Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Nat Immunol ; 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982286
2.
Nat Immunol ; 20(8): 1071-1082, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31263277

RESUMO

Systemic lupus erythematosus (SLE) is characterized by the expansion of extrafollicular pathogenic B cells derived from newly activated naive cells. Although these cells express distinct markers, their epigenetic architecture and how it contributes to SLE remain poorly understood. To address this, we determined the DNA methylomes, chromatin accessibility profiles and transcriptomes from five human B cell subsets, including a newly defined effector B cell subset, from subjects with SLE and healthy controls. Our data define a differentiation hierarchy for the subsets and elucidate the epigenetic and transcriptional differences between effector and memory B cells. Importantly, an SLE molecular signature was already established in resting naive cells and was dominated by enrichment of accessible chromatin in motifs for AP-1 and EGR transcription factors. Together, these factors acted in synergy with T-BET to shape the epigenome of expanded SLE effector B cell subsets. Thus, our data define the molecular foundation of pathogenic B cell dysfunction in SLE.


Assuntos
Subpopulações de Linfócitos B/patologia , Metilação de DNA/genética , Epigênese Genética/genética , Lúpus Eritematoso Sistêmico/genética , Subpopulações de Linfócitos B/imunologia , Montagem e Desmontagem da Cromatina/fisiologia , Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Fator de Transcrição AP-1/genética , Transcriptoma/genética
3.
Cell ; 166(1): 88-101, 2016 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-27293190

RESUMO

Antibodies to DNA and chromatin drive autoimmunity in systemic lupus erythematosus (SLE). Null mutations and hypomorphic variants of the secreted deoxyribonuclease DNASE1L3 are linked to familial and sporadic SLE, respectively. We report that DNASE1L3-deficient mice rapidly develop autoantibodies to DNA and chromatin, followed by an SLE-like disease. Circulating DNASE1L3 is produced by dendritic cells and macrophages, and its levels inversely correlate with anti-DNA antibody response. DNASE1L3 is uniquely capable of digesting chromatin in microparticles released from apoptotic cells. Accordingly, DNASE1L3-deficient mice and human patients have elevated DNA levels in plasma, particularly in circulating microparticles. Murine and human autoantibody clones and serum antibodies from human SLE patients bind to DNASE1L3-sensitive chromatin on the surface of microparticles. Thus, extracellular microparticle-associated chromatin is a potential self-antigen normally digested by circulating DNASE1L3. The loss of this tolerance mechanism can contribute to SLE, and its restoration may represent a therapeutic opportunity in the disease.


Assuntos
Autoanticorpos/imunologia , Micropartículas Derivadas de Células/química , Cromatina/imunologia , DNA/imunologia , Endodesoxirribonucleases/genética , Lúpus Eritematoso Sistêmico/imunologia , Animais , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Endodesoxirribonucleases/deficiência , Endodesoxirribonucleases/metabolismo , Humanos , Células Jurkat , Lúpus Eritematoso Sistêmico/enzimologia , Lúpus Eritematoso Sistêmico/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
Nat Immunol ; 16(7): 755-65, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26006014

RESUMO

Acute systemic lupus erythematosus (SLE) courses with surges of antibody-secreting cells (ASCs) whose origin, diversity and contribution to serum autoantibodies remain unknown. Here, deep sequencing, proteomic profiling of autoantibodies and single-cell analysis demonstrated highly diversified ASCs punctuated by clones expressing the variable heavy-chain region VH4-34 that produced dominant serum autoantibodies. A fraction of ASC clones contained autoantibodies without mutation, a finding consistent with differentiation outside the germinal centers. A substantial ASC segment was derived from a distinct subset of newly activated naive cells of considerable clonality that persisted in the circulation for several months. Thus, selection of SLE autoreactivities occurred during polyclonal activation, with prolonged recruitment of recently activated naive B cells. Our findings shed light on the pathogenesis of SLE, help explain the benefit of agents that target B cells and should facilitate the design of future therapies.


Assuntos
Diversidade de Anticorpos/imunologia , Células Produtoras de Anticorpos/imunologia , Autoanticorpos/imunologia , Proliferação de Células , Lúpus Eritematoso Sistêmico/imunologia , Doença Aguda , Sequência de Aminoácidos , Diversidade de Anticorpos/genética , Células Produtoras de Anticorpos/metabolismo , Autoanticorpos/genética , Autoanticorpos/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Sequência de Bases , Células Clonais/imunologia , Células Clonais/metabolismo , Citometria de Fluxo , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/metabolismo , Vacinas contra Influenza/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Dados de Sequência Molecular , Proteoma/análise , Proteoma/imunologia , Proteômica/métodos , Homologia de Sequência de Aminoácidos , Análise de Célula Única/métodos , Espectrometria de Massas em Tandem , Toxoide Tetânico/imunologia
5.
Immunity ; 43(1): 132-45, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26187412

RESUMO

Antibody responses to viral infections are sustained for decades by long-lived plasma cells (LLPCs). However, LLPCs have yet to be characterized in humans. Here we used CD19, CD38, and CD138 to identify four PC subsets in human bone marrow (BM). We found that the CD19(-)CD38(hi)CD138(+) subset was morphologically distinct, differentially expressed PC-associated genes, and exclusively contained PCs specific for viral antigens to which the subjects had not been exposed for more than 40 years. Protein sequences of measles- and mumps-specific circulating antibodies were encoded for by CD19(-)CD38(hi)CD138(+) PCs in the BM. Finally, we found that CD19(-)CD38(hi)CD138(+) PCs had a distinct RNA transcriptome signature and human immunoglobulin heavy chain (VH) repertoire that was relatively uncoupled from other BM PC subsets and probably represents the B cell response's "historical record" of antigenic exposure. Thus, our studies define human LLPCs and provide a mechanism for the life-long maintenance of anti-viral antibodies in the serum.


Assuntos
Anticorpos Antivirais/imunologia , Células da Medula Óssea/imunologia , Vírus do Sarampo/imunologia , Vírus da Caxumba/imunologia , Plasmócitos/imunologia , ADP-Ribosil Ciclase 1/metabolismo , Adulto , Idoso , Anticorpos Antivirais/sangue , Antígenos CD19/metabolismo , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , RNA Mensageiro/genética , Sindecana-1/metabolismo , Adulto Jovem
6.
Immunol Rev ; 292(1): 76-89, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31755562

RESUMO

The maintenance of immunological tolerance of B lymphocytes is a complex and critical process that must be implemented as to avoid the detrimental development of autoreactivity and possible autoimmunity. Murine models have been invaluable to elucidate many of the key components in B-cell tolerance; however, translation to human homeostatic and pathogenic immune states can be difficult to assess. Functional autoreactive, flow cytometric, and single-cell cloning assays have proven to be critical in deciphering breaks in B-cell tolerance within autoimmunity; however, newer approaches to assess human B-cell tolerance may prove to be vital in the further exploration of underlying tolerance defects. In this review, we supply a comprehensive overview of human immune tolerance checkpoints with associated mechanisms of enforcement, and highlight current and future methodologies which are likely to benefit future studies into the mechanisms that become defective in human autoimmune conditions.


Assuntos
Autoantígenos/imunologia , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Tolerância Imunológica/imunologia , Animais , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Ativação Linfocitária/imunologia
7.
Immunol Rev ; 284(1): 120-131, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29944759

RESUMO

Understanding antibody repertoires and in particular, the properties and fates of B cells expressing potentially pathogenic antibodies is critical to define the mechanisms underlying multiple immunological diseases including autoimmune and allergic conditions as well as transplant rejection. Moreover, an integrated knowledge of the antibody repertoires expressed by B cells and plasma cells (PC) of different functional properties and longevity is essential to develop new therapeutic strategies, better biomarkers for disease segmentation, and new assays to measure restoration of B-cell tolerance or, at least, of normal B-cell homeostasis. Reaching these goals, however, will require a more precise phenotypic, functional and molecular definition of B-cell and PC populations, and a comprehensive analysis of the antigenic reactivity of the antibodies they express. While traditionally hampered by technical and ethical limitations in human experimentation, new technological advances currently enable investigators to address these questions in a comprehensive fashion. In this review, we shall discuss these concepts as they apply to the study of Systemic Lupus Erythematosus.


Assuntos
Autoanticorpos/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Tolerância Imunológica/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária/imunologia , Humanos , Lúpus Eritematoso Sistêmico/patologia
8.
BMC Genomics ; 21(Suppl 9): 583, 2020 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-32900378

RESUMO

BACKGROUND: B cell affinity maturation enables B cells to generate high-affinity antibodies. This process involves somatic hypermutation of B cell immunoglobulin receptor (BCR) genes and selection by their ability to bind antigens. Lineage trees are used to describe this microevolution of B cell immunoglobulin genes. In a lineage tree, each node is one BCR sequence that mutated from the germinal center and each directed edge represents a single base mutation, insertion or deletion. In BCR sequencing data, the observed data only contains a subset of BCR sequences in this microevolution process. Therefore, reconstructing the lineage tree from experimental data requires algorithms to build the tree based on partially observed tree nodes. RESULTS: We developed a new algorithm named Grow Lineages along Minimum Spanning Tree (GLaMST), which efficiently reconstruct the lineage tree given observed BCR sequences that correspond to a subset of the tree nodes. Through comparison using simulated and real data, GLaMST outperforms existing algorithms in simulations with high rates of mutation, insertion and deletion, and generates lineage trees with smaller size and closer to ground truth according to tree features that highly correlated with selection pressure. CONCLUSIONS: GLaMST outperforms state-of-art in reconstruction of the BCR lineage tree in both efficiency and accuracy. Integrating it into existing BCR sequencing analysis frameworks can significant improve lineage tree reconstruction aspect of the analysis.


Assuntos
Centro Germinativo , Receptores de Antígenos de Linfócitos B , Algoritmos , Linfócitos B , Mutação , Receptores de Antígenos de Linfócitos B/genética
9.
J Immunol ; 192(3): 906-18, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24379124

RESUMO

Inappropriate activation of type I IFN plays a key role in the pathogenesis of autoimmune disease, including systemic lupus erythematosus (SLE). In this study, we report the presence of IFN activation in SLE bone marrow (BM), as measured by an IFN gene signature, increased IFN regulated chemokines, and direct production of IFN by BM-resident cells, associated with profound changes in B cell development. The majority of SLE patients had an IFN signature in the BM that was more pronounced than the paired peripheral blood and correlated with both higher autoantibodies and disease activity. Pronounced alterations in B cell development were noted in SLE in the presence of an IFN signature with a reduction in the fraction of pro/pre-B cells, suggesting an inhibition in early B cell development and an expansion of B cells at the transitional stage. These B cell changes strongly correlated with an increase in BAFF and APRIL expression in the IFN-high BM. Furthermore, we found that BM neutrophils in SLE were prime producers of IFN-α and B cell factors. In NZM lupus-prone mice, similar changes in B cell development were observed and mediated by IFN, given abrogation in NZM mice lacking type-I IFNR. BM neutrophils were abundant, responsive to, and producers of IFN, in close proximity to B cells. These results indicate that the BM is an important but previously unrecognized target organ in SLE with neutrophil-mediated IFN activation and alterations in B cell ontogeny and selection.


Assuntos
Subpopulações de Linfócitos B/imunologia , Medula Óssea/imunologia , Interferon Tipo I/fisiologia , Lúpus Eritematoso Sistêmico/imunologia , Linfopoese/imunologia , Neutrófilos/imunologia , Adulto , Animais , Fator Ativador de Células B/biossíntese , Fator Ativador de Células B/genética , Subpopulações de Linfócitos B/patologia , Medula Óssea/metabolismo , Quimiocinas/biossíntese , Quimiocinas/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Humanos , Interferon Tipo I/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Pessoa de Meia-Idade , Neutrófilos/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Regulação para Cima/imunologia
10.
Int J Cancer ; 136(6): 1371-80, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25099163

RESUMO

The development of endemic Burkitt's lymphoma (eBL) is closely associated with Epstein-Barr virus (EBV) infection and holoendemic malaria infections. The role of EBV in the development of malignancy has been studied in depth, but there is still little known about the mechanisms by which malaria affects Burkitt's lymphomagenesis. Activation induced cytidine deaminase (AID) expression is necessary for the introduction of c-myc translocations that are characteristic of BL, but a link between AID and EBV or malaria is unclear. To determine whether frequency of malaria exposure leads to increased AID expression in peripheral blood mononuclear cells (PBMC) we examined two cohorts of children in western Kenya with endemic and sporadic malaria transmission dynamics. High frequency of malaria exposure led to increased expression of AID, which coincided with decreases in the IgM(+) memory B cells. In the children from the malaria endemic region, the presence of a detectible EBV viral load was associated with higher AID expression compared to children with undetectable EBV, but this effect was not seen in children with sporadic exposure to malaria. This study demonstrates that intensity of malaria transmission correlates with AID expression levels in the presence of EBV suggesting that malaria and EBV infection have a synergistic effect on the development of c-myc translocations and BL.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B/enzimologia , Linfoma de Burkitt/etiologia , Citidina Desaminase/fisiologia , Herpesvirus Humano 4/isolamento & purificação , Malária/complicações , Linfoma de Burkitt/imunologia , Pré-Escolar , Citidina Desaminase/sangue , Citidina Desaminase/genética , Doenças Endêmicas , Humanos , Memória Imunológica , Lactente , Malária/epidemiologia , RNA Mensageiro/análise , Carga Viral
11.
J Immunol ; 188(12): 5944-53, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22593620

RESUMO

CD23(+)CD21(high)CD1d(high) B cells in inflamed nodes (Bin cells) accumulate in the lymph nodes (LNs) draining inflamed joints of the TNF-α-transgenic mouse model of rheumatoid arthritis and are primarily involved in the significant histological and functional LN alterations that accompany disease exacerbation in this strain. In this study, we investigate the origin and function of Bin cells. We show that adoptively transferred GFP(+) sorted mature follicular B (FoB) cells home preferentially to inflamed LNs of TNF-α-transgenic mice where they rapidly differentiate into Bin cells, with a close correlation with the endogenous Bin fraction. Bin cells are also induced in wild-type LNs after immunization with T-dependent Ags and display a germinal center phenotype at higher rates compared with FoB cells. Furthermore, we show that Bin cells can capture and process Ag-immune complexes in a CD21-dependent manner more efficiently than can FoB cells, and they express greater levels of MHC class II and costimulatory Ags CD80 and CD86. We propose that Bin cells are a previously unrecognized inflammation-induced B cell population with increased Ag capture and activation potential, which may facilitate normal immune responses but may contribute to autoimmunity when chronic inflammation causes their accumulation and persistence in affected LNs.


Assuntos
Artrite Experimental/imunologia , Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Linfonodos/citologia , Ativação Linfocitária/imunologia , Transferência Adotiva , Animais , Apresentação de Antígeno/imunologia , Antígenos CD1d/imunologia , Antígenos CD1d/metabolismo , Artrite Reumatoide/imunologia , Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/citologia , Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Inflamação/imunologia , Inflamação/metabolismo , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Complemento 3d/imunologia , Receptores de Complemento 3d/metabolismo , Receptores de IgE/imunologia , Receptores de IgE/metabolismo
12.
Autoimmunity ; 57(1): 2356089, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38770919

RESUMO

Autoimmune hepatitis (AIH) is a chronic, inflammatory liver disease of unknown aetiology which requires lifelong immunosuppression. Most therapeutic and outcome studies of AIH have been conducted predominantly in Caucasian (European Ancestry, EA) cohorts, with the exclusion of African American (AA) patients due to inadequate sample size. It is known that AA patients have a severe phenotype of autoimmune diseases and demonstrate a poor response to conventional medical therapy. Understanding cellular and molecular pathways which determine AIH severity and progression in AA patients is likely to lead to the discovery of novel, personalised and better tolerated therapies. The aim of the study is to determine the distinct effector B cell phenotypes which contribute to disease severity and progression of AIH in AA children as compared to their EA cohorts. PBMCs were isolated from blood samples collected from patients visiting Children's Healthcare of Atlanta (CHOA) and were grouped into AA, (n = 12), EA, (n = 11) and controls (n = 12) and were processed for flow cytometry. Markers of B cell development, maturation and activation were assessed namely CD19, CD21, IgD, CD27, CD38, CD11c, CD24, CD138. AA children with AIH demonstrated an expansion of CD19 + ve, Activated Naïve (aN), (CD19+ IgD-/CD27- Double Negative (DN2) ([CD19+/IgD-/CD27++CD38++) cells. Plasmablasts were significantly higher along with Signalling Lymphocytic activation molecule F7 (SLAMF7). Unswitched memory [CD19+] IgD+CD27+ (USM) B cells were significantly contracted in AA patients with AIH. B cell phenotyping reveals a distinct profile in AA AIH patients with a major skewing towards the expansion of effector pathways which have been previously characterised in severe SLE in AA patients. These results suggest that the quantification and therapeutic target of B cell pathway could contribute substantially to the clinical approach to AIH especially in the AA population.


Assuntos
Linfócitos B , Hepatite Autoimune , Imunoglobulina D , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral , Humanos , Hepatite Autoimune/imunologia , Hepatite Autoimune/sangue , Hepatite Autoimune/patologia , Hepatite Autoimune/diagnóstico , Imunoglobulina D/imunologia , Imunoglobulina D/metabolismo , Criança , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Masculino , Feminino , Adolescente , Linfócitos B/imunologia , Linfócitos B/metabolismo , Pré-Escolar , Imunofenotipagem , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Idade de Início , Biomarcadores
13.
Blood ; 118(13): 3591-602, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21821704

RESUMO

The follicular lymphoma (FL) T-cell microenvironment plays a critical role in the biology of this disease. We therefore determined the lineage, differentiation state, and functional potential of FL-infiltrating CD4(+) T-helper cells (T(H)) compared with reactive and normal lymph node (NLN) T(H) cells. Relative to NLNs, FL cells have decreased proportions of naive and central memory but increased proportions of effector memory T(H) cells. We further show differences in the distribution and anatomical localization of CXCR5(+) T(H) populations that, on the basis of transcription factor analysis, include both regulatory and follicular helper T cells. On Staphylococcus enterotoxin-B stimulation, which stimulates T cells through the T-cell receptor, requires no processing by APCs, and can overcome regulator T cell-mediated suppression, the proportion of uncommitted primed precursor cells, as well as T(H)2 and T(H)17 cells is higher in FL cells than in reactive lymph nodes or NLNs. However, the proportion of T(H)1 and polyfunctional T(H) cells (producing multiple cytokines simultaneously) is similar in FL cells and NLNs. These data suggest that, although T(H)-cell differentiation in FL is skewed compared with NLNs, FL T(H) cells should have the same intrinsic ability to elicit antitumor effector responses as NLN T(H) cells when tumor suppressive mechanisms are attenuated.


Assuntos
Diferenciação Celular/imunologia , Linfonodos/imunologia , Linfócitos do Interstício Tumoral/fisiologia , Linfoma Folicular/imunologia , Linfócitos T Auxiliares-Indutores/fisiologia , Diferenciação Celular/genética , Análise por Conglomerados , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Humanos , Memória Imunológica/genética , Memória Imunológica/fisiologia , Linfonodos/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfoma Folicular/genética , Linfoma Folicular/metabolismo , Linfoma Folicular/patologia , Análise em Microsséries , Proteínas Proto-Oncogênicas c-bcl-6 , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
14.
J Immunol ; 186(8): 4640-8, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21398610

RESUMO

B cell anergy represents an important mechanism of peripheral immunological tolerance for mature autoreactive B cells that escape central tolerance enforced by receptor editing and clonal deletion. Although well documented in mice, the extent of its participation in human B cell tolerance remains to be fully established. In this study, we characterize the functional behavior of strictly defined human naive B cells separated on the basis of their surface IgM (sIgM) expression levels. We demonstrate that cells with lower sIgM levels (IgM(lo)) are impaired in their ability to flux calcium in response to either anti-IgM or anti-IgD cross-linking and contain a significantly increased frequency of autoreactive cells compared with naive B cells with higher levels of sIgM. Phenotypically, in healthy subjects, IgM(lo) cells are characterized by the absence of activation markers, reduction of costimulatory molecules (CD19 and CD21), and increased levels of inhibitory CD22. Functionally, IgM(lo) cells display significantly weaker proliferation, impaired differentiation, and poor Ab production. In aggregate, the data indicate that hyporesponsiveness to BCR cross-linking associated with sIgM downregulation is present in a much larger fraction of all human naive B cells than previously reported and is likely to reflect a state of anergy induced by chronic autoantigen stimulation. Finally, our results indicate that in systemic lupus erythematosus patients, naive IgM(lo) cells display increased levels of CD95 and decreased levels of CD22, a phenotype consistent with enhanced activation of autoreactive naive B cells in this autoimmune disease.


Assuntos
Autoantígenos/imunologia , Linfócitos B/imunologia , Anergia Clonal/imunologia , Imunoglobulina M/imunologia , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Anti-Idiotípicos/metabolismo , Antígenos CD19/imunologia , Antígenos CD19/metabolismo , Linfócitos B/metabolismo , Cálcio/metabolismo , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Citometria de Fluxo , Humanos , Imunoglobulina D/imunologia , Imunoglobulina M/metabolismo , Transporte de Íons/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Receptores de Complemento 3d/imunologia , Receptores de Complemento 3d/metabolismo , Tolerância a Antígenos Próprios/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Receptor fas/imunologia , Receptor fas/metabolismo
15.
J Immunol ; 186(9): 5514-21, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21441455

RESUMO

Surges of serum Abs after immunization and infection are highly specific for the offending Ag, and recent studies demonstrate that vaccines induce transient increases in circulating Ab-secreting cells (ASCs). These ASCs are highly enriched but not universally specific for the immunizing Ag, suggesting that a fraction of these ASCs could arise from polyclonal bystander stimulation of preexisting memory cells to unrelated Ags. This model is proposed to explain maintenance of long-lived serological memory in the absence of Ag exposure. To test this model, we measure the ability of respiratory syncytial virus and influenza virus infection or immunizations to influenza virus, tetanus toxoid, hepatitis B Ag, and human papillomavirus to stimulate bystander memory cells specific for other major environmental Ags that represent a large fraction of the preexisting memory B compartment. Bystander or nonspecific ASC responses to respiratory syncytial virus and tetanus could not be detected above the background levels in healthy adults, despite the presence of circulating memory B cells specific for the corresponding Ags. Nonspecific ASC responses in the healthy subjects and cord blood samples were similar. In contrast, both vaccination and infection induce massive expansion of circulating Ag-specific ASCs without significant increases in the frequencies of ASCs against unrelated Ags. Hence, nonspecific stimulation of memory B cells is unlikely to contribute to the mechanisms of long-term serological memory against major human pathogens. Additionally, high specificity of circulating ASCs after antigenic challenge highlights the diagnostic value of interrogating ASCs as an ideal single-time-point diagnostic immune surrogate for serology during acute infection.


Assuntos
Células Produtoras de Anticorpos/imunologia , Efeito Espectador/imunologia , Memória Imunológica , Infecções Respiratórias/imunologia , Vacinação , Viroses/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos Virais/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adulto Jovem
16.
Nat Commun ; 14(1): 1388, 2023 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-36941260

RESUMO

Anti-dsDNA antibodies are pathogenically heterogeneous, implying distinct origins and antigenic properties. Unexpectedly, during the clinical and molecular characterization of autoantibodies to the endonuclease DNase1L3 in patients with systemic lupus erythematosus (SLE), we identified a subset of neutralizing anti-DNase1L3 antibodies previously catalogued as anti-dsDNA. Based on their variable heavy-chain (VH) gene usage, these antibodies can be divided in two groups. One group is encoded by the inherently autoreactive VH4-34 gene segment, derives from anti-DNase1L3 germline-encoded precursors, and gains cross-reactivity to dsDNA - and some additionally to cardiolipin - following somatic hypermutation. The second group, originally defined as nephritogenic anti-dsDNA antibodies, is encoded by diverse VH gene segments. Although affinity maturation results in dual reactivity to DNase1L3 and dsDNA, their binding efficiencies favor DNase1L3 as the primary antigen. Clinical, transcriptional and monoclonal antibody data support that cross-reactive anti-DNase1L3/dsDNA antibodies are more pathogenic than single reactive anti-dsDNA antibodies. These findings point to DNase1L3 as the primary target of a subset of antibodies classified as anti-dsDNA, shedding light on the origin and pathogenic heterogeneity of antibodies reactive to dsDNA in SLE.


Assuntos
Lúpus Eritematoso Sistêmico , Humanos , Lúpus Eritematoso Sistêmico/genética , Autoanticorpos , Anticorpos Antinucleares/genética , DNA/metabolismo , Anticorpos Monoclonais , Endodesoxirribonucleases/genética
17.
Clin Immunol ; 142(3): 237-42, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22284392

RESUMO

Cell signaling initiated by the B cell receptor is critical to normal development of B lymphocytes, most notably at the transitional B cell stage. Inhibition of this signaling pathway with the syk inhibitor, fostamatinib, has produced significant efficacy in lymphoid malignancies and autoimmune conditions. Here, we demonstrate that short-term use of fostamatinib impairs B lymphocyte development at the transitional stage without affecting mature B cell populations. Additionally, IL-10 producing B cells remained relatively constant throughout the treatment period. These findings provide insight into the mechanism of action of B cell receptor inhibition in autoimmune disease. As the development of agents targeting B cell receptor signaling proceeds, monitoring for long-term consequences as well as functional evaluation of B cell subsets may further improve our understanding of this rapidly growing class of novel agents.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Oxazinas/farmacologia , Células Precursoras de Linfócitos B/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Piridinas/farmacologia , Aminopiridinas , Humanos , Interleucina-10/biossíntese , Interleucina-10/imunologia , Morfolinas , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/imunologia , Pirimidinas , Quinase Syk
18.
Curr Opin Rheumatol ; 24(5): 451-6, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22871954

RESUMO

PURPOSE OF REVIEW: Last year was marked by important clinical and mechanistic studies that improved our understanding of B-cell immunotherapy for systemic lupus erythematosus (SLE) and Sjogren's syndrome. Here, we will highlight the most relevant studies published in the last 18 months. RECENT FINDINGS: The highlight of the year was the approval of belimumab on the basis of two major trials. On the flip side, the disappointing results of rituximab in lupus nephritis provided a clinical and mechanistic counterpoint in SLE. Still, major limitations in the LUpus Nephritis Assessment with Rituximab (LUNAR) trial, positive subset analysis and new open studies and registries continue to provide hope for and major insights into the use of B-cell depletion. In Sjogren's syndrome, the role of B-cell depletion has been further investigated, both for glandular and extraglandular manifestations of the disease with mixed results in a disease in which outcomes are notoriously hard to measure. SUMMARY: The approval of anti-B cell activating factor therapy and an increasing body of open studies with rituximab as well as subset studies and secondary analysis of the Efficacy and Safety of Rituximab in Moderately-to-Severely Active Systemic Lupus Erythematosus (EXPLORER) and LUNAR trials provide hope for B-cell immunotherapy and significant insight into its mechanisms of action and utilization in a selected subset of patients. Ongoing clinical trials of other B-cell targeting agents are eagerly anticipated.


Assuntos
Linfócitos B/imunologia , Imunoterapia/métodos , Lúpus Eritematoso Sistêmico/terapia , Síndrome de Sjogren/terapia , Anticorpos Monoclonais Murinos/uso terapêutico , Linfócitos B/patologia , Sobrevivência Celular/imunologia , Ensaios Clínicos como Assunto , Humanos , Imunossupressores/uso terapêutico , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/terapia , Depleção Linfocítica , Rituximab , Síndrome de Sjogren/imunologia , Síndrome de Sjogren/patologia
19.
J Immunol ; 184(11): 6142-50, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20435928

RESUMO

Anti-CD20 B cell depletion therapy (BCDT) is very effective for some patients with rheumatoid arthritis (RA); however the pathogenic role of B lymphocytes in RA and the primary targets of BCDT are unknown. The human TNF transgenic (hTNF-Tg) mouse model of RA displays a chronic, progressive disease that spreads from distal to proximal joints and is generally considered to be adaptive immune system independent. We have previously reported that knee arthritis in hTNF-Tg mice is accompanied by structural and functional changes of the adjoining popliteal lymph node (PLN), detectable by contrast-enhanced magnetic resonance imaging. To better understand these changes, in this paper we show that onset of knee synovitis and focal erosions are paralleled by PLN contraction and accumulation of large numbers of B cells in the lymphatic sinus spaces within the node. Flow cytometry from TNF-Tg mice 2, 4-5, and 8-12 mo old demonstrated that B cell accumulation in the PLN follows ankle arthritis, but commences before knee disease, and involves early expansion of CD21(hi), CD23(+), IgM(hi), CD1d(+), activation marker-negative, polyclonal B cells that are found to be specifically restricted to lymph nodes draining inflamed, arthritic joints. The same B cell population also accumulates in PLNs of K/BxN mice with autoantigen-dependent arthritis. Strikingly, we show that BCDT ameliorates hTNF-Tg disease and clears follicular and CD21(hi), CD23(+) B cells from the PLNs. On the basis of these findings, we propose a model whereby B cells contribute to arthritis in mice, and possibly RA, by directly affecting the structure, composition, and function of joint-draining lymph nodes.


Assuntos
Anticorpos Monoclonais/farmacologia , Antirreumáticos/farmacologia , Artrite Experimental/imunologia , Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Linfonodos/imunologia , Animais , Anticorpos Monoclonais Murinos , Artrite Experimental/tratamento farmacológico , Artrite Experimental/patologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Separação Celular , Citometria de Fluxo , Humanos , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Complemento 3d/imunologia , Receptores de IgE/imunologia , Rituximab , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
20.
Life Sci Alliance ; 5(3)2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34952892

RESUMO

Antibody secreting cells (ASCs) circulate after vaccination and infection and migrate to the BM where a subset known as long-lived plasma cells (LLPCs) persists and secrete antibodies for a lifetime. The mechanisms by which circulating ASCs become LLPCs are not well elucidated. Here, we show that human blood ASCs have distinct morphology, transcriptomes, and epigenetics compared with BM LLPCs. Compared with blood ASCs, BM LLPCs have decreased nucleus/cytoplasm ratio but increased endoplasmic reticulum and numbers of mitochondria. LLPCs up-regulate pro-survival genes MCL1, BCL2, and BCL-XL while simultaneously down-regulating pro-apoptotic genes HRK1, CASP3, and CASP8 Consistent with reduced gene expression, the pro-apoptotic gene loci are less accessible in LLPCs. Of the pro-survival genes, only BCL2 is concordant in gene up-regulation and loci accessibility. Using a novel in vitro human BM mimetic, we show that blood ASCs undergo similar morphological and molecular changes that resemble ex vivo BM LLPCs. Overall, our study demonstrates that early-minted blood ASCs in the BM microniche must undergo morphological, transcriptional, and epigenetic changes to mature into apoptotic-resistant LLPCs.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica , Impressão Genômica , Plasmócitos/citologia , Plasmócitos/metabolismo , Adolescente , Adulto , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Apoptose/genética , Biomarcadores , Sobrevivência Celular , Feminino , Heterogeneidade Genética , Histocitoquímica , Humanos , Imunofenotipagem , Masculino , Pessoa de Meia-Idade , Plasmócitos/imunologia , Plasmócitos/ultraestrutura , Fatores de Tempo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA