Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Front Immunol ; 15: 1372927, 2024.
Article in English | MEDLINE | ID: mdl-38742105

ABSTRACT

The parasitic helminth Schistosoma mansoni is a potent inducer of type 2 immune responses by stimulating dendritic cells (DCs) to prime T helper 2 (Th2) responses. We previously found that S. mansoni soluble egg antigens (SEA) promote the synthesis of Prostaglandin E2 (PGE2) by DCs through ERK-dependent signaling via Dectin-1 and Dectin-2 that subsequently induces OX40L expression, licensing them for Th2 priming, yet the ligands present in SEA involved in driving this response and whether specific targeting of PGE2 synthesis by DCs could affect Th2 polarization are unknown. We here show that the ability of SEA to bind Dectin-2 and drive ERK phosphorylation, PGE2 synthesis, OX40L expression, and Th2 polarization is impaired upon cleavage of high-mannose glycans by Endoglycosidase H treatment. This identifies high-mannose glycans present on glycoproteins in SEA as important drivers of this signaling axis. Moreover, we find that OX40L expression and Th2 induction are abrogated when microsomal prostaglandin E synthase-1 (mPGES) is selectively inhibited, but not when a general COX-1/2 inhibitor is used. This shows that the de novo synthesis of PGE2 is vital for the Th2 priming function of SEA-stimulated DCs as well as points to the potential existence of other COX-dependent lipid mediators that antagonize PGE2-driven Th2 polarization. Lastly, specific PGE2 inhibition following immunization with S. mansoni eggs dampened the egg-specific Th cell response. In summary, our findings provide new insights in the molecular mechanisms underpinning Th2 induction by S. mansoni and identify druggable targets for potential control of helminth driven-Th2 responses.


Subject(s)
Antigens, Helminth , Dendritic Cells , Dinoprostone , Lectins, C-Type , Mannose , Polysaccharides , Schistosoma mansoni , Th2 Cells , Animals , Schistosoma mansoni/immunology , Dinoprostone/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Lectins, C-Type/metabolism , Lectins, C-Type/immunology , Mannose/metabolism , Mannose/immunology , Mice , Polysaccharides/immunology , Polysaccharides/metabolism , Antigens, Helminth/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Ovum/immunology , Ovum/metabolism , Mice, Inbred C57BL , OX40 Ligand/metabolism
2.
Cell Mol Gastroenterol Hepatol ; 17(1): 107-117, 2024.
Article in English | MEDLINE | ID: mdl-37696392

ABSTRACT

BACKGROUND & AIMS: Schistosomiasis is one of the most prominent parasite-induced infectious diseases, affecting more than 250 million people. Schistosoma mansoni causes metabolic exhaustion and a strong redox imbalance in the liver, causing parenchymal damage, and may predispose for cancer. We investigated whether oxidative stress provokes hepatocellular proliferation upon S. mansoni infection. METHODS: The cell cycle, replication stress response, and proliferation were analyzed on transcriptional and protein levels in the livers of S. mansoni-infected hamsters and by mechanistic gain- and loss-of-function experiments in human hepatoma cells. Major results were validated in human biopsy specimens of S. mansoni-infected patients. RESULTS: S. mansoni infection induced licensing factors of DNA replication and cell-cycle checkpoint cyclins in parallel with a DNA damage response in hamster hepatocytes. Moreover, even unisexual infection without egg effects, as a reflection of a chronic inflammatory process, resulted in a moderate activation of several cell-cycle markers. S. mansoni soluble egg antigens induced proliferation of human hepatoma cells that could be abolished by reduced glutathione. CONCLUSIONS: Our data suggest that hepatocellular proliferation is triggered by S. mansoni egg-induced oxidative stress.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Schistosomiasis mansoni , Cricetinae , Animals , Humans , Schistosoma mansoni/physiology , Schistosomiasis mansoni/metabolism , Oxidative Stress , Cell Proliferation
3.
JHEP Rep ; 5(2): 100625, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36590323

ABSTRACT

Background & Aims: Schistosomiasis is a parasitic infection which affects more than 200 million people globally. Schistosome eggs, but not the adult worms, are mainly responsible for schistosomiasis-specific morbidity in the liver. It is unclear if S. mansoni eggs consume host metabolites, and how this compromises the host parenchyma. Methods: Metabolic reprogramming was analyzed by matrix-assisted laser desorption/ionization mass spectrometry imaging, liquid chromatography with high-resolution mass spectrometry, metabolite quantification, confocal laser scanning microscopy, live cell imaging, quantitative real-time PCR, western blotting, assessment of DNA damage, and immunohistology in hamster models and functional experiments in human cell lines. Major results were validated in human biopsies. Results: The infection with S. mansoni provokes hepatic exhaustion of neutral lipids and glycogen. Furthermore, the distribution of distinct lipid species and the regulation of rate-limiting metabolic enzymes is disrupted in the liver of S. mansoni infected animals. Notably, eggs mobilize, incorporate, and store host lipids, while the associated metabolic reprogramming causes oxidative stress-induced DNA damage in hepatocytes. Administration of reactive oxygen species scavengers ameliorates these deleterious effects. Conclusions: Our findings indicate that S. mansoni eggs completely reprogram lipid and carbohydrate metabolism via soluble factors, which results in oxidative stress-induced cell damage in the host parenchyma. Impact and implications: The authors demonstrate that soluble egg products of the parasite S. mansoni induce hepatocellular reprogramming, causing metabolic exhaustion and a strong redox imbalance. Notably, eggs mobilize, incorporate, and store host lipids, while the metabolic reprogramming causes oxidative stress-induced DNA damage in hepatocytes, independent of the host's immune response. S. mansoni eggs take advantage of the host environment through metabolic reprogramming of hepatocytes and enterocytes. By inducing DNA damage, this neglected tropical disease might promote hepatocellular damage and thus influence international health efforts.

4.
Front Immunol ; 12: 750160, 2021.
Article in English | MEDLINE | ID: mdl-34712239

ABSTRACT

Epidermolysis bullosa acquisita (EBA) is an autoimmune blistering disorder characterized and caused by autoantibodies against type VII collagen (COL7). Although it has been noticed that EBA in both patients and mice is associated with an increased scratching, it is not clear whether and how the scratching contributes to disease manifestation. Hence, we here aimed to validate this clinical observation and also to investigate the potential contribution of increased scratching in EBA pathogenesis in mice. Longitudinal assessment of scratching behavior revealed an increased frequency of scratching as early as 12 hours after injection of anti-COL7 IgG into the skin of mice. Subsequently, scratching events became even more frequent in mice. In contrast, mice injected with a control antibody showed an unaltered scratching behavior throughout the observation period. Based on these observations, we hypothesized that mechanical irritation may promote the induction of inflammation in experimental EBA. To challenge this assumption, the local anesthetic dyclonine hydrochloride was topically applied before injection of anti-COL7 IgG. Dyclonine hydrochloride reduced the scratching events and impaired clinical disease manifestation. In therapeutic experimental settings, i.e. administration of the local anesthetic 24 hours after injection of anti-COL7 IgG, dyclonine hydrochloride only inhibited the scratching behavior, but had no significant effect on clinical disease development. In addition, eosinophils were detected in the skin before the injection of anti-COL7 IgG and significantly increased 48 hours after the antibody injection. Collectively, our results suggest that scratching behavior contributes to the initiation phase of disease manifestation in experimental EBA.


Subject(s)
Anesthetics, Local/administration & dosage , Epidermolysis Bullosa Acquisita/drug therapy , Propiophenones/administration & dosage , Administration, Topical , Animals , Collagen Type VII/immunology , Disease Models, Animal , Female , Immunoglobulin G/administration & dosage , Mice, Inbred BALB C
5.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34196299

ABSTRACT

The inflammatory response after myocardial infarction (MI) is a precisely regulated process that greatly affects subsequent remodeling. Here, we show that basophil granulocytes infiltrated infarcted murine hearts, with a peak occurring between days 3 and 7. Antibody-mediated and genetic depletion of basophils deteriorated cardiac function and resulted in enhanced scar thinning after MI. Mechanistically, we found that basophil depletion was associated with a shift from reparative Ly6Clo macrophages toward increased numbers of inflammatory Ly6Chi monocytes in the infarcted myocardium. Restoration of basophils in basophil-deficient mice by adoptive transfer reversed this proinflammatory phenotype. Cellular alterations in the absence of basophils were accompanied by lower cardiac levels of IL-4 and IL-13, two major cytokines secreted by basophils. Mice with basophil-specific IL-4/IL-13 deficiency exhibited a similarly altered myeloid response with an increased fraction of Ly6Chi monocytes and aggravated cardiac function after MI. In contrast, IL-4 induction in basophils via administration of the glycoprotein IPSE/α-1 led to improved post-MI healing. These results in mice were corroborated by the finding that initially low counts of blood basophils in patients with acute MI were associated with a worse cardiac outcome after 1 year, characterized by a larger scar size. In conclusion, we show that basophils promoted tissue repair after MI by increasing cardiac IL-4 and IL-13 levels.


Subject(s)
Basophils/immunology , Interleukin-13/immunology , Interleukin-4/immunology , Myocardial Infarction/immunology , Animals , Basophils/pathology , Basophils/physiology , Disease Models, Animal , Humans , Interleukin-13/deficiency , Interleukin-13/genetics , Interleukin-4/deficiency , Interleukin-4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , ST Elevation Myocardial Infarction/immunology , ST Elevation Myocardial Infarction/pathology , ST Elevation Myocardial Infarction/physiopathology
6.
PLoS Negl Trop Dis ; 15(1): e0008814, 2021 01.
Article in English | MEDLINE | ID: mdl-33465071

ABSTRACT

Infections by schistosomes result in granulomatous lesions around parasite eggs entrapped within the host tissues. The host and parasite determinants of the Schistosoma mansoni egg-induced granulomatous response are areas of active investigation. Some studies in mice implicate Tumor Necrosis Factor (TNF) produced in response to the infection whereas others fail to find a role for it. In addition, in the mouse model, the S. mansoni secreted egg antigen omega-1 is found to induce granulomas but the underlying mechanism remains unknown. We have recently developed the zebrafish larva as a model to study macrophage recruitment and granuloma formation in response to Schistosoma mansoni eggs. Here we use this model to investigate the mechanisms by which TNF and omega-1 shape the early granulomatous response. We find that TNF, specifically signaling through TNF receptor 1, is not required for macrophage recruitment to the egg and granuloma initiation but does mediate granuloma enlargement. In contrast, omega-1 mediates initial macrophage recruitment, with this chemotactic activity being dependent on its RNase activity. Our findings further the understanding of the role of these host- and parasite-derived factors and show that they impact distinct facets of the granulomatous response to the schistosome egg.


Subject(s)
Granuloma/etiology , Helminth Proteins/immunology , Schistosoma mansoni/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antigens, Helminth/immunology , Glycoproteins/immunology , Granuloma/immunology , Larva , Macrophages/immunology , Mutation , Ovum/immunology , Receptors, Tumor Necrosis Factor, Type I/genetics , Ribonucleases , Schistosomiasis mansoni/immunology , Tumor Necrosis Factor-alpha/genetics , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/parasitology
7.
Sci Rep ; 10(1): 22373, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33361772

ABSTRACT

Schistosomiasis (bilharzia) is a neglected tropical disease caused by parasitic flatworms of the genus Schistosoma, with considerable morbidity in parts of the Middle East, South America, Southeast Asia, in sub-Saharan Africa, and particularly also in Europe. The WHO describes an increasing global health burden with more than 290 million people threatened by the disease and a potential to spread into regions with temperate climates like Corsica, France. The aim of our study was to investigate the influence of S. mansoni infection on colorectal carcinogenic signaling pathways in vivo and in vitro. S. mansoni infection, soluble egg antigens (SEA) and the Interleukin-4-inducing principle from S. mansoni eggs induce Wnt/ß-catenin signaling and the protooncogene c-Jun as well as downstream factor Cyclin D1 and markers for DNA-damage, such as Parp1 and γH2a.x in enterocytes. The presence of these characteristic hallmarks of colorectal carcinogenesis was confirmed in colon biopsies from S. mansoni-infected patients demonstrating the clinical relevance of our findings. For the first time it was shown that S. mansoni SEA may be involved in the induction of colorectal carcinoma-associated signaling pathways.


Subject(s)
Antigens, Helminth/immunology , Colon , Eggs , Proto-Oncogene Proteins c-jun/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Wnt Signaling Pathway/immunology , Animals , Colon/immunology , Colon/parasitology , Cricetinae , Female , Humans
8.
Hepatology ; 72(2): 626-641, 2020 08.
Article in English | MEDLINE | ID: mdl-30053321

ABSTRACT

Clinical data have provided evidence that schistosomiasis can promote hepatocellular carcinogenesis. c-Jun and STAT3 are critical regulators of liver cancer development and progression. The aim of the present study was to investigate the hepatocellular activation of c-Jun and STAT3 by Schistosoma mansoni infection. Expression and function of c-Jun and STAT3 as well as proliferation and DNA repair were analyzed by western blotting, electrophoretic mobility-shift assay, and immunohistochemistry in liver of S. mansoni-infected hamsters, Huh7 cells, primary hepatocytes, and human liver biopsies. Hepatocellular activation of c-Jun was demonstrated by nuclear translocation of c-Jun, enhanced phosphorylation (Ser73), and AP-1/DNA-binding in response to S. mansoni infection. Nuclear c-Jun staining pattern around lodged eggs without ambient immune reaction, and directionally from granuloma to the central veins, suggested that substances released from schistosome eggs were responsible for the observed effects. In addition, hepatocytes with c-Jun activation show cell activation and DNA double-strand breaks. These findings from the hamster model were confirmed by analyses of human biopsies from patients with schistosomiasis. Cell culture experiments finally demonstrated that activation of c-Jun and STAT3 as well as DNA repair were induced by an extract from schistosome eggs (soluble egg antigens) and culture supernatants of live schistosome egg (egg-conditioned medium), and in particular by IPSE/alpha-1, the major component secreted by live schistosome eggs. The permanent activation of hepatocellular carcinoma-associated proto-oncogenes such as c-Jun and associated transcription factors including STAT3 by substances released from tissue-trapped schistosome eggs may be important factors contributing to the development of liver cancer in S. mansoni-infected patients. Therefore, identification and therapeutic targeting of the underlying pathways is a useful strategy to prevent schistosomiasis-associated carcinogenesis.


Subject(s)
Antigens, Helminth/physiology , Carcinoma, Hepatocellular , Hepatocytes , Liver Neoplasms , Ovum/immunology , Proto-Oncogene Proteins c-jun/physiology , STAT3 Transcription Factor/physiology , Schistosoma mansoni/immunology , Animals , Antigens, Helminth/metabolism , Carcinoma, Hepatocellular/genetics , Cricetinae , Female , Humans , Liver Neoplasms/genetics , Ovum/metabolism
9.
Front Immunol ; 9: 2614, 2018.
Article in English | MEDLINE | ID: mdl-30487793

ABSTRACT

Countries with a high incidence of helminth infections are characterized by high morbidity and mortality to infections with intracellular pathogens such as Salmonella. Some patients with Salmonella-Schistosoma co-infections develop a so-called "chronic septicemic salmonellosis," with prolonged fever and enlargement of the liver and spleen. These effects are most likely due to the overall immunoregulatory activities of schistosomes such as induction of Tregs, Bregs, alternatively activated macrophages, and degradation of antibodies. However, detailed underlying mechanisms are not very well investigated. Here, we show that intraperitoneal application of live Schistosoma mansoni eggs prior to infection with Salmonella Typhimurium in mice leads to an impairment of IFN-γ and IL-17 responses together with a higher bacterial load compared to Salmonella infection alone. S. mansoni eggs were found in granulomas in the visceral peritoneum attached to the colon. Immunohistological staining revealed IPSE/alpha-1, a glycoprotein secreted from live schistosome eggs, and recruited basophils around the eggs. Noteworthy, IPSE/alpha-1 is known to trigger IL-4 and IL-13 release from basophils which in turn is known to suppress Th1/Th17 responses. Therefore, our data support a mechanism of how schistosomes impair a protective immune response against Salmonella infection and increase our understanding of helminth-bacterial co-infections.


Subject(s)
Basophils/immunology , Egg Proteins/metabolism , Granuloma/pathology , Helminth Proteins/metabolism , Peritoneum/pathology , Salmonella Infections/immunology , Salmonella typhimurium/physiology , Schistosoma mansoni/physiology , Schistosomiasis mansoni/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Bacterial Load , Cells, Cultured , Coinfection , Cytokines/metabolism , Eggs , Humans , Immunomodulation , Mice , Mice, Inbred C57BL
10.
Front Immunol ; 9: 2293, 2018.
Article in English | MEDLINE | ID: mdl-30364177

ABSTRACT

Schistosomes control inflammation in their hosts via highly effective mechanisms such as induction of Tregs, Bregs, and alternatively activated macrophages (AAMs). Notably, IPSE/alpha-1, the major secretory product from Schistosoma mansoni eggs, triggers basophils to release interleukin (IL)-4 and IL-13. Both cytokines are essential for AAM induction, suggesting an important role for IPSE/alpha-1 in inflammation control. Here, we show by in vitro co-culture experiments that IPSE/alpha-1-induced basophil IL-4/IL-13 inhibited pro-inflammatory cytokine release from human LPS-activated monocytes. This effect was cell/cell contact-independent but dependent on IL-4, since it was abrogated in the presence of anti-IL-4 antibodies. Importantly, the IPSE/alpha-1-induced IL-4/IL-13 release from basophils was amplified in the presence of LPS. Moreover, monocytes co-cultured in the presence of LPS with IPSE/alpha-1-stimulated basophils adopted an AAM-like phenotype as assessed by elevated expression of CD206 and CD209. The putative in vivo relevance of these findings was supported by immunohistological staining of S. mansoni-infected murine tissue revealing close physical contact between IPSE/alpha-1 and basophils in schistosome egg granulomas. Taken together, we found that IPSE/alpha-1 dampens inflammatory cytokine responses by triggering basophil IL-4/IL-13, in particular in the context of TLR activation, thereby turning inflammatory monocytes into anti-inflammatory AAMs. This might represent a mechanism used by schistosomes to control inflammation in the host.


Subject(s)
Antigens, Helminth/immunology , Basophils/immunology , Basophils/metabolism , Cytokines/metabolism , Inflammation Mediators/metabolism , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Humans , Immunoglobulin E/immunology , Interleukin-13/metabolism , Interleukin-4/metabolism , Mice , Monocytes/immunology , Monocytes/metabolism , Recombinant Proteins/metabolism , Schistosomiasis mansoni/parasitology
11.
PLoS Biol ; 16(4): e2005504, 2018 04.
Article in English | MEDLINE | ID: mdl-29668708

ABSTRACT

The molecular mechanisms through which dendritic cells (DCs) prime T helper 2 (Th2) responses, including those elicited by parasitic helminths, remain incompletely understood. Here, we report that soluble egg antigen (SEA) from Schistosoma mansoni, which is well known to drive potent Th2 responses, triggers DCs to produce prostaglandin E2 (PGE2), which subsequently-in an autocrine manner-induces OX40 ligand (OX40L) expression to license these DCs to drive Th2 responses. Mechanistically, SEA was found to promote PGE2 synthesis through Dectin-1 and Dectin-2, and via a downstream signaling cascade involving spleen tyrosine kinase (Syk), extracellular signal-regulated kinase (ERK), cytosolic phospholipase A2 (cPLA2), and cyclooxygenase 1 and 2 (COX-1 and COX-2). In addition, this pathway was activated independently of the actions of omega-1 (ω-1), a previously described Th2-priming glycoprotein present in SEA. These findings were supported by in vivo murine data showing that ω-1-independent Th2 priming by SEA was mediated by Dectin-2 and Syk signaling in DCs. Finally, we found that Dectin-2-/-, and to a lesser extent Dectin-1-/- mice, displayed impaired Th2 responses and reduced egg-driven granuloma formation following S. mansoni infection, highlighting the physiological importance of this pathway in Th2 polarization during a helminth infection. In summary, we identified a novel pathway in DCs involving Dectin-1/2-Syk-PGE2-OX40L through which Th2 immune responses are induced.


Subject(s)
Dendritic Cells/immunology , Dinoprostone/immunology , Lectins, C-Type/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Th2 Cells/immunology , Animals , Antigens, Helminth/immunology , Antigens, Helminth/pharmacology , Autocrine Communication , Cell Differentiation , Cyclooxygenase 1/genetics , Cyclooxygenase 1/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Dendritic Cells/drug effects , Dendritic Cells/parasitology , Dinoprostone/metabolism , Enterotoxins/pharmacology , Gene Expression Regulation , Humans , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , MAP Kinase Signaling System , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , OX40 Ligand , Phospholipases A2/genetics , Phospholipases A2/immunology , Primary Cell Culture , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/pathology , Syk Kinase/genetics , Syk Kinase/immunology , Th2 Cells/drug effects , Th2 Cells/parasitology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology
12.
Int J Parasitol ; 48(5): 345-357, 2018 04.
Article in English | MEDLINE | ID: mdl-29510117

ABSTRACT

Previous studies have shown that schistosome infection can protect against allergic symptoms, but the underlying mechanisms are still not fully understood. Here we have shown that rabbit IgG antibodies raised against Schistosoma mansoni soluble egg antigens (SmSEA) are cross-reactive with a wide array of molecules in Timothy grass pollen (TGP) and birch tree pollen (BTP). Five of the cross-reactive pollen molecules (two from TGP and three from BTP) were selected randomly and identified by tandem mass spectrometric (TMS) analysis to be, respectively, the TGP allergens Phl p 1 and Phl p 5b, and BTP glutathione S-transferase (GST), and the BTP allergens Bet v 1 and Bet v 6.0102. Rabbit anti-SmSEA IgG antibodies that cross-reacted with each of the five allergens were found to be reactive with three major S. mansoni egg antigens, IPSE/alpha-1, omega-1 and kappa-5. Pairwise alignment of the amino acid sequences of each of the five TMS-identified pollen allergens with each of the three egg antigens revealed a low level of amino acid sequence identity. Further experiments indicated that the schistosome antigen/allergen cross-reactivity was mostly due to similar glycans present in helminths and plants, but not in mammals: so called cross-reactive carbohydrate determinants (CCDs). Previously, CCDs have been implicated in the cross-reactivity between many plants and invertebrates. Furthermore, pollen-induced anti-CCD IgGs have been found in sera of patients undergoing allergen-specific immunotherapy (SIT) and implicated in the treatment of the allergy. Thus, our finding provides not only possible explanations for the allergy-protective effect of helminth/schistosome infections as explained by the hygiene hypothesis, but also a potential starting point for improved SIT.


Subject(s)
Allergens/immunology , Betula , Phleum , Pollen/immunology , Schistosoma mansoni/immunology , Animals , Antibodies , Antibodies, Helminth , Epitopes , Hygiene Hypothesis , Immunoglobulin G , Mice , Periodic Acid , Plant Extracts , Polysaccharides
13.
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
14.
J Allergy Clin Immunol ; 140(2): 356-368, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28479330

ABSTRACT

Glycan-specific IgE antibodies cross-react with highly similar or even identical carbohydrate structures on a variety of different natural allergens, the so-called cross-reactive carbohydrate determinants (CCDs). In clinical practice CCDs often interfere with the specificity of in vitro allergy diagnostics, thus impairing allergy therapy decisions for individual patients. Strikingly, these IgE antibodies directed against CCDs often do not cause clinically relevant allergy symptoms. On the other hand, the IgE-binding glycan allergen galactose-α-(1,3)-galactose (α-Gal) is associated with IgE-mediated delayed anaphylaxis in meat allergy. The reason for this discrepancy is not known. The discovery of α-Gal stimulated new discussions and investigations regarding the relevance of anti-glycan IgE for allergic diseases. In this review the effect of glycans and glycan-specific IgE on sensitization to allergens and allergy diagnosis is described. Because parasite infections elicit a similar immunologic environment as allergic diseases, the association of glycan-specific antibodies against parasite glycoproteins with glycan structures on allergens is discussed.


Subject(s)
Allergens/immunology , Immunoglobulin E/immunology , Polysaccharides/immunology , Cross Reactions , Humans , Hypersensitivity/immunology , Immune Tolerance , Th2 Cells/immunology
15.
Sci Rep ; 7: 45910, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28393916

ABSTRACT

Helminth parasites control host-immune responses by secreting immunomodulatory glycoproteins. Clinical trials and mouse model studies have demonstrated the potential of helminth-derived glycoproteins for the treatment of immune-related diseases, like allergies and autoimmune diseases. Studies are however hampered by the limited availability of native parasite-derived proteins. Moreover, recombinant protein production systems have thus far been unable to reconstitute helminth-like glycosylation essential for the functionality of some helminth glycoproteins. Here we exploited the flexibility of the N-glycosylation machinery of plants to reconstruct the helminth glycoproteins omega-1 and kappa-5, two major constituents of immunomodulatory Schistosoma mansoni soluble egg antigens. Fine-tuning transient co-expression of specific glycosyltransferases in Nicotiana benthamiana enabled the synthesis of Lewis X (LeX) and LDN/LDN-F glycan motifs as found on natural omega-1 and kappa-5, respectively. In vitro and in vivo evaluation of the introduction of native LeX motifs on plant-produced omega-1 confirmed that LeX on omega-1 contributes to the glycoprotein's Th2-inducing properties. These data indicate that mimicking the complex carbohydrate structures of helminths in plants is a promising strategy to allow targeted evaluation of therapeutic glycoproteins for the treatment of inflammatory disorders. In addition, our results offer perspectives for the development of effective anti-helminthic vaccines by reconstructing native parasite glycoprotein antigens.


Subject(s)
Glycoproteins/immunology , Helminth Proteins/immunology , Nicotiana/immunology , Schistosoma mansoni/immunology , Animals , Antibodies, Helminth/genetics , Antibodies, Helminth/immunology , Antibodies, Helminth/metabolism , Antigens, Helminth/genetics , Antigens, Helminth/immunology , Antigens, Helminth/metabolism , Egg Proteins/genetics , Egg Proteins/immunology , Egg Proteins/metabolism , Gene Expression/immunology , Genetic Engineering , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Helminth Proteins/genetics , Helminth Proteins/metabolism , Immunomodulation/genetics , Immunomodulation/immunology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Schistosoma mansoni/genetics , Schistosoma mansoni/metabolism , Nicotiana/genetics , Nicotiana/metabolism , Vaccines/immunology
16.
PLoS One ; 11(7): e0159542, 2016.
Article in English | MEDLINE | ID: mdl-27467385

ABSTRACT

IgG antibodies produced by rabbits immunized against S. mansoni antigens cross-reacted with aqueous soluble constituents of a variety of allergens. The antibody cross-reactivity was largely sensitive to degradation by treatment of the target antigens with sodium meta-periodate, suggesting the cross-reactivity was due to carbohydrate determinants that were common to both the schistosome and the allergens (CCDs). The reaction between the rabbit antibodies and a 43 kDa molecule in a rubber latex extract was analysed further: tandem mass spectrometry identified the latex molecule as allergen Hev b 7. Rabbit anti-schistosome IgG antibodies purified by acid-elution from solid-phase latex Hev b 7 reacted with the S. mansoni egg antigens IPSE/alpha-1 and kappa-5 and cercarial antigens SPO-1 and a fatty acid-binding protein. Moreover, purified anti-S. mansoni egg, latex cross-reactive antibodies reacted with antigenic constituents of some fruits, a result of potential relevance to the latex-fruit syndrome of allergic reactions. We propose that IgG anti-schistosome antibodies that cross-react with allergens may be able to block IgE-induced allergic reactions and thus provide a possible explanation for the hygiene hypothesis.


Subject(s)
Antigens, Plant/immunology , Antigens, Protozoan/immunology , Carbohydrates/immunology , Cross Reactions , Plant Proteins/immunology , Schistosoma mansoni/immunology , Animals , Electrophoresis, Polyacrylamide Gel , Rabbits , Tandem Mass Spectrometry
17.
Glycobiology ; 25(12): 1465-79, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26347524

ABSTRACT

During the complex lifecycle of Schistosoma mansoni, a large variety of glycans is expressed. To many of these glycans, antibodies are induced by the infected host and some might be targets for vaccines or diagnostic tests. Spatial changes in glycan expression during schistosome development are largely unexplored. To study the surface-exposed glycans during the important initial stages of infection, we analyzed the binding of a panel of anti-glycan monoclonal antibodies (mAbs) to cercariae and schistosomula up to 72 h after transformation by immunofluorescence microscopy. The mAb specificity toward their natural targets was studied using a microarray containing a wide range of schistosomal N-glycans, O-glycans and glycosphingolipid glycans. With the exception of GalNAcß1-4(Fucα1-3)GlcNAc (LDN-F), mono- and multifucosylated GalNAcß1-4GlcNAc (LDN)-motifs were exposed at the surface of all developmental stages studied. Multifucosylated LDN-motifs were present on cercarial glycocalyx-derived O-glycans as well as cercarial glycolipids. In contrast, the Galß1-4(Fucα1-3)GlcNAc (Lewis X) and LDN-F-motifs, also expressed on cercarial glycolipids, and in addition on a range of cercarial N- and O-glycans, became surface expressed only after transformation of cercariae to schistosomula. In line with the documented shedding of the O-glycan-rich cercarial glycocalyx after transformation these observations suggest that surface accessible multifucosylated LDN-motifs are mostly expressed by O-glycans in cercariae, but principally by glycosphingolipids in schistosomula. We hypothesize that these temporal changes in surface exposure of glycan antigens are relevant to the interaction with the host during the initial stages of infection with schistosomes and discuss the potential of these glycan antigens as intervention targets.


Subject(s)
Cercaria/immunology , Glycocalyx/immunology , Polysaccharides/immunology , Schistosoma mansoni/immunology , Animals , Antibodies, Helminth/immunology , Antibodies, Monoclonal/immunology , Schistosoma mansoni/growth & development
18.
Int J Parasitol ; 45(13): 809-13, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26385440

ABSTRACT

The T2 ribonuclease omega-1 is a powerful Th2-inducing factor secreted by the eggs of the blood fluke Schistosoma mansoni. Omega-1 can modulate pattern recognition receptor-induced inflammatory signatures and alter antigen presentation by dendritic cells. Recent findings have suggested that component(s) contained in or secreted by S. mansoni eggs (soluble egg antigen) can also enhance IL-1ß secretion by dendritic cells stimulated with pattern recognition receptor ligands. Here we show that omega-1 enhances IL-1ß secretion in macrophages stimulated with Toll-like receptor 2 ligand, and propose omega-1 as the factor in soluble egg antigen capable of regulating inflammasome activity. This effect is dependent on the C-type lectin receptor Dectin-1, caspase-8 and the ASC inflammasome adaptor protein, highlighting the ability of omega-1 to regulate multiple pattern recognition receptor signalling pathways. These mechanistic insights into manipulation of host immunity by a parasite product have implications for the design of anti-inflammatory therapeutic drugs.


Subject(s)
Antigens, Helminth/metabolism , Egg Proteins/metabolism , Endoribonucleases/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Macrophages, Peritoneal/immunology , Schistosoma mansoni/immunology , Animals , Caspase 8/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endoribonucleases/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Macrophages, Peritoneal/metabolism , Mice , Schistosoma mansoni/enzymology , Th2 Cells/immunology
19.
J Biol Chem ; 290(36): 22111-26, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26163514

ABSTRACT

The IL-4-inducing principle from Schistosoma mansoni eggs (IPSE/α-1), the major secretory product of eggs from the parasitic worm S. mansoni, efficiently triggers basophils to release the immunomodulatory key cytokine interleukin-4. Activation by IPSE/α-1 requires the presence of IgE on the basophils, but the detailed molecular mechanism underlying activation is unknown. NMR and crystallographic analysis of IPSEΔNLS, a monomeric IPSE/α-1 mutant, revealed that IPSE/α-1 is a new member of the ßγ-crystallin superfamily. We demonstrate that this molecule is a general immunoglobulin-binding factor with highest affinity for IgE. NMR binding studies of IPSEΔNLS with the 180-kDa molecule IgE identified a large positively charged binding surface that includes a flexible loop, which is unique to the IPSE/α-1 crystallin fold. Mutational analysis of amino acids in the binding interface showed that residues contributing to IgE binding are important for IgE-dependent activation of basophils. As IPSE/α-1 is unable to cross-link IgE, we propose that this molecule, by taking advantage of its unique IgE-binding crystallin fold, activates basophils by a novel, cross-linking-independent mechanism.


Subject(s)
Antigens, Helminth/metabolism , Basophils/metabolism , Crystallins/immunology , Egg Proteins/metabolism , Helminth Proteins/metabolism , Immunoglobulin E/metabolism , Amino Acid Sequence , Animals , Antigens, Helminth/chemistry , Antigens, Helminth/genetics , Binding Sites/genetics , Blotting, Western , Chromatography, Gel , Crystallins/genetics , Crystallins/metabolism , Crystallography, X-Ray , Egg Proteins/chemistry , Egg Proteins/genetics , Helminth Proteins/chemistry , Helminth Proteins/genetics , Humans , Immunoglobulin E/chemistry , Interleukin-4/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Interaction Mapping , Protein Structure, Secondary , Protein Structure, Tertiary , Schistosoma mansoni/genetics , Schistosoma mansoni/metabolism , Sequence Homology, Amino Acid
20.
PLoS Negl Trop Dis ; 9(3): e0003593, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25774883

ABSTRACT

BACKGROUND: Schistosomiasis is a serious health problem especially in developing countries and affects more than 243 million people. Only few anthelmintic drugs are available up to now. A major obstacle for drug treatment is the different developmental stages and the varying host compartments during worm development. Anthelmintic drugs have been tested mainly on adult schistosomes or freshly transformed cercariae. Knowledge concerning the larval stages is lacking. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we used in vitro-grown schistosomula (aged between 2 to 14 days) to investigate drug effects of the three anthelmintics praziquantel, artemether, and oxamniquine. Further, we analyzed the antibody accessibility of two exemplary schistosome antigens SmCD59a and SmKK7, before and after drug treatment. Our results demonstrated that praziquantel applied at a concentration of 1 µM inhibited development of all life stages. Application of 10 µM praziquantel led to dramatic morphological changes of all schistosomula. Artemether at 1 and 10 µM had differential effects depending on whether it was applied to 2-day as compared to 7- and 14-day schistosomula. While 2-day schistosomula were not killed but inhibited from further development, severe morphological damage was seen in 7- and 14-day schistosomula. Oxamniquine (1 and 10 µM) led to severe morphological impairment in all life stages. Analyzing the accessibility of the antigens SmCD59a and SmKK7 before drug treatment showed no antibody binding on living intact schistosomula. However, when schistosomula were treated with anthelmintics, both antigens became exposed on the larvae. Oxamniquine turned out to be most effective in promoting antibody binding to all schistosomula stages. CONCLUSION: This study has revealed marked differences in anthelmintic drug effects against larvae. Drug treatment increases surface antigen presentation and renders larvae accessible to antibody attack.


Subject(s)
Anthelmintics/pharmacology , Antigens, Helminth/analysis , CD59 Antigens/analysis , Schistosoma mansoni/drug effects , Animals , Artemether , Artemisinins/pharmacology , Humans , Male , Praziquantel/pharmacology , Rats , Rats, Inbred BN , Rats, Inbred F344 , Schistosoma mansoni/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...