Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 12(5): 1201-1211, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31417161

RESUMO

Uncontrolled interferon γ (IFNγ)-mediated T-cell responses to commensal microbiota are a driver of inflammatory bowel disease (IBD). Interleukin-10 (IL-10) is crucial for controlling these T-cell responses, but the precise mechanism of inhibition remains unclear. A better understanding of how IL-10 exerts its suppressive function may allow identification of individuals with suboptimal IL-10 function among the heterogeneous population of IBD patients. Using cells from patients with an IL10RA deficiency or STAT3 mutations, we demonstrate that IL-10 signaling in monocyte-derived dendritic cells (moDCs), but not T cells, is essential for controlling IFNγ-secreting CD4+ T cells. Deficiency in IL-10 signaling dramatically increased IL-1ß release by moDCs. IL-1ß boosted IFNγ secretion by CD4+ T cells either directly or indirectly by stimulating moDCs to secrete IL-12. As predicted a signature of IL-10 dysfunction was observed in a subgroup of pediatric IBD patients having higher IL-1ß expression in activated immune cells and macroscopically affected intestinal tissue. In agreement, reduced IL10RA expression was detected in peripheral blood mononuclear cells and a subgroup of pediatric IBD patients exhibited diminished IL-10 responsiveness. Our data unveil an important mechanism by which IL-10 controls IFNγ-secreting CD4+ T cells in humans and identifies IL-1ß as a potential classifier for a subgroup of IBD patients.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Transdução de Sinais , Adolescente , Comunicação Celular , Criança , Suscetibilidade a Doenças , Humanos , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/terapia
2.
Mucosal Immunol ; 12(2): 479-490, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30542112

RESUMO

Breach of tolerance to gluten leads to the chronic small intestinal enteropathy celiac disease. A key event in celiac disease development is gluten-dependent infiltration of activated cytotoxic intraepithelial lymphocytes (IELs), which cytolyze epithelial cells causing crypt hyperplasia and villous atrophy. The mechanisms leading to gluten-dependent small intestinal IEL infiltration and activation remain elusive. We have demonstrated that under homeostatic conditions in mice, gluten drives the differentiation of anti-inflammatory T cells producing large amounts of the immunosuppressive cytokine interleukin-10 (IL-10). Here we addressed whether this dominant IL-10 axis prevents gluten-dependent infiltration of activated cytotoxic IEL and subsequent small intestinal enteropathy. We demonstrate that IL-10 regulation prevents gluten-induced cytotoxic inflammatory IEL infiltration. In particular, IL-10 suppresses gluten-induced accumulation of a specialized population of cytotoxic CD4+CD8αα+ IEL (CD4+ CTL) expressing Tbx21, Ifng, and Il21, and a disparate non-cytolytic CD4+CD8α- IEL population expressing Il17a, Il21, and Il10. Concomitantly, IL-10 suppresses gluten-dependent small intestinal epithelial hyperproliferation and upregulation of stress-induced molecules on epithelial cells. Remarkably, frequencies of granzyme B+CD4+CD8α+ IEL are increased in pediatric celiac disease patient biopsies. These findings demonstrate that IL-10 is pivotal to prevent gluten-induced small intestinal inflammation and epithelial damage, and imply that CD4+ CTL are potential new players into these processes.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Doença Celíaca/imunologia , Interleucina-10/metabolismo , Mucosa Intestinal/imunologia , Linfócitos Intraepiteliais/imunologia , Animais , Morte Celular , Diferenciação Celular , Movimento Celular , Criança , Citotoxicidade Imunológica , Glutens/imunologia , Granzimas/metabolismo , Homeostase , Humanos , Tolerância Imunológica , Interleucina-10/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
3.
Clin Exp Immunol ; 192(2): 224-232, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29319177

RESUMO

We hypothesize that T cells such as interleukin (IL)-21+ B cell lymphoma 6 (BCL6)+ T follicular helper cells can regulate B cell-mediated immunity within the allograft during acute T cell-mediated rejection; this process may feed chronic allograft rejection in the long term. To investigate this mechanism, we determined the presence and activation status of organized T and B cells in so-called ectopic lymphoid structures (ELSs) in different types of acute renal allograft rejection. Biopsies showing the following primary diagnosis were included: acute/active antibody-mediated rejection, C4d+ (a/aABMR), acute T cell-mediated rejection grade I (aTCMRI) and acute T cell-mediated rejection grade II (aTCMRII). Paraffin sections were stained for T cells (CD3 and CD4), B cells (CD20), follicular dendritic cells (FDCs, CD23), activated B cells (CD79A), immunoglobulin (Ig)D, cell proliferation (Ki67) and double immunofluorescent stainings for IL-21 and BCL6 were performed. Infiltrates of T cells were detected in all biopsies. In aTCMRI, B cells formed aggregates surrounded by T cells. In these aggregates, FDCs, IgD and Ki67 were detected, suggesting the presence of ELSs. In contrast, a/aABMR and aTCMRII showed diffuse infiltrates of T and B cells but no FDCs and IgD. IL-21 was present in all biopsies. However, co-localization with BCL6 was observed mainly in aTCMRI biopsies. In conclusion, ELSs with an activated phenotype are found predominantly in aTCMRI where T cells co-localize with B cells. These findings suggest a direct pathway of B cell alloactivation at the graft site during T cell mediated rejection.


Assuntos
Linfócitos B/imunologia , Rejeição de Enxerto/imunologia , Transplante de Rim , Linfócitos T Auxiliares-Indutores/imunologia , Estruturas Linfoides Terciárias/imunologia , Adulto , Idoso , Biópsia , Células Dendríticas Foliculares/imunologia , Feminino , Humanos , Interleucinas/metabolismo , Antígeno Ki-67/análise , Rim/patologia , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Estudos Retrospectivos , Transplante Homólogo
4.
Paediatr Drugs ; 20(1): 19-28, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29079905

RESUMO

Antibodies directed to tumour necrosis factor-α (TNF-α) are very effective in treating paediatric Crohn's disease (CD). Over the last few years, research has provided important new insights into how to optimise this treatment's effectiveness. Research on predictors for anti-TNF treatment responsiveness has revealed potential markers, but data on their accuracy in paediatric CD patients are lagging behind. Also, new evidence has become available on the safety profile of anti-TNF antibodies that suggests the assumed increased malignancy risk seen in patients on anti-TNF and thiopurine combination treatment may be linked more to thiopurine use and not to anti-TNF treatment. In addition, the early results of CT-P13, an infliximab biosimilar, in CD patients confirm the expected similarity with its originator. Thus, the effectiveness of anti-TNF antibody treatment is slowly improving, its malignancy risk is lower than assumed, and its costs are reduced by the introduction of equally effective biosimilars. Together, these trends allow for a more prominent role for anti-TNF antibodies in future treatment of paediatric CD.


Assuntos
Medicamentos Biossimilares/uso terapêutico , Doença de Crohn/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adolescente , Medicamentos Biossimilares/farmacologia , Criança , Pré-Escolar , Doença de Crohn/patologia , Humanos , Resultado do Tratamento
5.
Nat Commun ; 8(1): 620, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931816

RESUMO

CD103+CD11b+ dendritic cells (DCs) are unique to the intestine, but the factors governing their differentiation are unclear. Here we show that transforming growth factor receptor 1 (TGFßR1) has an indispensable, cell intrinsic role in the development of these cells. Deletion of Tgfbr1 results in markedly fewer intestinal CD103+CD11b+ DCs and a reciprocal increase in the CD103-CD11b+ dendritic cell subset. Transcriptional profiling identifies markers that define the CD103+CD11b+ DC lineage, including CD101, TREM1 and Siglec-F, and shows that the absence of CD103+CD11b+ DCs in CD11c-Cre.Tgfbr1 fl/fl mice reflects defective differentiation from CD103-CD11b+ intermediaries, rather than an isolated loss of CD103 expression. The defect in CD103+CD11b+ DCs is accompanied by reduced generation of antigen-specific, inducible FoxP3+ regulatory T cells in vitro and in vivo, and by reduced numbers of endogenous Th17 cells in the intestinal mucosa. Thus, TGFßR1-mediated signalling may explain the tissue-specific development of these unique DCs.Developmental cues for the different dendritic cell (DC) subsets in the intestine are yet to be defined. Here the authors show that TGFßR1 signalling is needed for development of CD103+CD11b+ intestinal DCs from CD103-CD11b+ cells and that they contribute to the generation of Th17 and regulatory T cells.


Assuntos
Diferenciação Celular/genética , Células Dendríticas/imunologia , Mucosa Intestinal/imunologia , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Antígenos CD/imunologia , Antígeno CD11b/imunologia , Linhagem da Célula , Colite/imunologia , Células Dendríticas/citologia , Imunidade nas Mucosas , Cadeias alfa de Integrinas/imunologia , Mucosa Intestinal/citologia , Intestinos/citologia , Intestinos/imunologia , Linfopoese/genética , Camundongos , Camundongos Knockout , Receptor do Fator de Crescimento Transformador beta Tipo I , Linfócitos T Reguladores/citologia , Células Th17/citologia
6.
Mucosal Immunol ; 10(3): 635-649, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27579860

RESUMO

Celiac disease is caused by inflammatory T-cell responses against the insoluble dietary protein gliadin. We have shown that, in humanized mice, oral tolerance to deamidated chymotrypsin-digested gliadin (CT-TG2-gliadin) is driven by tolerogenic interferon (IFN)-γ- and interleukin (IL)-10-secreting type 1 regulatory T-like cells (Tr1-like cells) generated in the spleen but not in the mesenteric lymph nodes. We aimed to uncover the mechanisms underlying gliadin-specific Tr1-like-cell differentiation and hypothesized that proteolytic gliadin degradation by splenic macrophages is a decisive step in this process. In vivo depletion of macrophages caused reduced differentiation of splenic IFN-γ- and IL-10-producing Tr1-like cells after CT-TG2-gliadin but not gliadin peptide feed. Splenic macrophages, rather than dendritic cells, constitutively expressed increased mRNA levels of the endopeptidase Cathepsin D; macrophage depletion significantly reduced splenic Cathepsin D expression in vivo and Cathepsin D efficiently degraded recombinant γ-gliadin in vitro. In response to CT-TG2-gliadin uptake, macrophages enhanced the expression of Il27p28, a cytokine that favored differentiation of gliadin-specific Tr1-like cells in vitro, and was previously reported to increase Cathepsin D activity. Conversely, IL-27 neutralization in vivo inhibited splenic IFN-γ- and IL-10-secreting Tr1-like-cell differentiation after CT-TG2-gliadin feed. Our data infer that endopeptidase mediated gliadin degradation by macrophages and concomitant IL-27 production drive differentiation of splenic gliadin-specific Tr1-like cells.


Assuntos
Doença Celíaca/imunologia , Gliadina/metabolismo , Interleucina-27/metabolismo , Macrófagos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Neutralizantes/metabolismo , Catepsina E/metabolismo , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Glutens/imunologia , Antígenos HLA-DQ/genética , Humanos , Tolerância Imunológica , Interferon gama/metabolismo , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos SCID , Proteólise , Receptores de Antígenos de Linfócitos T/genética , Células Th1/imunologia
7.
Mucosal Immunol ; 9(4): 894-906, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26577569

RESUMO

Tolerance to harmless exogenous antigens is the default immune response in the gastrointestinal tract. Although extensive studies have demonstrated the importance of the mesenteric lymph nodes (MLNs) and intestinal CD103(+) dendritic cells (DCs) in driving small intestinal tolerance to protein antigen, the structural and immunological basis of colonic tolerance remain poorly understood. We show here that the caudal and iliac lymph nodes (ILNs) are inductive sites for distal colonic immune responses and that colonic T cell-mediated tolerance induction to protein antigen is initiated in these draining lymph nodes and not in MLNs. In agreement, colonic tolerance induction was not altered by mesenteric lymphadenectomy. Despite tolerance development, CD103(+)CD11b(+) DCs, which are the major migratory DC population in the MLNs, and the tolerance-related retinoic acid-generating enzyme RALDH2 were virtually absent from the ILNs. Administration of ovalbumin (OVA) to the distal colon did increase the number of CD11c(+)MHCII(hi) migratory CD103(-)CD11b(+) and CD103(+)CD11b(-) DCs in the ILNs. Strikingly, colonic tolerance was intact in Batf3-deficient mice specifically lacking CD103(+)CD11b(-) DCs, suggesting that CD103(-) DCs in the ILNs are sufficient to drive tolerance induction after protein antigen encounter in the distal colon. Altogether, we identify different inductive sites for small intestinal and colonic T-cell responses and reveal that distinct cellular mechanisms are operative to maintain tolerance at these sites.


Assuntos
Colo/imunologia , Células Dendríticas/imunologia , Intestino Delgado/imunologia , Linfonodos/imunologia , Linfócitos T/imunologia , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Animais , Antígenos CD/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Antígeno CD11b/metabolismo , Feminino , Veia Ilíaca/anatomia & histologia , Tolerância Imunológica , Cadeias alfa de Integrinas/metabolismo , Excisão de Linfonodo , Linfonodos/anatomia & histologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Repressoras/genética
8.
BMJ Open Gastroenterol ; 3(1): e000123, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28090335

RESUMO

INTRODUCTION: Crohn's disease (CD) is a chronic inflammatory disease predominantly affecting the gastrointestinal tract. CD usually requires lifelong medication and is accompanied by severe complications, such as fistulae and strictures, resulting in surgery. Infliximab (IFX) is very effective for treating paediatric patients with CD, but is currently only registered for therapy refractory patients-the so-called step-up strategy. We hypothesise that using IFX first-line, that is, top-down, will give more mucosal healing, fewer relapses, less complications, need for surgery and hospitalisation. METHODS AND ANALYSIS: This international multicentre open-label randomised controlled trial includes children, aged 3-17 years, with new-onset, untreated CD with moderate-to-severe disease activity (weighted Paediatric Crohn's Disease Activity Index (wPCDAI)>40). Eligible patients will be randomised to top-down or step-up treatment. Top-down treatment consists of 5 IFX infusions combined with azathioprine (AZA). After these 5 infusions, patients will continue AZA. Patients randomised to step-up will receive standard induction treatment, either oral prednisolone or exclusive enteral nutrition, combined with AZA as maintenance treatment. The primary outcome is clinical remission (wPCDAI<12.5) at 52 weeks without need for additional CD-related therapy or surgery. Total follow-up is 5 years. Secondary outcomes include clinical disease activity, mucosal healing by endoscopy (at week 10 and optionally week 52), faecal calprotectin, growth, quality of life, medication use and adverse events. ETHICS AND DISSEMINATION: Conducted according to the Declaration of Helsinki and Good Clinical Practice. Medical-ethical approval will be obtained for each site. TRIAL REGISTRATION NUMBER: NCT02517684; Pre-results.

10.
Mucosal Immunol ; 6(6): 1202-13, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23571506

RESUMO

Celiac disease (CD) is caused by inflammatory CD4(+) T-cell responses to dietary gluten. It is unclear whether interleukin (IL)-21 and IL-17A contribute to CD onset and lesion severity; therefore, we investigated IL-21 and IL-17A expression in biopsies from pediatric CD patients with different histopathological scores. High numbers of IL-21-producing cells were observed in pediatric CD lesions, even Marsh 1-2 lesions, whereas increased numbers of IL-17 secreting cells were not observed. Intraepithelial lymphocytes, CD4(+) T cells and also neutrophils secreted IL-21. Flow cytometry of lamina propria cells revealed a large population of IL-21- and interferon-γ (IFN-γ)-secreting CD3(+) T cells that did not secrete IL-17A. Adult CD patient biopsies also contained high numbers of IL-21-positive cells; however, enhanced numbers of IL-17-positive cells were observed in a small subgroup of patients with severe lesions. As duodenal tissue damage increases contact with microbe-associated molecular patterns, we hypothesized that microbial sensing by Toll-like receptors (TLRs) modulates T cell-derived cytokine secretion. Costimulation with TLR3 ligands during polyclonal T-cell activation significantly increased IL-21 secretion, whereas TLR2 ligands selectively enhanced IL-17A. These results demonstrate that an IL-17A-independent increase in IL-21 production by CD4(+) T cells is characteristic of pediatric CD. We hypothesize that incidental IL-17 secretion is caused by tissue damage rather than gluten-specific responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Doença Celíaca/imunologia , Interleucina-17/metabolismo , Interleucinas/metabolismo , Mucosa/imunologia , Adulto , Separação Celular , Células Cultivadas , Criança , Progressão da Doença , Citometria de Fluxo , Glutens/imunologia , Humanos , Interleucina-17/genética , Interleucinas/genética , Intestino Delgado/patologia , Ativação Linfocitária , Mucosa/patologia , Receptor 2 Toll-Like/metabolismo , Receptor 3 Toll-Like/metabolismo , Regulação para Cima
11.
Clin Microbiol Infect ; 19(2): E106-12, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23173866

RESUMO

Due to molecular mimicry, Campylobacter jejuni lipo-oligosaccharides can induce a cross-reactive antibody response to nerve gangliosides, which leads to Guillain-Barré syndrome (GBS). Cross-reactive antibodies to ganglioside GQ1b are strongly associated with oculomotor weakness in GBS and its variant, Miller Fisher syndrome (MFS). Antigen recognition is a crucial first step in the induction of a cross-reactive antibody response, and it has been shown that GQ1b-like epitopes expressed on the surface of C. jejuni are recognized by sialic acid-binding immunoglobulin-like lectin-7 (Siglec-7). We aimed to determine the epitope specificity of C. jejuni binding to Siglec-7, and correlate the outcome to disease symptoms in GBS and MFS patients. Using a well-defined GBS/MFS-associated C. jejuni strain collection, which included three sialic acid knockout strains, we found that Siglec-7 exclusively binds to C. jejuni strains that express terminal disialylated ganglioside mimics. When serological and diagnostic patient records were correlated with the Siglec-7-binding properties, we observed an association between Siglec-7 binding and the presence of anti-GQ1b antibodies in patient serum. In addition, Siglec-7 binding was associated with oculomotor weakness in GBS and MFS patients. Lipo-oligosaccharide-specific binding of C. jejuni to Siglec-7 may be an initiating event in immune recognition and presentation, and lead to anti-GQ1b antibody production and the development of ocular weakness in GBS or MFS.


Assuntos
Antígenos de Bactérias/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Autoanticorpos/sangue , Campylobacter jejuni/química , Campylobacter jejuni/patogenicidade , Síndrome de Guillain-Barré/patologia , Lectinas/metabolismo , Ácidos Siálicos/metabolismo , Campylobacter jejuni/genética , Técnicas de Inativação de Genes , Síndrome de Guillain-Barré/imunologia , Humanos , Músculos Oculomotores/fisiopatologia , Ligação Proteica
12.
Allergy ; 66(4): 478-90, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21143239

RESUMO

The term oral (or mucosal) tolerance has been classically defined as the suppression of T- and B-cell responses to an antigen by prior administration of the antigen by the oral route. In recent years, it has become clear that both innate and acquired regulatory immune responses are essential for the development of oral tolerance. As such, mucosal microenvironmental factors such as transforming growth factor- ß, prostaglandins but also dietary vitamin A create conditioning of an adaptive regulatory T-cell response that suppresses subsequent antigen-specific responses. Particular resident subsets of antigen presenting dendritic cells are pivotal to convey conditioning signals next to the presentation of antigen. This review discusses the primary mechanisms of adaptive regulatory T-cell induction to ingested soluble protein antigen. However, we also discuss the limitations of our knowledge with respect to understanding the very common food hypersensitivity Celiac disease caused by an aberrant adaptive immune response to the food protein gluten.


Assuntos
Imunidade Adaptativa/imunologia , Hipersensibilidade Alimentar/imunologia , Tolerância Imunológica/imunologia , Imunidade nas Mucosas/imunologia , Linfócitos T/imunologia , Animais , Antígenos/imunologia , Humanos , Proteínas/imunologia
13.
Mucosal Immunol ; 2(3): 254-64, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19262503

RESUMO

Oral intake of protein leads to tolerance through the induction of regulatory T cells (Tr cells) in mesenteric lymph nodes (MLNs). Here we show that the inhibition of cyclooxygenase-2 (COX-2) in vivo suppressed oral tolerance and was associated with enhanced differentiation of interleukin (IL)-4-producing T cells and reduced Foxp3(+) Tr-cell differentiation in MLN. As a result, the functional suppressive capacity of these differentiated mucosal T cells was lost. IL-4 was causally related to loss of tolerance as treatment of mice with anti-IL-4 antibodies during COX-2 inhibition restored tolerance. Dendritic cells (DCs) in the MLN differentially expressed COX-2 and reductionist experiments revealed that selective inhibition of the enzyme in these cells inhibited Foxp3(+) Tr-cell differentiation in vitro. Importantly, the inhibition of COX-2 in MLN-DC caused increased GATA-3 expression and enhanced IL-4 release by T cells, which was directly related to impaired Tr-cell differentiation. These data provide crucial insights into the mechanisms driving de novo Tr-cell induction and tolerance in the intestine.


Assuntos
Ciclo-Oxigenase 2/imunologia , Células Dendríticas/enzimologia , Interleucina-4/imunologia , Mucosa Intestinal/imunologia , Linfócitos T Reguladores/imunologia , Animais , Ácido Araquidônico/farmacologia , Diferenciação Celular/fisiologia , Células Cultivadas , Ciclo-Oxigenase 2/biossíntese , Inibidores de Ciclo-Oxigenase/farmacologia , Citocinas/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Tolerância Imunológica , Interleucina-4/antagonistas & inibidores , Interleucina-4/biossíntese , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Nitrobenzenos/farmacologia , Ovalbumina/imunologia , Sulfonamidas/farmacologia , Linfócitos T Reguladores/citologia
16.
Allergy ; 60(12): 1530-6, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16266386

RESUMO

Although as pretreatment oral tolerance is a potent means to achieve systemic suppression, its application in ongoing disease is controversial. Here we propose that availability of naive T cells may critically determine whether immunological tolerance is achieved during ongoing antigenic reactivity. Infusion of naive antigen-specific T cells into mice directly prior to eliciting a secondary Th2 response induces these naive cells to actively engage in the antigenic response despite presence of established memory. Naive antigen-specific T-cells divided faster, produced more interleukin (IL)-2, IL-4 and IL-5 and enhanced immunoglobulin E (IgE) release during a secondary Th2 response, compared with naive T cells that were infused prior to a primary response. Despite such contribution by new cohorts of naive T cells co-infusion of mucosal Tr together with naive T cells could suppress enhanced IgE release during a secondary Th2 response. We conclude that naive T cells contribute to a secondary Th2 response and although they can still be suppressed in the presence of sufficient numbers of mucosal Tr, they may interfere with potential therapeutic application of mucosal tolerance.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Hipersensibilidade/imunologia , Hipersensibilidade/fisiopatologia , Terapia de Imunossupressão , Linfócitos T Reguladores/imunologia , Animais , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Mucosa/imunologia , Receptores de Antígenos de Linfócitos T/genética , Células Th2/imunologia
17.
Allergy ; 59(11): 1211-8, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15461604

RESUMO

Induction of intranasal tolerance prevents the body from eliciting unwanted immune responses against harmless antigens that enter the body through the nasal mucosa. To study the intrinsic capacities of the cervical, nose draining lymph nodes (CLN), which are essential for tolerance induction, genes that are differentially expressed in CLN and not in peripheral lymph nodes (PLN) were characterized. The gene that is predominantly overexpressed in CLN codes for IgG2b. This is confirmed by a higher percentage of IgG2b+ B220+ cells in CLN compared with any PLN. However, this predominance of IgG2b-positive B cells in the CLN is not specific for the lymph node itself but rather determined by the region drained by lymph nodes at the cervical site, as transplanted PLN at these locations show a comparable predominance. It was demonstrated that IgG2b, when compared with IgG1, led to differential activation of dendritic cells (DC) through Fc receptor signalling. The results point to a unique local combination of cells and factors in the nose draining CLN leading to highly specialized immune reactivity. The results point out that predominance of a distinct IgG isotype in a lymphoid environment may lead to highly specialized immune reactivity.


Assuntos
Tolerância Imunológica/imunologia , Imunoglobulina G/imunologia , Linfonodos/imunologia , Mucosa Nasal/imunologia , Animais , Linfócitos B/imunologia , Células Dendríticas/imunologia , Expressão Gênica/imunologia , Tolerância Imunológica/genética , Imunoglobulina G/biossíntese , Imunoglobulina G/genética , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Pescoço , Receptores Fc/genética , Receptores Fc/imunologia , Transdução de Sinais/imunologia
18.
Vet Immunol Immunopathol ; 76(1-2): 125-35, 2000 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10973691

RESUMO

The aim of this study was to investigate the effects of a porcine reproductive and respiratory syndrome virus (PRRSV) infection on the development of the immune response after pseudorabies virus (PRV) vaccination in pigs. Pigs were intranasally inoculated with the European PRRSV strain, Lelystad virus ter Huurne, and were vaccinated intramuscularly with PRV 2 weeks later (LV-PRV group). Control pigs were vaccinated with PRV only (PRV group). Eight weeks after PRV vaccination, pigs from both groups were challenged intranasally with wild-type PRV. We measured the lymphoproliferative, and the cytolytic responses to PRV of peripheral blood mononuclear cells (PBMC), isolated from blood samples. In addition, serum samples were examined for antibodies against PRV and LV. One week after PRV vaccination, PBMC proliferated abundantly to PRV in both groups. However, in the LV-PRV group the lymphoproliferative response declined after 1 week, whereas, in the PRV group, the lymphoproliferative response was high for 3 weeks and declined thereafter (P<0.05). After challenge, the lymphoproliferative response was 1 week earlier and was consistently and significantly higher in the PRV group than in the LV-PRV group. The PRV-specific killing was higher at 3 weeks after PRV vaccination and 5 weeks after PRV challenge 19+/-3 and 24+/-6%, respectively, in the PRV group, compared to 7+/-4 and 6+/-9%, respectively, in the LV-PRV group (P<0.05). However, later after vaccination and challenge the cytolytic response was identical in both groups. The antibody titre against PRV developed equally in both groups. After challenge, no PRV virus was isolated from both groups. From these results we conclude that, although PRRSV infection did cause changes in the time course of the T-lymphocyte response after PRV vaccination, PRRSV infection did not inhibit the development of vaccine-induced protection after PRV.


Assuntos
Anticorpos Antivirais/biossíntese , Herpesvirus Suídeo 1/imunologia , Síndrome Respiratória e Reprodutiva Suína/imunologia , Administração Intranasal , Animais , Linhagem Celular , Citotoxicidade Imunológica , Tolerância Imunológica , Ativação Linfocitária , Vírus da Síndrome Respiratória e Reprodutiva Suína , Suínos , Porco Miniatura , Vacinas Virais/imunologia
19.
Infect Immun ; 68(8): 4666-72, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10899871

RESUMO

The aim of this study was to investigate the effect of interleukin-10 (IL-10) on the course of Listeria monocytogenes infection in naive and immune mice. Treatment with IL-10 during the course of a primary infection significantly decreased the number of bacteria in the spleen and did not affect the number in the liver. During a secondary infection in immune mice treated with IL-10, the number of bacteria was significantly lower in the spleen but significantly higher in the liver in comparison to mock-treated immune mice. IL-10 treatment during a primary Listeria infection decreased the concentration of gamma interferon (IFN-gamma) in plasma and the toxoplasmastatic activity of macrophages, whereas it increased the percentage of mildly CD3-positive T cells in the spleen. During a secondary infection, the concentration of IFN-gamma in plasma was decreased on day 1 but remained unaffected during later days of infection. From these results, we conclude that IL-10 has different effects on the proliferation of L. monocytogenes in the spleen and liver during primary and secondary Listeria infections.


Assuntos
Interleucina-10/imunologia , Listeriose/imunologia , Fígado/microbiologia , Baço/microbiologia , Animais , Anticorpos Monoclonais/farmacologia , Complexo CD3/análise , Divisão Celular/efeitos dos fármacos , Feminino , Interferon gama/sangue , Interleucina-10/uso terapêutico , Listeriose/tratamento farmacológico , Fígado/citologia , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos CBA , Receptores de Antígenos de Linfócitos T alfa-beta , Baço/citologia , Subpopulações de Linfócitos T , Células Th1
20.
J Gen Virol ; 81(Pt 6): 1529-37, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10811936

RESUMO

We previously observed that pseudorabies virus (PRV)-induced, cell-mediated cytolysis in pigs includes killing by natural killer (NK) cells. We also observed that IL-2 stimulation in vitro of naive PBMC expands porcine NK cells. The purpose of this study was to compare the phenotypes of the cytolytic subsets stimulated in vitro by PRV and by IL-2. PBMC were isolated from blood of PRV-immune and naive pigs and stimulated in vitro with PRV or IL-2. After 6 days, the frequency of various lymphocyte subsets in these cultured PBMC was determined by flow cytometry: the cells were separated with a magnet-activated cell sorter and the cytolytic activity of the separated populations was determined. When lymphocytes were separated and analysed with FACScan, the following lymphocyte subsets were discriminated: CD6(+) CD8(bright+) CD4(-) (CTL phenotype), CD6(+) CD8(dull+) CD4(+) (the fraction containing memory T helper cells), CD6(+) CD8(-) CD4(+) (T helper cell phenotype), CD6(-) CD8(dull+) CD4(-) gammadelta-T(+) ( gammadelta-T cell phenotype), CD6(-) CD8(dull+) CD4(-) gammadelta-T(-) (NK phenotype) and CD6(-) CD8(-) CD4(-) gammadelta-T(-) or gammadelta-T(+). Flow cytometry analysis demonstrated that PRV stimulation of immune PBMC resulted in the occurrence of more CD6(+) CD8(+) and CD4(+) CD8(+) and fewer CD6(-) CD8(+) and gammadelta-T(+) CD8(+) lymphocytes than IL-2 stimulation of naive PBMC (P<0.05). It was demonstrated further that killing by PRV-stimulated PBMC was mediated mainly by CD6(+) CD8(+) T lymphocytes. Killing by IL-2-stimulated PBMC was mediated mainly by CD6(-) CD8(+) T lymphocytes. These results demonstrate that both natural killing and killing by classical PRV-specific CTL were detected in PRV-immune pigs, whereas IL-2 stimulation of PBMC isolated from naive pigs mainly induced natural killing.


Assuntos
Citotoxicidade Imunológica/imunologia , Herpesvirus Suídeo 1/imunologia , Subpopulações de Linfócitos/classificação , Animais , Anticorpos Monoclonais , Antígenos CD/análise , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Separação Celular/métodos , Células Cultivadas , Imunofenotipagem , Interleucina-2/farmacologia , Leucócitos Mononucleares/citologia , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Linfócitos/citologia , Suínos , Porco Miniatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...