Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
Genes Immun ; 16(6): 378-87, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-25996526

RÉSUMÉ

Inflammatory immune disorders such as inflammatory bowel disease and multiple sclerosis are major health problems. Currently, the intestinal whipworm Trichuris suis is being explored in clinical trials to reduce inflammation in these diseases; however, the mechanisms by which the parasite affects the host immune system are not known. Here we determined the effects of T. suis soluble products (SPs) on Toll-like receptor-4 (TLR4)-stimulated human dendritic cells (DCs) using Illumina bead chip gene arrays. Pathway analysis of lipopolysaccharide-stimulated DCs with or without T. suis treatment showed that co-stimulation with T. suis SPs resulted in a downregulation of both the myeloid differentiation primary response gene 88-dependent and the TIR-domain-containing adaptor-inducing interferon-ß-dependent signalling pathways triggered by TLR4. These data were verified using quantitative real-time PCR of several key genes within these pathways and/or defining their protein levels. In addition, T. suis SPs induce Rab7b, a negative regulator of TLR4 signalling that interferes with its trafficking, which coincided with a reduced surface expression of TLR4. These data indicate that the mechanism by which T. suis SPs reduce inflammatory responses is through suppression of both TLR4 signalling and surface expression on DCs.


Sujet(s)
Cellules dendritiques/parasitologie , Récepteur de type Toll-4/métabolisme , Trichuris/immunologie , Protéines G rab/métabolisme , Animaux , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Régulation négative , Humains , Inflammation/immunologie , Inflammation/parasitologie , Inflammation/thérapie , Lipopolysaccharides/pharmacologie , Réaction de polymérisation en chaine en temps réel , Transduction du signal , Transcriptome , Protéines Rab7 liant le GTP
2.
Genes Immun ; 15(7): 477-86, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-25008860

RÉSUMÉ

Recent clinical trials in patients with inflammatory diseases like multiple sclerosis (MS) or inflammatory bowel disease (IBD) have shown the beneficial effects of probiotic helminth administration, although the underlying mechanism of action remains largely unknown. Potential cellular targets may include innate immune cells that propagate inflammation in these diseases, like pro-inflammatory macrophages. We here investigated the effects of the helminth Trichuris suis soluble products (SPs) on the phenotype and function of human inflammatory (granulocyte-macrophage colony-stimulating factor (GM-CSF)-differentiated) macrophages. Interestingly, we here show that T. suis SPs potently skew inflammatory macrophages into a more anti-inflammatory state in a Toll-like receptor 4 (TLR4)-dependent manner, and less effects are seen when stimulating macrophages with TLR2 or -3 ligands. Gene microarray analysis of GM-CSF-differentiated macrophages further revealed that many TLR4-induced inflammatory mediators, including interleukin (IL)-12B, CCL1 and CXCL9, are downregulated by T. suis SPs. In particular, we observed a strong reduction in the expression and function of P2RX7, a purinergic receptor involved in macrophage inflammation, leading to reduced IL-1ß secretion. In conclusion, we show that T. suis SPs suppress a broad range of inflammatory pathways in GM-CSF-differentiated macrophages in a TLR4-dependent manner, thereby providing enhanced mechanistic insight into the therapeutic potential of this helminth for patients with inflammatory diseases.


Sujet(s)
Protéines d'helminthes/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme , Trichuris/immunologie , Animaux , Cellules cultivées , Chimiokine CCL1/génétique , Chimiokine CCL1/métabolisme , Chimiokine CXCL9/génétique , Chimiokine CXCL9/métabolisme , Facteur de stimulation des colonies de granulocytes et de macrophages/métabolisme , Protéines d'helminthes/immunologie , Humains , Immunité innée , Inflammation/immunologie , Inflammation/métabolisme , Sous-unité p40 de l'interleukine-12/génétique , Sous-unité p40 de l'interleukine-12/métabolisme , Macrophages/immunologie , Récepteurs purinergiques P2X7/génétique , Récepteurs purinergiques P2X7/métabolisme , Trichuris/composition chimique
3.
Vascul Pharmacol ; 56(5-6): 297-305, 2012.
Article de Anglais | MEDLINE | ID: mdl-22387744

RÉSUMÉ

In this review we compare expression studies on monocyte subsets as an example to show the integrated possibilities of molecular databases and bioinformatic analysis tools. Monocytes have been recognized as cells with great plasticity and differentiation potential that play a pivotal role in revascularization processes, i.e. angiogenesis and arteriogenesis. To gain more insight in the relevant developmental programs, we compared the full-genome mRNA expression profiles of several distinct human monocyte subpopulations previously identified based on surface marker expression. These included classical and non-classical, M1 and M2 macrophages, circulating angiogenic cells (CAC), and non-monocyte-derived endothelial colony-forming cells (ECFC). Their transcriptional profiles revealed distinct and overlapping gene expression signatures and pathways reminiscent of utilization of transcription factors driving polarization into the different monocytic phenotypes. Hierarchical cluster analysis revealed that CAC are most related to M2 macrophages and unstimulated macrophages, and to a lesser extent to classical monocytes, and are quite distinct from M1 macrophages and ECFC. Analysis of the promoter region of CAC-expressed genes suggests that in particular the ETS family of transcription factors is important in CAC development. These analyses show the power of combining multiple datasets with existing databases on biological knowledge, to interpret full genome expression data.


Sujet(s)
Biologie informatique/méthodes , Monocytes/métabolisme , Néovascularisation physiologique/physiologie , Différenciation cellulaire , Analyse de regroupements , Interprétation statistique de données , Bases de données factuelles , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes/physiologie , Génome humain , Humains , Macrophages/métabolisme
4.
Genes Immun ; 12(6): 466-72, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-21471993

RÉSUMÉ

Interferon-ß (IFNß) therapy is effective in approximately half of the patients with relapsing-remitting multiple sclerosis (RRMS). Clinical non-responders were characterized by an increased expression of IFN response genes before the start of therapy, and a lack of a pharmacologically induced increase in IFN response gene activity. Because Interferon Regulatory Factor 5 (IRF5) is a master regulator of IFN-activity, we carried out a candidate gene study of IRF5 gene variants in relation to the pharmacological and clinical response upon IFNß treatment. We found that patients with the IRF5 rs2004640-TT and rs47281420-AA genotype exerted a poor pharmacological response to IFNß compared with patients carrying the respective G-alleles (P=0.0006 and P=0.0023, respectively). Moreover, patients with the rs2004640-TT genotype developed more magnetic resonance imaging (MRI)-based T2 lesions during IFNß treatment (P=0.003). Accordingly, an association between MRI-based non-responder status and rs2004640-TT genotype was observed (P=0.010). For the rs4728142-AA genotype a trend of an association with more T2 lesions during IFNß treatment and MRI-based non-responder status was observed (P=0.103 and P=0.154, respectively). The clinical relevance of the rs2004640-TT genotype was validated in an independent cohort wherein a shorter time to first relapse was found (P=0.037). These findings suggest a role for IRF5 gene variation in the pharmacological and clinical outcome of IFNß therapy that might have relevance as biomarker to predict the response to IFNß in multiple sclerosis.


Sujet(s)
Facteurs de régulation d'interféron/génétique , Interféron bêta/usage thérapeutique , Sclérose en plaques récurrente-rémittente/traitement médicamenteux , Adulte , Marqueurs biologiques , Études de cohortes , Femelle , Expression des gènes , Variation génétique , Génotype , Humains , Interféron bêta/pharmacologie , Imagerie par résonance magnétique , Mâle , Sclérose en plaques récurrente-rémittente/génétique , Polymorphisme de nucléotide simple , Résultat thérapeutique
5.
Genes Immun ; 11(8): 622-9, 2010 Dec.
Article de Anglais | MEDLINE | ID: mdl-20555356

RÉSUMÉ

To provide insight into the pharmacological changes in the peripheral blood (PB) molecular profile induced by tumor necrosis factor (TNF)-blockade in patients with rheumatoid arthritis (RA), blood was obtained in PAXgene tubes from 33 RA patients before and 1 month after TNF-blocking therapy (infliximab). From 15 randomly chosen patients pre- and post-treatment gene expression profiles were determined. The remaining 18 RA patients served as validation cohort. A group-based paired analysis of the gene expression profiles from the post- vs pre-treatment samples revealed a signature of genes significantly regulated by TNF-blockade. Downregulated genes reflected several biological pathways such as inflammation, angiogenesis, B- and T-cell activation. Further analysis revealed that the pharmacological response signature was significantly regulated in all treated patients, irrespective of clinical response, which is indicative for the presence of an active TNF pathway in all RA patients. The data imply that all patients carried features of TNF bioactivity irrespective of clinical response. These results favor a model for the parallel presence of TNF-dependent and TNF-independent pathways in the individual RA patient. Clinical response status to TNF-blockade may be dependent on the relative contribution of TNF-independent effector pathways.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Antirhumatismaux/pharmacologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/génétique , Régulation de l'expression des gènes , Adulte , Sujet âgé , Polyarthrite rhumatoïde/sang , Polyarthrite rhumatoïde/anatomopathologie , Cellules sanguines/métabolisme , Études cas-témoins , Femelle , Analyse de profil d'expression de gènes , Humains , Infliximab , Mâle , Adulte d'âge moyen , Pharmacogénétique , Transduction du signal/génétique , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Facteur de nécrose tumorale alpha/métabolisme
6.
Genes Immun ; 10(3): 210-8, 2009 Apr.
Article de Anglais | MEDLINE | ID: mdl-19129850

RÉSUMÉ

The objective of this study was to identify molecular profiles that may distinguish clinical subtypes in systemic sclerosis (SSc). Large-scale gene expression profiling was performed on peripheral blood (PB) from 12 SSc patients and 6 healthy individuals. Significance analysis of microarrays, two-way hierarchical cluster analysis and PANTHER (Protein ANalysis THrough Evolutionary Relationships) ontology classification were used to analyze the data. Quantitative PCR was applied for validation in a cohort of 43 SSc patients. The results show that the expression of genes involved in immune defense, cell cycle and signal transduction was significantly elevated in PB of SSc patients (n=12) compared with healthy individuals (n=6). SSc patients could be stratified into subgroups based on differential expression of genes induced by type I interferon (IFN) and genes involved in antimicrobial (AM) activity. Differential expression of type I IFN or AM signature genes was validated and extended in an independent cohort of 31 patients by quantitative PCR. Low expression of IFN response genes was associated with the presence of anti-centromere antibodies, whereas increased expression was associated with the appearance of digital ulcers. In conclusion, patients with SSc can be classified on the basis of differential expression of immune defense genes. Differences in the activity of the type I IFN response program stratify patients into two clinically relevant subgroups.


Sujet(s)
Anticorps antinucléaires/immunologie , Centromère/immunologie , Interféron de type I/génétique , Sclérodermie systémique/génétique , Ulcère cutané/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études de cohortes , Régulation négative/génétique , Régulation négative/immunologie , Femelle , Doigts , Analyse de profil d'expression de gènes , Humains , Interféron de type I/immunologie , Mâle , Adulte d'âge moyen , Sclérodermie systémique/classification , Sclérodermie systémique/immunologie , Ulcère cutané/immunologie , Régulation positive/génétique , Régulation positive/immunologie
7.
Genes Immun ; 9(1): 16-22, 2008 Jan.
Article de Anglais | MEDLINE | ID: mdl-17928867

RÉSUMÉ

Rheumatoid arthritis (RA) is a heterogeneous disease with unknown etiology. Here we aimed to distinguish RA subtypes based on peripheral blood (PB) gene expression profiles in comparison with a pathogen-response transcriptional program. PB was obtained from 35 RA patients and 15 healthy individuals. For expression profiling we used DNA microarrays. A combined cluster analysis of RA and control samples together with samples from a viral infection model revealed that the gene expression profile of a subgroup of RA patients (RA(A)) was reminiscent to that of poxvirus-infected macaques. Statistical analysis, followed by Gene Ontology analysis of the RA(A) patients confirmed that these patients form a distinct group, with activation of several host defense mechanisms that resemble a common host-pathogen response. Analysis of the promoter region of genes that were overexpressed in the RA(A) patients, revealed an enrichment of transcription factor binding sites for NF kappaB and interferon-activated transcription factors. Moreover, this subgroup of RA patients expressed significantly increased titers of anti-cyclic citrullinated peptide antibodies. We conclude that activation of a host-pathogen response defines a subgroup of RA patients characterized by increased autoreactivity against citrullinated proteins.


Sujet(s)
Polyarthrite rhumatoïde/classification , Polyarthrite rhumatoïde/génétique , Analyse de profil d'expression de gènes , Agranulocytes/métabolisme , Facteurs de transcription/métabolisme , Animaux , Polyarthrite rhumatoïde/sang , Études cas-témoins , Analyse de regroupements , Femelle , Régulation de l'expression des gènes , Gènes viraux , Interactions hôte-parasite , Humains , Agranulocytes/cytologie , Macaca/virologie , Mâle , Adulte d'âge moyen , Séquençage par oligonucléotides en batterie , Régions promotrices (génétique) , Variole , Facteurs de transcription/génétique
8.
Ann Rheum Dis ; 66(8): 1008-14, 2007 Aug.
Article de Anglais | MEDLINE | ID: mdl-17223656

RÉSUMÉ

BACKGROUND: Rheumatoid arthritis (RA) is a heterogeneous disease with unknown cause. AIM: To identify peripheral blood (PB) gene expression profiles that may distinguish RA subtypes. METHODS: Large-scale expression profiling by cDNA microarrays was performed on PB from 35 patients and 15 healthy individuals. Differential gene expression was analysed by significance analysis of microarrays (SAM), followed by gene ontology analysis of the significant genes. Gene set enrichment analysis was applied to identify pathways relevant to disease. RESULTS: A substantially raised expression of a spectrum of genes involved in immune defence was found in the PB of patients with RA compared with healthy individuals. SAM analysis revealed a highly significant elevated expression of interferon (IFN) type I regulated genes in patients with RA compared with healthy individuals, which was confirmed by gene ontology and pathway analysis, suggesting that this pathway was activated systemically in RA. A quantitative analysis revealed that increased expression of IFN-response genes was characteristic of approximately half of the patients (IFN(high) patients). Application of pathway analysis revealed that the IFN(high) group was largely different from the controls, with evidence for upregulated pathways involved in coagulation and complement cascades, and fatty acid metabolism, while the IFN(low) group was similar to the controls. CONCLUSION: The IFN type I signature defines a subgroup of patients with RA, with a distinct biomolecular phenotype, characterised by increased activity of the innate defence system, coagulation and complement cascades, and fatty acid metabolism.


Sujet(s)
Polyarthrite rhumatoïde/génétique , Analyse de profil d'expression de gènes , Interféron de type I/génétique , Séquençage par oligonucléotides en batterie , Régulation positive , Adulte , Polyarthrite rhumatoïde/immunologie , Coagulation sanguine/génétique , Activation du complément/génétique , Femelle , Humains , Mâle , Adulte d'âge moyen
9.
Genes Immun ; 7(6): 522-31, 2006 Sep.
Article de Anglais | MEDLINE | ID: mdl-16837931

RÉSUMÉ

Given the heterogeneous nature of multiple sclerosis (MS), we applied DNA microarray technology to determine whether variability is reflected in peripheral blood (PB) cells. In this study, we studied whole-blood gene expression profiles of 29 patients with relapsing-remitting MS (RRMS) and 25 age- and sex-matched healthy controls. We used microarrays with a complexity of 43K cDNAs. The data were analyzed using sophisticated pathway-level analysis in order to provide insight into the deregulated peripheral immune response programs in MS. We found a remarkable elevated expression of a spectrum of genes known to be involved in immune defense in the PB of MS patients compared to healthy individuals. Cluster analysis revealed that the increased expression of these genes was characteristic for approximately half of the patients. In addition, the gene signature in this group of patients was comparable with a virus response program. We conclude that the transcriptional signature of the PB cells reflects the heterogeneity of MS and defines a sub-population of RRMS patients, who exhibit an activated immune defense program that resembles a virus response program, which is supportive for a link between viruses and MS.


Sujet(s)
Sclérose en plaques récurrente-rémittente/génétique , Sclérose en plaques récurrente-rémittente/immunologie , Études cas-témoins , Analyse de regroupements , Régulation de l'expression des gènes , Hétérogénéité génétique , Humains , Interféron de type I/immunologie , Interféron de type I/métabolisme , Sclérose en plaques récurrente-rémittente/sang , Séquençage par oligonucléotides en batterie , Infections à Poxviridae/génétique , Transduction du signal , Régulation positive
10.
Tissue Antigens ; 63(6): 538-46, 2004 Jun.
Article de Anglais | MEDLINE | ID: mdl-15140029

RÉSUMÉ

Interleukin-12 (IL-12) is a potent inducer of interferon-gamma production by T cells and is a major factor for the development of T-helper 1 (Th1) cells. It exerts its biological effects through binding to the IL-12 receptor (IL-12R), a heterodimer composed of a 1 and a beta2 subunits. The signaling beta2 chain is expressed on Th1 cells and to a lesser extent on Th0 cells, but not on Th2 cells, rendering these latter cells unresponsive to IL-12. Polymorphisms in the coding region of the IL-12Rbeta2 gene were shown to be associated with atopic disease. Here, we analyzed the 5'-regulatory region of the human IL-12Rbeta2 gene by denaturing high-performance liquid chromatography (Transgenomic WAVE system, San Jose, CA). We found five novel single-nucleotide polymorphisms (SNPs) in the proximal 1.2 kb IL-12Rbeta2 promoter region, i.e. -237C/T, -465A/G, -1023A/G, -1033T/C, and -1035A/G. SNP -465A/G is of particular interest as it determines the integrity of a GATA consensus site. By functional comparison of both -465 alleles in transient transfection assays, we show that promoter activity is increased in case of the -465G allele, disrupting the intact GATA site. Comparison of the prevalence of -465A/G SNP alleles in small cohorts of allergic asthmatic and healthy control individuals provided no evidence for an altered distribution in the asthmatic population. In conclusion, we have identified a novel polymorphic GATA site that may affect transciptional activity of the human IL-12Rbeta2 gene under GATA3-mediated, Th2-polarizing conditions.


Sujet(s)
Régions promotrices (génétique) , Récepteurs aux interleukines/génétique , Région 5' flanquante , Asthme/génétique , Séquence nucléotidique , Séquence consensus , Humains , Données de séquences moléculaires , Polymorphisme de nucléotide simple , Récepteurs à l'interleukine-12
11.
Ann Rheum Dis ; 63(3): 233-9, 2004 Mar.
Article de Anglais | MEDLINE | ID: mdl-14962955

RÉSUMÉ

BACKGROUND: Expression of signal transducer and activator of transcription 1 (STAT1), the mediator of interferon (IFN) signalling, is raised in synovial tissue (ST) from patients with rheumatoid arthritis (RA). OBJECTIVES: To determine the extent to which this pathway is activated by phosphorylation in RA synovium. Additionally, to investigate the cellular basis of STAT1 activation in RA ST. METHODS: ST specimens from 12 patients with RA and 14 disease controls (patients with osteoarthritis and reactive arthritis) were analysed by immunohistochemistry, using antibodies to STAT1, tyrosine phosphorylated STAT1, and serine phosphorylated STAT1. Lysates of cultured fibroblast-like synoviocytes stimulated with IFNbeta were analysed by western blotting. Phenotypic characterisation of cells expressing STAT1 in RA ST was performed by double immunolabelling for STAT1 and CD3, CD22, CD55, or CD68. RESULTS: Raised levels of total STAT1 protein and both its activated tyrosine and serine phosphorylated forms were seen in RA synovium as compared with controls. STAT1 was predominantly abundant in T and B lymphocytes in focal inflammatory infiltrates and in fibroblast-like synoviocytes in the intimal lining layer. Raised levels of STAT1 are sustained in cultured RA compared with OA fibroblast-like synoviocytes, and STAT1 serine and tyrosine phosphorylation is rapidly induced upon stimulation with IFNbeta. CONCLUSION: These results demonstrate activation of the STAT1 pathway in RA synovium by raised STAT1 protein expression and concomitantly increased tyrosine (701) and serine (727) phosphorylation. High expression of STAT1 is intrinsic to RA fibroblast-like synoviocytes in the intimal lining layer, whereas activation of the pathway by phosphorylation is an active process.


Sujet(s)
Polyarthrite rhumatoïde/métabolisme , Protéines de liaison à l'ADN/analyse , Transduction du signal/physiologie , Membrane synoviale/métabolisme , Transactivateurs/analyse , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Arthrite réactionnelle/métabolisme , Technique de Western/méthodes , Cellules cultivées , Femelle , Humains , Immunohistochimie/méthodes , Interféron bêta/pharmacologie , Mâle , Microscopie de fluorescence , Adulte d'âge moyen , Arthrose/métabolisme , Phosphorylation , Facteur de transcription STAT-1 , Activation chimique
12.
Genes Immun ; 4(3): 187-96, 2003 Apr.
Article de Anglais | MEDLINE | ID: mdl-12700593

RÉSUMÉ

Rheumatoid arthritis (RA) is a heterogeneous disease. We used cDNA microarray technology to subclassify RA patients and disclose disease pathways in rheumatoid synovium. Hierarchical clustering of gene expression data identified two main groups of tissues (RA-I and RA-II). A total of 121 genes were significantly higher expressed in the RA-I tissues, whereas 39 genes were overexpressed in the RA-II tissues. Among the 121 genes overexpressed in RA-I tissues, a relative majority of nine genes are located on chromosome 6p21.3. An interpretation of biological processes that take place revealed that the gene expression profile in RA-I tissues is indicative for an adaptive immune response. The RA-II group showed expression of genes suggestive for fibroblast dedifferentiation. Within the RA-I group, two subgroups could be distinguished; the RA-Ia group showed predominantly immune-related gene activity, while the RA-Ib group showed an additional higher activity of genes indicative for the classical pathway of complement activation. All tissues except the RA-Ia subgroup showed elevated expression of genes involved in tissue remodeling. These results confirm the heterogeneous nature of RA and suggest the existence of distinct pathogenic mechanisms that contribute to RA. The differences in expression profiles provide opportunities to stratify patients based on molecular criteria.


Sujet(s)
Polyarthrite rhumatoïde/génétique , Membrane synoviale/métabolisme , Polyarthrite rhumatoïde/classification , Polyarthrite rhumatoïde/métabolisme , Cartographie chromosomique , Chromosomes humains de la paire 6 , Fibroblastes/métabolisme , Analyse de profil d'expression de gènes , Humains , Séquençage par oligonucléotides en batterie , Phylogenèse
13.
Genes Immun ; 3(7): 436-9, 2002 Nov.
Article de Anglais | MEDLINE | ID: mdl-12424628

RÉSUMÉ

IL-13 is strongly implicated in the development of asthma and chronic obstructive pulmonary disease (COPD). We previously identified an IL-13 promoter polymorphism (-1055 C to T) that is associated with allergic asthma. We now report an increased frequency of the -1055 T allele in COPD patients compared to healthy controls (P=0.002) and compared to a second control group consisting of smoking individuals with normal lung function (P=0.01). A closely linked IL-13 exon polymorphism is present at normal allelic frequencies (P=0.3 and 0.4, respectively). In addition, we observed a normal distribution of two IL-4 polymorphisms at positions -590 and +33 (P=0.2 and 0.9, respectively). These results could implicate a functional role for the IL-13 promoter polymorphism in the enhanced risk to develop COPD.


Sujet(s)
Prédisposition génétique à une maladie , Interleukine-13/génétique , Régions promotrices (génétique) , Broncho-pneumopathie chronique obstructive/génétique , Adulte , Sujet âgé , Femelle , Humains , Immunoglobuline E/sang , Mâle , Adulte d'âge moyen , Polymorphisme génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE