Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Mucosal Immunol ; 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38570140

RÉSUMÉ

Sublingual allergen immunotherapy (SLIT) is an emerging treatment option for allergic asthma and a potential disease-modifying strategy for asthma prevention. The key cellular events leading to such long-term tolerance remain to be fully elucidated. We administered prophylactic SLIT in a mouse model of house dust mite (HDM)-driven allergic asthma. HDM extract was sublingually administered over 3 weeks followed by intratracheal sensitization and intranasal challenges with HDM. Prophylactic SLIT prevented allergic airway inflammation and hyperreactivity with a low lab-to-lab variation. The HDM-specific T helper (Th)2 (cluster of differentiation 4 Th) response was shifted by SLIT toward a regulatory and Th17 response in the lung and mediastinal lymph node. By using Derp1-specific cluster of differentiation 4+ T cells (1-DER), we found that SLIT blocked 1-DER T cell recruitment to the mediastinal lymph node and dampened IL-4 secretion following intratracheal HDM sensitization. Sublingually administered Derp1 protein activated 1-DER T cells in the cervical lymph node via chemokine receptor7+ migratory dendritic cells (DC). DCs migrating from the oral submucosa to the cervical lymph node after SLIT-induced Foxp3+ regulatory T cells. When mice were sensitized with HDM, prior prophylactic SLIT increased Derp1 specific regulatory T cells (Tregs) and lowered Th2 recruitment in the lung. By using Foxp3-diphtheria toxin receptor mice, Tregs were found to contribute to the immunoregulatory prophylactic effect of SLIT on type 2 immunity. These findings in a mouse model suggest that DC-mediated functional Treg induction in oral mucosa draining lymph nodes is one of the driving mechanisms behind the disease-modifying effect of prophylactic SLIT.

2.
Immunol Rev ; 296(1): 169-190, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32594569

RÉSUMÉ

Therapeutic vaccination offers great promise as an intervention for a diversity of infectious and non-infectious conditions. Given that most chronic health conditions are thought to have an immune component, vaccination can at least in principle be proposed as a therapeutic strategy. Understanding the nature of protective immunity is of vital importance, and the progress made in recent years in defining the nature of pathological and protective immunity for a range of diseases has provided an impetus to devise strategies to promote such responses in a targeted manner. However, in many cases, limited progress has been made in clinical adoption of such approaches. This in part results from a lack of safe and effective vaccine adjuvants that can be used to promote protective immunity and/or reduce deleterious immune responses. Although somewhat simplistic, it is possible to divide therapeutic vaccine approaches into those targeting conditions where antibody responses can mediate protection and those where the principal focus is the promotion of effector and memory cellular immunity or the reduction of damaging cellular immune responses as in the case of autoimmune diseases. Clearly, in all cases of antigen-specific immunotherapy, the identification of protective antigens is a vital first step. There are many challenges to developing therapeutic vaccines beyond those associated with prophylactic diseases including the ongoing immune responses in patients, patient heterogeneity, and diversity in the type and stage of disease. If reproducible biomarkers can be defined, these could allow earlier diagnosis and intervention and likely increase therapeutic vaccine efficacy. Current immunomodulatory approaches related to adoptive cell transfers or passive antibody therapy are showing great promise, but these are outside the scope of this review which will focus on the potential for adjuvanted therapeutic active vaccination strategies.


Sujet(s)
Adjuvants immunologiques , Immunomodulation , Vaccination , Vaccins/immunologie , Vaccins/usage thérapeutique , Animaux , Production d'anticorps/immunologie , Auto-immunité , Prise en charge de la maladie , Humains , Immunité cellulaire , Immunité humorale , Thérapie moléculaire ciblée , Résultat thérapeutique , Vaccination/méthodes , Vaccins/administration et posologie
4.
Int J Nanomedicine ; 12: 8377-8388, 2017.
Article de Anglais | MEDLINE | ID: mdl-29200850

