Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Allergy Clin Immunol ; 148(3): 867-875.e4, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33662368

RESUMO

BACKGROUND: IL-33 is an emerging key factor in development of allergic diseases. The IL-33 receptor (suppressor of tumorigenicity [ST2]) is a differentially expressed gene in pathogenic TH2 cells, but its role in T-cell effector function has not been elucidated. OBJECTIVE: We investigated the role of IL-33 in modulating circulating allergen-specific T-cell responses. We hypothesized that selective ST2 expression on allergen-specific CD4+ T cells would confer susceptibility to the effects of IL-33. METHODS: PBMCs from subjects with food allergy, inhalant allergy, and no allergy were obtained on the basis of clinical history and serum IgE level. A T-cell receptor-dependent CD154 upregulation assay and direct peptide major histocompatibility complex class II tetramer staining were used to profile allergen-specific CD4+ T cells by flow cytometry. Allergen-specific CD4+ T cell cytokine production was evaluated during IL-33 exposure. ST2 expression was also tracked by using a 2-color flow-based assay. RESULTS: ST2 expression on peripheral allergen-specific CD4+ T cells was confined to subjects with allergy and restricted to TH2A cells. Comparison between direct peptide major histocompatibility complex class II tetramer staining and the CD154 functional assay identified ST2 as a marker of TH2A cell activation. IL-33 exposure enhanced IL-4 and IL-5 secretion in allergen-reactive TH2A cells. Allergen-induced ST2 expression on peripheral CD4+ T cells can be used to track allergen-reactive TH2A cells from donors with allergy. CONCLUSION: ST2 expression on circulating CD4+ T cells represents a transient phenotype associated with TH2A cell activation, allowing these cells to sense locally elicited tissue cytokines. IL-33 selectively amplifies pathogenic TH2 cell effector functions, suggesting a tissue checkpoint that may regulate adaptive allergic immunity.


Assuntos
Hipersensibilidade/imunologia , Interleucina-33/imunologia , Células Th2/imunologia , Células Epiteliais/imunologia , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/imunologia , Receptores de Interleucina-17/imunologia , Transdução de Sinais
2.
JCI Insight ; 4(22)2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31723064

RESUMO

BACKGROUNDIL-33, found in high levels in participants with allergic disorders, is thought to mediate allergic reactions. Etokimab, an anti-IL-33 biologic, has previously demonstrated a good safety profile and favorable pharmacodynamic properties in many clinical studies.METHODSIn this 6-week placebo-controlled phase 2a study, we evaluated the safety and the ability of a single dose of etokimab to desensitize peanut-allergic adults. Participants received either etokimab (n = 15) or blinded placebo (n = 5). Clinical tests included oral food challenges and skin prick tests at days 15 and 45. Blood samples were collected for IgE levels and measurement of ex vivo peanut-stimulated T cell cytokine production.RESULTSEfficacy measurements for active vs. placebo participants at the day 15 and 45 food challenge (tolerating a cumulative 275 mg of peanut protein, which was the food challenge outcome defined in this paper) demonstrated, respectively, 73% vs. 0% (P = 0.008) to 57% vs. 0% (ns). The etokimab group had fewer adverse events compared with placebo. IL-4, IL-5, IL-9, IL-13, and ST2 levels in CD4+ T cells were reduced in the active vs. placebo arm upon peanut-induced T cell activation (P = 0.036 for IL-13 and IL-9 at day 15), and peanut-specific IgE was reduced in active vs. placebo (P = 0.014 at day 15).CONCLUSIONThe phase 2a results suggest etokimab is safe and well tolerated and that a single dose of etokimab could have the potential to desensitize peanut-allergic participants and possibly reduce atopy-related adverse events.TRIAL REGISTRATIONClinicalTrials.gov NCT02920021.FUNDINGThis work was supported by NIH grant R01AI140134, AnaptysBio, the Hartman Vaccine Fund, and the Sean N. Parker Center for Allergy and Asthma Research at Stanford University.


Assuntos
Anticorpos Monoclonais , Interleucina-33/antagonistas & inibidores , Hipersensibilidade a Amendoim , Adolescente , Adulto , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Células Cultivadas , Citocinas/metabolismo , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Hipersensibilidade a Amendoim/tratamento farmacológico , Hipersensibilidade a Amendoim/imunologia , Placebos , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Adulto Jovem
3.
Sci Transl Med ; 11(515)2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31645451

RESUMO

Targeted inhibition of cytokine pathways provides opportunities to understand fundamental biology in vivo in humans. The IL-33 pathway has been implicated in the pathogenesis of atopy through genetic and functional associations. We investigated the role of IL-33 inhibition in a first-in-class phase 2a study of etokimab (ANB020), an IgG1 anti-IL-33 monoclonal antibody, in patients with atopic dermatitis (AD). Twelve adult patients with moderate to severe AD received a single systemic administration of etokimab. Rapid and sustained clinical benefit was observed, with 83% achieving Eczema Area and Severity Index 50 (EASI50), and 33% EASI75, with reduction in peripheral eosinophils at day 29 after administration. We noted significant reduction in skin neutrophil infiltration after etokimab compared with placebo upon skin challenge with house dust mite, reactivity to which has been implicated in the pathogenesis of AD. We showed that etokimab also inhibited neutrophil migration to skin interstitial fluid in vitro. Besides direct effects on neutrophil migration, etokimab revealed additional unexpected CXCR1-dependent effects on IL-8-induced neutrophil migration. These human in vivo findings confirm an IL-33 upstream role in modulating skin inflammatory cascades and define the therapeutic potential for IL-33 inhibition in human diseases, including AD.


Assuntos
Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/metabolismo , Interleucina-33/metabolismo , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Movimento Celular/efeitos dos fármacos , Dermatite Atópica/imunologia , Eczema/imunologia , Eczema/metabolismo , Líquido Extracelular , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-12/metabolismo , Interleucina-33/imunologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Receptores de Interleucina-8A/metabolismo , Pele/efeitos dos fármacos , Pele/imunologia , Pele/metabolismo
5.
PLoS One ; 8(10): e78483, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24205242

RESUMO

Type-1 diabetes (T1D) is an autoimmune disease targeting insulin-producing beta cells, resulting in dependence on exogenous insulin. To date, significant efforts have been invested to develop immune-modulatory therapies for T1D treatment. Previously, IL-2 immunotherapy was demonstrated to prevent and reverse T1D at onset in the non-obese diabetic (NOD) mouse model, revealing potential as a therapy in early disease stage in humans. In the NOD model, IL-2 deficiency contributes to a loss of regulatory T cell function. This deficiency can be augmented with IL-2 or antibody bound to IL-2 (Ab/IL-2) therapy, resulting in regulatory T cell expansion and potentiation. However, an understanding of the mechanism by which reconstituted regulatory T cell function allows for reversal of diabetes after onset is not clearly understood. Here, we describe that Ab/IL-2 immunotherapy treatment, given at the time of diabetes onset in NOD mice, not only correlated with reversal of diabetes and expansion of Treg cells, but also demonstrated the ability to significantly increase beta cell proliferation. Proliferation appeared specific to Ab/IL-2 immunotherapy, as anti-CD3 therapy did not have a similar effect. Furthermore, to assess the effect of Ab/IL-2 immunotherapy well after the development of diabetes, we tested the effect of delaying treatment for 4 weeks after diabetes onset, when beta cells were virtually absent. At this late stage after diabetes onset, Ab/IL-2 treatment was not sufficient to reverse hyperglycemia. However, it did promote survival in the absence of exogenous insulin. Proliferation of beta cells could not account for this improvement as few beta cells remained. Rather, abnormal insulin and glucagon dual-expressing cells were the only insulin-expressing cells observed in islets from mice with established disease. Thus, these data suggest that in diabetic NOD mice, beta cells have an innate capacity for regeneration both early and late in disease, which is revealed through IL-2 immunotherapy.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Insulina/imunologia , Interleucina-2/imunologia , Animais , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animais de Doenças , Feminino , Glucagon/metabolismo , Imunoterapia/métodos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Regeneração/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
6.
Gut ; 61(12): 1693-700, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22595313

RESUMO

OBJECTIVE: The authors tested whether the anti-interleukin (IL)-17A monoclonal antibody secukinumab was safe and effective for the treatment of active Crohn's disease. DESIGN: In a double-blind, randomised, placebo-controlled proof-of-concept study, 59 patients with moderate to severe Crohn's disease (Crohn's Disease Activity Index (CDAI) ≥220 to ≤450) were assigned in a 2:1 ratio to 2×10 mg/kg intravenous secukinumab or placebo. The primary end point, addressed by bayesian statistics augmented with historical placebo information, was the probability that secukinumab reduces the CDAI by ≥50 points more than placebo at week 6. Ancillary analyses explored associations of 35 candidate genetic polymorphisms and faecal calprotectin response. RESULTS: 59 patients (39 secukinumab, 20 placebo, mean baseline CDAI 307 and 301, respectively) were recruited. 18/59 (31%) patients discontinued prematurely (12/39 (31%) secukinumab, 6/20 (30%) placebo), 10/59 (17%) due to insufficient therapeutic effect (8/39 (21%) secukinumab, 2/20 (10%) placebo). Fourteen serious adverse events occurred in 10 patients (seven secukinumab, three placebo); 20 infections, including four local fungal infections, were seen on secukinumab versus none on placebo. Primary end point analysis estimated <0.1% probability (CDAI (SD) =33.9 (19.7), 95% credible interval -4.9 to 72.9) that secukinumab reduces CDAI by ≥50 points more than placebo. Secondary area under the curve analysis (weeks 4-10) showed a significant difference (mean ΔCDAI=49; 95% CI (2 to 96), p=0.043) in favour of placebo. Post hoc subgroup analysis showed that unfavourable responses on secukinumab were driven by patients with elevated inflammatory markers (CRP≥10 mg/l and/or faecal calprotectin≥200 ng/ml; mean ΔCDAI=62; 95% CI (-1 to 125), p=0.054 in favour of placebo). Absence of the minor allele of tumour necrosis factor-like ligand 1A was strongly associated with lack of response measured by baseline-adjusted changes in calprotectin at week 6 (p=0.00035 Bonferroni-corrected). CONCLUSIONS: Blockade of IL-17A was ineffective and higher rates of adverse events were noted compared with placebo. CLINICAL TRIAL REGISTRATION: This trial was registered at ClinicalTrial.gov with the number NCT01009281.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Doença de Crohn/tratamento farmacológico , Adolescente , Adulto , Idoso , Anticorpos Monoclonais Humanizados , Teorema de Bayes , Biomarcadores/metabolismo , Doença de Crohn/genética , Método Duplo-Cego , Esquema de Medicação , Feminino , Marcadores Genéticos , Humanos , Infusões Intravenosas , Interleucina-17/antagonistas & inibidores , Interleucina-17/genética , Complexo Antígeno L1 Leucocitário/metabolismo , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Índice de Gravidade de Doença , Falha de Tratamento , Adulto Jovem
7.
Gut ; 59(3): 311-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19951908

RESUMO

BACKGROUND: An unresolved question in coeliac disease is to understand how some toxic gliadin peptides, in particular p31-43, can initiate an innate response and lead to tissue transglutaminase (TG2) upregulation in coeliac intestine and gliadin sensitive epithelial cell lines. Aim We addressed whether the epithelial uptake of p31-43 induces an intracellular pro-oxidative envoronment favouring TG2 activation and leading to the innate immune response. METHODS: The time course of intracellular delivery to lysosomes of p31-43, palpha-2 or palpha-9 gliadin peptides was analysed in T84 and Caco-2 epithelial cells. The effects of peptide challenge on oxidative stress, TG2 and peroxisome proliferator-activated receptor (PPAR)gamma ubiquitination and p42/44-mitogen activated protein (MAP) kinase or tyrosine phosphorylation were investigated in cell lines and cultured coeliac disease biopsies with/without anti-oxidant treatment or TG2 gene silencing by immunoprecipitation, western blot, confocal microscopy and Fluorenscence Transfer Resonance Energy (FRET) analysis. RESULTS: After 24 h of challenge p31-43, but not palpha-2 or palpha-9, is still retained within LAMP1-positive perinuclear vesicles and leads to increased levels of reactive oxygen species (ROS) that inhibit TG2 ubiquitination and lead to increases of TG2 protein levels and activation. TG2 induces cross-linking, ubiquitination and proteasome degradation of PPARgamma. Treatment with the antioxidant EUK-134 as well as TG2 gene silencing restored PPARgamma levels and reversed all monitored signs of innate activation, as indicated by the dramatic reduction of tyrosine and p42/p44 phosphorylation. CONCLUSION: p31-43 accumulation in lysosomes leads to epithelial activation via the ROS-TG2 axis. TG2 works as a rheostat of ubiquitination and proteasome degradation and drives inflammation via PPARgamma downregulation.


Assuntos
Doença Celíaca/metabolismo , Gliadina/metabolismo , Lisossomos/metabolismo , PPAR gama/metabolismo , Fragmentos de Peptídeos/metabolismo , Transglutaminases/fisiologia , Adolescente , Adulto , Regulação para Baixo/fisiologia , Células Epiteliais/metabolismo , Proteínas de Ligação ao GTP , Humanos , Mucosa Intestinal/metabolismo , Técnicas de Cultura de Órgãos , Estresse Oxidativo/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína 2 Glutamina gama-Glutamiltransferase , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas , Ubiquitina/metabolismo , Adulto Jovem
8.
J Immunol ; 180(11): 7697-705, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18490773

RESUMO

Cystic fibrosis (CF), the most common life-threatening inherited disease in Caucasians, is due to mutations in the CF transmembrane conductance regulator (CFTR) gene and is characterized by airways chronic inflammation and pulmonary infections. The inflammatory response is not secondary to the pulmonary infections. Indeed, several studies have shown an increased proinflammatory activity in the CF tissues, regardless of bacterial infections, because inflammation is similarly observed in CFTR-defective cell lines kept in sterile conditions. Despite recent studies that have indicated that CF airway epithelial cells can spontaneously initiate the inflammatory cascade, we still do not have a clear insight of the molecular mechanisms involved in this increased inflammatory response. In this study, to understand these mechanisms, we investigated ex vivo cultures of nasal polyp mucosal explants of CF patients and controls, CFTR-defective IB3-1 bronchial epithelial cells, C38 isogenic CFTR corrected, and 16HBE normal bronchial epithelial cell lines. We have shown that a defective CFTR induces a remarkable up-regulation of tissue transglutaminase (TG2) in both tissues and cell lines. The increased TG2 activity leads to functional sequestration of the anti-inflammatory peroxisome proliferator-activated receptor gamma and increase of the classic parameters of inflammation, such as TNF-alpha, tyrosine phosphorylation, and MAPKs. Specific inhibition of TG2 was able to reinstate normal levels of peroxisome proliferator-activated receptor-gamma and dampen down inflammation both in CF tissues and CFTR-defective cells. Our results highlight an unpredicted central role of TG2 in the mechanistic pathway of CF inflammation, also opening a possible new wave of therapies for sufferers of chronic inflammatory diseases.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Inflamação/metabolismo , PPAR gama/metabolismo , Transglutaminases/metabolismo , Linhagem Celular , Fibrose Cística/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Regulação para Baixo , Ativação Enzimática , Células Epiteliais/citologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Desacetilase 6 de Histona , Histona Desacetilases/imunologia , Histona Desacetilases/metabolismo , Humanos , Inflamação/imunologia , Proteína 2 Glutamina gama-Glutamiltransferase , Ubiquitina/imunologia , Ubiquitina/metabolismo
9.
Immunology ; 125(2): 178-83, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18422560

RESUMO

Crohn's disease (CD) is a chronic inflammatory disease of the gastrointestinal tract. Its pathogenesis is not completely understood, though the prevailing model is that the intestinal flora drives a strong intestinal T helper 1 (Th1)/Th17 type immune response and inflammation in the genetically susceptible host. This leads to overly aggressive T-cell responses to normal bacteria causing tissue damage. Intestinal homeostasis and maintenance of tolerance to harmless antigens in the intestine has been shown to be maintained by CD4+ CD25+ T regulatory cells (Treg) in animal models of inflammatory bowel diseases. Here we investigated whether Infliximab, a chimeric monoclonal antibody directed against tumour necrosis factor (TNF)-alpha shown to be highly effective in the treatment of CD, has any effect on mucosal CD4+ CD25+ (FOXP3+) Tregs. Colonic mucosal biopsies from children with active Crohn's disease treated in vivo with Infliximab and controls were analysed to determine FOXP3 expression by immunofluorescence and reverse transcription-polymerase chain reaction. We observed that FOXP3+ T cells were significantly reduced in mucosa of CD patients with active disease compared with controls and restored to normal following Infliximab treatment. The frequency of FOXP3+ cells and mRNA expression was significantly increased in CD mucosa from patients treated in vivo with Infliximab compared with CD patients treated with conventional therapies. In conclusion, we show that Infliximab treatment does not solely neutralize soluble TNF-alpha, but also affects activation and possibly expansion of mucosal regulatory T cells. We suggest that anti TNF-alpha immunotherapy can also restore mucosal homeostasis in Crohn's disease.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doença de Crohn/tratamento farmacológico , Fatores de Transcrição Forkhead/biossíntese , Linfócitos T Reguladores/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Criança , Colo/imunologia , Doença de Crohn/imunologia , Fatores de Transcrição Forkhead/genética , Humanos , Infliximab , Mucosa Intestinal/imunologia , Contagem de Linfócitos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Linfócitos T Reguladores/imunologia
10.
Clin Nutr ; 26(6): 799-805, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17719701

RESUMO

BACKGROUND & AIMS: Celiac disease is a condition in which genetically predisposed people have an autoimmune reaction to gluten proteins found in all wheat types and closely related cereals such as barley and rye. This reaction causes the formation of autoantibodies and the destruction of the villi in the small intestine, which results in malabsorption of nutrientsand other gluten-induced autoimmune diseases. Sorghum is a cereal grain with potential to be developed into an important crop for human food products. The flour produced from white sorghum hybrids is light in color and has a bland, neutral taste that does not impart unusual colors or flavors to food products. These attributes make it desirable for use in wheat-free food products. While sorghum is considered as a safe food for celiac patients, primarily due to its relationship to maize, no direct testing has been conducted on its safety for gluten intolerance. Therefore studies are needed to assess its safety and tolerability in celiac patients. Thus the aim of the present study was to assess safety and tolerability of sorghum flour products in adult celiac disease patients, utilizing an in vitro and in vivo challenge. RESULTS: Sorghum protein digests did not elicit any morphometric or immunomediated alteration of duodenal explants from celiac patients. Patients fed daily for 5 days with sorghum-derived food product did not experience gastrointestinal or non-gastrointestinal symptoms and the level of anti-transglutaminase antibodies was unmodified at the end of the 5-days challenge. CONCLUSIONS: Sorghum-derived products did not show toxicity for celiac patients in both in vitro and in vivo challenge. Therefore sorghum can be considered safe for people with celiac disease.


Assuntos
Doença Celíaca/dietoterapia , Qualidade de Produtos para o Consumidor , Tecnologia de Alimentos , Sorghum/química , Paladar , Adulto , Comportamento do Consumidor , Feminino , Farinha/análise , Análise de Alimentos , Glutens/administração & dosagem , Glutens/efeitos adversos , Humanos , Masculino , Triticum/efeitos adversos
11.
J Immunol ; 179(1): 372-81, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17579058

RESUMO

We analyzed the autologous NK cell interaction with gliadin-presenting dendritic cells. Gliadin is the known Ag priming the celiac disease (CD) pathogenesis. We demonstrate that gliadin prevents immature dendritic cells (iDCs) elimination by NK cells. Furthermore, cooperation between human NK cells-iDCs and T cells increases IFN-gamma production of anti-gliadin immune response. Gliadin fractions were analyzed for their capability to stabilize HLA-E molecules. The alpha and omega fractions conferred the protection from NK cell lysis to iDCs and increased their HLA-E expression. Gliadin pancreatic enzyme digest and a peptide derived from gliadin alpha increased HLA-E levels on murine RMA-S/HLA-E-transfected cells. Analysis of HLA-E expression in the small intestinal mucosa of gluten-containing diet celiac patients and organ culture experiments confirmed the in vitro data.


Assuntos
Comunicação Celular/imunologia , Membrana Celular/metabolismo , Células Dendríticas/metabolismo , Gliadina/farmacologia , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/metabolismo , Adolescente , Adulto , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Criança , Citotoxicidade Imunológica/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Feminino , Antígenos HLA/biossíntese , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Imunofenotipagem , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Lactoglobulinas/fisiologia , Masculino , Camundongos , Técnicas de Cultura de Órgãos , Peptídeos/fisiologia , Antígenos HLA-E
12.
PLoS One ; 2(4): e387, 2007 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-17440622

RESUMO

Epicutaneous immunization is a potential non-invasive technique for antigen-specific immune-modulation. Topical application of protein antigens to barrier-disrupted skin induces potent antigen-specific immunity with a strong Th2-bias. In this study, we investigate whether the autoimmune inflammatory response of chronic collagen-induced arthritis (CCIA) in DBA/1-TCR-beta Tg mice can be modified by epicutaneous immunization. We show that epicutaneous immunization with type II collagen (CII) inhibited development and progression of CCIA and, importantly, also ameliorated ongoing disease as indicated by clinical scores of disease severity, paw swelling and joints histology. Treated mice show reduced CII-driven T cell proliferation and IFN-gamma production, as well as significantly lower levels of CII-specific IgG2a serum antibodies. In contrast, CII-driven IL-4 production and IgE antibody levels were increased consistent with skewing of the CII response from Th1 to Th2 in treated mice. IL-4 production in treated mice was inversely correlated with disease severity. Moreover, T cells from treated mice inhibited proliferation and IFN-gamma production by T cells from CCIA mice, suggesting induction of regulatory T cells that actively inhibit effector responses in arthritic mice. The levels of CD4(+)CD25(+) T cells were however not increased following epicutaneous CII treatment. Together, these results suggest that epicutaneous immunization may be used as an immune-modulating procedure to actively re-programme pathogenic Th1 responses, and could have potential as a novel specific and simple treatment for chronic autoimmune inflammatory diseases such as rheumatoid arthritis.


Assuntos
Artrite Experimental/prevenção & controle , Colágeno Tipo II/administração & dosagem , Pele/imunologia , Animais , Anticorpos/sangue , Artrite Experimental/imunologia , Proliferação de Células , Colágeno Tipo II/imunologia , Progressão da Doença , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos DBA , Linfócitos T/imunologia
13.
Diabetes ; 55(12): 3439-45, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17130490

RESUMO

Type 1 diabetes is caused by adaptive immune responses, but innate immunity is important because monocytes infiltrate islets. Activated monocytes express cyclooxygenase (COX)-2, promoting prostaglandin-E(2) (PGE(2)) secretion, whereas COX-1 expression is constitutive. We aimed to define monocyte COX expression in type 1 diabetes basally and after lipopolysaccharide (LPS) stimulation. Isolated CD14(+) monocytes were analyzed for COX mRNA and protein expression from identical twins (discordant for type 1 diabetes) and control subjects. Basal monocyte COX mRNA, protein expression, and PGE(2) secretion were normal in type 1 diabetic subjects. After LPS, twins and control subjects showed a COX mRNA isoform switch with decreased COX-1 mRNA (P < 0.01), increased COX-2 mRNA (P < 0.01), and increased COX-2 protein expression (P < 0.01). Compared with control subjects, both diabetic and nondiabetic twins showed greater LPS-induced downregulation of monocyte COX-1 mRNA (P = 0.02), reduced upregulation of COX-2 mRNA and protein (P < 0.03), and greater inhibition by the COX-2 inhibitor di-isopropylfluorophosphate (DFP) of monocyte PGE(2) (P < 0.007). We demonstrate an alteration in monocyte COX mRNA expression as well as monocyte COX-2 and PGE(2) production after LPS in type 1 diabetic patients and their nondiabetic twins. Because COX-2 response to LPS is proinflammatory, an inherited reduced response would predispose to chronic inflammatory diseases such as type 1 diabetes.


Assuntos
Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Diabetes Mellitus Tipo 1/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Monócitos/enzimologia , Animais , Sequência de Bases , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Ciclo-Oxigenase 1/sangue , Ciclo-Oxigenase 2/sangue , Diabetes Mellitus Tipo 1/sangue , Humanos , Camundongos , Dados de Sequência Molecular , Monócitos/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Diabetes Metab Res Rev ; 22(5): 367-75, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16572491

RESUMO

BACKGROUND: Recent studies in animal models indicate a role for natural killer (NK) cells in the protection against type 1 diabetes. In humans, a reduction of NK cell numbers has been reported in identical twins discordant for type 1 diabetes, irrespective of whether they have the disease. Here we have tested whether the activation and expansion of human NK cells with lipopolysaccharide (LPS) reveals differences between these twins. METHODS: Proportions of CD56(+) NK cells and T-cells and Va24Vb11(+) NK-T cells from diabetic and non-diabetic twins was assessed before and after activation using flow cytometry. NK receptor usage was monitored by PCR and flow cytometry. RESULTS: The profile of the expressed Killer Cell immunoglobulin-like receptor (KIR) repertoire (using mRNA) in freshly isolated NK cells was identical in pairs of identical twins, despite marked variation among individual twins as well as controls. Basal numbers of CD56(+) and CD94(+) (CD3(-) and CD3(+)) cells and Valpha24(+)Vbeta11(+) NK-T cells were similarly strongly correlated between identical twins (p < 0.006 for all correlations). Following LPS stimulation, the pattern of KIR mRNA expression remained unaltered in twins and the proportion of NK cells and Valpha24(+)Vbeta11(+) NK-T cells remained correlated between pairs of twins. However, there was a significant reduction in the proportion of CD56(+) cells and CD94(+) cells (whether defined as CD3(-) or CD3(+)) responding to LPS in the diabetic compared to the non-diabetic twin (p = 0.031 and 0.025, respectively). CONCLUSION: This reduction in NK cell expansion in response to LPS in patients with type 1 diabetes is consistent with a non-genetically determined alteration in the innate immune response either predisposing to or resulting from the disease.


Assuntos
Antígeno CD56/imunologia , Diabetes Mellitus Tipo 1/imunologia , Células Matadoras Naturais/imunologia , Técnicas de Cultura de Células , Doenças em Gêmeos , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Ativação Linfocitária , Proteínas Monoméricas de Ligação ao GTP/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gêmeos Monozigóticos
15.
Gastroenterology ; 129(5): 1400-13, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16285941

RESUMO

BACKGROUND & AIMS: In celiac disease (CD), transglutaminase type II (TG2) has 2 fundamental roles: (1) as the autoantigen recognized by highly specific autoantibodies and (2) the modifier of pathogenic gliadin T-cell epitopes. It follows that inhibition of TG2 might represent an attractive strategy to curb the toxic action of gliadin. Here we studied the validity of this strategy using the organ culture approach. METHODS: Duodenal biopsy specimens from 30 treated patients with CD, 33 untreated patients with CD, and 24 controls were cultured with or without gliadin peptides p31-43, palpha-9, and deamidated palpha-9 for 20 minutes, 3 hours, and 24 hours. In 31 patients with CD and 16 controls, TG2 inhibitor R283 or anti-TG CUB 7402 or anti-surface TG2 (6B9) mAbs were used in cultures. T84 cells were also cultured with or without peptides with or without TG inhibitors. Mucosal modifications after culture were assessed by immunofluorescence, in situ detection of TG activity, confocal microscopy, and fluorescence-activated cell sorter analysis. RESULTS: The enzymatic inhibition of TG2 only controlled gliadin-specific T-cell activation. The binding of surface TG2 contained gliadin-specific T-cell activation and p31-43-induced actin rearrangement, epithelial phosphorylation, and apoptosis, both in organ cultures and T84 cells. CONCLUSIONS: These data indicate a novel and unexpected biological role for surface TG2 in the pathogenesis of CD suggesting a third role for TG2 in CD. These results have a specific impact for celiac disease, with wider implications indicating a novel biologic function of TG2 with possible repercussions in other diseases.


Assuntos
Doença Celíaca/metabolismo , Doença Celíaca/patologia , Proteínas de Ligação ao GTP/metabolismo , Transglutaminases/metabolismo , Adulto , Anticorpos Monoclonais , Antígenos de Superfície/metabolismo , Apoptose , Biópsia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Doença Celíaca/imunologia , Linhagem Celular , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Proteínas de Ligação ao GTP/imunologia , Gliadina/metabolismo , Humanos , Mucosa Intestinal/enzimologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Intestino Delgado/enzimologia , Intestino Delgado/imunologia , Intestino Delgado/patologia , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Proteína 2 Glutamina gama-Glutamiltransferase , Especificidade por Substrato , Transglutaminases/imunologia , Regulação para Cima
17.
Arthritis Res Ther ; 7(3): 118-20, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15899062

RESUMO

CD4+CD25+ T regulatory cells are avidly studied because they modulate immune responses. Their possible role in autoimmunity and more specifically in rheumatoid arthritis (RA) has been highlighted by a string of reports, one of which is in the last issue of Arthritis Research & Therapy. There are, however, key questions that have not yet been addressed before their use can be considered as a real therapeutic option. The first is the actual, in a clinical setting, efficacy of Treg to treat active chronic autoimmune diseases such as RA. The second is how we can practically deliver their therapeutic activity in patients. Once these points have been addressed we will have a new and potentially very effective 'magic bullet' for the treatment of chronic autoimmune diseases.


Assuntos
Artrite Experimental/imunologia , Artrite Experimental/terapia , Linfócitos T Reguladores/fisiologia , Linfócitos T Reguladores/transplante , Animais , Artrite Experimental/tratamento farmacológico , Humanos
18.
Mol Immunol ; 42(8): 913-8, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15829281

RESUMO

In celiac disease (CD) we have the prototype of an immune mediated response dominated by the activation of the adaptive immune system and in particular of CD4+ HLA class II restricted T cells. Various seminal studies have established the precise mechanism of how antigen (prolamine) specific activation of CD4+ mucosal T cells occurs. Thus, CD is a condition in which T cells and their activation is the essential hinge in the pathogenic process. These functional studies have provided the explanation for the genetic association between CD and certain HLA alleles (HLA DQ2 and DQ8). These genetic, molecular and functional studies have permitted the clarification of a powerful Th1 dominated pro-inflammatory response that characterises the small intestine of active CD patients. Despite this unassailable set of information and reports there are some intriguing points that have been raised by a series of studies which have indicated that CD is not only defined by an aberrant prolamine-induced activation of the adaptive immune system. New evidence and re-assessments of old studies, point to a more complex pathogenic cascade, which may help to unravel some of the residual obscure points of CD pathogenesis. Here, we outline the current concepts that indicate a direct involvement of the adaptive immune system and we discuss all the evidence supporting a direct activation of the innate immune system by fragments of prolamines, which are not recognized T cell epitopes and how they could influence CD. The gliadin-induced activation of the 'innate' immune system might also have a significant role in the induction and persistence of many CD complications and most definitively for the most aggressive one, namely mucosal T cell lymphomas. We further suggest a novel way to harness the unwanted immune response to toxic prolamine, and thus indicate new potential therapeutic strategies to treat or at least control CD.


Assuntos
Doença Celíaca/imunologia , Gliadina/imunologia , Imunidade Inata/imunologia , Animais , Doença Celíaca/etiologia , Doença Celíaca/terapia , Antígenos HLA/imunologia , Humanos
19.
J Immunol ; 174(6): 3256-63, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15749856

RESUMO

In prion diseases, such as variant Creutzfeldt-Jakob disease normal cellular prion protein (PrPC), a largely alpha-helical structure is converted to an abnormal conformational isoform (PrPSc) that shows an increase in beta-sheet content. Similarly, the recombinant form of PrPC (ralpha-PrP) can be converted to a conformation dominated by beta-sheet (rbeta-PrP) by reduction and mild acidification in vitro, a process that may mimic in vivo conversion following PrPC internalization during recycling. Despite PrPSc accumulation and prion propagation in the lymphoreticular system before detectable neuroinvasion, no Ab response to PrP has been detected, probably due to immune tolerance. To investigate how the immune system may respond to alpha- and beta-PrP, we immunized Prnp(0/0) mice that are not tolerant of PrP with ralpha-PrP and rbeta-PrP. In this study, we show that although T cells stimulated by these differently folded conformers PrP recognize similar immunodominant epitopes (residues 111-130 and 191-210) the cytokine profile in response to ralpha- and rbeta-PrP was different. Challenge with ralpha-PrP elicited a strong response of IL-5 and IL-10, whereas rbeta-PrP led to an early increased production of IFN-gamma. In addition, immunization with ralpha-PrP led to production of predominantly IgG1 isotype Ab in the sera, whereas after immunization with rbeta-PrP, IgG2b was significantly produced. Thus, both humoral and cellular responses to these differently folded isoforms of the same protein are different, indicating a possible involvement of Th1 and Th2 pathway activation. These differences may be exploitable diagnostically and therapeutically for prion diseases, such as variant Creutzfeldt-Jakob disease.


Assuntos
Doenças Priônicas/imunologia , Príons/química , Príons/imunologia , Conformação Proteica , Animais , Anticorpos Monoclonais/biossíntese , Citocinas/biossíntese , Humanos , Imunização , Epitopos Imunodominantes/química , Isotipos de Imunoglobulinas/biossíntese , Técnicas In Vitro , Ativação Linfocitária , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Doenças Priônicas/metabolismo , Estrutura Secundária de Proteína , Linfócitos T/imunologia
20.
J Immunol ; 174(1): 557-63, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15611283

RESUMO

Abs can tune the responses of Ag-specific T cells by influencing the nature of the epitope repertoire displayed by APCs. We explored the interaction between human self-reactive T cells and human monoclonal autoantibodies from combinatorial Ig-gene libraries derived from autoimmune thyroiditis patients and specific for the main autoantigen thyroid peroxidase (TPO). All human mAbs extensively influenced the T cell epitope repertoire recognized by different TPO-specific T cell clones. The action of the human mAbs was complex, because sometimes the same Ab suppressed or enhanced the epitopes recognized by the 10 different TPO-specific T cell clones. The human mAbs could modulate the epitope repertoire when TPO was added exogenously and when expressed constitutively on the surface of APCs. However, they could not unmask an immunodominant cryptic TPO epitope. In this study, we show that human autoantibodies influence the activity of self-reactive T cells and prove their relevance in concealing or exposing epitopes recognized by self-reactive T cells. However, our results further stress the biological significance of the immunodominant cryptic epitope we have defined and its potential importance in the evolution of autoimmunity.


Assuntos
Anticorpos Monoclonais/imunologia , Autoanticorpos/imunologia , Epitopos de Linfócito T/imunologia , Iodeto Peroxidase/imunologia , Tireoidite Autoimune/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Clonais , Mapeamento de Epitopos , Biblioteca Gênica , Humanos , Iodeto Peroxidase/genética , Camundongos , Tireoidite Autoimune/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...