Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Immunol ; 201(7): 1865-1874, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30127086

ABSTRACT

The role of regulatory B cells (Bregs) in modulating immune responses and maintaining tolerance are well established. However, how cytokines present during immune responses affect Breg growth and function are not as well defined. Previously, our laboratory reported IL-5- and mCD40L-expressing fibroblast (mCD40L-Fb) stimulation induced IL-10 production from murine B cells. The current study investigated the phenotype and functional relevance of IL-10- producing B cells from this culture. We found IL-5/mCD40L-Fb stimulation induced IL-10 production exclusively from CD5+ splenic B cells of naive mice. After stimulation, the resulting IL-10+ B cells displayed markers of multiple reported Breg phenotypes. Interestingly, when investigating effects of IL-4 (a critical TH2 cytokine) on IL-5/mCD40L-Fb-induced IL-10 production, we found IL-4 inhibited IL-10 production in a STAT6-dependent manner. Upon adoptive transfer, CD5+ B cells previously stimulated with IL-5/mCD40L-Fb were able to reduce development of OVA-induced allergic airway disease in mice. Using B cells from IL-10 mutant mice differentiated by IL-5/mCD40L-Fb, we found protection from allergic airway disease development was dependent on the IL-10 production from the transferred B cells. Bregs have been shown to play crucial roles in the immune tolerance network, and understanding stimuli that modulate their growth and function may be key in development of future treatments for diseases of immune dysregulation.


Subject(s)
Asthma/immunology , B-Lymphocytes, Regulatory/immunology , Cytokines/metabolism , Respiratory Hypersensitivity/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , CD40 Ligand/metabolism , Cells, Cultured , Disease Models, Animal , Female , Humans , Immune Tolerance , Immunomodulation , Mice , Mice, Inbred C57BL , Ovalbumin/immunology
2.
PLoS Pathog ; 13(7): e1006539, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28753651

ABSTRACT

Infection with the helminth Schistosoma (S.) mansoni drives the development of interleukin (IL)-10-producing regulatory B (Breg) cells in mice and man, which have the capacity to reduce experimental allergic airway inflammation and are thus of high therapeutic interest. However, both the involved antigen and cellular mechanisms that drive Breg cell development remain to be elucidated. Therefore, we investigated whether S. mansoni soluble egg antigens (SEA) directly interact with B cells to enhance their regulatory potential, or act indirectly on B cells via SEA-modulated macrophage subsets. Intraperitoneal injections of S. mansoni eggs or SEA significantly upregulated IL-10 and CD86 expression by marginal zone B cells. Both B cells as well as macrophages of the splenic marginal zone efficiently bound SEA in vivo, but macrophages were dispensable for Breg cell induction as shown by macrophage depletion with clodronate liposomes. SEA was internalized into acidic cell compartments of B cells and induced a 3-fold increase of IL-10, which was dependent on endosomal acidification and was further enhanced by CD40 ligation. IPSE/alpha-1, one of the major antigens in SEA, was also capable of inducing IL-10 in naïve B cells, which was reproduced by tobacco plant-derived recombinant IPSE. Other major schistosomal antigens, omega-1 and kappa-5, had no effect. SEA depleted of IPSE/alpha-1 was still able to induce Breg cells indicating that SEA contains more Breg cell-inducing components. Importantly, SEA- and IPSE-induced Breg cells triggered regulatory T cell development in vitro. SEA and recombinant IPSE/alpha-1 also induced IL-10 production in human CD1d+ B cells. In conclusion, the mechanism of S. mansoni-induced Breg cell development involves a direct targeting of B cells by SEA components such as the secretory glycoprotein IPSE/alpha-1.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Egg Proteins/immunology , Helminth Proteins/immunology , Ovum/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Egg Proteins/genetics , Female , Helminth Proteins/genetics , Humans , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Schistosoma mansoni/genetics , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/parasitology
4.
J Infect Dis ; 210(8): 1207-16, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-24795476

ABSTRACT

BACKGROUND: Chronic schistosome infections are associated with T-cell hyporesponsiveness and a strong regulatory network. Murine studies have shown that schistosome infections can induce regulatory CD1d(hi) B cells, which inhibit inflammatory responses. Here, we evaluated the influence of regulatory B cells (Bregs) on T-cell cytokines in vitro in human schistosomiasis. METHODS: Gabonese young adults were recruited from areas where Schistosoma haematobium (S.h) infections were high or low endemic. The study participants were categorized as infected or uninfected from an high endemic area or uninfected from a low endemic (nonendemic) area. Their B cells were studied for Breg subset markers and cocultured with allogenic anti-CD3-stimulated CD4(+) T cells, followed by T-cell cytokine analysis. RESULTS: A greater percentage of B cells from S. haematobium-infected donors expressed cytoplasmic interleukin 10 (IL-10) and membrane-bound latency-associated peptide/transforming growth factor ß1, compared with uninfected donors. T cells produced less interferon γ, interleukin 4, and interleukin 17 upon coculture with B cells from schistosome-infected individuals only, while the conversion to CD25(hi)FoxP3(+) and the percentage of IL-10(+) T cells was enhanced. Interestingly, depletion of the prominent IL-10-producing B-cell subset, CD1d(hi) cells, resulted in less IL-10(+) T cells in the S. haematobium-infected group, while levels of FoxP3(+) regulatory T cells remained unaffected. CONCLUSIONS: Schistosomes can induce functional Bregs in humans that may be instrumental in general T-cell hyporesponsiveness and may contribute to the increased regulatory milieu found in schistosomiasis.


Subject(s)
Antigens, CD1/metabolism , B-Lymphocytes, Regulatory/metabolism , Cytokines/classification , Interleukin-10/metabolism , Schistosomiasis haematobia/metabolism , T-Lymphocytes/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Gabon/epidemiology , Gene Expression Regulation/immunology , Humans , Interleukin-10/genetics , Schistosoma haematobium , Schistosomiasis haematobia/epidemiology , Schistosomiasis haematobia/immunology
6.
J Allergy Clin Immunol ; 128(4): 733-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21684587

ABSTRACT

Chronic helminth infections are often associated with a reduced prevalence of inflammatory disorders, including allergic diseases. Helminths influence the host immune system by downregulating T-cell responses; the cytokine IL-10 appears to play a central role in this process. Over the last decade, evidence has emerged toward a new regulatory cell type: IL-10-producing B cells, capable of regulating immunity and therefore termed regulatory B cells. Initially, regulatory B cells have been described in autoimmunity models where they dampen inflammation, but recently they were also found in several helminth infection models. Importantly, regulatory B cells have recently been identified in humans, and it has been suggested that patients suffering from autoimmunity have an impaired regulatory B-cell function. As such, it is of therapeutic interest to study the conditions in which IL-10-producing B cells can be induced. Chronic helminth infections appear to hold promise in this context as emerging evidence suggests that helminth-induced regulatory B cells strongly suppress allergic inflammation. In this review, we will discuss the conditions under which regulatory B cells are present, leading to a state of tolerance, as well as the conditions where their absence or functional impairment leads to exacerbated disease. We will summarize their phenotypic characteristics and their mechanisms of action and elaborate on possible mechanisms whereby regulatory B cells can be induced or expanded, as this may open novel avenues for the treatment of inflammatory diseases, such as allergic asthma.


Subject(s)
B-Lymphocyte Subsets/immunology , Helminthiasis/immunology , Helminths/immunology , Hypersensitivity/immunology , Immune Tolerance , Animals , Autoimmune Diseases/immunology , Chronic Disease , Humans , Inflammation/immunology , Interleukin-10/immunology , T-Lymphocytes/immunology
7.
Methods Mol Biol ; 2270: 149-178, 2021.
Article in English | MEDLINE | ID: mdl-33479898

ABSTRACT

B lymphocytes make several contributions to immune regulation including production of antibodies with regulatory properties, release of immune suppressive cytokines, and expression of death-inducing ligands. A role for Fas ligand (FasL)-expressing "killer" B cells in regulating T helper (TH) cell survival and chronic inflammation has been demonstrated in animal models of schistosome worm and other infections, asthma, autoimmune arthritis, and type 1 diabetes. FasL+ B cells were also capable of inducing immune tolerance in a male-to-female transplantation model. Interestingly, populations of B cells found in the spleen and lungs of naïve mice constitutively expresses FasL and have potent killer function against TH cells that is antigen-specific and FasL-dependent. Epstein-Barr virus-transformed human B cells constitutively express FasL and package it into exosomes that co-express MHC Class II molecules and have killer function against antigen-specific TH cells. FasL+ exosomes with markers of B-cell lineage are abundant in the spleen of naïve mice. Killer B cells therefore represent a novel target for immune modulation in many disease settings. Our laboratory has published methods of characterizing FasL+ B cells and inducing their proliferation in vitro. This updated chapter will describe methods of identifying and expanding killer B cells from mice, detecting FasL expression in B cells, extracting FasL+ exosomes from spleen and culture supernatants, and performing functional killing assays against antigen-specific TH cells.


Subject(s)
Exosomes/metabolism , Fas Ligand Protein/isolation & purification , Flow Cytometry/methods , Animals , Antigens/metabolism , Apoptosis/immunology , B-Lymphocytes/immunology , Cytokines/metabolism , Fas Ligand Protein/metabolism , Female , Histocompatibility Antigens Class II/metabolism , Immune Tolerance , Interleukin-10/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Spleen/cytology , fas Receptor/metabolism
8.
PLoS One ; 11(11): e0166255, 2016.
Article in English | MEDLINE | ID: mdl-27829065

ABSTRACT

Cigarette smoking is the main risk factor associated with chronic obstructive pulmonary disease (COPD), and contributes to COPD development and progression by causing epithelial injury and inflammation. Whereas it is known that cigarette smoke (CS) may affect the innate immune function of airway epithelial cells and epithelial repair, this has so far not been explored in an integrated design using mucociliary differentiated airway epithelial cells. In this study, we examined the effect of whole CS exposure on wound repair and the innate immune activity of mucociliary differentiated primary bronchial epithelial cells, upon injury induced by disruption of epithelial barrier integrity or by mechanical wounding. Upon mechanical injury CS caused a delayed recovery in the epithelial barrier integrity and wound closure. Furthermore CS enhanced innate immune responses, as demonstrated by increased expression of the antimicrobial protein RNase 7. These differential effects on epithelial repair and innate immunity were both mediated by CS-induced oxidative stress. Overall, our findings demonstrate modulation of wound repair and innate immune responses of injured airway epithelial cells that may contribute to COPD development and progression.


Subject(s)
Immunity, Innate/drug effects , Respiratory Mucosa/drug effects , Smoking/adverse effects , Blotting, Western , Bronchi/cytology , Bronchi/drug effects , Cells, Cultured , ErbB Receptors/physiology , Humans , MAP Kinase Signaling System/physiology , Microscopy, Fluorescence , Oxidative Stress/drug effects , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/immunology , Ribonucleases/metabolism , Signal Transduction/physiology , Wound Healing/drug effects
9.
Methods Mol Biol ; 1190: 127-41, 2014.
Article in English | MEDLINE | ID: mdl-25015278

ABSTRACT

Toll-like receptors (TLRs) are key components for the recognition of microorganisms, for the initiation of innate immunity, and for promoting adaptive immune responses. TLR signaling in B cells, in addition to B cell receptor or CD40 ligation, plays an important role in B cell differentiation and activation. In contrast, various infectious agents and/or TLR ligands can also prime B cells to induce tolerance and downregulate inflammatory reactions; those B cells are called regulatory B (Breg) cells and are characterized by a dominant IL-10 production. Several studies have suggested that Breg cells are impaired in patients with autoimmune diseases and allergic asthma. However, the role for TLR ligands in the induction of Breg cells as a potential therapy for some of these inflammatory diseases has not yet been investigated. Here, we provide detailed instructions on how to analyze and validate cytokine production in human and mouse B cells in response to various TLR ligands. Furthermore, we describe an assay to investigate the suppressive properties of TLR-induced B cells to confirm their regulatory B cell status.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Cytokines/immunology , Toll-Like Receptors/immunology , Animals , Antigens, CD19/analysis , Antigens, CD19/immunology , B-Lymphocytes, Regulatory/cytology , Cell Separation/methods , Coculture Techniques/methods , Cytokines/analysis , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry/methods , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/immunology , Humans , Immunity, Innate , Interleukin-10/analysis , Interleukin-10/immunology , Mice , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptors/analysis
10.
PLoS One ; 9(12): e114630, 2014.
Article in English | MEDLINE | ID: mdl-25531674

ABSTRACT

BACKGROUND: Children who have been exposed in utero to maternal filarial infection are immunologically less responsive to filarial antigens, have less pathology, and are more susceptible to acquire infection than offspring of uninfected mothers. Moreover children from filaria infected mothers have been shown to be less responsive to vaccination as a consequence of an impairment of their immune response. However, it is not well known how in utero exposure to parasite antigens affects cellular immune responses. METHODOLOGY: Here, 30 pregnant women were examined for the presence of microfilaria of Loa loa and Mansonella perstans in peripheral blood. At delivery, cord blood mononuclear cells (CBMC) were obtained and the CD4+T cells were phenotyped by expression of the transcription factors Tbet, RORγt, and FOXP3. RESULTS: No significant difference was observed between newborns from infected versus uninfected mothers in the frequencies of total CD4+T cells and CD4+T cells subsets including CD4+Tbet+, CD4+RORγt+ T and CD4+CD25hiFOXP3+ T cells. However, there was a negative association between CD4+CD25hiFOXP3+T cells and CD4+Tbet+ as well as CD4+RORγt+ T cells in the infected group only (B = -0.242, P = 0.002; B = -0.178, P = 0.013 respectively). CONCLUSION: Our results suggest that filarial infection during pregnancy leads to an expansion of functionally active regulatory T cells that keep TH1 and TH17 in check.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Fetal Blood/cytology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Microfilariae/isolation & purification , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Box Domain Proteins/metabolism , Adult , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Female , Humans , Infant, Newborn , Loa/growth & development , Loa/physiology , Mansonella/growth & development , Mansonella/physiology , Mothers , Pregnancy , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
11.
PLoS Negl Trop Dis ; 7(3): e2094, 2013.
Article in English | MEDLINE | ID: mdl-23505586

ABSTRACT

Antibody responses are thought to play an important role in control of Schistosoma infections, yet little is known about the phenotype and function of B cells in human schistosomiasis. We set out to characterize B cell subsets and B cell responses to B cell receptor and Toll-like receptor 9 stimulation in Gabonese schoolchildren with Schistosoma haematobium infection. Frequencies of memory B cell (MBC) subsets were increased, whereas naive B cell frequencies were reduced in the schistosome-infected group. At the functional level, isolated B cells from schistosome-infected children showed higher expression of the activation marker CD23 upon stimulation, but lower proliferation and TNF-α production. Importantly, 6-months after 3 rounds of praziquantel treatment, frequencies of naive B cells were increased, MBC frequencies were decreased and with the exception of TNF-α production, B cell responsiveness was restored to what was seen in uninfected children. These data show that S. haematobium infection leads to significant changes in the B cell compartment, both at the phenotypic and functional level.


Subject(s)
B-Lymphocyte Subsets/immunology , Schistosoma haematobium/immunology , Schistosomiasis/immunology , Adolescent , Animals , Anthelmintics/therapeutic use , Antibodies, Helminth/blood , Child , Female , Gabon , Humans , Male , Praziquantel/therapeutic use , Schistosomiasis/drug therapy , Time Factors
12.
PLoS One ; 7(2): e30883, 2012.
Article in English | MEDLINE | ID: mdl-22347409

ABSTRACT

Chronic helminth infections, such as schistosomes, are negatively associated with allergic disorders. Here, using B cell IL-10-deficient mice, Schistosoma mansoni-mediated protection against experimental ovalbumin-induced allergic airway inflammation (AAI) was shown to be specifically dependent on IL-10-producing B cells. To study the organs involved, we transferred B cells from lungs, mesenteric lymph nodes or spleen of OVA-infected mice to recipient OVA-sensitized mice, and showed that both lung and splenic B cells reduced AAI, but only splenic B cells in an IL-10-dependent manner. Although splenic B cell protection was accompanied by elevated levels of pulmonary FoxP3(+) regulatory T cells, in vivo ablation of FoxP3(+) T cells only moderately restored AAI, indicating an important role for the direct suppressory effect of regulatory B cells. Splenic marginal zone CD1d(+) B cells proved to be the responsible splenic B cell subset as they produced high levels of IL-10 and induced FoxP3(+) T cells in vitro. Indeed, transfer of CD1d(+) MZ-depleted splenic B cells from infected mice restored AAI. Markedly, we found a similarly elevated population of CD1d(hi) B cells in peripheral blood of Schistosoma haematobium-infected Gabonese children compared to uninfected children and these cells produced elevated levels of IL-10. Importantly, the number of IL-10-producing CD1d(hi) B cells was reduced after anti-schistosome treatment. This study points out that in both mice and men schistosomes have the capacity to drive the development of IL-10-producing regulatory CD1d(hi) B cells and furthermore, these are instrumental in reducing experimental allergic inflammation in mice.


Subject(s)
Antigens, CD1d/immunology , B-Lymphocytes/immunology , Hypersensitivity/pathology , Interleukin-10/immunology , Schistosoma/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/parasitology , Child , Gabon/epidemiology , Helminths , Humans , Hypersensitivity/parasitology , Inflammation/immunology , Inflammation/parasitology , Interleukin-10/deficiency , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL