Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
FASEB J ; 35(2): e21331, 2021 02.
Article in English | MEDLINE | ID: mdl-33476078

ABSTRACT

Type 2 immunity plays an essential role in the maintenance of metabolic homeostasis and its disruption during obesity promotes meta-inflammation and insulin resistance. Infection with the helminth parasite Schistosoma mansoni and treatment with its soluble egg antigens (SEA) induce a type 2 immune response in metabolic organs and improve insulin sensitivity and glucose tolerance in obese mice, yet, a causal relationship remains unproven. Here, we investigated the effects and underlying mechanisms of the T2 ribonuclease omega-1 (ω1), one of the major S mansoni immunomodulatory glycoproteins, on metabolic homeostasis. We show that treatment of obese mice with plant-produced recombinant ω1, harboring similar glycan motifs as present on the native molecule, decreased body fat mass, and improved systemic insulin sensitivity and glucose tolerance in a time- and dose-dependent manner. This effect was associated with an increase in white adipose tissue (WAT) type 2 T helper cells, eosinophils, and alternatively activated macrophages, without affecting type 2 innate lymphoid cells. In contrast to SEA, the metabolic effects of ω1 were still observed in obese STAT6-deficient mice with impaired type 2 immunity, indicating that its metabolic effects are independent of the type 2 immune response. Instead, we found that ω1 inhibited food intake, without affecting locomotor activity, WAT thermogenic capacity or whole-body energy expenditure, an effect also occurring in leptin receptor-deficient obese and hyperphagic db/db mice. Altogether, we demonstrate that while the helminth glycoprotein ω1 can induce type 2 immunity, it improves whole-body metabolic homeostasis in obese mice by inhibiting food intake via a STAT6-independent mechanism.


Subject(s)
Eating , Endoribonucleases/therapeutic use , Glycoproteins/therapeutic use , Helminth Proteins/therapeutic use , Obesity/drug therapy , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Cells, Cultured , Endoribonucleases/pharmacology , Glycoproteins/pharmacology , Helminth Proteins/pharmacology , Locomotion , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Schistosoma mansoni/enzymology , T-Lymphocytes, Helper-Inducer/drug effects , Thermogenesis , Nicotiana/genetics , Nicotiana/metabolism
2.
Parasitology ; 149(3): 306-313, 2022 03.
Article in English | MEDLINE | ID: mdl-34736550

ABSTRACT

Assays which enable the detection of schistosome gut-associated circulating anodic (CAA) and cathodic (CCA) antigen in serum or urine are increasingly used as a diagnostic tool for schistosome infection. However, little is known about the production and clearance of these circulating antigens in relation to the sex and reproductive maturity of the parasite. Here we describe CAA and CCA excretion patterns by exploring a mouse model after exposure to 36 male-only, female-only and mixed (male/female) Schistosoma mansoni cercariae. We found that serum and urine CAA levels, analysed at 3 weeks intervals, peaked at 6 weeks post-infection. Worms recovered after perfusion at 14 weeks were cultured ex vivo. Male parasites excreted more circulating antigens than females, in the mouse model as well as ex vivo. In mixed infections (supporting egg production), serum CAA levels correlated to the number of recovered worms, whereas faecal egg counts or Schistosoma DNA in stool did not. No viable eggs and no inflammation were seen in the livers from mice infected with female worms only. Ex vivo, CAA levels were higher than CCA levels. Our study confirms that CAA levels reflect worm burden and allows detection of low-level single-sex infections.


Subject(s)
Parasites , Schistosomiasis mansoni , Animals , Antibodies, Helminth , Antigens, Helminth , Female , Male , Parasite Egg Count , Schistosoma mansoni , Schistosomiasis mansoni/diagnosis
3.
Eur J Immunol ; 49(8): 1226-1234, 2019 08.
Article in English | MEDLINE | ID: mdl-31099896

ABSTRACT

The helminth Schistosoma mansoni (S. mansoni) induces a network of regulatory immune cells, including interleukin (IL)-10-producing regulatory B cells (Bregs). However, the signals required for the development and activation of Bregs are not well characterized. Recent reports suggest that helminths induce type I interferons (IFN-I), and that IFN-I drive the development of Bregs in humans. We therefore assessed the role of IFN-I in the induction of Bregs by S. mansoni. Mice chronically infected with S. mansoni or i.v. injected with S. mansoni soluble egg antigen (SEA) developed a systemic IFN-I signature. Recombinant IFN-α enhanced IL-10 production by Bregs stimulated with S. mansoni SEA in vitro, while not activating Bregs by itself. IFN-I signaling also supported ex vivo IL-10 production by SEA-primed Bregs but was dispensable for activation of S. mansoni egg-induced Bregs in vivo. These data indicate that although IFN-I can serve as a coactivator for Breg IL-10 production, they are unlikely to participate in the development of Bregs in response to S. mansoni eggs.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Interferon Type I/metabolism , Schistosoma mansoni/physiology , Schistosomiasis mansoni/immunology , Animals , Antigens, Helminth/immunology , Cells, Cultured , Disease Models, Animal , Eggs , Female , Flow Cytometry , Humans , Interleukin-10/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Signal Transduction
4.
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
6.
J Infect Dis ; 218(7): 1142-1146, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29905805

ABSTRACT

To accelerate the development of novel vaccines for schistosomiasis, we set out to develop a human model for Schistosoma mansoni infection in healthy volunteers. During natural infections, female schistosomes produce eggs that give rise to morbidity. Therefore, we produced single-sex, male Schistosoma mansoni cercariae for human infection without egg production and associated pathology. Cercariae were produced in their intermediate snail hosts in accordance with the principles of good manufacturing practice (GMP). The application of GMP principles to an unconventional production process is a showcase for the controlled production of complex live challenge material in the European Union or under Food and Drug Administration guidance.


Subject(s)
Schistosoma mansoni/immunology , Schistosomiasis mansoni/prevention & control , Schistosomiasis/prevention & control , Snails/parasitology , Animals , Cercaria , Humans , Male , Schistosomiasis/parasitology , Schistosomiasis mansoni/parasitology
7.
Parasite Immunol ; 40(10): e12579, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30107039

ABSTRACT

Chronic helminth infection with Schistosoma (S.) mansoni protects against allergic airway inflammation (AAI) in mice and is associated with reduced Th2 responses to inhaled allergens in humans, despite the presence of schistosome-specific Th2 immunity. Schistosome eggs strongly induce type 2 immunity and allow to study the dynamics of Th2 versus regulatory responses in the absence of worms. Treatment with isolated S. mansoni eggs by i.p. injection prior to induction of AAI to ovalbumin (OVA)/alum led to significantly reduced AAI as assessed by less BAL and lung eosinophilia, less cellular influx into lung tissue, less OVA-specific Th2 cytokines in lungs and lung-draining mediastinal lymph nodes and less circulating allergen-specific IgG1 and IgE antibodies. While OVA-specific Th2 responses were inhibited, treatment induced a strong systemic Th2 response to the eggs. The protective effect of S. mansoni eggs was unaltered in µMT mice lacking mature (B2) B cells and unaffected by Treg cell depletion using anti-CD25 blocking antibodies during egg treatment and allergic sensitization. Notably, prophylactic egg treatment resulted in a reduced influx of pro-inflammatory, monocyte-derived dendritic cells into lung tissue of allergic mice following challenge. Altogether, S. mansoni eggs can protect against the development of AAI, despite strong egg-specific Th2 responses.


Subject(s)
Antibodies, Protozoan/blood , Asthma/prevention & control , Ovum/immunology , Schistosoma mansoni/immunology , Allergens/immunology , Alum Compounds/pharmacology , Animals , Antibodies, Protozoan/immunology , Asthma/immunology , Cytokines/immunology , Disease Models, Animal , Eosinophilia/prevention & control , Female , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Inflammation/pathology , Interleukin-2 Receptor alpha Subunit , Lung/immunology , Lung/parasitology , Lung/pathology , Mice, Inbred C57BL , Ovalbumin/immunology , Ovalbumin/pharmacology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
8.
FASEB J ; 29(7): 3027-39, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25852044

ABSTRACT

Chronic low-grade inflammation associated with obesity contributes to insulin resistance and type 2 diabetes. Helminth parasites are the strongest natural inducers of type 2 immune responses, and short-lived infection with rodent nematodes was reported to improve glucose tolerance in obese mice. Here, we investigated the effects of chronic infection (12 weeks) with Schistosoma mansoni, a helminth that infects millions of humans worldwide, on whole-body metabolic homeostasis and white adipose tissue (WAT) immune cell composition in high-fat diet-induced obese C57BL/6 male mice. Our data indicate that chronic helminth infection reduced body weight gain (-62%), fat mass gain (-89%), and adipocyte size; lowered whole-body insulin resistance (-23%) and glucose intolerance (-16%); and improved peripheral glucose uptake (+25%) and WAT insulin sensitivity. Analysis of immune cell composition by flow cytometry and quantitative PCR (qPCR) revealed that S. mansoni promoted strong increases in WAT eosinophils and alternatively activated (M2) macrophages. Importantly, injections with S. mansoni-soluble egg antigens (SEA) recapitulated the beneficial effect of parasite infection on whole-body metabolic homeostasis and induced type 2 immune responses in WAT and liver. Taken together, we provide novel data suggesting that chronic helminth infection and helminth-derived molecules protect against metabolic disorders by promoting a T helper 2 (Th2) response, eosinophilia, and WAT M2 polarization.


Subject(s)
Antigens, Helminth/administration & dosage , Insulin Resistance/immunology , Obesity/complications , Obesity/immunology , Schistosomiasis mansoni/complications , Schistosomiasis mansoni/immunology , Adipose Tissue, White/immunology , Adipose Tissue, White/pathology , Animals , Chronic Disease , Diet, High-Fat/adverse effects , Disease Models, Animal , Eosinophils/immunology , Female , Glucose Tolerance Test , Humans , Insulin Resistance/physiology , Liver/immunology , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Schistosoma mansoni/immunology , Schistosomiasis mansoni/metabolism , Th2 Cells/immunology
9.
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
10.
JCI Insight ; 8(11)2023 06 08.
Article in English | MEDLINE | ID: mdl-37140993

ABSTRACT

Obesity-associated metabolic inflammation drives the development of insulin resistance and type 2 diabetes, notably through modulating innate and adaptive immune cells in metabolic organs. The nutrient sensor liver kinase B1 (LKB1) has recently been shown to control cellular metabolism and T cell priming functions of DCs. Here, we report that hepatic DCs from high-fat diet-fed (HFD-fed) obese mice display increased LKB1 phosphorylation and that LKB1 deficiency in DCs (CD11cΔLKB1) worsened HFD-driven hepatic steatosis and impaired glucose homeostasis. Loss of LKB1 in DCs was associated with increased expression of Th17-polarizing cytokines and accumulation of hepatic IL-17A+ Th cells in HFD-fed mice. Importantly, IL-17A neutralization rescued metabolic perturbations in HFD-fed CD11cΔLKB1 mice. Mechanistically, deficiency of the canonical LKB1 target AMPK in HFD-fed CD11cΔAMPKα1 mice recapitulated neither the hepatic Th17 phenotype nor the disrupted metabolic homeostasis, suggesting the involvement of other and/or additional LKB1 downstream effectors. We indeed provide evidence that the control of Th17 responses by DCs via LKB1 is actually dependent on both AMPKα1 salt-inducible kinase signaling. Altogether, our data reveal a key role for LKB1 signaling in DCs in protection against obesity-induced metabolic dysfunctions by limiting hepatic Th17 responses.


Subject(s)
AMP-Activated Protein Kinases , Diabetes Mellitus, Type 2 , Mice , Animals , AMP-Activated Protein Kinases/metabolism , Interleukin-17/metabolism , Diabetes Mellitus, Type 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Obesity/metabolism , Liver/metabolism , Homeostasis , Dendritic Cells/metabolism
11.
PLoS Negl Trop Dis ; 17(6): e0011344, 2023 06.
Article in English | MEDLINE | ID: mdl-37363916

ABSTRACT

During chronic schistosome infections, a complex regulatory network is induced to regulate the host immune system, in which IL-10-producing regulatory B (Breg) cells play a significant role. Schistosoma mansoni soluble egg antigens (SEA) are bound and internalized by B cells and induce both human and mouse IL-10 producing Breg cells. To identify Breg-inducing proteins in SEA, we fractionated SEA by size exclusion chromatography and found 6 fractions able to induce IL-10 production by B cells (out of 18) in the high, medium and low molecular weight (MW) range. The high MW fractions were rich in heavily glycosylated molecules, including multi-fucosylated proteins. Using SEA glycoproteins purified by affinity chromatography and synthetic glycans coupled to gold nanoparticles, we investigated the role of these glycan structures in inducing IL-10 production by B cells. Then, we performed proteomics analysis on active low MW fractions and identified a number of proteins with putative immunomodulatory properties, notably thioredoxin (SmTrx1) and the fatty acid binding protein Sm14. Subsequent splenic murine B cell stimulations and hock immunizations with recombinant SmTrx1 and Sm14 showed their ability to dose-dependently induce IL-10 production by B cells both in vitro and in vivo. Identification of unique Breg cells-inducing molecules may pave the way to innovative therapeutic strategies for inflammatory and auto-immune diseases.


Subject(s)
B-Lymphocytes, Regulatory , Metal Nanoparticles , Schistosomiasis mansoni , Humans , Animals , Mice , Schistosoma mansoni , Schistosomiasis mansoni/prevention & control , Interleukin-10/genetics , Gold , Immunologic Factors , Thioredoxins/genetics , Antigens, Helminth
12.
EBioMedicine ; 97: 104832, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37837930

ABSTRACT

BACKGROUND: A controlled human infection model for schistosomiasis (CHI-S) can speed up vaccine development and provides insight into early immune responses following schistosome exposure. Recently, we established CHI-S model using single-sex male-only Schistosoma mansoni (Sm) cercariae in Schistosoma-naïve individuals. Given important differences in antigenic profile and human immune responses to schistosomes of different sex, we pioneered a single-sex female-only CHI-S model for future use in vaccine development. METHODS: We exposed 13 healthy, Schistosoma-naïve adult participants to 10 (n = 3) or 20 (n = 10) female cercariae and followed for 20 weeks, receiving treatment with praziquantel (PZQ) 60 mg/kg at week 8 and 12 after exposure. FINDINGS: The majority (11/13) participants reported rash and/or itch at the site of exposure, 5/13 had transient symptoms of acute schistosomiasis. Exposure to 20 cercariae led to detectable infection, defined as serum circulating anodic antigen levels >1.0 pg/mL, in 6/10 participants. Despite two rounds of PZQ treatment, 4/13 participants showed signs of persistent infection. Additional one- or three-day PZQ treatment (1 × 60 mg/kg and 3 × 60 mg/kg) or artemether did not result in cure, but over time three participants self-cured. Antibody, cellular, and cytokine responses peaked at week 4 post infection, with a mixed Th1, Th2, and regulatory profile. Cellular responses were (most) discriminative for symptoms. INTERPRETATION: Female-only infections exhibit similar clinical and immunological profiles as male-only infections but are more resistant to PZQ treatment. This limits future use of this model and may have important implications for disease control programs. FUNDING: European Union's Horizon 2020 (grant no. 81564).


Subject(s)
Anthelmintics , Schistosomiasis mansoni , Adult , Animals , Humans , Male , Female , Schistosomiasis mansoni/drug therapy , Healthy Volunteers , Schistosoma mansoni , Praziquantel/pharmacology , Praziquantel/therapeutic use , Cytokines , Anthelmintics/pharmacology , Anthelmintics/therapeutic use
13.
Cell Rep ; 40(1): 111032, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35793635

ABSTRACT

How mechanistic target of rapamycin complex 1 (mTORC1), a key regulator of cellular metabolism, affects dendritic cell (DC) metabolism and T cell-priming capacity has primarily been investigated in vitro, but how mTORC1 regulates this in vivo remains poorly defined. Here, using mice deficient for mTORC1 component raptor in DCs, we find that loss of mTORC1 negatively affects glycolytic and fatty acid metabolism and maturation of conventional DCs, particularly cDC1s. Nonetheless, antigen-specific CD8+ T cell responses to infection are not compromised and are even enhanced following skin immunization. This is associated with increased activation of Langerhans cells and a subpopulation of EpCAM-expressing cDC1s, of which the latter show an increased physical interaction with CD8+ T cells in situ. Together, this work reveals that mTORC1 limits CD8+ T cell priming in vivo by differentially orchestrating the metabolism and immunogenicity of distinct antigen-presenting cell subsets, which may have implications for clinical use of mTOR inhibitors.


Subject(s)
CD8-Positive T-Lymphocytes , Mechanistic Target of Rapamycin Complex 1 , Skin , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Signal Transduction , Skin/immunology , Skin/metabolism
14.
Methods Mol Biol ; 2270: 235-261, 2021.
Article in English | MEDLINE | ID: mdl-33479902

ABSTRACT

Toll-like receptors (TLRs) are pattern recognition receptors (PRRs), which constitute key components in the recognition of pathogens, thereby initiating innate immune responses and promoting adaptive immune responses. In B cells, TLR ligation is important for their activation and, together with CD40, for their differentiation. TLR ligands are also strong promoters of regulatory B (Breg)-cell development, by enhancing the production of IL-10 and their capacity to induce tolerance. In inflammatory diseases, such as autoimmunity or allergies, Breg-cell function is often impaired, while in chronic infections, such as with helminths, or cancer, Breg-cell function is boosted. Following pathogen exposure, B cells can respond directly by producing cytokines and/or IgM (innate response) and develop into various memory B (Bmem)-cell subsets with class-switched immunoglobulin receptors. Depending on the disease state or chronic infection conditions, various Breg subsets can be recognized as well. Currently, a large array of surface markers is known to distinguish between these large range of B-cell subsets. In recent years, the development of mass cytometers and spectral flow cytometry has allowed for high-dimensional detection of up to 48 markers, including both surface and intracellular/intranuclear markers. Therefore, this novel technology is highly suitable to provide a comprehensive overview of Bmem/Breg-cell subsets in different disease states and/or in clinical intervention trials. Here, we provide detailed instructions of the steps necessary to obtain high-quality data for high-dimensional analysis of multiple human Breg-cell subsets using various TLR ligands.


Subject(s)
B-Lymphocytes, Regulatory/cytology , Flow Cytometry/methods , Toll-Like Receptors/immunology , Autoimmunity/immunology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Cell Differentiation/immunology , Humans , Immune Tolerance/immunology , Immunity, Innate/immunology , Interleukin-10/immunology , Lymphocyte Activation/immunology
15.
J Extracell Vesicles ; 9(1): 1753420, 2020.
Article in English | MEDLINE | ID: mdl-32489529

ABSTRACT

Helminths like Schistosoma mansoni release excretory/secretory (E/S) products that modulate host immunity to enable infection. Extracellular vesicles (EVs) are among these E/S products, yet molecular mechanisms and functionality of S. mansoni EV interaction with host immune cells is unknown. Here we demonstrate that EVs released by S. mansoni schistosomula are internalised by human monocyte-derived dendritic cells (moDCs). Importantly, we show that this uptake was mainly mediated via DC-SIGN (CD209). Blocking DC-SIGN almost completely abrogated EV uptake, while blocking mannose receptor (MR, CD206) or dendritic cell immunoreceptor (DCIR, CLEC4A) had no effect on EV uptake. Mass spectrometric analysis of EV glycans revealed the presence of surface N-glycans with terminal Galß1-4(Fucα1-3)GlcNAc (LewisX) motifs, and a wide array of fucosylated lipid-linked glycans, including LewisX, a known ligand for DC-SIGN. Stimulation of moDCs with schistosomula EVs led to increased expression of costimulatory molecules CD86 and CD80 and regulatory surface marker PD-L1. Furthermore, schistosomula EVs increased expression of IL-12 and IL-10 by moDCs, which was partly dependent on the interaction with DC-SIGN. These results provide the first evidence that glycosylation of S. mansoni EVs facilitates the interaction with host immune cells and reveals a role for DC-SIGN and EV-associated glycoconjugates in parasite-induced immune modulation.

16.
Nat Med ; 26(3): 326-332, 2020 03.
Article in English | MEDLINE | ID: mdl-32066978

ABSTRACT

Schistosomiasis treatment relies on the use of a single drug, praziquantel, which is insufficient to control transmission in highly endemic areas1. Novel medicines and vaccines are urgently needed2,3. An experimental human model for schistosomiasis could accelerate the development of these products. We performed a dose-escalating clinical safety trial in 17 volunteers with male Schistosoma mansoni cercariae, which do not produce eggs (clinicaltrials.gov NCT02755324), at the Leiden University Medical Center, the Netherlands. The primary endpoints were adverse events and infectivity. We found a dose-related increase in adverse events related to acute schistosomiasis syndrome, which occurred in 9 of 17 volunteers. Overall, 5 volunteers (all 3 of the high dose group and 2 of 11 of the medium dose group) reported severe adverse events. Worm-derived circulating anodic antigen, the biomarker of the primary infection endpoint, peaked in 82% of volunteers at 3-10 weeks following exposure. All volunteers showed IgM and IgG1 seroconversion and worm-specific cytokine production by CD4+ T cells. All volunteers were cured with praziquantel provided at 12 weeks after exposure. Infection with 20 Schistosoma mansoni cercariae led to severe adverse events in 18% of volunteers and high infection rates. This infection model paves the way for fast-track product development for treatment and prevention of schistosomiasis.


Subject(s)
Antiparasitic Agents/therapeutic use , Models, Biological , Schistosoma mansoni/physiology , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/immunology , Vaccines/immunology , Adolescent , Adult , Animals , Antigens, Helminth/blood , Antigens, Helminth/immunology , Antiparasitic Agents/pharmacology , Cytokines/blood , Female , Humans , Immunity, Humoral/drug effects , Immunoglobulin M/blood , Male , Middle Aged , Praziquantel/pharmacology , Praziquantel/therapeutic use , Schistosoma mansoni/drug effects , Schistosomiasis mansoni/blood , Schistosomiasis mansoni/microbiology , Young Adult
17.
Cell Res ; 29(5): 406-419, 2019 05.
Article in English | MEDLINE | ID: mdl-30940876

ABSTRACT

Liver Kinase B1 (LKB1) plays a key role in cellular metabolism by controlling AMPK activation. However, its function in dendritic cell (DC) biology has not been addressed. Here, we find that LKB1 functions as a critical brake on DC immunogenicity, and when lost, leads to reduced mitochondrial fitness and increased maturation, migration, and T cell priming of peripheral DCs. Concurrently, loss of LKB1 in DCs enhances their capacity to promote output of regulatory T cells (Tregs) from the thymus, which dominates the outcome of peripheral immune responses, as suggested by increased resistance to asthma and higher susceptibility to cancer in CD11cΔLKB1 mice. Mechanistically, we find that loss of LKB1 specifically primes thymic CD11b+ DCs to facilitate thymic Treg development and expansion, which is independent from AMPK signalling, but dependent on mTOR and enhanced phospholipase C ß1-driven CD86 expression. Together, our results identify LKB1 as a critical regulator of DC-driven effector T cell and Treg responses both in the periphery and the thymus.


Subject(s)
Dendritic Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes, Regulatory/metabolism , AMP-Activated Protein Kinases , Animals , Asthma/immunology , Asthma/pathology , B7-2 Antigen/metabolism , CD11b Antigen/metabolism , CD11c Antigen/deficiency , CD11c Antigen/genetics , Cell Line, Tumor , Dendritic Cells/cytology , Disease Models, Animal , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Phospholipase C beta/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Signal Transduction , T-Lymphocytes, Regulatory/cytology , TOR Serine-Threonine Kinases/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
18.
Sci Rep ; 7(1): 10372, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871201

ABSTRACT

In humans and murine models of malaria, intradermal immunization (ID-I) with genetically attenuated sporozoites that arrest in liver induces lower protective immunity than intravenous immunization (IV-I). It is unclear whether this difference is caused by fewer sporozoites migrating into the liver or by suboptimal hepatic and injection site-dependent immune responses. We therefore developed a Plasmodium yoelii immunization/boost/challenge model to examine parasite liver loads as well as hepatic and lymph node immune responses in protected and unprotected ID-I and IV-I animals. Despite introducing the same numbers of genetically attenuated parasites in the liver, ID-I resulted in lower sterile protection (53-68%) than IV-I (93-95%). Unprotected mice developed less sporozoite-specific CD8+ and CD4+ effector T-cell responses than protected mice. After immunization, ID-I mice showed more interleukin-10-producing B and T cells in livers and skin-draining lymph nodes, but fewer hepatic CD8 memory T cells and CD8+ dendritic cells compared to IV-I mice. Our results indicate that the lower protection efficacy obtained by intradermal sporozoite administration is not linked to low hepatic parasite numbers as presumed before, but correlates with a shift towards regulatory immune responses. Overcoming these immune suppressive responses is important not only for live-attenuated malaria vaccines but also for other live vaccines administered in the skin.


Subject(s)
Liver/parasitology , Malaria/immunology , Malaria/parasitology , Parasite Load , Plasmodium/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Humans , Immunization , Life Cycle Stages , Liver/immunology , Lymphocyte Count , Mice , Parasitemia/parasitology , Plasmodium falciparum/immunology
19.
PLoS One ; 8(3): e59822, 2013.
Article in English | MEDLINE | ID: mdl-23527272

ABSTRACT

It is currently unknown how mucosal adjuvants cause induction of secretory immunoglobulin A (IgA), and how T cell-dependent (TD) or -independent (TI) pathways might be involved. Mucosal dendritic cells (DCs) are the primary antigen presenting cells driving TI IgA synthesis, by producing a proliferation-inducing ligand (APRIL), B cell activating factor (BAFF), Retinoic Acid (RA), TGF-ß or nitric oxide (NO). We hypothesized that the mucosal adjuvant Cholera Toxin subunit B (CTB) could imprint non-mucosal DCs to induce IgA synthesis, and studied the mechanism of its induction. In vitro, CTB-treated bone marrow derived DCs primed for IgA production by B cells without the help of T cells, yet required co-signaling by different Toll-like receptor (TLR) ligands acting via the MyD88 pathway. CTB-DC induced IgA production was blocked in vitro or in vivo when RA receptor antagonist, TGF-ß signaling inhibitor or neutralizing anti-TGF-ß was added, demonstrating the involvement of RA and TGF-ß in promoting IgA responses. There was no major involvement for BAFF, APRIL or NO. This study highlights that synergism between CTB and MyD88-dependent TLR signals selectively imprints a TI IgA-inducing capacity in non-mucosal DCs, explaining how CTB acts as an IgA promoting adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cholera Toxin/pharmacology , Dendritic Cells/metabolism , Immunoglobulin A/biosynthesis , Transforming Growth Factor beta/metabolism , Tretinoin/metabolism , Analysis of Variance , Animals , B-Lymphocytes/metabolism , Cell Culture Techniques , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
20.
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