Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Autoimmun ; 147: 103243, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38788537

RESUMO

OBJECTIVES: Autoreactive B cells and interferon (IFN) signature are hallmarks of primary sjögren's syndrome (pSS), but how IFN signaling pathways influence autoantibody production and clinical manifestations remain unclear. More detailed studies hold promise for improved diagnostic methodologies and personalized treatment. METHODS: We analyzed peripheral blood T and B cell subsets from 34 pSS patients and 38 healthy donors (HDs) at baseline and upon stimulation regarding their expression levels of type I and II IFN signaling molecules (STAT1/2, IRF1, IRF9). Additionally, we investigated how the levels of these molecules correlated with serological and clinical characteristics and performed ROC analysis. RESULTS: Patients showed elevated IFN pathway molecules, including STAT1, STAT2 and IRF9 among most T and B cell subsets. We found a reduced ratio of phosphorylated STAT1 and STAT2 in patients in comparison to HDs, although B cells from patients were highly responsive by increased phosphorylation upon IFN stimulation. Correlation matrices showed further interrelations between STAT1, IRF1 and IRF9 in pSS. Levels of STAT1 and IRF9 in T and B cells correlated with the IFN type I marker Siglec-1 (CD169) on monocytes. High levels of STAT1 and IRF9 within pSS B cells were significantly associated with hypergammaglobulinemia as well as anti-SSA/anti-SSB autoantibodies. Elevated STAT1 levels were found in patients with extraglandular disease and could serve as a biomarker for this subgroup (p < 0.01). Notably, IRF9 levels in T and B cells correlated with EULAR Sjögren's syndrome disease activity index (ESSDAI). CONCLUSION: Here, we provide evidence that in active pSS patients, enhanced IFN signaling incl. unphosphorylated STAT1 and STAT2 with IRFs entertain chronic T and B cell activation. Furthermore, increased STAT1 levels candidate as biomarker of extraglandular disease, while IRF9 levels can serve as biomarker for disease activity.


Assuntos
Biomarcadores , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Fator de Transcrição STAT1 , Síndrome de Sjogren , Humanos , Síndrome de Sjogren/imunologia , Síndrome de Sjogren/diagnóstico , Síndrome de Sjogren/metabolismo , Fator de Transcrição STAT1/metabolismo , Feminino , Fosforilação , Pessoa de Meia-Idade , Masculino , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Idoso , Adulto , Linfócitos B/imunologia , Linfócitos B/metabolismo , Autoanticorpos/imunologia , Autoanticorpos/sangue , Transdução de Sinais , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
2.
Nat Commun ; 15(1): 4182, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38755157

RESUMO

Bone marrow plasma cells (BMPC) are the correlate of humoral immunity, consistently releasing antibodies into the bloodstream. It remains unclear if BMPC reflect different activation environments or maturation of their precursors. Here we define human BMPC heterogeneity and track the recruitment of antibody-secreting cells (ASC) from SARS-CoV-2 vaccine immune reactions to the bone marrow (BM). Trajectories based on single-cell transcriptomes and repertoires of peripheral and BM ASC reveal sequential colonisation of BMPC compartments. In activated B cells, IL-21 suppresses CD19 expression, indicating that CD19low-BMPC are derived from follicular, while CD19high-BMPC originate from extrafollicular immune reactions. In primary immune reactions, both CD19low- and CD19high-BMPC compartments are populated. In secondary immune reactions, most BMPC are recruited to CD19high-BMPC compartments, reflecting their origin from extrafollicular reactivations of memory B cells. A pattern also observable in vaccinated-convalescent individuals and upon diphtheria/tetanus/pertussis recall-vaccination. Thus, BMPC diversity reflects the evolution of a given humoral immune response.


Assuntos
Antígenos CD19 , Medula Óssea , Interleucinas , Plasmócitos , Humanos , Plasmócitos/imunologia , Interleucinas/imunologia , Interleucinas/metabolismo , Medula Óssea/imunologia , Antígenos CD19/imunologia , Antígenos CD19/metabolismo , Imunidade Humoral/imunologia , COVID-19/imunologia , COVID-19/virologia , SARS-CoV-2/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/citologia , Análise de Célula Única , Adulto , Linfócitos B/imunologia , Células Produtoras de Anticorpos/imunologia , Feminino , Masculino , Vacinação , Pessoa de Meia-Idade , Vacina contra Difteria, Tétano e Coqueluche/imunologia
3.
Sci Rep ; 13(1): 22587, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38114596

RESUMO

Inflammation conditions are associated with autism spectrum disorder (ASD) and cerebral palsy (CP), primarily observed in the peripheral immune system. However, the extent of neuro-inflammation and neuro-immune dysregulation remains poorly studied. In this study, we analyzed the composition of cerebrospinal fluid (CSF) to uncover the inflammatory mediators driving the neuro-immune system in ASD and CP patients. Our findings revealed that ASD patients had elevated levels of four inflammatory cytokines (TNF-α, IL-4, IL-21, and BAFF) compared to controls, while CP patients exhibited increased levels of eight inflammatory cytokines (IFN-γ, GM-CSF, TNF-α, IL-2, IL-4, IL-6, IL-17A and IL-12), one anti-inflammatory cytokine (IL-10), and five growth factors (GFs) (NGF-ß, EGF, GDF-15, G-CSF and BMP-9) compared to both controls and ASD patients. Additionally, intrathecal infusion of autologous bone marrow mononuclear cells (BMMNCs) led to a slight decrease in TGF-ß and GDF-15 levels in the CSF of ASD and CP patients, respectively. Our study provides new insights into the molecular composition of CSF in ASD and CP patients, with the potential to develop more effective diagnosis methods and improved treatment for these diseases.Clinical trial registration CSF samples used in this study are from clinical trials NCT03225651, NCT05307536, NCT02569775, NCT03123562, NCT02574923, NCT05472428 and previous reports [7, 9, 17-19].


Assuntos
Transtorno do Espectro Autista , Paralisia Cerebral , Humanos , Fator 15 de Diferenciação de Crescimento , Fator de Necrose Tumoral alfa/metabolismo , Mediadores da Inflamação , Doenças Neuroinflamatórias , Interleucina-4 , Citocinas/metabolismo , Inflamação/metabolismo
4.
Mol Ther Nucleic Acids ; 30: 621-632, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36514352

RESUMO

Human B lymphocytes are attractive targets for immunotherapies in autoantibody-mediated diseases. Gene editing technologies could provide a powerful tool to determine gene regulatory networks regulating B cell differentiation into plasma cells, and identify novel therapeutic targets for prevention and treatment of autoimmune disorders. Here, we describe a new approach that uses CRISPR-Cas9 technology to target genes in primary human B cells in vitro for identifying plasma cell regulators. We found that sgRNA and Cas9 components can be efficiently delivered into primary human B cells through RD114-pseudotyped retroviral vectors. Using this system, we achieved approximately 80% of gene knockout efficiency. We disrupted expression of a triad of transcription factors, IRF4, PRDM1, and XBP1, and showed that human B cell survival and plasma cell differentiation are severely impaired. Specifically, that IRF4, PRDM1, and XBP1 were expressed at different stages during plasma cell differentiation, IRF4, PRDM1, and XBP1-targeted B cells failed to progress to the pre-plasmablast, plasma cell state, and plasma cell survival, respectively. Our method opens a new avenue to study gene functions in primary human B cells and identify novel plasma cell regulators for therapeutic applications.

5.
Arthritis Rheumatol ; 74(9): 1556-1568, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35507291

RESUMO

OBJECTIVE: Altered composition of the B cell compartment in the pathogenesis of systemic lupus erythematosus (SLE) is characterized by expanded plasmablast and IgD-CD27- double-negative B cell populations. Previous studies showed that double-negative B cells represent a heterogeneous subset, and further characterization is needed. METHODS: We analyzed 2 independent cohorts of healthy donors and SLE patients, using a combined approach of flow cytometry (for 16 healthy donors and 28 SLE patients) and mass cytometry (for 18 healthy donors and 24 SLE patients) and targeted RNA-Seq analysis. To compare B cell subset formation during the acute immune response versus that during autoimmune disease, we investigated healthy donors at various time points after receipt of the BNT162b2 messenger RNA COVID-19 vaccine and patients with acute SARS-CoV-2 infection, using flow cytometry. RESULTS: We found that IgD-CD27+ switched and atypical IgD-CD27- memory B cells, the levels of which were increased in SLE patients, represented heterogeneous populations composed of 3 different subsets each. CXCR5+CD19intermediate , CXCR5-CD19high , and CXCR5-CD19low populations were found in the switched memory and double-negative compartments, suggesting the relatedness of IgD-CD27+ and IgD-CD27- B cells. We characterized a hitherto unknown and antigen-experienced CXCR5-CD19low subset that was enhanced in SLE patients, had a plasmablast phenotype with diminished B cell receptor responsiveness, and expressed CD38, CD95, CD71, PRDM1, XBP1, and IRF4. Levels of CXCR5-CD19low subsets were increased and correlated with plasmablast frequencies in SLE patients and in healthy donors who received BNT162b2, suggesting their interrelationship and contribution to plasmacytosis. The detection of CXCR5-CD19low B cells among both CD27+ and CD27- populations calls into question the role of CD27 as a reliable marker of B cell differentiation. CONCLUSION: Our data suggest that CXCR5-CD19low B cells are precursors of plasmablasts. Thus, cotargeting this subset may have therapeutic value in SLE.


Assuntos
Subpopulações de Linfócitos B , COVID-19 , Lúpus Eritematoso Sistêmico , Antígenos CD19/genética , Antígenos CD19/metabolismo , Subpopulações de Linfócitos B/metabolismo , Vacina BNT162 , Vacinas contra COVID-19 , Humanos , Imunoglobulina D , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Fenótipo , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , SARS-CoV-2
6.
Front Immunol ; 13: 873217, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35464469

RESUMO

Antibody-secreting cells (ASCs) contribute to immunity through production of antibodies and cytokines. Identification of specific markers of ASC would allow selective targeting of these cells in several disease contexts. Here, we performed an unbiased, large-scale protein screening, and identified twelve new molecules that are specifically expressed by murine ASCs. Expression of these markers, particularly CD39, CD81, CD130, and CD326, is stable and offers an improved resolution for ASC identification. We accessed their expression in germ-free conditions and in T cell deficient mice, showing that at least in part their expression is controlled by microbial- and T cell-derived signals. Further analysis of lupus mice revealed the presence of a subpopulation of LAG-3- plasma cells, co-expressing high amounts of CD39 and CD326 in the bone marrow. This population was IgM+ and correlated with IgM anti-dsDNA autoantibodies in sera. Importantly, we found that CD39, CD81, CD130, and CD326 are also expressed by human peripheral blood and bone marrow ASCs. Our data provide innovative insights into ASC biology and function in mice and human, and identify an intriguing BM specific CD39++CD326++ ASC subpopulation in autoimmunity.


Assuntos
Medula Óssea , Plasmócitos , Animais , Anticorpos Antinucleares , Células Produtoras de Anticorpos , Biomarcadores/metabolismo , Medula Óssea/metabolismo , Humanos , Imunoglobulina M , Camundongos , Plasmócitos/metabolismo
7.
J Invest Dermatol ; 142(3 Pt B): 811-822, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34955289

RESUMO

B lymphocytes play a central role in immunity owing to their unique antibody-producing capacity that provides protection against certain infections and during vaccination. In autoimmune diseases, B cells can gain pathogenic relevance through autoantibody production, antigen presentation, and proinflammatory cytokine secretion. Recent data indicate that B and plasma cells can function as regulators through the production of immunoregulatory cytokines and/or employing checkpoint molecules. In this study, we review the key findings that define subsets of B and plasma cells with pathogenic and protective functions in autoimmunity. In addition to harsh B-cell depletion, we discuss the strategies that have the potential to reinstall the balance of pathogenic and protective B cells with the potential of more specific and personalized therapies.


Assuntos
Doenças Autoimunes , Plasmócitos , Autoimunidade , Linfócitos B , Citocinas , Humanos
8.
Methods Mol Biol ; 2270: 47-59, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33479892

RESUMO

B cells are primarily known for their capacity to differentiate into antibody-secreting cells (ASCs). ASCs are usually viewed as terminally differentiated cells sharing a unique phenotype. However, it lately became evident that ASCs exist in a variety of subsets differing by their lifespan, anatomic location, and immunological function. Thus, ASCs can exist as long-lived plasma cells (LLPC) that can persist for years in a nonproliferating state within particular niches in the bone marrow (BM), or as short-lived plasma cells (SLPC) that are primarily found in secondary lymphoid organs or inflamed tissues and wane upon the termination of the associated immune response. Another layer of ASC diversity was uncovered with the discovery of their capacity to produce various pro- or anti-inflammatory cytokines. Notably, a subset of natural regulatory plasma cells characterized by the distinctive expression of the inhibitory receptor lymphocyte activation gene (LAG)-3 and a unique capacity to produce interleukin (IL)-10 upon stimulation was recently identified. Here, we describe how to immunophenotypically characterize murine plasma cells as well as how to isolate them using cell sorting, with a special focus on these recently described natural regulatory plasma cells.


Assuntos
Citometria de Fluxo/métodos , Imunofenotipagem/métodos , Plasmócitos/imunologia , Animais , Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Medula Óssea/imunologia , Células da Medula Óssea/citologia , Citocinas/metabolismo , Imunidade Humoral/imunologia , Memória Imunológica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia
9.
Front Mol Biosci ; 7: 119, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32671095

RESUMO

Exosomes are nano-scale and closed membrane vesicles which are promising for therapeutic applications due to exosome-enclosed therapeutic molecules such as DNA, small RNAs, proteins and lipids. Recently, it has been demonstrated that mesenchymal stem cell (MSC)-derived exosomes have capacity to regulate many biological events associated with wound healing process, such as cell proliferation, cell migration and blood vessel formation. This study investigated the regenerative potentials for cutaneous tissue, in regard to growth factors associated with wound healing and skin cell proliferation and migration, by exosomes released from primary MSCs originated from bone marrow (BM), adipose tissue (AD), and umbilical cord (UC) under serum- and xeno-free condition. We found crucial wound healing-mediated growth factors, such as vascular endothelial growth factor A (VEGF-A), fibroblast growth factor 2 (FGF-2), hepatocyte growth factor (HGF), and platelet-derived growth factor BB (PDGF-BB) in exosomes derived from all three MSC sources. However, expression levels of these growth factors in exosomes were influenced by MSC origins, especially transforming growth factor beta (TGF-ß) was only detected in UCMSC-derived exosomes. All exosomes released by three MSCs sources induced keratinocyte and fibroblast proliferation and migration; and, the induction of cell migration is a dependent manner with the higher dose of exosomes was used (20 µg), the faster migration rate was observed. Additionally, the influences of exosomes on cell proliferation and migration was associated with exosome origins and also target cells of exosomes that the greatest induction of primary dermal fibroblasts belongs to BMMSC-derived exosomes and keratinocytes belongs to UCMSC-derived exosomes. Data from this study indicated that BMMSCs and UCMSCs under clinical condition secreted exosomes are promising to develop into therapeutic products for wound healing treatment.

10.
J Invest Dermatol ; 139(12): 2477-2487, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31152755

RESUMO

Antibodies are key to cutaneous host defense and inflammation. Despite their importance, the mechanisms by which skin antibodies are sustained are poorly described. Here, we identified that, in addition to antibody production in lymphoid tissues, plasma cells reside in healthy mouse and human skin. In naïve mice, IgM was the predominant isotype produced in skin. Skin plasma cells developed independently of T cells and microbiota. Importantly, chronic skin inflammation promoted the massive accumulation of IgM-secreting cells, and cutaneous immunization directed both T cell-dependent and -independent antigen-specific IgM-secreting cells into skin. Unlike their counterparts in lymphoid tissues, cutaneous IgM-secreting cells were completely dependent on survival factors such as a proliferation-inducing ligand or B cell-activating factor, which were constitutively expressed and upregulated during inflammation in skin. Our data support a model in which skin plasma cells supply natural and adaptive IgM to the cutaneous environment, thereby supporting homeostatic skin barrier functions and providing defense against pathogen intrusion. Our results are also of potential relevance for manipulation of cutaneous plasma cells in inflammatory skin diseases or cutaneous plasma cell malignancies.


Assuntos
Linfócitos B/imunologia , Imunidade Celular , Imunoglobulina M/imunologia , Inflamação/imunologia , Pele/imunologia , Linfócitos T/imunologia , Animais , Formação de Anticorpos , Linfócitos B/metabolismo , Doença Crônica , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos , Plasmócitos/imunologia , Plasmócitos/metabolismo , Valores de Referência , Pele/patologia , Linfócitos T/metabolismo
11.
Immunity ; 49(1): 120-133.e9, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30005826

RESUMO

B lymphocytes can suppress immunity through interleukin (IL)-10 production in infectious, autoimmune, and malignant diseases. Here, we have identified a natural plasma cell subset that distinctively expresses the inhibitory receptor LAG-3 and mediates this function in vivo. These plasma cells also express the inhibitory receptors CD200, PD-L1, and PD-L2. They develop from various B cell subsets in a B cell receptor (BCR)-dependent manner independently of microbiota in naive mice. After challenge they upregulate IL-10 expression via a Toll-like receptor-driven mechanism within hours and without proliferating. This function is associated with a unique transcriptome and epigenome, including the lowest amount of DNA methylation at the Il10 locus compared to other B cell subsets. Their augmented accumulation in naive mutant mice with increased BCR signaling correlates with the inhibition of memory T cell formation and vaccine efficacy after challenge. These natural regulatory plasma cells may be of broad relevance for disease intervention.


Assuntos
Antígenos CD/genética , Expressão Gênica , Interleucina-10/biossíntese , Plasmócitos/imunologia , Animais , Antígenos CD/imunologia , Subpopulações de Linfócitos B/imunologia , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Interleucina-10/genética , Ativação Linfocitária , Masculino , Camundongos , Plasmócitos/fisiologia , Receptores de Antígenos de Linfócitos B/metabolismo , Salmonelose Animal/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Receptores Toll-Like/metabolismo , Regulação para Cima/genética , Vacinas/imunologia , Proteína do Gene 3 de Ativação de Linfócitos
13.
PLoS One ; 9(8): e105421, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25136908

RESUMO

Clinical trial results demonstrating that B-cell depletion substantially reduces new relapses in patients with multiple sclerosis (MS) have established that B cells play a role in the pathophysiology of MS relapses. The same treatment appears not to impact antibodies directed against the central nervous system, which underscores the contribution of antibody-independent functions of B cells to disease activity. One mechanism by which B cells are now thought to contribute to MS activity is by over-activating T cells, including through aberrant expression of B cell pro-inflammatory cytokines. However, the mechanisms underlying the observed B cell cytokine dysregulation in MS remain unknown. We hypothesized that aberrant expression of particular microRNAs might be involved in the dysregulated pro-inflammatory cytokine responses of B cells of patients with MS. Through screening candidate microRNAs in activated B cells of MS patients and matched healthy subjects, we discovered that abnormally increased secretion of lymphotoxin and tumor necrosis factor α by MS B cells is associated with abnormally increased expression of miR-132. Over-expression of miR-132 in normal B cells significantly enhanced their production of lymphotoxin and tumor necrosis factor α. The over-expression of miR-132 also suppressed the miR-132 target, sirtuin-1. We confirmed that pharmacological inhibition of sirtuin-1 in normal B cells induces exaggerated lymphotoxin and tumor necrosis factor α production, while the abnormal production of these cytokines by MS B cells can be normalized by resveratrol, a sirtuin-1 activator. These results define a novel miR-132-sirtuin-1 axis that controls pro-inflammatory cytokine secretion by human B cells, and demonstrate that a dysregulation of this axis underlies abnormal pro-inflammatory B cell cytokine responses in patients with MS.


Assuntos
Linfócitos B/metabolismo , MicroRNAs/metabolismo , Esclerose Múltipla/metabolismo , Sirtuína 1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Linfócitos B/imunologia , Estudos de Casos e Controles , Feminino , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Ativação Linfocitária/imunologia , Linfotoxina-alfa/imunologia , Linfotoxina-alfa/metabolismo , Masculino , MicroRNAs/imunologia , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Sirtuína 1/imunologia , Fator de Necrose Tumoral alfa/imunologia
14.
Curr Top Microbiol Immunol ; 380: 69-92, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25004814

RESUMO

B cells are usually considered primarily for their unique capacity to produce antibodies after differentiation into plasma cells. In addition to their roles as antibody-producing cells, it has become apparent during the last 10 years that B cells also perform important functions in immunity through the production of cytokines. In particular, it was shown that B cells could negatively regulate immunity through provision of interleukin (IL)-10 during autoimmune and infectious diseases in mice. Here, we review data on the suppressive functions of B cells in mice with particular emphasis on the signals controlling the acquisition of such suppressive functions by B cells, the phenotype of the B cells involved in the negative regulation of immunity, and the processes targeted by this inhibitory circuit. Finally, we discuss the possibility that human B cells might also perform similar inhibitory functions through the provision of IL-10, and review data suggesting that such B cell-mediated regulatory activities might be impaired in patients with autoimmune diseases.


Assuntos
Linfócitos B/imunologia , Interleucina-10/fisiologia , Animais , Encefalomielite Autoimune Experimental/imunologia , Humanos , Doenças Inflamatórias Intestinais/imunologia , Listeriose/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium
15.
Eur J Immunol ; 44(5): 1251-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24615065

RESUMO

B-cell depletion can improve disease in some patients with rheumatoid arthritis or multiple sclerosis, indicating the pathogenic contribution of B cells to autoimmunity. However, studies in mice have demonstrated that B cells have immunosuppressive functions as well, with IL-10 being a critical mediator of B-cell-mediated suppression. IL-10-secreting B cells have been shown to promote disease remission in some mouse models of autoimmune disorders. Human B cells also produce IL-10, and evidence is accumulating that human IL-10-producing B cells might inhibit immunity. There is considerable interest in identifying the phenotype of B cells providing IL-10 in a suppressive manner, which would facilitate the analysis of the molecular mechanisms controlling this B-cell property. Here, we review current knowledge on the B-cell subpopulations found to provide suppressive functions in mice, considering both the pathological context in which they were identified and the signals that control their induction. We discuss the phenotype of B cells that have IL-10-dependent regulatory activities in mice, which leads us to propose that antibody-secreting cells are, in some cases at least, the major source of B-cell-derived regulatory IL-10 in vivo. Anti-inflammatory cytokine production by antibody-secreting cells offers a novel mechanism for the coordination of innate and humoral immune responses.


Assuntos
Artrite Reumatoide/imunologia , Linfócitos B Reguladores/imunologia , Interleucina-10/metabolismo , Esclerose Múltipla/imunologia , Plasmócitos/imunologia , Animais , Artrite Reumatoide/patologia , Linfócitos B Reguladores/patologia , Humanos , Imunidade Humoral , Imunidade Inata , Camundongos , Esclerose Múltipla/patologia , Plasmócitos/patologia
16.
Curr Opin Immunol ; 28: 77-83, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24637161

RESUMO

B lymphocytes have a unique role as antibody-producing cells. Antibodies are key mediators of humoral immunity against infections, and are thought to account for the protection afforded by successful vaccines. B cells can also secrete cytokines and subsequently regulate immune responses mediated by T and innate cells. Remarkably, recent studies identified plasma blasts/plasma cells as the main types of activated B cells producing the cytokines interleukin (IL)-10, IL-35, tumor necrosis factor (TNF)-α, IL-17, and GM-CSF in various contexts in mice. Here, we discuss these observations, which suggest the existence of various subsets of plasma blast/plasma cells distinguishable through their cytokine expression pattern.


Assuntos
Linfócitos B/imunologia , Diferenciação Celular , Citocinas/imunologia , Plasmócitos/imunologia , Animais , Linfócitos B/citologia , Citocinas/biossíntese , Humanos , Óxido Nítrico Sintase Tipo II/imunologia , Plasmócitos/citologia , Trypanosoma cruzi/imunologia
17.
Nature ; 507(7492): 366-370, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24572363

RESUMO

B lymphocytes have critical roles as positive and negative regulators of immunity. Their inhibitory function has been associated primarily with interleukin 10 (IL-10) because B-cell-derived IL-10 can protect against autoimmune disease and increase susceptibility to pathogens. Here we identify IL-35-producing B cells as key players in the negative regulation of immunity. Mice in which only B cells did not express IL-35 lost their ability to recover from the T-cell-mediated demyelinating autoimmune disease experimental autoimmune encephalomyelitis (EAE). In contrast, these mice displayed a markedly improved resistance to infection with the intracellular bacterial pathogen Salmonella enterica serovar Typhimurium as shown by their superior containment of the bacterial growth and their prolonged survival after primary infection, and upon secondary challenge, compared to control mice. The increased immunity found in mice lacking IL-35 production by B cells was associated with a higher activation of macrophages and inflammatory T cells, as well as an increased function of B cells as antigen-presenting cells (APCs). During Salmonella infection, IL-35- and IL-10-producing B cells corresponded to two largely distinct sets of surface-IgM(+)CD138(hi)TACI(+)CXCR4(+)CD1d(int)Tim1(int) plasma cells expressing the transcription factor Blimp1 (also known as Prdm1). During EAE, CD138(+) plasma cells were also the main source of B-cell-derived IL-35 and IL-10. Collectively, our data show the importance of IL-35-producing B cells in regulation of immunity and highlight IL-35 production by B cells as a potential therapeutic target for autoimmune and infectious diseases. This study reveals the central role of activated B cells, particularly plasma cells, and their production of cytokines in the regulation of immune responses in health and disease.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Imunidade/imunologia , Interleucinas/metabolismo , Infecções por Salmonella/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD40/imunologia , Feminino , Humanos , Interleucina-10/metabolismo , Interleucinas/imunologia , Ativação Linfocitária , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Camundongos , Plasmócitos/imunologia , Plasmócitos/metabolismo , Infecções por Salmonella/microbiologia , Linfócitos T/imunologia , Receptor 4 Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...