Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1241474, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37638026

RESUMO

B cells are key pathogenic drivers of chronic inflammation in rheumatoid arthritis (RA). There is limited understanding of the relationship between synovial B cell subsets and pathogenic antibody secreting cells (ASCs). This knowledge is crucial for the development of more targeted B-cell depleting therapies. While CD11c+ double-negative 2 (DN2) B cells have been suggested as an ASC precursor in lupus, to date there is no proven link between the two subsets in RA. We have used both single-cell gene expression and BCR sequencing to study synovial B cells from patients with established RA, in addition to flow cytometry of circulating B cells. To better understand the differentiation patterns within the diseased tissue, a combination of RNA-based trajectory inference and clonal lineage analysis of BCR relationships were used. Both forms of analysis indicated that DN2 B cells serve as a major precursors to synovial ASCs. This study advances our understanding of B cells in RA and reveals the origin of pathogenic ASCs in the RA synovium. Given the significant role of DN2 B cells as a progenitor to pathogenic B cells in RA, it is important to conduct additional research to investigate the origins of DN2 B cells in RA and explore their potential as therapeutic targets in place of the less specific pan-B cells depletion therapies currently in use.


Assuntos
Artrite Reumatoide , Subpopulações de Linfócitos B , Humanos , Plasmócitos , Linfócitos B , Células Produtoras de Anticorpos
2.
Front Immunol ; 11: 395, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32265907

RESUMO

B cells are critical for promoting autoimmunity and the success of B cell depletion therapy in rheumatoid arthritis (RA) confirms their importance in driving chronic inflammation. Whilst disease specific autoantibodies are useful diagnostically, our understanding of the pathogenic B cell repertoire remains unclear. Defining it would lead to novel insights and curative treatments. To address this, we have undertaken the largest study to date of over 150 RA patients, utilizing next generation sequencing (NGS) to analyze up to 200,000 BCR sequences per patient. The full-length antigen-binding variable region of the heavy chain (IgGHV) of the IgG B cell receptor (BCR) were sequenced. Surprisingly, RA patients do not express particular clonal expansions of B cells at diagnosis. Rather they express a polyclonal IgG repertoire with a significant increase in BCRs that have barely mutated away from the germline sequence. This pattern remains even after commencing disease modifying therapy. These hypomutated BCRs are expressed by TNF-alpha secreting IgG+veCD27-ve B cells, that are expanded in RA peripheral blood and enriched in the rheumatoid synovium. A similar B cell repertoire is expressed by patients with Sjögren's syndrome. A rate limiting step in the initiation of autoimmunity is the activation of B cells and this data reveals that a sizeable component of the human autoimmune B cell repertoire consists of polyclonal, hypomutated IgG+ve B cells, that may play a critical role in driving chronic inflammation.


Assuntos
Artrite Reumatoide/imunologia , Autoimunidade , Linfócitos B/imunologia , Genes de Imunoglobulinas , Imunoglobulina G/genética , Subpopulações de Linfócitos/imunologia , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Autoanticorpos/imunologia , Linhagem da Célula , Células Clonais , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imunoglobulina G/análise , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Ativação Linfocitária , Receptores de Antígenos de Linfócitos B/genética , Síndrome de Sjogren/imunologia , Síndrome de Sjogren/patologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/análise , Fator de Necrose Tumoral alfa/metabolismo
3.
Oncoimmunology ; 8(8): 1608106, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31413918

RESUMO

Immunization of patients with autologous, ex vivo matured dendritic cell (DC) preparations, in order to prime antitumor T-cell responses, is the focus of intense research. Despite progress and approval of clinical approaches, significant enhancement of these personalized immunotherapies is urgently needed to improve efficacy. We show that immunotherapeutic murine and human DC, generated in the presence of the antimicrobial host defense peptide LL-37, have dramatically enhanced expansion and differentiation of cells with key features of the critical CD103+/CD141+ DC subsets, including enhanced cross-presentation and co-stimulatory capacity, and upregulation of CCR7 with improved migratory capacity. These LL-37-DC enhanced proliferation, activation and cytokine production by CD8+ (but not CD4+) T cells in vitro and in vivo. Critically, tumor antigen-presenting LL-37-DC increased migration of primed, activated CD8+ T cells into established squamous cell carcinomas in mice, and resulted in tumor regression. This advance therefore has the potential to dramatically enhance DC immunotherapy protocols.

4.
Inflamm Bowel Dis ; 24(10): 2113-2122, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29718255

RESUMO

Background: Due to common evolutionary origins, mitochondrial DNA (mtDNA) shares many similarities with immunogenic bacterial DNA. MtDNA is recognized as a pro-inflammatory damage-associated molecular pattern (DAMP) with a pathogenic role in several inflammatory diseases. We hypothesised that mtDNA is released during active disease, serving as a key pro-inflammatory factor in inflammatory bowel disease (IBD). Methods: Between 2014 and 2015, we collected plasma separated within 2 hours of sampling from 97 prospectively recruited IBD patients (67 ulcerative colitis [UC] and 30 Crohn's disease [CD]) and 40 non-IBD controls. We measured circulating mtDNA using quantitative polymerase chain reaction (amplifying mitochondria COXIII/ND2 genes) and also in mouse colitis induced by dextran sulfate-sodium (DSS). We used a mass spectometry approach to detect free plasma mitochondrial formylated peptides. Furthermore, we examined for mitochondrial damage using electron microscopy (EM) and TLR9 expression, the target for mtDNA, in human intestinal IBD mucosa. Results: Plasma mtDNA levels were increased in UC and CD (both P < 0.0001) compared with non-IBD controls. These levels were significantly correlated to blood (C-reactive protein, albumin, white cell count), clinical and endoscopic markers of severity, and disease activity. In active UC, we identified 5 mitochondrial formylated peptides (the most abundant being fMMYALF with known chemoattractant function) in plasma. We observed mitochondrial damage in inflamed UC mucosa and significantly higher fecal MtDNA levels (vs non-IBD controls [P < 0.0001]), which supports gut mucosal mitochondrial DAMP release as the primary source. In parallel, plasma mtDNA levels increased during induction of acute DSS colitis and were associated with more severe colitis (P < 0.05). In active IBD, TLR9+ lamina propria inflammatory cells were significantly higher in UC and CD compared with controls (P < 0.05). Conclusions: We present the first evidence to show that mtDNA is released during active IBD. MtDNA is a potential mechanistic biomarker, and our data point to mtDNA-TLR9 as a therapeutic target in IBD. 10.1093/ibd/izy095_videoizy095.video5776747659001.


Assuntos
Alarminas/metabolismo , Biomarcadores/análise , Colite Ulcerativa/patologia , Colite/patologia , Doença de Crohn/patologia , DNA Mitocondrial/genética , Adulto , Animais , Colite/induzido quimicamente , Colite/genética , Colite/metabolismo , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Doença de Crohn/genética , Doença de Crohn/metabolismo , Sulfato de Dextrana/toxicidade , Feminino , Seguimentos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos
5.
Eur J Immunol ; 47(4): 658-664, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28102569

RESUMO

Human ß-defensin 3 (hBD3) is a cationic antimicrobial peptide with potent bactericidal activity in vitro. HBD3 is produced in response to pathogen challenge and can modulate immune responses. The amplified recognition of self-DNA by human plasmacytoid dendritic cells has been previously reported, but we show here that hBD3 preferentially enhances the response to bacterial DNA in mouse Flt-3 induced dendritic cells (FLDCs) and in human peripheral blood mononuclear cells. We show the effect is mediated through TLR9 and although hBD3 significantly increases the cellular uptake of both E. coli and self-DNA in mouse FLDCs, only the response to bacterial DNA is enhanced. Liposome transfection also increases uptake of bacterial DNA and amplifies the TLR9-dependent response. In contrast to hBD3, lipofection of self-DNA enhances inflammatory signaling, but the response is predominantly TLR9-independent. Together, these data show that hBD3 has a role in the innate immune-mediated response to pathogen DNA, increasing inflammatory signaling and promoting activation of the adaptive immune system via antigen presenting cells including dendritic cells. Therefore, our data identify an additional immunomodulatory role for this copy-number variable defensin, of relevance to host defence against infection and indicate a potential for the inclusion of HBD3 in pathogen DNA-based vaccines.


Assuntos
Células Dendríticas/imunologia , Escherichia coli/imunologia , Leucócitos Mononucleares/imunologia , Receptor Toll-Like 9/metabolismo , beta-Defensinas/metabolismo , Animais , Células Cultivadas , DNA Bacteriano/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Toll-Like 9/genética
6.
Front Immunol ; 8: 1952, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29403471

RESUMO

The chronic autoimmune inflammatory diseases, systemic lupus erythematosus and Sjogren's syndrome, develop when tolerance to apoptotic cells (ACs) is lost. We have previously reported that this tolerance is maintained by innate-like, IL-10 secreting regulatory B cells. Two questions remained. First, do these regulatory B cells belong predominantly to a single subset of steady-state B cells and second, what is their specificity? We report here that innate-like B cells with markers characteristic for B1a cells (CD43+veCD19hiCD5+veIgMhiIgDlo) constitute 80% of splenic and 96% of peritoneal B cells that respond to ACs by secreting IL-10. AC responsive B1a cells secrete self-reactive natural antibodies (NAbs) and IL-10, which is augmented by toll-like receptor (TLR) 7 or TLR9 stimulation. In so doing, they both accelerate the clearance of dying cells by macrophages and inhibit their potential to mount proinflammatory immune responses. While B1a cells make prolonged contact with ACs, they do not require TIM1 or complement to mediate their regulatory function. In an animal model of neural inflammation (experimental autoimmune encephalomyelitis), just 105 activated B1a B cells was sufficient to restrain inflammation. Activated B1a B cells also induced antigen-specific T cells to secrete IL-10. Hence, regulatory B1a cells specifically recognize and augment tolerance to apoptotic self via IL-10 and NAbs; but once activated, can also prevent autoimmune mediated inflammation.

7.
Front Immunol ; 7: 590, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28018356

RESUMO

Plasmacytoid dendritic cells (pDCs) play a pivotal role in driving the autoimmune disease systemic lupus erythematosus, via the secretion of IFN-α in response to nuclear self-antigens complexed with autoantibodies. Apoptotic cells, generated at sites of inflammation or secondary lymphoid organs, are exposed to activated pDCs and also express the same nuclear antigens on their cell surface. Here, we show that in the absence of autoantibodies, activated pDCs directly respond to apoptotic cell-expressed chromatin complexes by secreting IL-10 and IL-6, which also induces T cells to secrete IL-10. Conversely, when activated by the viral mimetic CpG-A, apoptotic cells enhance their secretion of IFN-α. This study demonstrates that activated pDCs respond directly to apoptotic cells and may maintain tolerance via IL-10, or promote inflammation through secretion of IFN-α, depending on the inflammatory context.

8.
J Immunol ; 197(12): 4771-4779, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27807193

RESUMO

Neutrophils act as a first line of defense against bacterial and fungal infections, but they are also important effectors of acute and chronic inflammation. Genome-wide association studies have established that the gene encoding the protein tyrosine phosphatase nonreceptor 22 (PTPN22) makes an important contribution to susceptibility to autoimmune disease, notably rheumatoid arthritis. Although PTPN22 is most highly expressed in neutrophils, its function in these cells remains poorly characterized. We show in this article that neutrophil effector functions, including adhesion, production of reactive oxygen species, and degranulation induced by immobilized immune complexes, were reduced in Ptpn22-/- neutrophils. Tyrosine phosphorylation of Lyn and Syk was altered in Ptpn22-/- neutrophils. On stimulation with immobilized immune complexes, Ptpn22-/- neutrophils manifested reduced activation of key signaling intermediates. Ptpn22-/- mice were protected from immune complex-mediated arthritis, induced by the transfer of arthritogenic serum. In contrast, in vivo neutrophil recruitment following thioglycollate-induced peritonitis and in vitro chemotaxis were not affected by lack of PTPN22. Our data suggest an important role for PTPN22-dependent dephosphorylation events, which are required to enable full FcγR-induced activation, pointing to an important role for this molecule in neutrophil function.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/genética , Neutrófilos/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 22/metabolismo , Animais , Complexo Antígeno-Anticorpo , Artrite Experimental/genética , Adesão Celular , Degranulação Celular , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativação de Neutrófilo , Fosforilação , Polimorfismo Genético , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética , Espécies Reativas de Oxigênio/metabolismo , Receptores de IgG/metabolismo , Transdução de Sinais , Quinase Syk/metabolismo , Quinases da Família src/metabolismo
9.
Proc Natl Acad Sci U S A ; 113(16): 4350-5, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27044108

RESUMO

Neutrophils are the first and most numerous cells to arrive at the site of an inflammatory insult and are among the first to die. We previously reported that alpha defensins, released from apoptotic human neutrophils, augmented the antimicrobial capacity of macrophages while also inhibiting the biosynthesis of proinflammatory cytokines. In vivo, alpha defensin administration protected mice from inflammation, induced by thioglychollate-induced peritonitis or following infection withSalmonella entericaserovar Typhimurium. We have now dissected the antiinflammatory mechanism of action of the most abundant neutrophil alpha defensin, Human Neutrophil Peptide 1 (HNP1). Herein we show that HNP1 enters macrophages and inhibits protein translation without inducing the unfolded-protein response or affecting mRNA stability. In a cell-free in vitro translation system, HNP1 powerfully inhibited both cap-dependent and cap-independent mRNA translation while maintaining mRNA polysomal association. This is, to our knowledge, the first demonstration of a peptide released from one cell type (neutrophils) directly regulating mRNA translation in another (macrophages). By preventing protein translation, HNP1 functions as a "molecular brake" on macrophage-driven inflammation, ensuring both pathogen clearance and the resolution of inflammation with minimal bystander tissue damage.


Assuntos
Macrófagos/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Infecções por Salmonella/metabolismo , Salmonella typhimurium , alfa-Defensinas/metabolismo , Animais , Humanos , Macrófagos/patologia , Camundongos , Infecções por Salmonella/patologia , alfa-Defensinas/farmacologia
10.
Int Immunol ; 27(10): 505-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26306497

RESUMO

B cells are able to regulate immune responses through the secretion of IL-10 and other inhibitory cytokines, though no transcription factor that can define 'regulatory B cells' as a separate lineage has yet been found. Instead it is likely that this function arises as a result of the immune context in which B cells find themselves and the stimuli they perceive. However, some B cells found within the B1a and the marginal zone subsets have a greater propensity to produce IL-10 than others. What are the natural stimuli for these cells to induce immune regulation? We discuss the role that the recognition of autoantigens exposed by apoptotic cells plays in stimulating IL-10 production in mouse and human studies. This mechanism involves the recognition and uptake of self-antigens by autoreactive BCRs, for delivery to endocytic compartments, where apoptosis-derived DNA binds to TLR9, driving IL-10 production. These 'natural' regulatory B cells represent a way of maintaining tolerance to self. We discuss how this may operate in inflammatory lesions where there is an excess of apoptotic leukocytes and how this impacts on our understanding of autoimmune disease.


Assuntos
Artrite Reumatoide/imunologia , Linfócitos B Reguladores/imunologia , Linhagem da Célula/imunologia , Tolerância Imunológica , Interleucina-10/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Autoantígenos/genética , Autoantígenos/imunologia , Linfócitos B Reguladores/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/imunologia , Humanos , Interleucina-10/genética , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/imunologia , Transdução de Sinais , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia
11.
Nat Immunol ; 16(8): 819-828, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26147686

RESUMO

Fat-associated lymphoid clusters (FALCs) are a type of lymphoid tissue associated with visceral fat. Here we found that the distribution of FALCs was heterogeneous, with the pericardium containing large numbers of these clusters. FALCs contributed to the retention of B-1 cells in the peritoneal cavity through high expression of the chemokine CXCL13, and they supported B cell proliferation and germinal center differentiation during peritoneal immunological challenges. FALC formation was induced by inflammation, which triggered the recruitment of myeloid cells that expressed tumor-necrosis factor (TNF) necessary for signaling via the TNF receptors in stromal cells. Natural killer T cells (NKT cells) restricted by the antigen-presenting molecule CD1d were likewise required for the inducible formation of FALCs. Thus, FALCs supported and coordinated the activation of innate B cells and T cells during serosal immune responses.


Assuntos
Inflamação/imunologia , Gordura Intra-Abdominal/imunologia , Linfócitos/imunologia , Tecido Linfoide/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/imunologia , Quimiocina CXCL13/metabolismo , Citometria de Fluxo , Expressão Gênica/imunologia , Inflamação/genética , Inflamação/metabolismo , Gordura Intra-Abdominal/metabolismo , Linfócitos/metabolismo , Tecido Linfoide/citologia , Tecido Linfoide/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Células Mieloides/imunologia , Células Mieloides/metabolismo , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/imunologia , Células Estromais/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
12.
Ann Rheum Dis ; 74(1): 220-6, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24095938

RESUMO

INTRODUCTION: Inflammatory joint diseases such as rheumatoid arthritis are associated with local bone erosions and systemic bone loss, mediated by increased osteoclastic activity. The receptor activator of nuclear factor (NF) κB ligand (RANKL) plays a key role in mediating inflammation-induced bone loss, whereas tumour necrosis factor (TNF) plays a central role in the inflammatory process. Here we tested whether a recently identified class of small molecule inhibitors of RANKL signalling (ABD compounds) also affect TNF signalling and whether these compounds inhibit inflammation in an animal model of rheumatoid arthritis. METHODS: The inhibitory effects of the ABD compounds on TNF-induced signalling were tested in mouse macrophage cultures by western blotting and in an NFκB luciferase-reporter cell line. The anti-inflammatory effects of the compounds were tested in the mouse collagen-induced arthritis model of rheumatoid arthritis. RESULTS: The ABD compounds ABD328 and ABD345 both inhibited TNF-induced activation of the NFκB pathway and the extracellular signal-regulated kinase (ERK) and Jun kinase (JNK) mitogen activated protein kinases (MAPKs). When tested in the mouse collagen-induced arthritis model of rheumatoid arthritis, the compounds suppressed inflammatory arthritis, inhibited joint destruction and prevented systemic bone loss. Furthermore, one of the compounds (ABD328) showed oral activity. CONCLUSIONS: Here we describe a novel class of small molecule compounds that inhibit both RANKL- and TNF-induced NFκB and MAPK signalling in osteoclasts and macrophages, and inflammation and bone destruction in a mouse model of rheumatoid arthritis. These novel compounds therefore represent a promising new class of treatments for inflammatory diseases, such as rheumatoid arthritis.


Assuntos
Artrite Experimental/metabolismo , Artrite Reumatoide/metabolismo , Compostos de Bifenilo/farmacologia , Reabsorção Óssea/metabolismo , Hexanonas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Ligante RANK/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Necrose Tumoral/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Reabsorção Óssea/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , NF-kappa B/imunologia , NF-kappa B/metabolismo , Ligante RANK/imunologia , Ligante RANK/metabolismo , Fatores de Necrose Tumoral/imunologia
13.
J Immunol ; 193(6): 2984-93, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25114104

RESUMO

Helminth parasites remain one of the most common causes of infections worldwide, yet little is still known about the immune signaling pathways that control their expulsion. C57BL/6 mice are chronically susceptible to infection with the gastrointestinal helminth parasite Heligmosomoides polygyrus. In this article, we report that C57BL/6 mice lacking the adapter protein MyD88, which mediates signaling by TLRs and IL-1 family members, showed enhanced immunity to H. polygyrus infection. Alongside increased parasite expulsion, MyD88-deficient mice showed heightened IL-4 and IL-17A production from mesenteric lymph node CD4(+) cells. In addition, MyD88(-/-) mice developed substantial numbers of intestinal granulomas around the site of infection, which were not seen in MyD88-sufficient C57BL/6 mice, nor when signaling through the adapter protein TRIF (TIR domain-containing adapter-inducing IFN-ß adapter protein) was also ablated. Mice deficient solely in TLR2, TLR4, TLR5, or TLR9 did not show enhanced parasite expulsion, suggesting that these TLRs signal redundantly to maintain H. polygyrus susceptibility in wild-type mice. To further investigate signaling pathways that are MyD88 dependent, we infected IL-1R1(-/-) mice with H. polygyrus. This genotype displayed heightened granuloma numbers compared with wild-type mice, but without increased parasite expulsion. Thus, the IL-1R-MyD88 pathway is implicated in inhibiting granuloma formation; however, protective immunity in MyD88-deficient mice appears to be granuloma independent. Like IL-1R1(-/-) and MyD88(-/-) mice, animals lacking signaling through the type 1 IFN receptor (i.e., IFNAR1(-/-)) also developed intestinal granulomas. Hence, IL-1R1, MyD88, and type 1 IFN receptor signaling may provide pathways to impede granuloma formation in vivo, but additional MyD88-mediated signals are associated with inhibition of protective immunity in susceptible C57BL/6 mice.


Assuntos
Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Granuloma/genética , Granuloma/imunologia , Interleucina-17/biossíntese , Interleucina-4/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptores Tipo I de Interleucina-1/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Infecções por Strongylida/parasitologia , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Receptor 5 Toll-Like/genética , Receptor Toll-Like 9/genética
14.
J Immunol ; 191(6): 3128-38, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23935195

RESUMO

Long-lived plasma cells (LLPCs) that maintain humoral immunity to previously encountered Ags occupy a compartment in the bone marrow (BM). The rules and mechanisms by which cells enter (and leave) this compartment are poorly understood. We looked at what happens to the LLPC compartment and to plasma cell lifespan in general, in situations in which Ag stimulation and/or inflammation persist. We find that chronic Ag supply causes the generation of short-lived plasma cells in the local lymphoid organ, at the expense of any LLPC production. Furthermore, we find that inflammation caused by infection (mediated via TNF-α) causes a dramatic mobilization of LLPCs from the BM, with a concomitant reduction in circulating Ab levels against previously immunized Ags. These data are discussed in the context of the capacity of the BM LLPC compartment and competition for entry to it.


Assuntos
Homeostase/imunologia , Imunidade Humoral/imunologia , Inflamação/imunologia , Plasmócitos/imunologia , Animais , Antígenos/imunologia , Medula Óssea/imunologia , Quimiotaxia de Leucócito/imunologia , Citometria de Fluxo , Tecido Linfoide/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
PLoS One ; 8(1): e54640, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23349944

RESUMO

Mast cells are key initiators of allergic, anaphylactic and inflammatory reactions, producing mediators that affect vascular permeability, angiogenesis and fibrosis. Glucocorticoid pharmacotherapy reduces mast cell number, maturation and activation but effects at physiological levels are unknown. Within cells, glucocorticoid concentration is modulated by the 11ß-hydroxysteroid dehydrogenases (11ß-HSDs). Here we show expression and activity of 11ß-HSD1, but not 11ß-HSD2, in mouse mast cells with 11ß-HSD activity only in the keto-reductase direction, regenerating active glucocorticoids (cortisol, corticosterone) from inert substrates (cortisone, 11-dehydrocorticosterone). Mast cells from 11ß-HSD1-deficient mice show ultrastructural evidence of increased activation, including piecemeal degranulation and have a reduced threshold for IgG immune complex-induced mast cell degranulation. Consistent with reduced intracellular glucocorticoid action in mast cells, levels of carboxypeptidase A3 mRNA, a glucocorticoid-inducible mast cell-specific transcript, are lower in peritoneal cells from 11ß-HSD1-deficient than control mice. These findings suggest that 11ß-HSD1-generated glucocorticoids may tonically restrain mast cell degranulation, potentially influencing allergic, anaphylactic and inflammatory responses.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Corticosterona/biossíntese , Hidrocortisona/biossíntese , Mastócitos/enzimologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/deficiência , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Anafilaxia/enzimologia , Animais , Carboxipeptidases A/metabolismo , Degranulação Celular , Corticosterona/metabolismo , Expressão Gênica , Hidrocortisona/metabolismo , Hipersensibilidade/enzimologia , Inflamação/enzimologia , Camundongos
16.
Ann Rheum Dis ; 71(11): 1872-80, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22562975

RESUMO

OBJECTIVES: To establish the role and effect of glucocorticoids and the endogenous annexin A1 (AnxA1) pathway in inflammatory arthritis. METHODS: Ankle joint mRNA and protein expression of AnxA1 and its receptors were analysed in naive and arthritic mice by real-time PCR and immunohistochemistry. Inflammatory arthritis was induced with the K/BxN arthritogenic serum in AnxA1(+/+) and AnxA1(-/-) mice; in some experiments, animals were treated with dexamethasone (Dex) or with human recombinant AnxA1 or a protease-resistant mutant (termed SuperAnxA1). Readouts were arthritic score, disease incidence, paw oedema and histopathology, together with pro-inflammatory gene expression. RESULTS: All elements of the AnxA1 pathway could be detected in naive joints, with augmentation during ongoing disease, due to the infiltration of immune cells. No difference in arthritis intensity of profile could be observed between AnxA1(+/+) and AnxA1(-/-) mice. Treatment of mice with Dex (10 µg intraperitoneally daily from day 2) afforded potent antiarthritic effects highly attenuated in the knockouts: macroscopic changes were mirrored by histopathological findings and pro-inflammatory gene (eg, Nos2) expression. Presence of proteinase 3 mRNA in the arthritic joints led the authors to test AnxA1 and the mutant SuperAnxA1 (1 µg intraperitoneally daily in both cases from day 2), with the latter one being able to accelerate the resolving phase of the disease. CONCLUSION: AnxA1 is an endogenous determinant for the therapeutic efficacy of Dex in inflammatory arthritis. Such an effect can be partially mimicked by application of SuperAnxA1 which may represent the starting point for novel antiarthritic therapeutic strategies.


Assuntos
Anexina A1/fisiologia , Antirreumáticos/uso terapêutico , Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Dexametasona/uso terapêutico , Glucocorticoides/uso terapêutico , Animais , Anexina A1/química , Anexina A1/farmacologia , Artrite Experimental/genética , Artrite Experimental/metabolismo , Artrite Reumatoide/genética , Artrite Reumatoide/metabolismo , Modelos Animais de Doenças , Quimioterapia Combinada , Edema/tratamento farmacológico , Edema/patologia , Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteínas Mutantes/química , Proteínas Mutantes/farmacologia , Proteínas Recombinantes/farmacologia
17.
Infect Disord Drug Targets ; 12(3): 222-31, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22394172

RESUMO

B cells are once again gaining prominence as important programmers of CD4 T cell responses. With widespread use of B cell depletion therapy in the clinic, proving effective in treating diseases previously considered T cell-mediated, the time is right for a re-appraisal of the B cell. Though typically considered weak, Th2 driving APC, it is now clear that they are necessary for a robust and long-lived CD4 T cell response in many settings. The sphere of B cell influence extends well beyond that of simply antibody production; antigen presentation, cytokine secretion, costimulation and development of lymphoid tissue architecture are all critical aspects of B cell immunobiology, the absence of which has serious impacts for T cell priming and memory. The aim of this review is to look at non-antibody mediated B cell function and to ask how, where and when do B cells influence the CD4 T cell response?


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Animais , Formação de Anticorpos/imunologia , Citocinas/metabolismo , Humanos , Memória Imunológica , Células Th2/imunologia
18.
Proc Natl Acad Sci U S A ; 109(3): 887-92, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22207622

RESUMO

Intracellular protein complexes containing nucleic acids are common targets of autoantibodies in many autoimmune diseases. Central tolerance to these antigens is incomplete, yet nucleosomal DNA is expressed on the surface of cells dying by apoptosis. It is commonly believed that autoimmunity is prevented by the rapid uptake of apoptotic cells (ACs) by neighbors or professional phagocytes to which they deliver anti-inflammatory signals. Self-reactive, innate-like B cells contact and are selected by intracellular antigens expressed on ACs; however, how self-tolerance is maintained is not well understood. Here we report that IL-10 production by B cells, stimulated by contact with ACs, results from the engagement of Toll-like receptor 9 (TLR9) within the B cell after recognition of DNA-containing complexes on the surface of ACs. Until now, TLR9 ligation has been considered an inflammatory signal, but we have confirmed a hitherto unexpected immunoregulatory role by demonstrating the absence of the protective effect of ACs during experimental autoimmune encephalitis (EAE) in TLR9-deficient mice. Human circulating CD27(+) B cells also respond to DNA-bearing ACs, but not to DNase-treated cells, by secreting IL-10. Chronic autoimmune disease may arise if this tolerance mechanism is not reimposed after episodes of inflammation, or if the regulatory B-cell response is subverted.


Assuntos
Apoptose/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Comunicação Celular/imunologia , DNA/imunologia , Tolerância Imunológica/imunologia , Receptor Toll-Like 9/imunologia , Animais , Desoxirribonucleases/metabolismo , Humanos , Terapia de Imunossupressão , Interleucina-10/biossíntese , Interleucina-10/metabolismo , Camundongos , Muramidase/imunologia , Transdução de Sinais/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
19.
Endocrinology ; 153(1): 234-40, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22067318

RESUMO

Glucocorticoids profoundly influence immune responses, and synthetic glucocorticoids are widely used clinically for their potent antiinflammatory effects. Endogenous glucocorticoid action is modulated by the two isozymes of 11ß-hydroxysteroid dehydrogenase (11ß-HSD). In vivo, 11ß-HSD1 catalyzes the reduction of inactive cortisone or 11-dehydrocorticosterone into active cortisol or corticosterone, respectively, thereby increasing intracellular glucocorticoid levels. 11ß-HSD2 catalyzes the reverse reaction, inactivating intracellular glucocorticoids. Both enzymes have been postulated to modulate inflammatory responses. In the K/BxN serum transfer model of arthritis, 11ß-HSD1-deficient mice showed earlier onset and slower resolution of inflammation than wild-type controls, with greater exostoses in periarticular bone and, uniquely, ganglion cysts, consistent with greater inflammation. In contrast, K/BxN serum arthritis was unaffected by 11ß-HSD2 deficiency. In a distinct model of inflammation, thioglycollate-induced sterile peritonitis, 11ß-HSD1-deficient mice had more inflammatory cells in the peritoneum, but again 11ß-HSD2-deficient mice did not differ from controls. Additionally, compared with control mice, 11ß-HSD1-deficient mice showed greater numbers of inflammatory cells in pleural lavages in carrageenan-induced pleurisy with lung pathology consistent with slower resolution. These data suggest that 11ß-HSD1 limits acute inflammation. In contrast, 11ß-HSD2 plays no role in acute inflammatory responses in mice. Regulation of local 11ß-HSD1 expression and/or delivery of substrate may afford a novel approach for antiinflammatory therapy.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/deficiência , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Artrite Experimental/etiologia , Inflamação/etiologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Doença Aguda , Animais , Artrite Experimental/enzimologia , Artrite Experimental/patologia , Inflamação/enzimologia , Inflamação/genética , Articulações/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/etiologia , Peritonite/patologia , Pleurisia/etiologia , Pleurisia/patologia , Fatores de Tempo
20.
Eur J Immunol ; 40(10): 2677-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21038464

RESUMO

B cells are now acknowledged to play multiple roles in the immune response, in addition to making antibodies. Their role in regulating T-cell responses during inflammation has come into focus recently. Thus, IL-10 production by B cells has been shown to be important in limiting auto-reactive and pathogen-driven immune pathology; however, the exact identity of the regulatory B cells remains elusive; do they belong to a committed subset or can all B cells regulate given the appropriate inducing stimuli? A study in this issue of the European Journal of Immunology provides insight into the IL-10-producing B cells in humans, suggesting that many B cells have the capacity to make IL-10 when optimally stimulated via the BCR and TLR9. Despite producing significant amounts of inflammatory cytokines as well, these B cells suppress T-cell proliferation. This Commentary places this study in the context of what we think we know about regulatory B cells and more importantly highlights the questions we still need to answer.


Assuntos
Linfócitos B/imunologia , Interleucina-10/imunologia , Animais , Humanos , Ativação Linfocitária/imunologia , Camundongos , Receptores de Antígenos de Linfócitos B/imunologia , Receptores Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA