immunol ; 11: 736, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32435242

RESUMO

The origin and function of blood IgM+IgD+CD27+ B cells is controversial, and they are considered a heterogeneous population. Previous staining of circulating B cells of healthy donors with rotavirus fluorescent virus-like particles allowed us to differentiate two subsets of IgM+IgD+CD27+: IgMhi and IgMlo B cells. Here, we confirmed this finding and compared the phenotype, transcriptome, in vitro function, and Ig gene repertoire of these two subsets. Eleven markers phenotypically discriminated both subsets (CD1c, CD69, IL21R, CD27, MTG, CD45RB, CD5, CD184, CD23, BAFFR, and CD38) with the IgMhi phenotypically resembling previously reported marginal zone B cells and the IgMlo resembling both naïve and memory B cells. Transcriptomic analysis showed that both subpopulations clustered close to germinal center-experienced IgM only B cells with a Principal Component Analysis, but differed in expression of 78 genes. Moreover, IgMhi B cells expressed genes characteristic of previously reported marginal zone B cells. After stimulation with CpG and cytokines, significantly (p < 0.05) higher frequencies (62.5%) of IgMhi B cells proliferated, compared with IgMlo B cells (35.37%), and differentiated to antibody secreting cells (14.22% for IgMhi and 7.19% for IgMlo). IgMhi B cells had significantly (p < 0.0007) higher frequencies of mutations in IGHV and IGKV regions, IgMlo B cells had higher usage of IGHJ6 genes (p < 0.0001), and both subsets differed in their HCDR3 properties. IgMhi B cells shared most of their shared IGH clonotypes with IgM only memory B cells, and IgMlo B cells with IgMhi B cells. These results support the notion that differential expression of IgM and IgD discriminates two subpopulations of human circulating IgM+IgD+CD27+ B cells, with the IgMhi B cells having similarities with previously described marginal zone B cells that passed through germinal centers, and the IgMlo B cells being the least differentiated amongst the IgM+CD27+ subsets.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B/imunologia , Centro Germinativo/imunologia , Imunoglobulina D/metabolismo , Imunoglobulina M/metabolismo , Adulto , Perfilação da Expressão Gênica , Humanos , Imunoglobulina D/genética , Cadeias Pesadas de Imunoglobulinas/genética , Imunoglobulina M/genética , Região Variável de Imunoglobulina/genética , Imunofenotipagem , Fenótipo , Análise de Componente Principal , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
4.
J Immunol ; 204(12): 3262-3272, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32332108

RESUMO

The self-renewal ability is a unique property of fetal-derived innate-like B-1a lymphocytes, which survive and function without being replenished by bone marrow (BM) progenitors. However, the mechanism by which IgM-secreting mature B-1a lymphocytes self-renew is poorly understood. In this study, we showed that Bmi1 was critically involved in this process. Although Bmi1 is considered essential for lymphopoiesis, the number of mature conventional B cells was not altered when Bmi1 was deleted in the B cell lineage. In contrast, the number of peritoneal B-1a cells was significantly reduced. Peritoneal cell transfer assays revealed diminished self-renewal ability of Bmi1-deleted B-1a cells, which was restored by additional deletion of Ink4-Arf, the well-known target of Bmi1 Fetal liver cells with B cell-specific Bmi1 deletion failed to repopulate peritoneal B-1a cells, but not other B-2 lymphocytes after transplantation assays, suggesting that Bmi1 may be involved in the developmental process of B-1 progenitors to mature B-1a cells. Although Bmi1 deletion has also been shown to alter the microenvironment for hematopoietic stem cells, fat-associated lymphoid clusters, the reported niche for B-1a cells, were not impaired in Bmi1 -/- mice. RNA expression profiling suggested lysine demethylase 5B (Kdm5b) as another possible target of Bmi1, which was elevated in Bmi1-/- B-1a cells in a stress setting and might repress B-1a cell proliferation. Our work has indicated that Bmi1 plays pivotal roles in self-renewal and maintenance of fetal-derived B-1a cells.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Subpopulações de Linfócitos B/fisiologia , Medula Óssea/metabolismo , Linhagem da Célula/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Microambiente Celular/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Linfócitos/metabolismo , Linfócitos/fisiologia , Linfopoese/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID
5.
J Immunol ; 204(10): 2852-2863, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32253241

RESUMO

BCR signaling, involving phosphorylation of various downstream molecules, including kinases, lipases, and linkers, is crucial for B cell selection, survival, proliferation, and differentiation. Phosphoflow cytometry (phosphoflow) is a single-cell-based technique to measure phosphorylated intracellular proteins, providing a more quantitative read-out than Western blotting. Recent advances in phosphoflow basically allow simultaneous analysis of protein phosphorylation in B cell (sub)populations, without prior cell sorting. However, fixation and permeabilization procedures required for phosphoflow often affect cell surface epitopes or mAb conjugates, precluding the evaluation of the phosphorylation status of signaling proteins across different B cell subpopulations present in a single sample. In this study, we report a versatile phosphoflow protocol allowing extensive staining of B cell subpopulations in human peripheral blood or various anatomical compartments in the mouse, starting from freshly isolated or frozen cell suspensions. Both human and mouse B cell subpopulations showed different basal and BCR stimulation-induced phosphorylation levels of downstream signaling proteins. For example, peritoneal B-1 cells and splenic marginal zone B cells exhibited significantly increased basal (ex vivo) signaling and increased responsiveness to in vitro BCR stimulation compared with peritoneal B-2 cells and splenic follicular B cells, respectively. In addition, whereas stimulation with anti-IgM or anti-Igκ L chain Abs resulted in strong pCD79a and pPLCγ2 signals, IgD stimulation only induced CD79a but not pPLCγ2 phosphorylation. In summary, the protocol is user friendly and quantifies BCR-mediated phosphorylation with high sensitivity at the single-cell level, in combination with extensive staining to identify individual B cell development and differentiation stages.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B/fisiologia , Citometria de Fluxo/métodos , Receptores de Antígenos de Linfócitos B/metabolismo , Animais , Antígenos CD79/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Imunoglobulina D/metabolismo , Imunoglobulina M/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipase C gama/metabolismo , Fosforilação , Transdução de Sinais , Análise de Célula Única
6.
Int J Hematol ; 111(5): 634-641, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31797231

RESUMO

Chronic lymphocytic leukemia (CLL), the most frequent type of leukemia in adults, is a lymphoproliferative disease characterized by the clonal expansion of mature CD5+ B cells in peripheral blood, bone marrow, and secondary lymphoid tissues. Over the past decade, substantial advances have been made in understanding the pathogenesis of CLL, including the identification of recurrent mutations, and clarification of clonal architectures, transcriptome analyses, and the multistep leukemogenic process. The biology of CLL is now better understood. The present review focuses on recent insights into CLL leukemogenesis, emphasizing the role of genetic lesions, and the multistep process initiating from very immature hematopoietic stem cells. Finally, we also review progress in the study of human B1 B cells, the putative normal counterparts of CLL cells.


Assuntos
Subpopulações de Linfócitos B/imunologia , Leucemia Linfocítica Crônica de Células B , Subpopulações de Linfócitos B/fisiologia , Antígenos CD5 , Células-Tronco Hematopoéticas , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/imunologia , Mutação , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética , Receptor Notch1/genética , Complexo Shelterina , Proteínas de Ligação a Telômeros/genética
7.
Int J Hematol ; 111(5): 628-633, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31721035

RESUMO

B1 cells, which are distinct from conventional B cells, are a rare B lymphocyte subpopulation that plays a pivotal role in innate immunity. Extensive previous studies have revealed the functions and ontogeny of murine B1 cells, but the properties of human B1 cells have just begun to be uncovered over the past decade. The phenotype of human B1 cells has recently been proposed, facilitating further studies. Here, we review the latest knowledge on human B1 cells, especially their ontogeny. A previous study using xenotransplantation models showed that human hematopoietic stem cells (HSCs) derived from cord blood or adult bone marrow can produce B1 cells in vivo. A recent study by our group reported that human B1 cells in peripheral blood are derived from adult HSCs and persist for approximately 3 years in situ. These findings suggest that adult human HSCs have the ability to produce B1 cells and contribute to maintenance of the adult B1 cell pool in peripheral blood. Further understanding of human B1 cell functions and ontogeny may elucidate the pathogenesis of B cell malignancies and autoimmune diseases.


Assuntos
Subpopulações de Linfócitos B/imunologia , Animais , Subpopulações de Linfócitos B/fisiologia , Células da Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Sangue Fetal , Genes abl , Transplante de Células-Tronco Hematopoéticas , Hemoglobinúria Paroxística/imunologia , Xenoenxertos , Humanos , Imunidade Inata , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Fenótipo
8.
Transpl Immunol ; 56: 101224, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31325493

RESUMO

BACKGROUND: Primary graft dysfunction (PGD) is the leading cause of early mortality after lung transplantation. Anti-collagen type-V (col(V)) immunity has been observed in animal models of ischemia-reperfusion injury (IRI) and in PGD. We hypothesized that collagen type-V is an innate danger signal contributing to PGD pathogenesis. METHODS: Anti-col(V) antibody production was detected by flow cytometric assay following cultures of murine CD19+ splenic cells with col.(V). Responding murine B cells were phenotyped using surface markers. RNA-Seq analysis was performed on murine CD19+ cells. Levels of anti-col(V) antibodies were measured in 188 recipients from the Lung Transplant Outcomes Group (LTOG) after transplantation. RESULTS: Col(V) induced rapid production of anti-col(V) antibodies from murine CD19+ B cells. Subtype analysis demonstrated innate B-1 B cells bound col.(V). Col(V) induced a specific transcriptional signature in CD19+ B cells with similarities to, yet distinct from, B cell receptor (BCR) stimulation. Rapid de novo production of anti-col(V) Abs was associated with an increased incidence of clinical PGD after lung transplant. CONCLUSIONS: This study demonstrated that col.(V) is an rapidly recognized by B cells and has specific transcriptional signature. In lung transplants recipients the rapid seroconversion to anti-col(V) Ab is linked to increased risk of grade 3 PGD.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Colágeno Tipo V/imunologia , Rejeição de Enxerto/imunologia , Transplante de Pulmão , Adulto , Idoso , Animais , Formação de Anticorpos , Antígenos CD19/metabolismo , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Imunidade Inata , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Transcriptoma
10.
Elife ; 82019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31090539

RESUMO

Although B cells expressing the IFNγR or the IFNγ-inducible transcription factor T-bet promote autoimmunity in Systemic Lupus Erythematosus (SLE)-prone mouse models, the role for IFNγ signaling in human antibody responses is unknown. We show that elevated levels of IFNγ in SLE patients correlate with expansion of the T-bet expressing IgDnegCD27negCD11c+CXCR5neg (DN2) pre-antibody secreting cell (pre-ASC) subset. We demonstrate that naïve B cells form T-bethi pre-ASCs following stimulation with either Th1 cells or with IFNγ, IL-2, anti-Ig and TLR7/8 ligand and that IL-21 dependent ASC formation is significantly enhanced by IFNγ or IFNγ-producing T cells. IFNγ promotes ASC development by synergizing with IL-2 and TLR7/8 ligands to induce genome-wide epigenetic reprogramming of B cells, which results in increased chromatin accessibility surrounding IRF4 and BLIMP1 binding motifs and epigenetic remodeling of IL21R and PRDM1 loci. Finally, we show that IFNγ signals poise B cells to differentiate by increasing their responsiveness to IL-21.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Diferenciação Celular , Epigênese Genética , Interferon gama/metabolismo , Interleucinas/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo , Subpopulações de Linfócitos B/química , Subpopulações de Linfócitos B/efeitos dos fármacos , Redes Reguladoras de Genes , Humanos , Lúpus Eritematoso Sistêmico/patologia , Proteínas com Domínio T/análise
12.
Nutrients ; 11(4)2019 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-31018604

RESUMO

In patients with non-IgE-mediated milk allergy, a cellular mechanism of delayed-type hypersensitivity (DTH) is considered. Recent findings prove that cell-mediated reactions can be antigen-specifically inhibited by extracellular vesicles (EVs) carrying miRNA-150. We sought to establish a new mouse model of DTH to casein and test the possibility of antigen-specific suppression of the inflammatory reaction. To produce soluble antigenic peptides, casein was subjected to alkaline hydrolysis. DTH reaction to casein was induced in CBA, C57BL/6, and BALB/c mice by intradermal (id) injection of the antigen. Cells collected from spleens and lymph nodes were positively or negatively selected and transferred to naive recipients intravenously (iv). CBA mice were tolerized by iv injection of mouse erythrocytes conjugated with casein antigen and following id immunization with the same antigen. Suppressive EVs were harvested from cell cultures and serum of tolerized donors by means of ultrafiltration and ultracentrifugation for further therapeutic utilization. The newly established mouse model of DTH to casein was mediated by CD4+ Th1 cells and macrophages, while EVs produced by casein-tolerized animals effectively suppressed effector cell response, in an miRNA-150-dependent manner. Altogether, our observations contribute to the current understanding of non-IgE-mediated allergy to casein and of the possibilities to downregulate this reaction.


Assuntos
Caseínas/imunologia , Vesículas Extracelulares/química , Hipersensibilidade Tardia , MicroRNAs/metabolismo , Transferência Adotiva , Animais , Antígenos/imunologia , Subpopulações de Linfócitos B/fisiologia , Linfócitos T CD4-Positivos , Regulação da Expressão Gênica/imunologia , Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA
13.
Eur J Immunol ; 49(8): 1213-1225, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31034584

RESUMO

Among the family of regulatory B cells, the subset able to produce interleukin-10 (IL-10) is the most studied, yet its biology is still a matter of investigation. The DNA methylation profiling of the il-10 gene locus revealed a novel epigenetic signature characterizing murine B cells ready to respond through IL-10 synthesis: a demethylated region located 4.5 kb from the transcription starting site (TSS), that we named early IL10 regulatory region (eIL10rr). This feature allows to distinguish B cells that are immediately prone and developmentally committed to IL-10 production from those that require a persistent stimulation to exert an IL-10-mediated regulatory function. These late IL-10 producers are instead characterized by a delayed IL10 regulatory region (dIL10rr), a partially demethylated DNA portion located 9 kb upstream from the TSS. A demethylated region was also found in human IL-10-producing B cells and, very interestingly, in some B-cell malignancies, such as chronic lymphocytic leukemia and mantle cell lymphoma, characterized by an immunosuppressive microenvironment. Our findings define murine and human regulatory B cells as an epigenetically controlled functional state of mature B cell subsets and open a new perspective on IL-10 regulation in B cells in homeostasis and disease.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B Reguladores/fisiologia , Interleucina-10/metabolismo , Leucemia Linfocítica Crônica de Células B/genética , Linfoma de Célula do Manto/genética , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Diferenciação Celular , Metilação de DNA , Feminino , Perfilação da Expressão Gênica , Humanos , Tolerância Imunológica , Imunidade Humoral , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Microambiente Tumoral
14.
Clin Lab Med ; 39(1): 15-29, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30709503

RESUMO

B cells shape the alloimmune response through polarized subsets. These cells inhibit or promote immune responses by expressing suppressive or proinflammatory cytokines. Their summed activity dictates the influence of B cells on the alloimmune response. We review the evidence for regulatory B cells and effector B cells in mice and humans, discuss current limitations in their phenotypic identification, and discuss regulatory B cells as a signature for clinical renal allograft tolerance and predictive markers for allograft outcomes. We discuss the effects of therapeutic agents on regulatory B cells and potential approaches to augment their numbers as a therapeutic tool.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B Reguladores/fisiologia , Tolerância ao Transplante , Animais , Subpopulações de Linfócitos B/metabolismo , Linfócitos B Reguladores/metabolismo , Biomarcadores/metabolismo , Humanos , Camundongos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
15.
Front Immunol ; 9: 1996, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30271400

RESUMO

Chronic lymphocytic leukemia (CLL) can be divided into prognostically distinct subsets with stereotyped or non-stereotyped, mutated or unmutated B cell receptors (BCRs). Individual subsets vary in antigen specificity and origin, but the impact of antigenic pressure on the CLL BCR repertoire remains unknown. Here, we employed IgH.TEµ mice that spontaneously develop CLL, expressing mostly unmutated BCRs of which ~35% harbor VH11-2/Vκ14-126 and recognize phosphatidylcholine. Proportions of VH11/Vκ14-expressing CLL were increased in the absence of functional germinal centers in IgH.TEµ mice deficient for CD40L or activation-induced cytidine deaminase. Conversely, in vivo T cell-dependent immunization decreased the proportions of VH11/Vκ14-expressing CLL. Furthermore, CLL onset was accelerated by enhanced BCR signaling in Siglec-G-/- mice or in mice expressing constitutively active Bruton's tyrosine kinase. Transcriptional profiling revealed that VH11 and non-VH11 CLL differed in the upregulation of specific pathways implicated in cell signaling and metabolism. Interestingly, principal component analyses using the 148 differentially expressed genes revealed that VH11 and non-VH11 CLL clustered with BCR-stimulated and anti-CD40-stimulated B cells, respectively. We identified an expression signature consisting of 13 genes that were differentially expressed in a larger panel of T cell-dependent non-VH11 CLL compared with T cell-independent VH11/Vκ14 or mutated IgH.TEµ CLL. Parallel differences in the expression of these 13 signature genes were observed between heterogeneous and stereotypic human unmutated CLL. Our findings provide evidence for two distinct unmutated CLL subsets with a specific transcriptional signature: one is T cell-independent and B-1 cell-derived while the other arises upon antigen stimulation in the context of T-cell help.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B/fisiologia , Centro Germinativo/imunologia , Leucemia Linfocítica Crônica de Células B/genética , Mutação/genética , Receptores de Antígenos de Linfócitos B/genética , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Lectinas/genética , Leucemia Linfocítica Crônica de Células B/diagnóstico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Prognóstico , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico , Transdução de Sinais/genética
16.
J Immunol ; 201(8): 2203-2208, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30201809

RESUMO

In systemic lupus erythematosus (SLE), type I IFNs promote induction of type I IFN-stimulated genes (ISG) and can drive B cells to produce autoantibodies. Little is known about the expression of distinct type I IFNs in lupus, particularly high-affinity IFN-ß. Single-cell analyses of transitional B cells isolated from SLE patients revealed distinct B cell subpopulations, including type I IFN producers, IFN responders, and mixed IFN producer/responder clusters. Anti-Ig plus TLR3 stimulation of SLE B cells induced release of bioactive type I IFNs that could stimulate HEK-Blue cells. Increased levels of IFN-ß were detected in circulating B cells from SLE patients compared with controls and were significantly higher in African American patients with renal disease and in patients with autoantibodies. Together, the results identify type I IFN-producing and -responding subpopulations within the SLE B cell compartment and suggest that some patients may benefit from specific targeting of IFN-ß.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B/fisiologia , Negro ou Afro-Americano , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Insuficiência Renal Crônica/imunologia , Autoanticorpos/sangue , Circulação Sanguínea , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Interferon Tipo I/genética , Espaço Intracelular , Lúpus Eritematoso Sistêmico/diagnóstico , Lúpus Eritematoso Sistêmico/epidemiologia , Insuficiência Renal Crônica/diagnóstico , Insuficiência Renal Crônica/epidemiologia , Análise de Célula Única , Transcriptoma , Estados Unidos/epidemiologia
17.
Nat Immunol ; 19(9): 1013-1024, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30104629

RESUMO

Most adult B cell lymphomas originate from germinal center (GC) B cells, but it is unclear to what extent B cells in overt lymphoma retain the functional dynamics of GC B cells or are blocked at a particular stage of the GC reaction. Here we used integrative single-cell analysis of phenotype, gene expression and variable-region sequence of the immunoglobulin heavy-chain locus to track the characteristic human GC B cell program in follicular lymphoma B cells. By modeling the cyclic continuum of GC B cell transitional states, we identified characteristic patterns of synchronously expressed gene clusters. GC-specific gene-expression synchrony was lost in single lymphoma B cells. However, distinct follicular lymphoma-specific cell states co-existed within single patient biopsies. Our data show that lymphoma B cells are not blocked in a GC B cell state but might adopt new dynamic modes of functional diversity, which opens the possibility of novel definitions of lymphoma identity.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos B/fisiologia , Centro Germinativo/fisiologia , Região Variável de Imunoglobulina/genética , Linfoma de Células B/genética , Adulto , Diferenciação Celular , Células Cultivadas , Feminino , Regulação Neoplásica da Expressão Gênica , Centro Germinativo/patologia , Humanos , Linfoma de Células B/patologia , Masculino , Pessoa de Meia-Idade , Análise de Célula Única , Transcriptoma/genética
18.
Mol Immunol ; 94: 166-175, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29324237

RESUMO

The study is aimed to investigate the role of MDM2 in the pathogenesis of lupus nephritis (LN) in pediatric SLE (pSLE). We confirmed that MDM2 expression was increased in peripheral blood mononuclear cells (PBMCs) as well as renal specimen of SLE compared with that of controls by western blot and immunofluorescence staining. Percentage of apoptotic and necrotic CD4+T, CD8+T and B cells were detected by flow cytometry respectively and levels of plasma cell free DNA (cfDNA) were quantified in SLE and healthy controls (HC). We also proved that elevated apoptotic and necrotic CD4+T cells were the main cause for increased plasma levels of cfDNA in pSLE. Additionally, upon DNA transfection MDM2 increased while P53 and P21 decreased in human renal mesangial cells (HRMC), with concomitant increase in proliferation rate and proportion of cells in S phase, as demonstrated by cell proliferation assay and cell cycle analysis. However, MDM2 inhibition reversed the trend. Furthermore, percentage of switched memory B cells decreased and proportion of double negative B cells increased upon blockage of MDM2 in PBMC. In summary, our study provided the first evidence that DNA induction of MDM2 promotes proliferation of HRMC and alters peripheral B cells subsets in pSLE. Thus our study has not only elucidated the pathogenesis of MDM2 in pediatric LN but also provided a novel target for drug development. In conclusion, our data suggested that apoptosis, cfDNA and MDM2 could form a pathological axis in SLE, especially in pSLE.


Assuntos
Subpopulações de Linfócitos B/patologia , Proliferação de Células/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Células Mesangiais/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Subpopulações de Linfócitos B/fisiologia , Linfócitos B/patologia , Linfócitos B/fisiologia , Estudos de Casos e Controles , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Criança , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Lúpus Eritematoso Sistêmico/patologia , Contagem de Linfócitos , Células Mesangiais/patologia
20.
Microbes Infect ; 20(3): 196-204, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29203270

RESUMO

Microsporidia are intracellular pathogens that cause severe disease in immunocompromised humans and animals. We recently demonstrated that XID mice are more susceptible to Encephalitozoon cuniculi infection by intraperitoneal route, evidencing the role of B-1 cells in resistance against infection. The present study investigated the resistance and susceptibility against E. cuniculi oral infection, including the role of B-1 cells. BALB/c and BALB/c XID (B-1 cells deficient) mice were orally infected with E. cuniculi spores. No clinical symptoms were observed in infected animals; histopathology showed lymphoplasmocytic enteritis with degeneration of the apexes of the villi in all infected groups. Higher parasite burden was observed in infected BALB/c XID mice. In the spleen and peritoneum, all infected mice showed a decrease of lymphocytes, including CD8+ T cells, mostly in infected BALB/c XID mice. Adoptive transfer of B-1 cells (XID + B-1) was associated with a lower parasite burden. Pro-inflammatory cytokines (IFN-γ, TNF-α and IL-6) increased mostly in infected XID + B1 mice. Together, the present results showed that BALB/c XID mice infected by the oral route were more susceptible to encephalitozoonosis than BALB/c mice, demonstrating the B-1 cells importance in the control of the immune response against oral E. cuniculi infection.


Assuntos
Subpopulações de Linfócitos B/fisiologia , Linfócitos T CD8-Positivos/imunologia , Citocinas/sangue , Encephalitozoon cuniculi/fisiologia , Encefalitozoonose/imunologia , Regulação para Cima/imunologia , Transferência Adotiva , Animais , Subpopulações de Linfócitos B/imunologia , Citocinas/imunologia , Encefalitozoonose/microbiologia , Encefalitozoonose/patologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Baço/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/imunologia , Doenças por Imunodeficiência Combinada Ligada ao Cromossomo X/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...