Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Eur J Pharm Sci ; 192: 106670, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38070782

ABSTRACT

Aggregation has been widely described as a factor contributing to therapeutic antibody immunogenicity. Although production of high-affinity anti-drug antibodies depends on the activation of CD4 T lymphocytes, little is known about the T-cell response induced by antibody aggregates, especially for aggregates produced in mild conditions resulting from minor handling errors of vials. Large insoluble infliximab (IFX) aggregates produced in severe elevated temperature stress conditions have been previously shown to induce human monocyte-derived dendritic cell (moDC) maturation. We here showed that large IFX aggregates recruit in vitro a significantly higher number of CD4 T-cells compared to native IFX. Moreover, a larger array of T-cell epitopes encompassing the entire variable regions was evidenced compared to the native antibody. We then compared the responses of moDCs to different types of aggregates generated by submitting IFX to mild conditions of various times of incubation at an elevated temperature. Decreasing stress duration reduced aggregate size and quantity, and subsequently altered moDC activation. Of importance, IFX aggregates generated in mild conditions and not altering moDC phenotype generated an in vitro T-cell response with a higher frequency of CD4 T cells compared to native IFX. Moreover, cross-reactivity studies of aggregate-specific T cells showed that some T cells could recognize both native and aggregated IFX, while others responded only to IFX aggregates. Taken together, our results suggest that aggregation of antibodies in mild elevated temperature stress conditions is sufficient to alter moDC phenotype in a dose-dependent manner and to increase T-cell response.


Subject(s)
CD4-Positive T-Lymphocytes , Monocytes , Humans , Infliximab/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Temperature
2.
Eur J Immunol ; 54(2): e2250340, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37985174

ABSTRACT

Internalization and processing by antigen-presenting cells such as dendritic cells (DCs) are critical steps for initiating a T-cell response to therapeutic antibodies. Consequences are the production of neutralizing antidrug antibodies altering the clinical response, the presence of immune complexes, and, in some rare cases, hypersensitivity reactions. In recent years, significant progress has been made in the knowledge of cellular uptake mechanisms of antibodies in DCs. The uptake of antibodies could be directly related to their immunogenicity by regulating the quantity of materials entering the DCs in relation to antibody structure. Here, we summarize the latest insights into cellular uptake mechanisms and pathways in DCs. We highlight the approaches to study endocytosis, the impact of endocytosis routes on T-cell response, and discuss the link between how DCs internalize therapeutic antibodies and the potential mechanisms that could give rise to immunogenicity. Understanding these processes could help in developing assays to evaluate the immunogenicity potential of biotherapeutics.


Subject(s)
Antibodies , Dendritic Cells , Antibodies/metabolism , T-Lymphocytes , Endocytosis
3.
J Pharm Sci ; 110(2): 952-958, 2021 02.
Article in English | MEDLINE | ID: mdl-33220239

ABSTRACT

Sucrose is a commonly used stabilizing excipient in protein formulations. However, recent studies have indicated the presence of nanoparticulate impurities (NPIs) in the size range of 100-200 nm in pharmaceutical-grade sucrose. Furthermore, isolated NPIs have been shown to induce protein aggregation when added to monoclonal antibody formulations. Moreover, nanoparticles are popular vaccine delivery systems used to increase the immunogenicity of antigens. Therefore, we hypothesized that NPIs may have immunostimulatory properties. In this study, we evaluated the immunomodulatory effects of NPIs in presence and absence of trastuzumab in vitro with monocyte-derived dendritic cells (moDCs). Exposure of trastuzumab, the model IgG used in this study, to NPIs led to an increase in concentration of proteinaceous particles in the sub-micron range. When added to moDCs, the NPIs alone or in presence of trastuzumab did not affect cell viability or cytotoxicity. Moreover, no significant effect on the expression of surface markers, and cytokine and chemokine production was observed. Our findings showed, surprisingly, no evidence of any immunomodulatory activity of NPIs. As this study was limited to a single IgG formulation and to in vitro immunological read-outs, further work is required to fully understand the immunogenic potential of NPIs.


Subject(s)
Nanoparticles , Pharmaceutical Preparations , Antibodies, Monoclonal , Excipients , Sucrose
4.
J Immunol ; 205(9): 2351-2361, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32989091

ABSTRACT

The development of anti-drug Abs in response to biological products (BP) is a major drawback in the treatment of patients. Factors related to the patient, the treatment, and the product can influence BP immunogenicity. Among these factors, BP aggregates have been suggested to promote immunogenicity by acting as danger signals recognized by dendritic cells (DC) facilitating the establishment of an anti-BP CD4 T cell-dependent adaptive immune response leading to anti-drug Abs production. To date, little is known on the mechanism supporting the effect of aggregates on DCs and consequently on the T cell response. The aim of this work was to identify key signaling pathways involved in BP aggregate DC activation and T cell response. We generated aggregates by submitting infliximab (IFX), an immunogenic anti-TNF-α chimeric Ab, to heat stress. Our results showed that IFX aggregates were able to induce human monocyte-derived DC (moDC) maturation in a concentration-dependent manner. Aggregate-treated moDCs enhanced allogeneic T cell proliferation and IL-5, IL-9, and IL-13 production compared with native Ab-treated moDCs. We then investigated the implication of FcγRIIa and spleen tyrosine kinase (Syk) in DC activation and showed that they were both strongly implicated in moDC maturation induced by IFX aggregates. Indeed, we found that neutralization of FcγRIIa inhibited DC activation, and consequently, Syk inhibition led to a decrease in T cell proliferation and cytokine production in response to IFX aggregates. Taken together, our results bring new insight, to our knowledge, on how protein aggregates could induce DC and T cell activation via the FcγRIIa-Syk signaling pathway.


Subject(s)
Dendritic Cells/immunology , Infliximab/immunology , Lymphocyte Activation/immunology , Receptors, IgG/immunology , Syk Kinase/immunology , T-Lymphocytes/immunology , Cell Proliferation/physiology , Cells, Cultured , Humans , Monocytes/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology
5.
Front Immunol ; 11: 725, 2020.
Article in English | MEDLINE | ID: mdl-32431697

ABSTRACT

Patients treated with bioproducts (BPs) frequently develop anti-drug antibodies (ADAs) with potential neutralizing capacities leading to loss of clinical response or potential hypersensitivity reactions. Many factors can influence BP immunogenicity and could be related to the patient, the treatment, as well as to the product itself. Among these latter factors, it is now well accepted that BP aggregation is associated with an increased potential for immunogenicity, as aggregates seem to be correlated with ADA development. Moreover, the presence of high-affinity ADAs suggests a CD4 T-cell dependent adaptive immune response and therefore a pivotal role for antigen-presenting cells (APCs), such as dendritic cells (DCs). In this review, we address the in vitro methods developed to evaluate how monoclonal antibodies could trigger the immunization process by focusing on the role of aggregated antibodies in the establishment of this response. In particular, we will present the different cell-based assays that have been used to assess the potential of antibodies and their aggregates to modulate cellular mechanisms leading to activation and the biological parameters (cellular activation markers, proliferation and secreted molecules) that can be measured to evaluate the different cell activation stages and their consequences in the propagation of the immune response. Indeed, the use of such strategies could help evaluate the risk of BP immunogenicity and their role in mitigating this risk.


Subject(s)
Adaptive Immunity , Antibodies, Monoclonal/immunology , Biological Products/immunology , Models, Immunological , Protein Aggregates/immunology , Protein Aggregation, Pathological/immunology , Antibodies, Monoclonal/therapeutic use , Biological Products/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Humans , Immunization
6.
J Pharm Sci ; 109(1): 927-932, 2020 01.
Article in English | MEDLINE | ID: mdl-31520643

ABSTRACT

The presence of protein aggregates in biological products is suggested to promote immunogenicity, leading to the production of anti-drug antibodies with neutralizing capacities. This suggests a CD4+ T-cell dependent adaptive immune response, thus a pivotal role for antigen-presenting cells, such as dendritic cells (DCs). We previously showed that human growth hormone aggregates induced DC maturation, with notably an increase in CXCL10 production. DC phenotypic modifications were sufficient to promote allogeneic CD4+ T-cell proliferation with Th1 polarization. In this work, we identified the main intracellular signaling pathways involved in DC activation by human growth hormone aggregates, showing that aggregates induced p38 mitogen-activated protein kinase, extracellular signal-regulated kinase, and c-Jun N-terminal kinase phosphorylation, as well as nuclear factor κB subunit p65 nuclear translocation. Next, investigating the implication of Rho GTPases and phosphoinositide 3-kinase (PI3K) in activated DC showed that Rac1 and Cdc42 regulated the phosphorylation of MAP kinases, whereas PI3K was only implicated in c-Jun N-terminal kinase phosphorylation. Furthermore, we showed that Rac1 and PI3K pathways, but not Cdc42, regulated the production of CXCL10 via the MAP kinases and nuclear factor κB. Taken together, our results bring new insight on how protein aggregates could induce DC activation, leading to a better understanding of aggregates role in therapeutic proteins immunogenicity.


Subject(s)
Dendritic Cells/drug effects , Human Growth Hormone/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , rac1 GTP-Binding Protein/metabolism , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Dendritic Cells/enzymology , Dendritic Cells/immunology , Drug Compounding , Human Growth Hormone/chemistry , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Aggregates , Signal Transduction , Up-Regulation
7.
Front Immunol ; 10: 601, 2019.
Article in English | MEDLINE | ID: mdl-31001248

ABSTRACT

Therapeutic antibodies have the potential to induce immunogenicity leading to the development of anti-drug antibodies (ADA) that consequently may result in reduced serum drug concentrations, a loss of efficacy or potential hypersensitivity reactions. Among other factors, aggregated antibodies have been suggested to promote immunogenicity, thus enhancing ADA production. Dendritic cells (DC) are the most efficient antigen-presenting cell population and are crucial for the initiation of T cell responses and the subsequent generation of an adaptive immune response. This work focuses on the development of predictive in vitro assays that can monitor DC maturation, in order to determine whether drug products have direct DC stimulatory capabilities. To this end, four independent laboratories aligned a common protocol to differentiate human monocyte-derived DC (moDC) that were treated with either native or aggregated preparations of infliximab, natalizumab, adalimumab, or rituximab. These drug products were subjected to different forms of physical stress, heat and shear, resulting in aggregation and the formation of subvisible particles. Each partner developed and optimized assays to monitor diverse end-points of moDC maturation: measuring the upregulation of DC activation markers via flow cytometry, analyzing cytokine, and chemokine production via mRNA and protein quantification and identifying cell signaling pathways via quantification of protein phosphorylation. These study results indicated that infliximab, with the highest propensity to form aggregates when heat-stressed, induced a marked activation of moDC as measured by an increase in CD83 and CD86 surface expression, IL-1ß, IL-6, IL-8, IL-12, TNFα, CCL3, and CCL4 transcript upregulation and release of respective proteins, and phosphorylation of the intracellular signaling proteins Syk, ERK1/2, and Akt. In contrast, natalizumab, which does not aggregate under these stress conditions, induced no DC activation in any assay system, whereas adalimumab or rituximab aggregates induced only slight parameter variation. Importantly, the data generated in the different assay systems by each partner site correlated and supported the use of these assays to monitor drug-intrinsic propensities to drive maturation of DC. This moDC assay is also a valuable tool as an in vitro model to assess the intracellular mechanisms that drive DC activation by aggregated therapeutic proteins.


Subject(s)
Antibodies, Monoclonal/pharmacology , Dendritic Cells/drug effects , Biological Assay , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/metabolism , Humans
8.
Int J Pharm ; 537(1-2): 111-121, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29241702

ABSTRACT

The effect of high pressure homogenization (HPH) on the structure of ß-lactoglobulin (ß-lg) was studied by combining spectroscopic, chromatographic, and electrophoretic methods. The consequences of the resulting structure modifications on oil/water (O/W) interfacial properties were also assessed. Moderated HPH treatment (100 MPa/4 cycles) showed no significant modification of protein structure and interfacial properties. However, a harsher HPH treatment (300 MPa/5 cycles) induced structural transformation, mainly from ß-sheets to random coils, wide loss in lipocalin core, and protein aggregation via intermolecular disulfide bridges. HPH-modified ß-lg displayed higher surface hydrophobicity leading to a faster adsorption rate at the interface and an earlier formation of an elastic interfacial film at Cß-lg = 0.1 wt%. However, no modification of the interfacial properties was observed at Cß-lg = 1 wt%. At this protein concentration, the prior denaturation of ß-lg by HPH did not modify the droplet size of nanoemulsions prepared with these ß-lg solutions as the aqueous phases. A slightly increased creaming rate was however observed. The effects of HPH and heat denaturations appeared qualitatively similar, but with differences in their extent.


Subject(s)
Emulsions/chemistry , Lactoglobulins/chemistry , Adsorption/drug effects , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Oils/chemistry , Particle Size , Pressure , Protein Conformation, beta-Strand , Water/chemistry
9.
Front Immunol ; 8: 544, 2017.
Article in English | MEDLINE | ID: mdl-28555135

ABSTRACT

Particles possess huge specific surface area and therefore nanomaterials exhibit unique characteristics, such as special physical properties and chemical hyper-reactivity, which make them particularly attractive but also raise numerous questions concerning their safety. Interactions of nanomaterials with the immune system can potentially lead to immunosuppression, hypersensitivity (allergy), immunogenicity and autoimmunity, involving both innate and adaptive immune responses. Inherent physical and chemical NP characteristics may influence their immunotoxicity, i.e., the adverse effects that can result from exposure. This review will focus on the possible interaction of nanomaterials including protein aggregates with the innate immune system with specific emphasis on antigen-presenting cells, i.e., dendritic cells, macrophages and monocytes.

10.
Immunol Cell Biol ; 95(3): 306-315, 2017 03.
Article in English | MEDLINE | ID: mdl-27713394

ABSTRACT

Patients treated with therapeutic biological products (BP) frequently develop anti-drug antibodies (ADA) with potential neutralizing capacities leading to loss of clinical response or serious side effects. BP aggregates have been suggested to promote immunogenicity, thus enhancing ADA production. Dendritic cells (DC) are key effectors in T-cell and B-cell fates, and the subsequent generation of immunogenicity. The objective of this work was to determine if BP aggregates can participate to DC maturation and T-cell activation. We compared aggregates from three different proteins: human growth hormone (hGH), Rituximab, a chimeric anti-CD20 antibody and a serum-purified human IgG1. All three proteins underwent a stir stress, generating comparable populations of aggregated particles. Maturation of human monocyte-derived DC (moDC) upon exposure to native BPs or aggregates was evaluated in vitro. Results showed that hGH aggregates induced an increased expression of moDC co-stimulation markers, and augmented levels of IL-6, IL-8, IL-12p40, CCL2, CCL3, CCL4 and CXCL10. Both antibodies aggregates were also able to modify DC phenotype, but cytokine and chemokine productions were seen only with IL-6, IL-8, IL-12p40 and CXCL10. Aggregates-treated moDC enhanced allogenic T-cell proliferation and cytokines production, suggesting Th1 polarization with hGH, and mixed T-cell responses with antibodies aggregates. These results showed that BP aggregates provoked DC maturation, thus driving adaptive T-cell responses and polarization.


Subject(s)
Cell Polarity/drug effects , Dendritic Cells/cytology , Growth Hormone/pharmacology , Immunoglobulin G/pharmacology , Protein Aggregates , T-Lymphocytes/cytology , Cell Proliferation/drug effects , Chemokines/metabolism , Dendritic Cells/drug effects , Humans , Inflammation Mediators/metabolism , Phenotype , T-Lymphocytes/drug effects
11.
J Leukoc Biol ; 97(4): 737-49, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25673294

ABSTRACT

DCs are the first immune cells to be exposed to allergens, including chemical sensitizers, such as nickel, a human TLR4 agonist that induces DC maturation. In ACD, DCs can interact with PMNs that are recruited and activated, leading, in particular, to ectosome release. The objective of this work was to characterize the effects of PMN-Ect on DC functions in an ACD context. We first developed a standardized protocol to produce, characterize, and quantify ectosomes by use of human PLB-985 cells, differentiated into mature PMN (PLB-Ect). We then studied the in vitro effects of these purified ectosomes on human moDC functions in response to NiSO4 and to LPS, another TLR4 agonist. Confocal fluorescence microscopy showed that PLB-Ect was internalized by moDCs and localized in the lysosomal compartment. We then showed that PLB-Ect down-regulated NiSO4-induced moDC maturation, as witnessed by decreased expression of CD40, CD80, CD83, CD86, PDL-1, and HLA-DR and by decreased levels of IL-1ß, IL-6, TNF-α, and IL-12p40 mRNAs. These effects were related to p38MAPK and NF-κB down-regulation. However, no increase in pan-regulatory DC marker genes (GILZ, CATC, C1QA) was observed; rather, levels of effector DC markers (Mx1, NMES1) were increased. Finally, when these PLB-Ect + NiSO4-treated moDCs were cocultured with CD4(+) T cells, a Th2 cytokine profile seemed to be induced, as shown, in particular, by enhanced IL-13 production. Together, these results suggest that the PMN-Ect can modulate DC maturation in response to nickel, a common chemical sensitizer responsible for ADC.


Subject(s)
Allergens/immunology , Antigens, CD/biosynthesis , Cell-Derived Microparticles/physiology , Dendritic Cells/immunology , Dermatitis, Allergic Contact/immunology , Gene Expression Regulation/immunology , Lymphokines/biosynthesis , Myeloid Cells/immunology , Neutrophils/immunology , Nickel/immunology , Th2 Cells/cytology , Allergens/pharmacology , Antigens, CD/genetics , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Cell Differentiation , Coculture Techniques , Dendritic Cells/drug effects , Dermatitis, Allergic Contact/etiology , HLA-DR Antigens/biosynthesis , HLA-DR Antigens/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Liposomes , Lymphokines/genetics , Monocytes/cytology , Myeloid Cells/ultrastructure , Neutrophils/ultrastructure , Nickel/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology
12.
Infect Immun ; 80(5): 1891-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22371374

ABSTRACT

We recently documented the neutrophil response to enterovirulent diffusely adherent Escherichia coli expressing Afa/Dr fimbriae (Afa/Dr DAEC), using the human myeloid cell line PLB-985 differentiated into fully mature neutrophils. Upon activation, particularly during infections, neutrophils release neutrophil extracellular traps (NETs), composed of a nuclear DNA backbone associated with antimicrobial peptides, histones, and proteases, which entrap and kill pathogens. Here, using fluorescence microscopy and field emission scanning electron microscopy, we observed NET production by PLB-985 cells infected with the Afa/Dr wild-type (WT) E. coli strain C1845. We found that these NETs were able to capture, immobilize, and kill WT C1845 bacteria. We also developed a coculture model of human enterocyte-like Caco-2/TC7 cells and PLB-985 cells previously treated with WT C1845 and found, for the first time, that the F-actin cytoskeleton of enterocyte-like cells is damaged in the presence of bacterium-induced NETs and that this deleterious effect is prevented by inhibition of protease release. These findings provide new insights into the neutrophil response to bacterial infection via the production of bactericidal NETs and suggest that NETs may damage the intestinal epithelium, particularly in situations such as inflammatory bowel diseases.


Subject(s)
Adhesins, Escherichia coli/metabolism , Enterocytes/cytology , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Neutrophils/physiology , alpha-Defensins/metabolism , Adhesins, Escherichia coli/genetics , Cell Line , Coculture Techniques , Escherichia coli/ultrastructure , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Histones/metabolism , Humans , Peptide Hydrolases/metabolism
13.
Infect Immun ; 78(7): 2974-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20404079

ABSTRACT

The enterovirulent Escherichia coli strains potentially involved in inflammatory bowel diseases include diffusely adherent strains expressing Afa/Dr fimbriae (Afa/Dr DAEC). We have previously observed type 1 pilus-mediated interleukin-8 (IL-8) hyperproduction in infected neutrophils. As pathogen induction of host cell death programs and clearance of apoptotic infected cells are crucial for innate immune system homeostasis and host integrity, we examined modulation of neutrophil cell death by Afa/Dr DAEC. Using the human PLB-985 cell line differentiated into fully mature neutrophils, we found that the wild-type enterovirulent E. coli strain C1845 and the recombinant strain DH5alpha/pF1845 (expressing the fimbrial adhesin F1845) similarly induced time-dependent phosphatidylserine (PS) externalization, suggesting a major specific role of this virulence factor. Using small interfering RNA (siRNA) decay-accelerating factor (DAF)-transfected PLB-985 cells, we then showed that this PS externalization was triggered in part by glycosylphosphatidylinositol (GPI)-anchored DAF receptor engagement (leading to tyrosine kinase and protein kinase C activation) and that it required cytoskeleton and lipid raft architectural integrity. PS externalization under these conditions was not dependent on caspases, mitochondria, lysosomes, or reactive oxygen or nitrogen species. F1845-mediated PS externalization was sufficient to enable macrophage engulfment of infected differentiated PLB-985 cells. These findings provide new insights into the neutrophil response to Afa/Dr DAEC infection and highlight a new role for F1845 fimbriae. Interestingly, although apoptosis pathways were not engaged, C1845-infected PLB-985 cells displayed enhanced removal by macrophages, a process that may participate in the resolution of Afa/Dr DAEC infection and related inflammation.


Subject(s)
Enteropathogenic Escherichia coli/physiology , Escherichia coli Proteins/physiology , Fimbriae, Bacterial/physiology , Transcription Factors/physiology , Adhesins, Escherichia coli/physiology , Apoptosis/physiology , Bacterial Adhesion/physiology , Blotting, Western , Cell Line, Tumor , Escherichia coli Infections/microbiology , Fimbriae Proteins/physiology , Granulocytes/microbiology , Granulocytes/physiology , Humans , Lysosomes/microbiology , Lysosomes/physiology , Macrophages/microbiology , Macrophages/physiology , Neutrophils/microbiology , Neutrophils/physiology , Phagocytosis/physiology
14.
J Leukoc Biol ; 85(2): 310-21, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19015376

ABSTRACT

The innate immune response to enteropathogenic bacteria includes chemokine-induced polymorphonuclear neutrophil (PMN) migration across mucosal epithelia leading to bacterial clearance and resolution of infection. Among these bacteria, diffusely adherent Escherichia coli expressing Afa/Dr fimbriae (Afa/Dr DAEC), causing childhood diarrhea, can promote IL-8-dependent PMN transmigration across cultured intestinal epithelial cell monolayers via MAPK pathway activation. However, interactions between PMN and Afa/Dr DAEC are poorly documented and constitute the aim of the present study. Using the human PLB-985 cell line differentiated into fully mature PMN, we described the coordinated response to various E. coli. The rapid and strong release of reactive oxygen species and preformed intragranular mediators (myeloperoxidase and IL-8) is followed by a later TNF-alpha, IL-1beta, and IL-8 synthesis. The use of wild-type (IH11128, C1845, LF82), control (AAEC185), and recombinant (AAEC185 bearing Dr or F1845 fimbriae, AdLF82, or type 1 pili) bacterial strains allowed us to demonstrate that late IL-8 hyperproduction is triggered by type 1 pili but not by Dr or F1845 fimbriae; MAPKs (p38, ERK, Src) and NF-kappaB activations are implicated in this response. Thus, in the course of Afa/Dr DAEC intestinal infection, epithelium- and neutrophil-derived IL-8 could, at least in part, control the flow of neutrophils through the lamina propria. Afa/Dr DAEC-induced IL-8 hyperproduction by PMN might thus be important for inducing and perpetuating local inflammation, and this self-amplifying loop might play a role in the pathogenesis of inflammatory bowel diseases such as Crohn's disease.


Subject(s)
Cell Differentiation , Escherichia coli/immunology , Fimbriae, Bacterial/immunology , Interleukin-8/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Neutrophils/enzymology , src-Family Kinases/metabolism , Antigens, CD/immunology , Bacterial Adhesion , CD11b Antigen/immunology , CD18 Antigens/immunology , Cell Adhesion Molecules/immunology , Escherichia coli/cytology , GPI-Linked Proteins , Humans , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neutrophils/cytology , Neutrophils/microbiology , Peroxidase/metabolism , Respiratory Burst/immunology , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...