RÉSUMÉ

Immunotherapy by sublingual administration of allergens provides high patient compliance and has emerged as an alternative to subcutaneous immunotherapy for the treatment of IgE-associated allergic diseases. However, sublingual immunotherapy (SLIT) can cause adverse events. Development of allergen delivery systems enabling more efficient delivery and hence lower allergen load might reduce the adverse events. In the present study, we have investigated neutral and cationic liposomes as delivery systems of ovalbumin (OVA), as a model allergen, in an OVA-induced allergic airway inflammation model. We investigated the liposome carriers' ability to improve tolerance induction of antigens compared to the corresponding dose of free OVA. Mice were treated sublingually over 2 weeks with free or liposome encapsulated OVA followed by intraperitoneal injections and intranasal challenge. Mice sublingually treated with OVA-liposomes showed a significant reduction of airway eosinophilia and splenocyte proliferation in comparison to free OVA. A similar nonsignificant pattern was seen for OVA-specific IgE antibodies. In addition, reduced levels of interferon-γ and interleukin-5 were observed in spleen cell culture supernatants from OVA-liposome-treated mice compared to the sham-treated group. In conclusion, in vivo efficacy data showed that prophylactic SLIT with OVA-liposomes is significantly more effective in preventing allergic inflammation than the corresponding dose of free OVA.


Sujet(s)
Allergènes/administration et posologie , Systèmes de délivrance de médicaments , Immunothérapie sublinguale , Allergènes/immunologie , Animaux , Cytokines/métabolisme , Test ELISA , Femelle , Lipides/composition chimique , Liposomes , Souris de lignée BALB C , Ovalbumine/immunologie , Pneumopathie infectieuse/immunologie , Pneumopathie infectieuse/anatomopathologie , Pneumopathie infectieuse/prévention et contrôle , Rate/immunologie
5.
Int Arch Allergy Immunol ; 170(1): 22-34, 2016.
Article de Anglais | MEDLINE | ID: mdl-27287860

RÉSUMÉ

BACKGROUND: Evidence regarding sublingual immunotherapy (SLIT) efficacy and its good safety profile has been demonstrated with pollen and house dust mite (HDM) allergens in the treatment of airway allergies. In addition, the use of grass pollen presents a SLIT disease-modifying treatment for respiratory allergies. OBJECTIVES: The aim of this study was to demonstrate the efficacy of HDM-based SLIT in mouse models of allergic airway inflammation and to gain insights into the involved local immunological mechanisms. METHODS: Balb/c mice were sensitized/challenged with Dermatophagoides farinae (Der f) extract and underwent Der f-SLIT in prophylactic and therapeutic settings. The SLIT efficacy was assessed using lung function measurements, analysis of local inflammatory responses by bronchoalveolar lavage cell differentiation and lung histology. Humoral and cellular responses were monitored by ELISA, cytokine bead array and flow cytometry analyses. RESULTS: In a prophylactic setting, Der f-SLIT with 12 development units per dose reduced the eosinophil-dominated inflammatory response in the lung paralleled by a marked reduction in airway hyperresponsiveness. Local Th2 responses were prevented as demonstrated by significantly lower levels of IL-5 and IL-13. Additionally, SLIT-treated mice revealed a lower proportion of CD4-CD8- x03B3;δ cells and a higher frequency of CD8+CD25+IFNx03B3;+ T cells in the lungs compared to sham-treated mice. In a therapeutic setting, Der f-SLIT also resulted in reduced inflammatory responses in the lung. CONCLUSION: The efficacy of Der f-SLIT was demonstrated in prophylactic and therapeutic conditions using experimental mouse models of HDM-induced airway inflammation. A potential role of a so far underestimated lymphocyte subpopulation was also indicated.


Sujet(s)
Allergènes/immunologie , Antigènes de Dermatophagoides/immunologie , Asthme/immunologie , Asthme/anatomopathologie , Pyroglyphidae/immunologie , Immunothérapie sublinguale , Animaux , Asthme/thérapie , Liquide de lavage bronchoalvéolaire/cytologie , Liquide de lavage bronchoalvéolaire/immunologie , Cytokines/métabolisme , Modèles animaux de maladie humaine , Femelle , Immunoglobuline E/sang , Immunoglobuline E/immunologie , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Activation des lymphocytes/immunologie , Souris , Immunothérapie sublinguale/méthodes , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Résultat thérapeutique
6.
Mol Cell Proteomics ; 14(1): 191-204, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25389185

RÉSUMÉ

Allergenic proteins such as grass pollen and house dust mite (HDM) proteins are known to trigger hypersensitivity reactions of the immune system, leading to what is commonly known as allergy. Key allergenic proteins including sequence variants have been identified but characterization of their post-translational modifications (PTMs) is still limited. Here, we present a detailed PTM(1) characterization of a series of the main and clinically relevant allergens used in allergy tests and vaccines. We employ Orbitrap-based mass spectrometry with complementary fragmentation techniques (HCD/ETD) for site-specific PTM characterization by bottom-up analysis. In addition, top-down mass spectrometry is utilized for targeted analysis of individual proteins, revealing hitherto unknown PTMs of HDM allergens. We demonstrate the presence of lysine-linked polyhexose glycans and asparagine-linked N-acetylhexosamine glycans on HDM allergens. Moreover, we identified more complex glycan structures than previously reported on the major grass pollen group 1 and 5 allergens, implicating important roles for carbohydrates in allergen recognition and response by the immune system. The new findings are important for understanding basic disease-causing mechanisms at the cellular level, which ultimately may pave the way for instigating novel approaches for targeted desensitization strategies and improved allergy vaccines.


Sujet(s)
Allergènes/métabolisme , Antigènes de Dermatophagoides/métabolisme , Antigènes végétaux/métabolisme , Protéines végétales/métabolisme , Polyosides/métabolisme , Maturation post-traductionnelle des protéines , Betula , Spectrométrie de masse , Phleum , Pollen
7.
Ann Allergy Asthma Immunol ; 98(4): 366-72, 2007 Apr.
Article de Anglais | MEDLINE | ID: mdl-17458434

RÉSUMÉ

BACKGROUND: Many studies have demonstrated immunologic changes induced by sublingual immunotherapy (SLIT), but the definitive mechanism of action needs further investigation. OBJECTIVE: To study the immunologic response induced by SLIT in sensitized mice. METHODS: Timothy grass (Phleum pratense)-sensitized mice received SLIT for 2, 4, or 6 weeks at 3 different concentrations, including a buffer control. Serum samples and washes of the lungs (bronchoalveolar lavage [BAL]) and the nasal passages (nasal lavage [NAL]) were analyzed for allergen-specific antibodies. T cells were isolated from the spleen and cervical lymph nodes for the analysis of proliferation and cytokine production. RESULTS: Sublingual immunotherapy in sensitized mice resulted in a 30-fold increase in antigen specific IgA levels in BAL and NAL fluid compared with buffer-treated mice, whereas antigen specific IgE was undetectable in BAL and NAL fluid in animals treated with SLIT. Furthermore, IgA levels were proportional to the dose and duration of SLIT. Levels of specific IgA in serum correlated with levels in BAL and NAL fluid. Serum IgA levels were proportional to the duration of allergen exposure to the oral mucosa. Conversely, no changes in serum levels of IgE and IgG were induced by SLIT. Proliferation of T cells was increased in mice treated with SLIT compared with nontreated mice. CONCLUSION: High levels of IgA in serum and in BAL and NAL fluid of mice treated with SLIT demonstrate that SLIT induces a mucosal, nonallergic response in sensitized mice.


Sujet(s)
Immunothérapie , Phleum , Administration par voie sublinguale , Allergènes/immunologie , Allergènes/usage thérapeutique , Animaux , Femelle , Hypersensibilité/immunologie , Hypersensibilité/thérapie , Immunité muqueuse/immunologie , Immunoglobuline A/sang , Immunoglobuline A/immunologie , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Souris , Souris de lignée BALB C , Phleum/immunologie , Préparations à base de plantes/immunologie , Préparations à base de plantes/usage thérapeutique , Lymphocytes T/immunologie
8.
Am J Physiol Gastrointest Liver Physiol ; 292(6): G1630-40, 2007 Jun.
Article de Anglais | MEDLINE | ID: mdl-17347451

RÉSUMÉ

Previous studies have suggested that intestinal epithelial cells (IECs) have the capacity to function as nonprofessional antigen presenting cells that in the normal state preferentially activate CD8+ T cells. However, under pathological conditions, such as those found in inflammatory bowel disease (IBD), persistent activation of CD4+ T cells is seen. The aim of this study was to determine whether the IBD IECs contribute to CD4+ T cell activation. Freshly isolated human IECs were obtained from surgical specimens of patients with or without IBD and cocultured with autologous or allogeneic peripheral blood T lymphocytes. Cocultures of normal T cells and IECs derived from IBD patients resulted in the preferential activation of CD4+ T cell proliferation that was associated with significant IFN-gamma, but not IL-2, secretion. Cytokine secretion and CD4+ T cell proliferation was inhibited by pretreatment of the IBD IECs with the anti-DR MAb L243. In contrast, normal IECs stimulated the proliferation and cytokine secretion by CD4+ T cells to a significantly lesser degree than IBD IECs. Furthermore, blockade of human leukocyte antigen-DR had a lesser effect in the normal IEC-CD4+ T cell cocultures. We conclude that IECs can contribute to the ongoing CD4+ T cell activation seen in IBD. We suggest that the apparent differences between the secreted levels of IFN-gamma indicate that it may play a dual role in intestinal homeostasis, in which low levels contribute to physiological inflammation whereas higher levels are associated with an uncontrolled inflammatory state.


Sujet(s)
Lymphocytes T CD4+/immunologie , Communication cellulaire , Prolifération cellulaire , Cellules épithéliales/immunologie , Maladies inflammatoires intestinales/immunologie , Interféron gamma/métabolisme , Muqueuse intestinale/immunologie , Activation des lymphocytes , Anticorps monoclonaux , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/anatomopathologie , Cellules cultivées , Techniques de coculture , Rectocolite hémorragique/immunologie , Rectocolite hémorragique/métabolisme , Maladie de Crohn/immunologie , Maladie de Crohn/métabolisme , Cellules épithéliales/anatomopathologie , Antigènes HLA-DR/immunologie , Humains , Maladies inflammatoires intestinales/métabolisme , Maladies inflammatoires intestinales/anatomopathologie , Interleukine-10/métabolisme , Interleukine-2/métabolisme , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie
9.
J Immunol ; 175(6): 3577-83, 2005 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-16148101

RÉSUMÉ

Several cytokines derived from Th3 and Tr1 cells, including IL-10, are believed to regulate oral tolerance, but direct evidence is lacking. We have explored the potential role of IL-10 by generating transgenic (TG) mice with sustained hepatocyte-specific expression of rat IL-10. TG mice expressed rat IL-10 downstream of a transthyretin promoter, which led to serum levels that were increased 10- to 100-fold compared with normal animals. Animals were orally administered 1 mg of whole OVA for 5 consecutive days, with control animals receiving PBS. There were six animal groups: Either OVA or PBS were fed orally to rat IL-10 TG mice, non-TG wild-type mice without IL-10 administration, and non-TG wild-type mice administered rat IL-10 systemically. On day 8, all mice were immunized with two injections of OVA, and then analyzed on day 18. T cell proliferation responses were reduced by 65.8 +/- 14.3% after feeding of OVA in rIL-10 TG animals, compared with 39.4 +/- 15.6% in the non-TG mice (p = 0.02). Anti-OVA titers were expressed as fold increase over naive non-TG mice. After feeding, titers decreased by approximately 33% (from 3- to 2-fold) in TG animals and, to a lesser extent, in non-TG animals. IFN-gamma secretion by cultured popliteal lymphocytes decreased in TG animals by 83% after feeding and by 69% in non-TG animals. IL-4 secretion increased 4-fold in TG-fed mice, but did not significantly change in non-TG OVA-fed animals. In contrast to hepatic TG expression of rIL-10, systemic administration of rIL-10 had only a modest effect on tolerance. IL-10, when transgenically expressed in the liver enhances mucosal tolerance to an oral Ag.


Sujet(s)
Hépatocytes/immunologie , Tolérance immunitaire , Interleukine-10/métabolisme , Muqueuse de la bouche/immunologie , Administration par voie orale , Animaux , Anticorps/sang , Hépatocytes/métabolisme , Immunisation , Interleukine-10/sang , Interleukine-10/génétique , Interleukine-4/métabolisme , Souris , Souris transgéniques , Ovalbumine/administration et posologie , Ovalbumine/immunologie , Rats , Lymphocytes T/immunologie
10.
J Clin Invest ; 115(8): 2234-43, 2005 Aug.
Article de Anglais | MEDLINE | ID: mdl-16041410

RÉSUMÉ

To explore the requirement for M cells and the Peyer's patch (PP) in induction of oral tolerance and address the potential in vivo role of intestinal epithelial cells as nonprofessional APCs, we have attempted to induce tolerance in mice with ligated small bowel loops without M cells and Peyer's patches. A 2-centimeter section of vascularized small bowel was spliced away from the gut without disruption of the mesenteric attachments. We introduced OVA directly into the lumen of the loop prior to footpad immunization. By excising segments of bowel that contain PPs in some mice and segments without patches in others, we could study the necessity of the M cell and the underlying patch versus epithelial cells in induction of mucosal tolerance. We show that OVA-specific T cell proliferation and serum antibody responses are reduced in mice that have previously been given OVA both in PP-containing loops and in loops without patches. Furthermore, both high- and low-dose tolerance could be induced in the absence of PPs. Low-dose tolerance is associated with bystander suppression and requires IL-10, which indicates active suppression and the induction of regulatory cells. These data suggest that there is a critical role for components of the mucosal immune system other than PPs in antigen sampling and induction of oral tolerance.


Sujet(s)
Présentation d'antigène/immunologie , Cellules présentatrices d'antigène/immunologie , Tolérance immunitaire/immunologie , Immunité muqueuse , Intestin grêle/immunologie , Plaques de Peyer/immunologie , Animaux , Effet bystander/immunologie , Prolifération cellulaire , Interleukine-10/immunologie , Muqueuse intestinale/immunologie , Ligature , Souris , Souris de lignée BALB C , Ovalbumine/immunologie , Lymphocytes T/immunologie
11.
J Immunol ; 174(9): 5814-22, 2005 May 01.
Article de Anglais | MEDLINE | ID: mdl-15843585

RÉSUMÉ

Mucosal tolerance is believed to be partly mediated by regulatory T cells. Intestinal epithelial cells (IECs) may play an important role in the generation of such regulatory cells, because they are able to process and present Ag to T cells. Furthermore, we have previously demonstrated that IECs are able to generate regulatory CD8(+) T cells in vitro. In the present study, we have analyzed lamina propria (LP) lymphocytes for the presence of such regulatory CD8(+) T cells in normal individuals as well as in patients with inflammatory bowel disease (IBD). The results of the present study show that LP CD8(+) T cells derived from normal controls possess regulatory activity, whereas both unfractionated LP lymphocytes and purified LP CD4(+) T cells do not. The LP CD8(+) T cells suppress Ig production by pokeweed mitogen-stimulated PBMCs by 31-80%, in a cell contact-dependent manner. No significant difference in suppression between CD28(+) and CD28(-)CD8(+) LP T cells was observed. In contrast to CD8(+) T cells from normal LP, CD8(+) T cells isolated from LP of IBD patients, did not suppress Ig production by pokeweed mitogen-stimulated PBMC (five of six ulcerative colitis specimens; six of six Crohn's disease specimens). Furthermore, we demonstrate that the frequency of TCR Vbeta5.1-positive CD8(+) T cells, which we previously have demonstrated to be regulatory and to be expanded by IECs in vitro, is decreased in IBD LP compared with normal LP. In conclusion, this study demonstrates that CD8(+) T cells with regulatory activity are present in the LP of normal healthy individuals, but not in patients with IBD, suggesting that these cells might play an active role in mucosal tolerance.


Sujet(s)
Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/anatomopathologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/métabolisme , Communication cellulaire/immunologie , Cellules cultivées , Techniques de coculture , Cytokines/pharmacologie , Femelle , Humains , Immunoglobulines/biosynthèse , Maladies inflammatoires intestinales/métabolisme , Muqueuse intestinale/cytologie , Agranulocytes/immunologie , Agranulocytes/métabolisme , Activation des lymphocytes/immunologie , Numération des lymphocytes , Test de culture lymphocytaire mixte , Mâle , Adulte d'âge moyen , Récepteurs aux antigènes des cellules T/biosynthèse , Lymphocytes T régulateurs/cytologie , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme
12.
Gastroenterology ; 126(5): 1347-57, 2004 May.
Article de Anglais | MEDLINE | ID: mdl-15131796

RÉSUMÉ

BACKGROUND & AIMS: Previous studies have suggested that intestinal epithelial cells (IECs) may function as antigen-presenting cells for CD4+ and CD8+ T cells. However, these cells fail to express conventional costimulatory molecules (CD80, CD86), leading to the possibility that antigen presented by normal IECs could result in anergy. Other members of the B7 family have recently been identified. B7h interacts with inducible costimulator (ICOS) on T cells and provides a positive signal, whereas B7-H1 and B7-DC interact with PD-1 and transmit an inhibitory signal. Our aim was to determine whether IECs express novel B7 family members and whether these molecules play a role in IEC:T-cell interactions. METHODS: B7h and B7-H1 expression was assessed in isolated IECs and IEC lines. The functional role of B7h and B7-H1 in the interaction between IECs and T cells was assessed in coculture experiments using purified anti-B7h or B7-H1 monoclonal antibodies (mAbs), B7h immunoglobulin (Ig), or B7-H1 fusion proteins. RESULTS: B7h and B7-H1 messenger RNA was detected in IEC lines and IECs from healthy controls and patients with inflammatory bowel disease (IBD). IECs from patients with IBD but not healthy controls expressed B7h and B7-H1 protein on their surface. Proliferation of IEC-stimulated T cells was inhibited only by B7h immunoglobulin treatment, whereas interferon gamma secretion in these cocultures was inhibited by both anti-B7h mAb and B7h Ig. No difference was seen between IBD or normal IEC populations. CONCLUSIONS: These data suggest that the B7h-ICOS costimulatory pathway may be important in IEC:T-cell interactions.


Sujet(s)
Antigène CD80 , Protéines du sang/physiologie , Côlon/métabolisme , Muqueuse intestinale/métabolisme , Peptides/physiologie , Protéines/physiologie , Antigènes CD , Antigène CD274 , Protéines du sang/antagonistes et inhibiteurs , Protéines du sang/métabolisme , Division cellulaire , Lignée cellulaire tumorale , Techniques de coculture , Humains , Ligand de la protéine inductible de costimulation du lymphocyte T , Interféron gamma/métabolisme , Muqueuse intestinale/cytologie , Glycoprotéines membranaires , Peptides/antagonistes et inhibiteurs , Peptides/métabolisme , Protéines/antagonistes et inhibiteurs , Protéines/métabolisme , Lymphocytes T/cytologie
13.
Ann N Y Acad Sci ; 1029: 22-35, 2004 Dec.
Article de Anglais | MEDLINE | ID: mdl-15681740

RÉSUMÉ

Intestinal epithelial cells may play a role in the regulation of immune responses toward luminal antigens. We show that a subset of CD8(+) T cells undergoes oligoclonal expansion in the intestinal mucosa, probably through interaction with a unique complex expressed on epithelial cells, formed by a CEA subfamily member (gp180) and CD1d. This subset, which is regulatory in vitro, may play a role in the control of intestinal immune responses toward luminal antigens. A lack of expansion of these CD8(+) regulatory T cells, probably related to the defective expression of the gp180/CD1d complex, is observed in inflammatory bowel disease.


Sujet(s)
Lymphocytes T CD8+/immunologie , Immunité muqueuse , Muqueuse intestinale/immunologie , Lymphocytes/immunologie , Lignée cellulaire , Techniques de coculture , Fluorescéines , Colorants fluorescents , Humains , Activation des lymphocytes , Lymphocytes/anatomopathologie , Succinimides , Lymphocytes T/immunologie
14.
Eur J Gastroenterol Hepatol ; 15(3): 267-73, 2003 Mar.
Article de Anglais | MEDLINE | ID: mdl-12610322

RÉSUMÉ

OBJECTIVE: In acute stages of ulcerative colitis (UC), neutrophils migrate from the circulation into inflamed colonic tissue, initiated by yet unknown stimuli. The bacterial peptide N-formyl-methionyl-leucyl-phenylalanine (FMLP) is a component of the surface membrane of colonic bacteria such as Escherichia coli and stimulates Ca2+ influx into neutrophils, reflecting the fact that ionized calcium is an important secondary messenger for several neutrophil functions, including locomotion, phagocytosis and free oxygen radical production. Recent studies have revealed that Ca2+ dependent ICAM-1/beta 2-integrin mediated neutrophil migration is impaired in UC patients. The aim of the present work was to study the influx of Ca2+ into peripheral blood neutrophils of UC patients after exposure to FMLP and after binding of either beta 2-integrins or intercellular adhesion molecule-1 (ICAM-1). METHODS: The relative intracellular Ca2+ levels ([Ca2+]i ) were measured spectrofluorometrically in neutrophils isolated from eight UC patients and eight controls. The cells were exposed to 1 nm FMLP, 5 pm free ICAM-1, or antibodies binding ICAM-1 or the beta 2-integrins CD11a, CD11b, CD11c and CD18. RESULTS: A pronounced increase in [Ca2+]i was observed by exposure of cells to FMLP, and neutrophils from UC patients showed a consistent and significant delayed response as compared to cells from control subjects (P < 0.01). Antibody mediated cross-linking of CD18 triggered a small but detectable increase in [Ca2+]i, which did not differ between patients and controls. CONCLUSION: A delayed response to bacterial peptides appears to be a phenotypic trait for neutrophils of UC patients. A connection between FMLP stimulated Ca2+ influx and CD11/CD18 upregulation is discussed.


Sujet(s)
Calcium/sang , Rectocolite hémorragique/sang , N-Formyl-méthionyl-leucyl-phénylalanine/pharmacologie , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Adulte , Sujet âgé , Antigènes CD11/sang , Molécules d'adhérence cellulaire/sang , Mouvement cellulaire , Cellules cultivées , Cytosol/métabolisme , Humains , Mâle , Adulte d'âge moyen , Granulocytes neutrophiles/métabolisme , Facteurs temps , Régulation positive/effets des médicaments et des substances chimiques
15.
Gastroenterology ; 123(5): 1516-26, 2002 Nov.
Article de Anglais | MEDLINE | ID: mdl-12404227

RÉSUMÉ

BACKGROUND & AIMS: Regulatory T cells play a role in the control of immune responses in the intestinal mucosa and their absence may predispose to inflammatory bowel disease (IBD). We have previously shown that T cells activated by intestinal epithelial cells (IECs) are suppressive in function. Our goal was to characterize the phenotype and function of T cells proliferating after interaction with IECs. METHODS: Irradiated human IECs, isolated from normal resection specimens, were cultured with carboxy fluorescein succinimidyl ester (CFSE) labeled T cells. Flow cytometric analysis of T cells was performed at days 5-10. CD8+ T cells proliferating in culture with IECs were sorted and added to suppressive assays. RESULTS: The precursor frequency of T cells proliferating in response to IECs ranged from 0.3%-0.9%. Several subpopulations were shown to proliferate (CD8+CD28-/CD8+CD28+/CD4+CD25+), but one population (CD8+CD28-CD101+CD103+) appeared to be dependent on contact with the CD8 ligand gp180. After sorting, culture in the presence of interleukin (IL)-7 and IL-15 allowed for the generation of cell lines. IEC-activated CD8+ T cells, but not nonactivated CD8+ T cells, were suppressive in function. Suppression belonged to the CD101+CD103+ subset of IEC-activated CD8+ T cells and appeared to require cell contact. CD8+ lamina propria T cells also showed suppressive function, suggesting the presence of CD8+ regulatory T cells in the mucosa. CONCLUSIONS: IECs are able to induce the proliferation of a small fraction of CD8+ peripheral T cells. The CD8+CD28- subset of IEC-activated CD8+ T cells, which express CD101 and CD103, interacts with IECs through gp180 and has regulatory function.


Sujet(s)
Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/physiologie , Communication cellulaire/physiologie , Muqueuse intestinale/physiologie , Antigènes CD/métabolisme , Marqueurs biologiques , Division cellulaire/physiologie , Cellules cultivées , Techniques de coculture , Humains , Intégrines alpha/métabolisme , Muqueuse intestinale/cytologie , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/physiologie , Phénotype , Sous-populations de lymphocytes T/cytologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE