Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 582(7811): 259-264, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32499639

RESUMO

The synovium is a mesenchymal tissue composed mainly of fibroblasts, with a lining and sublining that surround the joints. In rheumatoid arthritis the synovial tissue undergoes marked hyperplasia, becomes inflamed and invasive, and destroys the joint1,2. It has recently been shown that a subset of fibroblasts in the sublining undergoes a major expansion in rheumatoid arthritis that is linked to disease activity3-5; however, the molecular mechanism by which these fibroblasts differentiate and expand is unknown. Here we identify a critical role for NOTCH3 signalling in the differentiation of perivascular and sublining fibroblasts that express CD90 (encoded by THY1). Using single-cell RNA sequencing and synovial tissue organoids, we found that NOTCH3 signalling drives both transcriptional and spatial gradients-emanating from vascular endothelial cells outwards-in fibroblasts. In active rheumatoid arthritis, NOTCH3 and Notch target genes are markedly upregulated in synovial fibroblasts. In mice, the genetic deletion of Notch3 or the blockade of NOTCH3 signalling attenuates inflammation and prevents joint damage in inflammatory arthritis. Our results indicate that synovial fibroblasts exhibit a positional identity that is regulated by endothelium-derived Notch signalling, and that this stromal crosstalk pathway underlies inflammation and pathology in inflammatory arthritis.


Assuntos
Artrite Reumatoide/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Receptor Notch3/metabolismo , Transdução de Sinais , Membrana Sinovial/patologia , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/patologia , Células Endoteliais/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Receptor Notch3/antagonistas & inibidores , Receptor Notch3/deficiência , Receptor Notch3/genética , Antígenos Thy-1/metabolismo
2.
Br J Cancer ; 130(10): 1659-1669, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38480935

RESUMO

BACKGROUND: Vestibular schwannomas (VSs) remain a challenge due to their anatomical location and propensity to growth. Macrophages are present in VS but their roles in VS pathogenesis remains unknown. OBJECTIVES: The objective was to assess phenotypic and functional profile of macrophages in VS with single-cell RNA sequencing (scRNAseq). METHODS: scRNAseq was carried out in three VS samples to examine characteristics of macrophages in the tumour. RT-qPCR was carried out on 10 VS samples for CD14, CD68 and CD163 and a panel of macrophage-associated molecules. RESULTS: scRNAseq revealed macrophages to be a major constituent of VS microenvironment with three distinct subclusters based on gene expression. The subclusters were also defined by expression of CD163, CD68 and IL-1ß. AREG and PLAUR were expressed in the CD68+CD163+IL-1ß+ subcluster, PLCG2 and NCKAP5 were expressed in CD68+CD163+IL-1ß- subcluster and AUTS2 and SPP1 were expressed in the CD68+CD163-IL-1ß+ subcluster. RT-qPCR showed expression of several macrophage markers in VS of which CD14, ALOX15, Interleukin-1ß, INHBA and Colony Stimulating Factor-1R were found to have a high correlation with tumour volume. CONCLUSIONS: Macrophages form an important component of VS stroma. scRNAseq reveals three distinct subsets of macrophages in the VS tissue which may have differing roles in the pathogenesis of VS.


Assuntos
Macrófagos , Neuroma Acústico , Análise de Sequência de RNA , Análise de Célula Única , Humanos , Neuroma Acústico/genética , Neuroma Acústico/patologia , Neuroma Acústico/metabolismo , Análise de Célula Única/métodos , Macrófagos/metabolismo , Macrófagos/patologia , Microambiente Tumoral/genética , Feminino , Masculino , Pessoa de Meia-Idade , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Antígenos de Diferenciação Mielomonocítica/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo
3.
Nature ; 542(7639): 110-114, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28150777

RESUMO

CD4+ T cells are central mediators of autoimmune pathology; however, defining their key effector functions in specific autoimmune diseases remains challenging. Pathogenic CD4+ T cells within affected tissues may be identified by expression of markers of recent activation. Here we use mass cytometry to analyse activated T cells in joint tissue from patients with rheumatoid arthritis, a chronic immune-mediated arthritis that affects up to 1% of the population. This approach revealed a markedly expanded population of PD-1hiCXCR5-CD4+ T cells in synovium of patients with rheumatoid arthritis. However, these cells are not exhausted, despite high PD-1 expression. Rather, using multidimensional cytometry, transcriptomics, and functional assays, we define a population of PD-1hiCXCR5- 'peripheral helper' T (TPH) cells that express factors enabling B-cell help, including IL-21, CXCL13, ICOS, and MAF. Like PD-1hiCXCR5+ T follicular helper cells, TPH cells induce plasma cell differentiation in vitro through IL-21 secretion and SLAMF5 interaction (refs 3, 4). However, global transcriptomics highlight differences between TPH cells and T follicular helper cells, including altered expression of BCL6 and BLIMP1 and unique expression of chemokine receptors that direct migration to inflamed sites, such as CCR2, CX3CR1, and CCR5, in TPH cells. TPH cells appear to be uniquely poised to promote B-cell responses and antibody production within pathologically inflamed non-lymphoid tissues.


Assuntos
Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Linfócitos B/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia , Artrite Reumatoide/sangue , Linfócitos B/patologia , Diferenciação Celular , Movimento Celular , Quimiocina CXCL13/metabolismo , Perfilação da Expressão Gênica , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interleucinas/metabolismo , Fatores Ativadores de Macrófagos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Receptor de Morte Celular Programada 1/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Receptores CXCR5/deficiência , Receptores CXCR5/metabolismo , Receptores de Quimiocinas/metabolismo , Proteínas Repressoras/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Líquido Sinovial/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo
4.
J Cell Sci ; 132(5)2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30745334

RESUMO

Mesenchymal stromal cells (MSCs) upregulate podoplanin at sites of infection, chronic inflammation and cancer. Here, we investigated the functional consequences of podoplanin expression on the migratory potential of MSCs and their interactions with circulating platelets. Expression of podoplanin significantly enhanced the migration of MSCs compared to MSCs lacking podoplanin. Rac-1 inhibition altered the membrane localisation of podoplanin and in turn significantly reduced MSC migration. Blocking Rac-1 activity had no effect on the migration of MSCs lacking podoplanin, indicating that it was responsible for regulation of migration through podoplanin. When podoplanin-expressing MSCs were seeded on the basal surface of a porous filter, they were able to capture platelets perfused over the uncoated apical surface and induce platelet aggregation. Similar microthrombi were observed when endothelial cells (ECs) were co-cultured on the apical surface. Confocal imaging shows podoplanin-expressing MSCs extending processes into the EC layer, and these processes could interact with circulating platelets. In both models, platelet aggregation induced by podoplanin-expressing MSCs was inhibited by treatment with recombinant soluble C-type lectin-like receptor 2 (CLEC-2; encoded by the gene Clec1b). Thus, podoplanin may enhance the migratory capacity of tissue-resident MSCs and enable novel interactions with cells expressing CLEC-2.


Assuntos
Plaquetas/fisiologia , Endotélio Vascular/fisiologia , Glicoproteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Trombose/metabolismo , Movimento Celular , Células Cultivadas , Endotélio Vascular/patologia , Humanos , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/genética , Microscopia Confocal , Comunicação Parácrina , Agregação Plaquetária , RNA Interferente Pequeno/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
5.
Ann Rheum Dis ; 78(2): 186-191, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30552174

RESUMO

OBJECTIVES: Controlled immune responses rely on integrated crosstalk between cells and their microenvironment. We investigated whether targeting proinflammatory signals from the extracellular matrix that persist during pathological inflammation provides a viable strategy to treat rheumatoid arthritis (RA). METHODS: Monoclonal antibodies recognising the fibrinogen-like globe (FBG) of tenascin-C were generated by phage display. Clones that neutralised FBG activation of toll-like receptor 4 (TLR4), without impacting pathogenic TLR4 activation, were epitope mapped by crystallography. Antibodies stained synovial biopsies of patients at different stages of RA development. Antibody efficacy in preventing RA synovial cell cytokine release, and in modulating collagen-induced arthritis in rats, was assessed. RESULTS: Tenascin-C is expressed early in the development of RA, even before disease diagnosis, with higher levels in the joints of people with synovitis who eventually developed RA than in people whose synovitis spontaneously resolved. Anti-FBG antibodies inhibited cytokine release by RA synovial cells and prevented disease progression and tissue destruction during collagen-induced arthritis. CONCLUSIONS: Early changes in the synovial microenvironment contribute to RA progression; blocking proinflammatory signals from the matrix can ameliorate experimental arthritis. These data highlight a new drug class that could offer early, disease-specific immune modulation in RA, without engendering global immune suppression.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Artrite Reumatoide/imunologia , Microambiente Celular/imunologia , Imunoterapia/métodos , Membrana Sinovial/imunologia , Animais , Anticorpos Monoclonais/imunologia , Artrite Experimental , Colágeno , Citocinas/metabolismo , Progressão da Doença , Fibrinogênio/imunologia , Humanos , Ratos , Tenascina/metabolismo , Receptor 4 Toll-Like/imunologia
6.
Eur J Immunol ; 45(8): 2299-311, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26036767

RESUMO

Soluble flagellin (sFliC) from Salmonella Typhimurium (STm) can induce a Th2 response to itself and coadministered antigens through ligation of TLR5. These properties suggest that sFliC could potentially modulate responses to Th1 antigens like live STm if both antigens are given concurrently. After coimmunization of mice with sFliC and STm there was a reduction in Th1 T cells (T-bet(+) IFN-γ(+) CD4 T cells) compared to STm alone and there was impaired clearance of STm. In contrast, there was no significant defect in the early extrafollicular B-cell response to STm. These effects are dependent upon TLR5 and flagellin expression by STm. The mechanism for these effects is not related to IL-4 induced to sFliC but rather to the effects of sFliC coimmunization on DCs. After coimmunization with STm and sFliC, splenic DCs had a lower expression of costimulatory molecules and profoundly altered kinetics of IL-12 and TNFα expression. Ex vivo experiments using in vivo conditioned DCs confirmed the effects of sFliC were due to altered DC function during a critical window in the coordinated interplay between DCs and naïve T cells. This has marked implications for understanding how limits in Th1 priming can be achieved during infection-induced, Th1-mediated inflammation.


Assuntos
Citocinas/imunologia , Células Dendríticas/imunologia , Flagelina/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Células Th1/imunologia , Animais , Citocinas/genética , Células Dendríticas/patologia , Flagelina/genética , Imunização , Camundongos , Camundongos Knockout , Infecções por Salmonella/genética , Infecções por Salmonella/prevenção & controle , Salmonella typhimurium/genética , Células Th1/patologia , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/imunologia
7.
Eur J Immunol ; 44(8): 2318-30, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24825601

RESUMO

The generation of immune cells from BM precursors is a carefully regulated process. This is essential to limit the potential for oncogenesis and autoimmunity yet protect against infection. How infection modulates this is unclear. Salmonella can colonize systemic sites including the BM and spleen. This resolving infection has multiple IFN-γ-mediated acute and chronic effects on BM progenitors, and during the first week of infection IFN-γ is produced by myeloid, NK, NKT, CD4(+) T cells, and some lineage-negative cells. After infection, the phenotype of BM progenitors rapidly but reversibly alters, with a peak ∼ 30-fold increase in Sca-1(hi) progenitors and a corresponding loss of Sca-1(lo/int) subsets. Most strikingly, the capacity of donor Sca-1(hi) cells to reconstitute an irradiated host is reduced; the longer donor mice are exposed to infection, and Sca-1(hi) c-kit(int) cells have an increased potential to generate B1a-like cells. Thus, Salmonella can have a prolonged influence on BM progenitor functionality not directly related to bacterial persistence. These results reflect changes observed in leucopoiesis during aging and suggest that BM functionality can be modulated by life-long, periodic exposure to infection. Better understanding of this process could offer novel therapeutic opportunities to modulate BM functionality and promote healthy aging.


Assuntos
Células da Medula Óssea/imunologia , Salmonelose Animal/imunologia , Células-Tronco/imunologia , Animais , Antígenos Ly/imunologia , Células da Medula Óssea/microbiologia , Células da Medula Óssea/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Homeostase/imunologia , Interferon gama/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Salmonella/imunologia , Salmonelose Animal/patologia , Células-Tronco/microbiologia , Células-Tronco/patologia
8.
J Immunol ; 189(12): 5745-54, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23152564

RESUMO

Mucosal immunity is poorly activated after systemic immunization with protein Ags. Nevertheless, induction of mucosal immunity in such a manner would be an attractive and simple way to overcome the intrinsic difficulties in delivering Ag to such sites. Flagellin from Salmonella enterica serovar Typhimurium (FliC) can impact markedly on host immunity, in part via its recognition by TLR5. In this study, we show that systemic immunization with soluble FliC (sFliC) drives distinct immune responses concurrently in the mesenteric lymph nodes (MLN) and the spleen after i.p. and s.c. immunization. In the MLN, but not the spleen, sFliC drives a TLR5-dependent recruitment of CD103(+) dendritic cells (DCs), which correlates with a diminution in CD103(+) DC numbers in the lamina propria. In the MLN, CD103(+) DCs carry Ag and are the major primers of endogenous and transgenic T cell priming. A key consequence of these interactions with CD103(+) DCs in the MLN is an increase in local regulatory T cell differentiation. In parallel, systemic sFliC immunization results in a pronounced switching of FliC-specific B cells to IgA in the MLN but not elsewhere. Loss of TLR5 has more impact on MLN than splenic Ab responses, reflected in an ablation of IgA, but not IgG, serum Ab titers. Therefore, systemic sFliC immunization targets CD103(+) DCs and drives distinct mucosal T and B cell responses. This offers a potential "Trojan horse" approach to modulate mucosal immunity by systemically immunizing with sFliC.


Assuntos
Antígenos CD/biossíntese , Células Dendríticas/imunologia , Flagelina/administração & dosagem , Flagelina/imunologia , Fatores de Transcrição Forkhead/biossíntese , Imunoglobulina A/biossíntese , Cadeias alfa de Integrinas/biossíntese , Linfonodos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Imunização , Linfonodos/citologia , Linfonodos/metabolismo , Mesentério , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mucosa/citologia , Mucosa/imunologia , Mucosa/metabolismo , Infecções por Salmonella/metabolismo , Infecções por Salmonella/patologia , Infecções por Salmonella/prevenção & controle , Salmonella typhimurium , Baço/citologia , Baço/imunologia , Baço/metabolismo , Linfócitos T Reguladores/metabolismo
9.
J Immunol ; 189(12): 5527-32, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23162127

RESUMO

Vaccination with purified capsular polysaccharide Vi Ag from Salmonella typhi can protect against typhoid fever, although the mechanism for its efficacy is not clearly established. In this study, we have characterized the B cell response to this vaccine in wild-type and T cell-deficient mice. We show that immunization with typhoid Vi polysaccharide vaccine rapidly induces proliferation in B1b peritoneal cells, but not in B1a cells or marginal zone B cells. This induction of B1b proliferation is concomitant with the detection of splenic Vi-specific Ab-secreting cells and protective Ab in Rag1-deficient B1b cell chimeras generated by adoptive transfer-induced specific Ab after Vi immunization. Furthermore, Ab derived from peritoneal B cells is sufficient to confer protection against Salmonella that express Vi Ag. Expression of Vi by Salmonella during infection did not inhibit the development of early Ab responses to non-Vi Ags. Despite this, the protection conferred by immunization of mice with porin proteins from Salmonella, which induce Ab-mediated protection, was reduced postinfection with Vi-expressing Salmonella, although protection was not totally abrogated. This work therefore suggests that, in mice, B1b cells contribute to the protection induced by Vi Ag, and targeting non-Vi Ags as subunit vaccines may offer an attractive strategy to augment current Vi-based vaccine strategies.


Assuntos
Antígenos de Bactérias/imunologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/microbiologia , Polissacarídeos Bacterianos/biossíntese , Salmonella typhi/imunologia , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/fisiologia , Antígenos de Bactérias/biossíntese , Subpopulações de Linfócitos B/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cavidade Peritoneal/citologia , Cavidade Peritoneal/microbiologia , Peritônio/citologia , Peritônio/imunologia , Peritônio/metabolismo , Polissacarídeos Bacterianos/imunologia , Porinas , Vacinas contra Salmonella/administração & dosagem , Vacinas contra Salmonella/biossíntese , Vacinas contra Salmonella/imunologia , Febre Tifoide/imunologia , Febre Tifoide/metabolismo , Febre Tifoide/prevenção & controle
10.
J Immunol ; 189(9): 4266-74, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22993205

RESUMO

Thymic atrophy is a frequent consequence of infection with bacteria, viruses, and parasites and is considered a common virulence trait between pathogens. Multiple reasons have been proposed to explain this atrophy, including premature egress of immature thymocytes, increased apoptosis, or thymic shutdown to prevent tolerance to the pathogen from developing. The severe loss in thymic cell number can reflect an equally dramatic reduction in thymic output, potentially reducing peripheral T cell numbers. In this study, we examine the relationship between systemic Salmonella infection and thymic function. During infection, naive T cell numbers in peripheral lymphoid organs increase. Nevertheless, this occurs despite a pronounced thymic atrophy caused by viable bacteria, with a peak 50-fold reduction in thymocyte numbers. Thymic atrophy is not dependent upon homeostatic feedback from peripheral T cells or on regulation of endogenous glucocorticoids, as demonstrated by infection of genetically altered mice. Once bacterial numbers fall, thymocyte numbers recover, and this is associated with increases in the proportion and proliferation of early thymic progenitors. During atrophy, thymic T cell maturation is maintained, and single-joint TCR rearrangement excision circle analysis reveals there is only a modest fall in recent CD4(+) thymic emigrants in secondary lymphoid tissues. Thus, thymic atrophy does not necessarily result in a matching dysfunctional T cell output, and thymic homeostasis can constantly adjust to systemic infection to ensure that naive T cell output is maintained.


Assuntos
Recuperação de Função Fisiológica/imunologia , Infecções por Salmonella/imunologia , Timo/imunologia , Timo/patologia , Animais , Atrofia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Camundongos , Infecções por Salmonella/patologia , Infecções por Salmonella/fisiopatologia , Salmonella typhimurium/imunologia , Timo/microbiologia
11.
J Immunol ; 186(4): 2365-71, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21217014

RESUMO

Nontyphoidal strains of Salmonella are a major cause of fatal bacteremia in Africa. Developing a vaccine requires an improved understanding of the relevant mechanisms of protective immunity, and the mouse model of Salmonella infection is useful for studying immunity to Salmonella in vivo. It is important to appreciate the similarities and differences between immunity to Salmonella in mice and men. Ab is important for protection against nontyphoidal Salmonella in both species, and we have previously found an important role for Ab in cell-free complement-mediated bactericidal activity against Salmonella in Africans. It is unclear whether this modality of immunity is relevant in the mouse model. C57BL/6, BALB/c, and C3H mice immunized with heat-killed Salmonella Typhimurium strains D23580 (African invasive strain) and SL1344 and live-attenuated strain SL3261 produced a Salmonella-specific Ab response. Sera from these mice deposited reduced levels of C3 on Salmonella compared with human sera and were unable to kill both wild-type and galE(-) rough mutant of D23580, indicating absent cell-free killing via classical and alternative complement pathways. Supplementing immune mouse sera with human complement enabled killing of Salmonella, whereas addition of human anti-Salmonella Ab to immune mouse sera had no effect. These findings indicate that mouse serum cannot effect [corrected] cell-free complement-dependent killing of Salmonella, because of the reduced mouse complement ability to kill these bacteria compared with human complement. This difference in Ab-dependent immunity to Salmonella in mice and men must be considered when applying findings from the mouse model of Salmonella disease and vaccination response to man.


Assuntos
Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/sangue , Atividade Bactericida do Sangue/imunologia , Proteínas do Sistema Complemento/deficiência , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Adulto , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Bacteriemia/epidemiologia , Bacteriemia/imunologia , Bacteriemia/microbiologia , Pré-Escolar , Via Clássica do Complemento/imunologia , Proteínas do Sistema Complemento/fisiologia , Modelos Animais de Doenças , Feminino , Humanos , Malaui/epidemiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Coelhos , Infecções por Salmonella/epidemiologia , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/classificação
12.
J Immunol ; 187(4): 1553-65, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21734076

RESUMO

Hematopoietic cells constitutively express CD31/PECAM1, a signaling adhesion receptor associated with controlling responses to inflammatory stimuli. Although expressed on CD4(+) T cells, its function on these cells is unclear. To address this, we have used a model of systemic Salmonella infection that induces high levels of T cell activation and depends on CD4(+) T cells for resolution. Infection of CD31-deficient (CD31KO) mice demonstrates that these mice fail to control infection effectively. During infection, CD31KO mice have diminished numbers of total CD4(+) T cells and IFN-γ-secreting Th1 cells. This is despite a higher proportion of CD31KO CD4(+) T cells exhibiting an activated phenotype and an undiminished capacity to prime normally and polarize to Th1. Reduced numbers of T cells reflected the increased propensity of naive and activated CD31KO T cells to undergo apoptosis postinfection compared with wild-type T cells. Using adoptive transfer experiments, we show that loss of CD31 on CD4(+) T cells alone is sufficient to account for the defective CD31KO T cell accumulation. These data are consistent with CD31 helping to control T cell activation, because in its absence, T cells have a greater propensity to become activated, resulting in increased susceptibility to become apoptotic. The impact of CD31 loss on T cell homeostasis becomes most pronounced during severe, inflammatory, and immunological stresses such as those caused by systemic Salmonella infection. This identifies a novel role for CD31 in regulating CD4 T cell homeostasis.


Assuntos
Apoptose/imunologia , Ativação Linfocitária/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Infecções por Salmonella/imunologia , Salmonella/imunologia , Células Th1/imunologia , Transferência Adotiva , Animais , Apoptose/genética , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Ativação Linfocitária/genética , Camundongos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Salmonella/genética , Infecções por Salmonella/genética
13.
Eur J Immunol ; 41(12): 3506-12, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21932446

RESUMO

Rapid production of neutralizing antibody can be critical for limiting the spread of infection. Such early antibody results when B-cell blasts mature directly to plasmablasts without forming germinal centers. These extrafollicular responses can involve Ig class switch recombination (CSR), producing antibody that can readily disseminate through infected tissues. The present study identifies the differentiation stage where CSR occurs in an extrafollicular response induced by 4-hydroxy-3-nitrophenyl acetyl (NP) conjugated to Ficoll (NP-Ficoll). To do this, we took advantage of the antigen dose dependency of CSR in this response. Thus, while both 30 and 1 µg NP-Ficoll induce plasmablasts, only the higher antigen dose induces CSR. Activation-induce cytidine deaminase (AID) is critical for CSR and in keeping with this a proportion of NP-specific B-cell blasts induced by 30 µg NP-Ficoll express AID. None of the B blasts responding to the non-CSR-inducing 1 µg dose of NP-Ficoll express AID. We confirmed that CSR occurs in B blasts by demonstrating the presence of rearranged heavy-chain transcripts in B blasts in the 30 µg response. CSR in this extrafollicular response is confined to B blasts, because NP-specific plasmablasts, identified by expressing CD138 and Blimp-1, no longer express AID and cannot undergo CSR.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Switching de Imunoglobulina/genética , Switching de Imunoglobulina/imunologia , Plasmócitos/imunologia , Animais , Citidina Desaminase/genética , Citidina Desaminase/imunologia , Ficoll/farmacologia , Imunoconjugados/imunologia , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Nitrofenóis/farmacologia , Fenilacetatos/farmacologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Sindecana-1/genética , Sindecana-1/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
14.
Eur J Immunol ; 41(9): 2654-65, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21630252

RESUMO

Control of intracellular Salmonella infection requires Th1 priming and IFN-γ production. Here, we show that efficient Th1 priming after Salmonella infection requires CD11c(+) CD11b(hi) F4/80(+) monocyte-derived dendritic cells (moDCs). In non-infected spleens, moDCs are absent from T-cell zones (T zones) of secondary lymphoid tissues, but by 24 h post-infection moDCs are readily discernible in these sites. The accumulation of moDCs is more dependent upon bacterial viability than bacterial virulence. Kinetic studies showed that moDCs were necessary to prime but not sustain Th1 responses, while ex vivo studies showed that antigen-experienced moDCs were sufficient to induce T-cell proliferation and IFN-γ production via a TNF-α-dependent mechanism. Importantly, moDCs and cDCs when co-cultured induced superior Th1 differentiation than either subset alone, and this activity was independent of TNF-α. Thus, optimal Th1 development to Salmonella requires the rapid accumulation of moDCs within T zones and their collaboration with cDCs.


Assuntos
Células Dendríticas/metabolismo , Infecções por Salmonella/imunologia , Salmonella/imunologia , Baço/patologia , Células Th1/metabolismo , Animais , Apresentação de Antígeno , Antígenos de Diferenciação/biossíntese , Antígeno CD11b/biossíntese , Antígeno CD11c/biossíntese , Comunicação Celular , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Interferon gama/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/patologia , Salmonella/patogenicidade , Células Th1/imunologia , Células Th1/microbiologia , Células Th1/patologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Eur J Immunol ; 41(6): 1606-18, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21469112

RESUMO

Clearance of disseminated Salmonella infection requires bacterial-specific Th1 cells and IFN-γ production, and Th1-promoting vaccines are likely to help control these infections. Consequently, vaccine design has focused on developing Th1-polarizing adjuvants or Ag that naturally induce Th1 responses. In this study, we show that, in mice, immunization with soluble, recombinant FliC protein flagellin (sFliC) induces Th2 responses as evidenced by Ag-specific GATA-3, IL-4 mRNA, and protein induction in CD62L(lo) CD4(+) T cells without associated IFN-γ production. Despite these Th2 features, sFliC immunization can enhance the development of protective Th1 immunity during subsequent Salmonella infection in an Ab-independent, T-cell-dependent manner. Salmonella infection in sFliC-immunized mice resulted in augmented Th1 responses, with greater bacterial clearance and increased numbers of IFN-γ-producing CD4(+) T cells, despite the early induction of Th2 features to sFliC. The augmented Th1 immunity after sFliC immunization was regulated by T-bet although T-bet is dispensable for primary responses to sFliC. These findings show that there can be flexibility in T-cell responses to some subunit vaccines. These vaccines may induce Th2-type immunity during primary immunization yet promote Th1-dependent responses during later infection. This suggests that designing Th1-inducing subunit vaccines may not always be necessary since this can occur naturally during subsequent infection.


Assuntos
Vacinas Bacterianas , Flagelina/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Proteínas com Domínio T/metabolismo , Células Th1/metabolismo , Células Th2/metabolismo , Animais , Carga Bacteriana , Células Cultivadas , Regulação da Expressão Gênica , Imunização , Interferon gama/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Salmonella/microbiologia , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/patogenicidade , Proteínas com Domínio T/genética , Proteínas com Domínio T/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T , Células Th1/imunologia , Células Th1/microbiologia , Células Th1/patologia , Células Th2/imunologia , Células Th2/microbiologia , Células Th2/patologia
16.
Proc Natl Acad Sci U S A ; 106(24): 9803-8, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19487686

RESUMO

Invasive nontyphoidal Salmonella (NTS), including Salmonella typhimurium (STm), are major yet poorly-recognized killers of infants in sub-Saharan Africa. Death in these children is usually associated with bacteremia, commonly in the absence of gastrointestinal symptoms. Evidence from humans and animal studies suggest that severe infection and bacteremia occur when specific Ab is lacking. Understanding how Ab responses to Salmonella are regulated will help develop vaccines against these devastating infections. STm induces atypical Ab responses characterized by prominent, accelerated, extrafollicular T-independent (TI) Ab against a range of surface antigens. These responses develop without concomitant germinal centers, which only appear as infection resolves. Here, we show STm rapidly induces a population of TI B220(+)CD5(-) B1b cells during infection and TI Ab from B1b cells targets the outer membrane protein (Omp) porins OmpC, OmpD and OmpF but not flagellin. When porins are used as immunogens they can ablate bacteremia and provide equivalent protection against STm as killed bacterial vaccine and this is wholly B cell-dependent. Furthermore Ab from porin-immunized chimeras, that have B1b cells, is sufficient to impair infection. Infecting with porin-deficient bacteria identifies OmpD, a protein absent from Salmonella Typhi, as a key target of Ab in these infections. This work broadens the recognized repertoire of TI protein antigens and highlights the importance of Ab from different B cell subsets in controlling STm infection. OmpD is a strong candidate vaccine target and may, in part, explain the lack of cross-protection between Salmonella Typhi and STm infections.


Assuntos
Anticorpos Antibacterianos/biossíntese , Porinas/imunologia , Salmonella/metabolismo , Animais , Linfócitos B/citologia , Sequência de Bases , Western Blotting , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Cavidade Peritoneal/citologia , Salmonella/imunologia
17.
Med ; 3(7): 481-518.e14, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35649411

RESUMO

BACKGROUND: Pro-inflammatory fibroblasts are critical for pathogenesis in rheumatoid arthritis, inflammatory bowel disease, interstitial lung disease, and Sjögren's syndrome and represent a novel therapeutic target for chronic inflammatory disease. However, the heterogeneity of fibroblast phenotypes, exacerbated by the lack of a common cross-tissue taxonomy, has limited our understanding of which pathways are shared by multiple diseases. METHODS: We profiled fibroblasts derived from inflamed and non-inflamed synovium, intestine, lungs, and salivary glands from affected individuals with single-cell RNA sequencing. We integrated all fibroblasts into a multi-tissue atlas to characterize shared and tissue-specific phenotypes. FINDINGS: Two shared clusters, CXCL10+CCL19+ immune-interacting and SPARC+COL3A1+ vascular-interacting fibroblasts, were expanded in all inflamed tissues and mapped to dermal analogs in a public atopic dermatitis atlas. We confirmed these human pro-inflammatory fibroblasts in animal models of lung, joint, and intestinal inflammation. CONCLUSIONS: This work represents a thorough investigation into fibroblasts across organ systems, individual donors, and disease states that reveals shared pathogenic activation states across four chronic inflammatory diseases. FUNDING: Grant from F. Hoffmann-La Roche (Roche) AG.


Assuntos
Artrite Reumatoide , Membrana Sinovial , Animais , Artrite Reumatoide/genética , Fibroblastos/metabolismo , Fenótipo , Células Estromais/metabolismo
18.
Front Immunol ; 12: 725641, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34512657

RESUMO

Fibroblast-like synoviocytes (FLS) play an important role in maintaining joint homeostasis and orchestrating local inflammatory processes. When activated during injury or inflammation, FLS undergo transiently increased bioenergetic and biosynthetic demand. We aimed to identify metabolic changes which occur early in inflammatory disease pathogenesis which might support sustained cellular activation in persistent inflammation. We took primary human FLS from synovial biopsies of patients with very early rheumatoid arthritis (veRA) or resolving synovitis, and compared them with uninflamed control samples from the synovium of people without arthritis. Metabotypes were compared using NMR spectroscopy-based metabolomics and correlated with serum C-reactive protein levels. We measured glycolysis and oxidative phosphorylation by Seahorse analysis and assessed mitochondrial morphology by immunofluorescence. We demonstrate differences in FLS metabolism measurable after ex vivo culture, suggesting that disease-associated metabolic changes are long-lasting. We term this phenomenon 'metabolic memory'. We identify changes in cell metabolism after acute TNFα stimulation across disease groups. When compared to FLS from patients with early rheumatoid arthritis, FLS from patients with resolving synovitis have significantly elevated mitochondrial respiratory capacity in the resting state, and less fragmented mitochondrial morphology after TNFα treatment. Our findings indicate the potential to restore cell metabotypes by modulating mitochondrial function at sites of inflammation, with implications for treatment of RA and related inflammatory conditions in which fibroblasts play a role.


Assuntos
Artrite Reumatoide/imunologia , Fibroblastos/imunologia , Inflamação/imunologia , Sinoviócitos/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Idoso , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Pessoa de Meia-Idade , Fosforilação Oxidativa , Análise de Regressão , Sinoviócitos/metabolismo , Sinoviócitos/patologia , Fator de Necrose Tumoral alfa/genética
19.
Nat Commun ; 11(1): 851, 2020 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-32051408

RESUMO

Lipopolysaccharide (LPS) O-antigen (O-Ag) is known to limit antibody binding to surface antigens, although the relationship between antibody, O-Ag and other outer-membrane antigens is poorly understood. Here we report, immunization with the trimeric porin OmpD from Salmonella Typhimurium (STmOmpD) protects against infection. Atomistic molecular dynamics simulations indicate this is because OmpD trimers generate footprints within the O-Ag layer sufficiently sized for a single IgG Fab to access. While STmOmpD differs from its orthologue in S. Enteritidis (SEn) by a single amino-acid residue, immunization with STmOmpD confers minimal protection to SEn. This is due to the OmpD-O-Ag interplay restricting IgG binding, with the pairing of OmpD with its native O-Ag being essential for optimal protection after immunization. Thus, both the chemical and physical structure of O-Ag are key for the presentation of specific epitopes within proteinaceous surface-antigens. This enhances combinatorial antigenic diversity in Gram-negative bacteria, while reducing associated fitness costs.


Assuntos
Anticorpos Antibacterianos/imunologia , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/imunologia , Imunização , Antígenos O/imunologia , Salmonella typhimurium/imunologia , Animais , Anticorpos Antibacterianos/sangue , Formação de Anticorpos , Especificidade de Anticorpos , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteção Cruzada , Modelos Animais de Doenças , Epitopos/química , Epitopos/imunologia , Imunoglobulina G/sangue , Camundongos , Modelos Moleculares , Antígenos O/química , Antígenos O/genética , Porinas/química , Porinas/genética , Porinas/imunologia , Conformação Proteica , Salmonelose Animal/imunologia , Salmonelose Animal/prevenção & controle , Análise de Sequência de Proteína
20.
Cell Rep ; 27(5): 1461-1471.e4, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-31042473

RESUMO

B cell lymphoma-6 (BCL6) is highly expressed in germinal center B cells, but how its expression is maintained is still not completely clear. Aryl hydrocarbon receptor interacting protein (AIP) is a co-chaperone of heat shock protein 90. Deletion of Aip in B cells decreased BCL6 expression, reducing germinal center B cells and diminishing adaptive immune responses. AIP was required for optimal AKT signaling in response to B cell receptor stimulation, and AIP protected BCL6 from ubiquitin-mediated proteasomal degradation by the E3-ubiquitin ligase FBXO11 by binding to the deubiquitinase UCHL1, thus helping to maintain the expression of BCL6. AIP was highly expressed in primary diffuse large B cell lymphomas compared to healthy tissue and other tumors. Our findings describe AIP as a positive regulator of BCL6 expression with implications for the pathobiology of diffuse large B cell lymphoma.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfoma Difuso de Grandes Células B/imunologia , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Proteínas F-Box/metabolismo , Feminino , Centro Germinativo/citologia , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Proteína-Arginina N-Metiltransferases/metabolismo , Proteólise , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA