Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.345
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 20(8): e1012457, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39186814

ABSTRACT

Chronic infection with Schistosoma mansoni parasites is associated with reduced allergic sensitization in humans, while schistosome eggs protects against allergic airway inflammation (AAI) in mice. One of the main secretory/excretory molecules from schistosome eggs is the glycosylated T2-RNAse Omega-1 (ω1). We hypothesized that ω1 induces protection against AAI during infection. Peritoneal administration of ω1 prior to sensitization with Ovalbumin (OVA) reduced airway eosinophilia and pathology, and OVA-specific Th2 responses upon challenge, independent from changes in regulatory T cells. ω1 was taken up by monocyte-derived dendritic cells, mannose receptor (CD206)-positive conventional type 2 dendritic cells (CD206+ cDC2), and by recruited peritoneal macrophages. Additionally, ω1 impaired CCR7, F-actin, and costimulatory molecule expression on myeloid cells and cDC2 migration in and ex vivo, as evidenced by reduced OVA+ CD206+ cDC2 in the draining mediastinal lymph nodes (medLn) and retainment in the peritoneal cavity, while antigen processing and presentation in cDC2 were not affected by ω1 treatment. Importantly, RNAse mutant ω1 was unable to reduce AAI or affect DC migration, indicating that ω1 effects are dependent on its RNAse activity. Altogether, ω1 hampers migration of OVA+ cDC2 to the draining medLn in mice, elucidating how ω1 prevents allergic airway inflammation in the OVA/alum mouse model.


Subject(s)
Cell Movement , Dendritic Cells , Ovalbumin , Schistosoma mansoni , Animals , Mice , Ovalbumin/immunology , Dendritic Cells/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Female , Mice, Inbred C57BL , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/prevention & control , Respiratory Hypersensitivity/parasitology , Th2 Cells/immunology , Inflammation/immunology
2.
PLoS Pathog ; 20(5): e1012268, 2024 May.
Article in English | MEDLINE | ID: mdl-38814989

ABSTRACT

The eggs of the blood fluke Schistosoma mansoni are the main cause of the clinical manifestations of chronic schistosomiasis. After laying, the egg "winners" attach to the endothelium of the mesenteric vein and, after a period of development, induce the growth of a small granuloma, which facilitates their passage to the intestinal lumen. Egg "losers" carried by the bloodstream to non-specific tissues also undergo full development and induce large granuloma formation, but their life ends there. Although these trapped eggs represent a dead end in the parasite life cycle, the vast majority of studies attempting to describe the biology of the S. mansoni eggs have studied these liver-trapped "losers" instead of migrating intestinal "winners". This raises the fundamental question of how these eggs differ. With robust comparative transcriptomic analysis performed on S. mansoni eggs isolated 7 weeks post infection, we show that gene expression is critically dependent on tissue localization, both in the early and late stages of development. While mitochondrial genes and venom allergen-like proteins are significantly upregulated in mature intestinal eggs, well-described egg immunomodulators IPSE/alpha-1 and omega-1, together with micro-exon genes, are predominantly expressed in liver eggs. In addition, several proteases and protease inhibitors previously implicated in egg-host interactions display clear tissue-specific gene expression patterns. These major differences in gene expression could be then reflected in the observed different ability of liver and intestinal soluble egg antigens to elicit host immune responses and in the shorter viability of miracidia hatched from liver eggs. Our comparative analysis provides a new perspective on the biology of parasite's eggs in the context of their development and tissue localization. These findings could contribute to a broader and more accurate understanding of parasite eggs interactions with the host, which have historically been often restricted to liver eggs and sometimes inaccurately generalized.


Subject(s)
Liver , Schistosoma mansoni , Schistosomiasis mansoni , Animals , Schistosoma mansoni/immunology , Schistosoma mansoni/genetics , Liver/parasitology , Liver/immunology , Liver/metabolism , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Mice , Ovum/metabolism , Ovum/immunology , Intestines/parasitology , Intestines/immunology , Antigens, Helminth/immunology , Helminth Proteins/genetics , Helminth Proteins/metabolism , Helminth Proteins/immunology , Female , Egg Proteins
3.
Immunity ; 45(1): 145-58, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27421703

ABSTRACT

Fibroproliferative diseases are driven by dysregulated tissue repair responses and are a major cause of morbidity and mortality because they affect nearly every organ system. Type 2 cytokine responses are critically involved in tissue repair; however, the mechanisms that regulate beneficial regeneration versus pathological fibrosis are not well understood. Here, we have shown that the type 2 effector cytokine interleukin-13 simultaneously, yet independently, directed hepatic fibrosis and the compensatory proliferation of hepatocytes and biliary cells in progressive models of liver disease induced by interleukin-13 overexpression or after infection with Schistosoma mansoni. Using transgenic mice with interleukin-13 signaling genetically disrupted in hepatocytes, cholangiocytes, or resident tissue fibroblasts, we have revealed direct and distinct roles for interleukin-13 in fibrosis, steatosis, cholestasis, and ductular reaction. Together, these studies show that these mechanisms are simultaneously controlled but distinctly regulated by interleukin-13 signaling. Thus, it may be possible to promote interleukin-13-dependent hepatobiliary expansion without generating pathological fibrosis. VIDEO ABSTRACT.


Subject(s)
Fatty Liver/immunology , Fibroblasts/immunology , Interleukin-13/metabolism , Liver Cirrhosis, Biliary/immunology , Liver/pathology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Bile Acids and Salts/biosynthesis , Cell Proliferation , Cells, Cultured , Fibrosis , Humans , Interleukin-13/genetics , Interleukin-13/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Th2 Cells/immunology
4.
Exp Parasitol ; 261: 108752, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38604301

ABSTRACT

AIMS: We have previously reported reduction of anti-type II collagen (IIC) IgG levels in collagen-induced arthritis (CIA) by Schistosoma mansoni (Sm) and Trichinella spiralis (Ts). To clarify the contribution of the impairment of humoral immunity to their anti-arthritic activities, we herein investigated the relationship between anti-IIC IgG levels and arthritic swelling in Sm- or Ts-infected mice. METHODS AND RESULTS: Male DBA/1J mice were infected with Sm cercariae or Ts muscle larvae prior to the IIC immunization. In the Sm-infected mice, paw swelling and anti-IIC IgG levels were continuously lower than those of non-infected control group. In contrast, arthritic swelling in the Ts-infected mice only decreased in the early phase of CIA progression, despite the continued impairment of anti-IIC IgG production throughout the experimental period. Correlation coefficients between residual paw swelling and anti-IIC IgG titers were similar or higher in the Sm group than in the control group, but were similar or lower in the Ts group than in the control group. CONCLUSION: The down-modulations of anti-IIC IgG levels by the two parasitic infections and the correlation analyses suggest that the anti-arthritic activity of Sm was primarily attributed to the modulation of IgG-independent arthritogenic mechanisms and secondarily to the impairment of anti-IIC IgG production. In contrast, Ts could alleviate CIA mainly via the impairment of antibody production.


Subject(s)
Arthritis, Experimental , Immunity, Humoral , Immunoglobulin G , Mice, Inbred DBA , Schistosoma mansoni , Schistosomiasis mansoni , Trichinella spiralis , Trichinellosis , Animals , Trichinella spiralis/immunology , Male , Mice , Immunoglobulin G/blood , Arthritis, Experimental/immunology , Schistosomiasis mansoni/immunology , Trichinellosis/immunology , Schistosoma mansoni/immunology , Collagen Type II/immunology , Antibodies, Helminth/blood
5.
Mem Inst Oswaldo Cruz ; 119: e240013, 2024.
Article in English | MEDLINE | ID: mdl-38896633

ABSTRACT

BACKGROUND: The impact of Schistosoma mansoni infection over the immune response and the mechanisms involved in pathogenesis are not yet completely understood. OBJECTIVES: This study aimed to evaluate the expression of innate immune receptors in three distinct mouse lineages (BALB/c, C57BL/6 and Swiss) during experimental S. mansoni infection with LE strain. METHODS: The parasite burden, intestinal tissue oogram and presence of hepatic granulomas were evaluated at 7- and 12-weeks post infection (wpi). The mRNA expression for innate Toll-like receptors, Nod-like receptors, their adaptor molecules, and cytokines were determined at 2, 7 and 12 wpi in the hepatic tissue by real-time quantitative polymerase chain reaction (qPCR). FINDINGS: Swiss mice showed 100% of survival, had lower parasite burden and intestinal eggs, while infected BALB/c and C57BL/6 presented 80% and 90% of survival, respectively, higher parasite burden and intestinal eggs. The three mouse lineages displayed distinct patterns in the expression of innate immune receptors, their adaptor molecules and cytokines, at 2 and 7 wpi. MAIN CONCLUSIONS: Our results suggest that the pathogenesis of S. mansoni infection is related to a dynamic early activation of innate immunity receptors and cytokines important for the control of developing worms.


Subject(s)
Cytokines , Immunity, Innate , Mice, Inbred BALB C , Mice, Inbred C57BL , Schistosomiasis mansoni , Animals , Schistosomiasis mansoni/immunology , Immunity, Innate/immunology , Cytokines/immunology , Mice , Schistosoma mansoni/immunology , Disease Models, Animal , Female , Toll-Like Receptors/immunology , Real-Time Polymerase Chain Reaction , Parasite Egg Count , Male , RNA, Messenger , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology
6.
Int J Mol Sci ; 25(17)2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39273153

ABSTRACT

BACKGROUND: Schistosomiasis is a parasitic infection that can cause pulmonary hypertension (PH). Th2 CD4 T cells are necessary for experimental Schistosoma-PH. However, if T cells migrate to the lung to initiate, the localized inflammation that drives vascular remodeling and PH is unknown. METHODS: Mice were sensitized to Schistosoma mansoni eggs intraperitoneally and then challenged using tail vein injection. FTY720 was administered, which blocks lymphocyte egress from lymph nodes. T cells were quantified using flow cytometry, PH severity via heart catheterization, and cytokine concentration through ELISA. RESULTS: FTY720 decreased T cells in the peripheral blood, and increased T cells in the mediastinal lymph nodes. However, FTY720 treatment resulted in no change in PH or type 2 inflammation severity in mice sensitized and challenged with S. mansoni eggs, and the number of memory and effector CD4 T cells in the lung parenchyma was also unchanged. Notably, intraperitoneal Schistosoma egg sensitization alone resulted in a significant increase in intravascular lymphocytes and T cells, including memory T cells, although there was no significant change in parenchymal cell density, IL-4 or IL-13 expression, or PH. CONCLUSION: Blocking T cell migration did not suppress PH following Schistosoma egg challenge. Memory CD4 T cells, located in the lung intravascular space following egg sensitization, appear sufficient to cause type 2 inflammation and PH.


Subject(s)
Hypertension, Pulmonary , Lung , Schistosoma mansoni , Animals , Mice , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/parasitology , Hypertension, Pulmonary/immunology , Lung/parasitology , Lung/immunology , Lung/pathology , Schistosoma mansoni/immunology , Fingolimod Hydrochloride/pharmacology , Female , CD4-Positive T-Lymphocytes/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/complications , Schistosomiasis mansoni/pathology , Disease Models, Animal , Interleukin-4/metabolism , Cytokines/metabolism , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Schistosomiasis/complications , Schistosomiasis/immunology , Schistosomiasis/parasitology
7.
PLoS Pathog ; 17(1): e1009198, 2021 01.
Article in English | MEDLINE | ID: mdl-33417618

ABSTRACT

Macrophages have a defined role in the pathogenesis of metabolic disease and cholesterol metabolism where alternative activation of macrophages is thought to be beneficial to both glucose and cholesterol metabolism during high fat diet induced disease. It is well established that helminth infection protects from metabolic disease, but the mechanisms underlying protection are not well understood. Here, we investigated the effects of Schistosoma mansoni infection and cytokine activation in the metabolic signatures of bone marrow derived macrophages using an approach that integrated transcriptomics, metabolomics, and lipidomics in a metabolic disease prone mouse model. We demonstrate that bone marrow derived macrophages (BMDM) from S. mansoni infected male ApoE-/- mice have dramatically increased mitochondrial respiration compared to those from uninfected mice. This change is associated with increased glucose and palmitate shuttling into TCA cycle intermediates, increased accumulation of free fatty acids, and decreased accumulation of cellular cholesterol esters, tri and diglycerides, and is dependent on mgll activity. Systemic injection of IL-4 complexes is unable to recapitulate either reductions in systemic glucose AUC or the re-programing of BMDM mitochondrial respiration seen in infected males. Importantly, the metabolic reprogramming of male myeloid cells is transferrable via bone marrow transplantation to an uninfected host, indicating maintenance of reprogramming in the absence of sustained antigen exposure. Finally, schistosome induced metabolic and bone marrow modulation is sex-dependent, with infection protecting male, but not female mice from glucose intolerance and obesity. Our findings identify a transferable, long-lasting sex-dependent reprograming of the metabolic signature of macrophages by helminth infection, providing key mechanistic insight into the factors regulating the beneficial roles of helminth infection in metabolic disease.


Subject(s)
Antigens/immunology , Cell Lineage , Macrophages/metabolism , Metabolic Diseases/prevention & control , Myeloid Cells/metabolism , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/metabolism , Animals , Cellular Reprogramming , Diet, High-Fat/adverse effects , Female , Lipid Metabolism , Macrophages/immunology , Macrophages/parasitology , Male , Metabolic Diseases/immunology , Metabolic Diseases/parasitology , Metabolome , Mice , Mice, Knockout, ApoE , Myeloid Cells/immunology , Myeloid Cells/parasitology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology
8.
Nat Immunol ; 12(6): 527-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21552267

ABSTRACT

Contributions by basophils to allergic and helminth immunity remain incompletely defined. Using sensitive interleukin 4 (Il4) reporter alleles, we demonstrate here that basophil IL-4 production occurs by a CD4(+) T cell-dependent process restricted to the peripheral tissues affected. We genetically marked and achieved specific deletion of basophils and found that basophils did not mediate T helper type 2 (T(H)2) priming in vivo. Two-photon imaging confirmed that basophils did not interact with antigen-specific T cells in lymph nodes but engaged in prolonged serial interactions with T cells in lung tissues. Although targeted deletion of IL-4 and IL-13 in either CD4(+) T cells or basophils had a minimal effect on worm clearance, deletion from both lineages demonstrated a nonredundant role for basophil cytokines in primary helminth immunity.


Subject(s)
Basophils/immunology , Interleukin-4/immunology , Lung/immunology , Strongylida Infections/immunology , Animals , Basophils/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Helminthiasis, Animal/immunology , Helminthiasis, Animal/metabolism , Helminthiasis, Animal/parasitology , Host-Parasite Interactions/immunology , Humans , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Liver/immunology , Liver/metabolism , Liver/parasitology , Lung/metabolism , Lung/parasitology , Lung Diseases, Parasitic/immunology , Lung Diseases, Parasitic/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Nippostrongylus/immunology , Nippostrongylus/physiology , Schistosoma mansoni/immunology , Schistosoma mansoni/physiology , Strongylida Infections/metabolism , Strongylida Infections/parasitology
9.
Proc Natl Acad Sci U S A ; 117(37): 23174-23181, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32868437

ABSTRACT

Schistosomiasis is among the most common parasitic diseases in the world, with over 142 million people infected in low- and middle-income countries. Measuring population-level transmission is centrally important in guiding schistosomiasis control programs. Traditionally, human Schistosoma mansoni infections have been detected using stool microscopy, which is logistically difficult at program scale and has low sensitivity when people have low infection burdens. We compared serological measures of transmission based on antibody response to S. mansoni soluble egg antigen (SEA) with stool-based measures of infection among 3,663 preschool-age children in an area endemic for S. mansoni in western Kenya. We estimated force of infection among children using the seroconversion rate and examined how it varied geographically and by age. At the community level, serological measures of transmission aligned with stool-based measures of infection (ρ = 0.94), and serological measures provided more resolution for between-community differences at lower levels of infection. Force of infection showed a clear gradient of transmission with distance from Lake Victoria, with 94% of infections and 93% of seropositive children in communities <1.5 km from the lake. Force of infection increased through age 3 y, by which time 65% (95% CI: 53%, 75%) of children were SEA positive in high-transmission communities-2 y before they would be reached by school-based deworming programs. Our results show that serologic surveillance platforms represent an important opportunity to guide and monitor schistosomiasis control programs, and that in high-transmission settings preschool-age children represent a key population missed by school-based deworming programs.


Subject(s)
Antibody Formation/immunology , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Schistosomiasis/immunology , Animals , Child, Preschool , Feces/parasitology , Female , Humans , Infant , Kenya , Male , Prevalence , Schistosomiasis/parasitology , Schistosomiasis mansoni/parasitology
10.
Proc Natl Acad Sci U S A ; 117(32): 19299-19309, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32737161

ABSTRACT

Schistosomes are parasitic flatworms that cause schistosomiasis, a neglected tropical disease affecting over 200 million people. Schistosomes develop multiple body plans while navigating their complex life cycle, which involves two different hosts: a mammalian definitive host and a molluscan intermediate host. Their survival and propagation depend upon proliferation and differentiation of stem cells necessary for parasite homeostasis and reproduction. Infective larvae released from snails carry a handful of stem cells that serve as the likely source of new tissues as the parasite adapts to life inside the mammalian host; however, the role of these stem cells during this critical life cycle stage remains unclear. Here, we characterize stem cell fates during early intramammalian development. Surprisingly, we find that the esophageal gland, an accessory organ of the digestive tract, develops before the rest of the digestive system is formed and blood feeding is initiated, suggesting a role in processes beyond nutrient uptake. To explore such a role, we examine schistosomes that lack the esophageal gland due to knockdown of a forkhead-box transcription factor, Sm-foxA, which blocks development and maintenance of the esophageal gland, without affecting the development of other somatic tissues. Intriguingly, schistosomes lacking the esophageal gland die after transplantation into naive mice, but survive in immunodeficient mice lacking B cells. We show that parasites lacking the esophageal gland are unable to lyse ingested immune cells within the esophagus before passing them into the gut. These results unveil an immune-evasion mechanism mediated by the esophageal gland, which is essential for schistosome survival and pathogenesis.


Subject(s)
Esophagus/parasitology , Immune Evasion , Schistosoma mansoni/immunology , Schistosomiasis mansoni/immunology , Animals , Esophagus/immunology , Female , Helminth Proteins/genetics , Helminth Proteins/immunology , Host-Parasite Interactions , Humans , Life Cycle Stages , Male , Mice , Schistosoma mansoni/genetics , Schistosoma mansoni/growth & development , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/physiopathology
11.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163381

ABSTRACT

Schistosoma mansoni uses different mechanisms to escape its host's immunity. Understanding the ability of memory T cells to withstand this pathogen's manipulation is important for the development of effective vaccines against this immunomodulatory pathogen. In this study, ovalbumin (OVA) transgenic S. mansoni is used as a tool to investigate whether fully differentiated Th1, Th2 and Th17 cells are able to withstand pathogen manipulation. Naïve T cells from OT-II T cell receptor transgenic mice with a specificity for OVA were differentiated into Th1, Th2, and Th17 polarised memory cells in vitro. These cells were adoptively transferred into recipient mice to investigate whether these polarised immune memory T cells are resilient in the face of pathogen-mediated manipulation. After transferring memory cells, mice were challenged with OVA-transduced S. mansoni eggs as well as wild-type controls. The in vitro differentiated Th1, Th2 and Th17 memory cells continued to produce the same cytokines when challenged by OVA-expressing S. mansoni eggs as to these they produced when transferred in vivo, suggesting that the Th phenotypes of the memory T cells remains unaltered in the face of stimulation by S. mansoni. The ability of memory T cells to remain resilient to manipulation by the parasite suggests that vaccines might be able to produce immune memory responses able to withstand S. mansoni immune manipulation and hence protect the host from infection.


Subject(s)
Immunity , Schistosoma mansoni/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Antigens/immunology , Cell Polarity , Cell Proliferation , Cytokines/metabolism , Female , Immunologic Memory , Lymph Nodes/metabolism , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Ovum/metabolism , Schistosomiasis mansoni/immunology , Spleen/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
12.
EMBO J ; 36(16): 2404-2418, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28716804

ABSTRACT

Type 2 inflammation is a defining feature of infection with parasitic worms (helminths), as well as being responsible for widespread suffering in allergies. However, the precise mechanisms involved in T helper (Th) 2 polarization by dendritic cells (DCs) are currently unclear. We have identified a previously unrecognized role for type I IFN (IFN-I) in enabling this process. An IFN-I signature was evident in DCs responding to the helminth Schistosoma mansoni or the allergen house dust mite (HDM). Further, IFN-I signaling was required for optimal DC phenotypic activation in response to helminth antigen (Ag), and efficient migration to, and localization with, T cells in the draining lymph node (dLN). Importantly, DCs generated from Ifnar1-/- mice were incapable of initiating Th2 responses in vivo These data demonstrate for the first time that the influence of IFN-I is not limited to antiviral or bacterial settings but also has a central role to play in DC initiation of Th2 responses.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/metabolism , Th2 Cells/immunology , Allergens/immunology , Animals , Mice , Mice, Knockout , Pyroglyphidae/immunology , Receptor, Interferon alpha-beta/deficiency , Schistosoma mansoni/immunology
13.
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
14.
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
15.
Parasitology ; 148(4): 420-426, 2021 04.
Article in English | MEDLINE | ID: mdl-33190646

ABSTRACT

This study aimed to evaluate the performance of the point-of-care circulating cathodic antigen (POC-CCA) test in a highly endemic area in Brazil, comparing it to the Kato-Katz (KK) technique for sensitivity, specificity and the intensity of the reaction of the test in relation to the parasitic load. The community in Sergipe, Brazil, participated in the study, providing three stool samples, one of urine (POC-CCA) and fingers tick blood sample was tested by enzyme-linked immunosorbent assay (ELISA). Sensitivity, specificity, positive predictive value, negative predictive value, accuracy, kappa coefficient and Spearman's correlation were calculated for the POC-CCA test using the KK as the reference. The prevalence of schistosomiasis by KK testing was 48.82%; POC-CCA (t+) 66.14%; POC-CCA (t-) 45.24%. ELISA results showed 100% agreement in individuals with high and moderate eggs per gram (EPG). POC-CCA presented good diagnostic performance in individuals with medium and high EPG, but there were a high number of false negatives in individuals with low intensity infections. As observed, POC-CCA-filter test improves accuracy and sensitivity compared to a conventional test.


Subject(s)
Antigens, Helminth/blood , Feces/parasitology , Schistosomiasis mansoni/diagnosis , Adolescent , Adult , Animals , Brazil/epidemiology , Child , Child, Preschool , Endemic Diseases , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Point-of-Care Testing , Prevalence , ROC Curve , Schistosoma mansoni/immunology , Schistosoma mansoni/isolation & purification , Schistosomiasis mansoni/epidemiology , Urine/parasitology , Young Adult
16.
Parasitol Res ; 120(3): 1011-1023, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33409634

ABSTRACT

Schistosomiasis still affects a lot of people in many developing countries. Reducing the disease dissemination has been the target of various studies. As methyl gallate has antioxidant properties, it is assumed that it can be a good candidate for stimulating the immune response of snails. So, the aim of this work is to investigate the potential of using methyl gallate as an immunostimulant to Biomphalaria alexandrina snails in order to prevent the development of invading miracidia into infective cercariae. The infected snails were exposed to three concentrations of methyl gallate for two periods: 24 and 72 h. The results indicated that the most effective concentration was the lowest one: 125 mg/L of methyl gallate for 72 h, as it reduced both infection rate and mean number of shed cercariae. Also, it increased the total number of snails' hemocytes in hemolymph, which were observed in head-foot region and digestive gland of treated snails surrounding degenerated sporocysts and cercariae. In addition, hydrogen peroxide showed its highest content in tissues of snails exposed to 125 mg/L of methyl gallate for 72 h. In conclusion, methyl gallate can be considered as one of the most promising immunostimulants of B. alexandrina snails against infection with Schistosoma mansoni.


Subject(s)
Adjuvants, Immunologic/pharmacology , Biomphalaria/drug effects , Biomphalaria/parasitology , Gallic Acid/analogs & derivatives , Schistosoma mansoni/immunology , Animals , Biomphalaria/immunology , Gallic Acid/pharmacology , Hemocytes/drug effects , Hemolymph/cytology , Hemolymph/drug effects , Immunity/drug effects , Oocysts/drug effects , Schistosoma mansoni/drug effects
17.
Infect Immun ; 88(8)2020 07 21.
Article in English | MEDLINE | ID: mdl-32341115

ABSTRACT

The parasites and eggs of helminths, including schistosomes, are associated with factors that can modulate the nature and outcomes of host immune responses, particularly enhancing type 2 immunity and impairing the effects of type 1 and type 17 immunity. The main species of schistosomes that cause infection in humans are capable of generating a microenvironment that allows survival of the parasite by evasion of the immune response. Schistosome infections are associated with beneficial effects on chronic immune disorders, including allergies, autoimmune diseases, and alloimmune responses. Recently, there has been increasing research interest in the role of schistosomes in immunoregulation during human infection, and the mechanisms underlying these roles continue to be investigated. Further studies may identify potential opportunities to develop new treatments for immune disease. In this review, we provide an update on the advances in our understanding of schistosome-associated modulation of the cells of the innate and adaptive immune systems as well as the potential role of schistosome-associated factors as therapeutic modulators of immune disorders, including allergies, autoimmune diseases, and transplant immunopathology. We also discuss potential opportunities for targeting schistosome-induced immunoregulation for future translation to the clinical setting.


Subject(s)
Autoimmune Diseases/therapy , Hypersensitivity/therapy , Immunologic Factors/therapeutic use , Schistosoma japonicum/immunology , Schistosoma mansoni/immunology , Schistosomiasis/therapy , Adaptive Immunity/drug effects , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/parasitology , Autoimmune Diseases/pathology , Hypersensitivity/immunology , Hypersensitivity/parasitology , Hypersensitivity/pathology , Immune Evasion , Immunity, Innate/drug effects , Immunomodulation , Immunotherapy/methods , Organ Transplantation/rehabilitation , Schistosoma japonicum/chemistry , Schistosoma mansoni/chemistry , Schistosomiasis/immunology , Schistosomiasis/parasitology , Schistosomiasis/pathology , Th1 Cells/immunology , Th1 Cells/parasitology , Th17 Cells/immunology , Th17 Cells/parasitology , Th2 Cells/immunology , Th2 Cells/parasitology , Zygote/chemistry , Zygote/immunology
18.
PLoS Pathog ; 14(2): e1006870, 2018 02.
Article in English | MEDLINE | ID: mdl-29425229

ABSTRACT

Protease inhibitors have important function during homeostasis, inflammation and tissue injury. In this study, we described the role of Schistosoma mansoni SmKI-1 serine protease inhibitor in parasite development and as a molecule capable of regulating different models of inflammatory diseases. First, we determine that recombinant (r) SmKI-1 and its Kunitz domain but not the C-terminal region possess inhibitory activity against trypsin and neutrophil elastase (NE). To better understand the molecular basis of NE inhibition by SmKI-1, molecular docking studies were also conducted. Docking results suggest a complete blockage of NE active site by SmKI-1 Kunitz domain. Additionally, rSmKI-1 markedly inhibited the capacity of NE to kill schistosomes. In order to further investigate the role of SmKI-1 in the parasite, we designed specific siRNA to knockdown SmKI-1 in S. mansoni. SmKI-1 gene suppression in larval stage of S. mansoni robustly impact in parasite development in vitro and in vivo. To determine the ability of SmKI-1 to interfere with neutrophil migration and function, we tested SmKI-1 anti-inflammatory potential in different murine models of inflammatory diseases. Treatment with SmKI-1 rescued acetaminophen (APAP)-mediated liver damage, with a significant reduction in both neutrophil recruitment and elastase activity. In the model of gout arthritis, this protein reduced neutrophil accumulation, IL-1ß secretion, hypernociception, and overall pathological score. Finally, we demonstrated the ability of SmKI-1 to inhibit early events that trigger neutrophil recruitment in pleural cavities of mice in response to carrageenan. In conclusion, SmKI-1 is a key protein in S. mansoni survival and it has the ability to inhibit neutrophil function as a promising therapeutic molecule against inflammatory diseases.


Subject(s)
Inflammation/metabolism , Leukocyte Elastase/metabolism , Neutrophils/drug effects , Schistosoma mansoni , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/pharmacology , Animals , Cells, Cultured , Female , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Neutrophils/physiology , Protein Binding , Schistosoma mansoni/immunology , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/metabolism
19.
Nat Immunol ; 9(1): 25-33, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18066066

ABSTRACT

The interleukin 4 receptor (IL-4R) is a central mediator of T helper type 2 (T(H)2)-mediated disease and associates with either the common gamma-chain to form the type I IL-4R or with the IL-13R alpha1 chain (IL-13Ralpha1) to form the type II IL-4R. Here we used Il13ra1-/- mice to characterize the distinct functions of type I and type II IL-4 receptors in vivo. In contrast to Il4ra-/- mice, which have weak T(H)2 responses, Il13ra1-/- mice had exacerbated T(H)2 responses. Il13ra1-/- mice showed much less mortality after infection with Schistosoma mansoni and much more susceptibility to Nippostrongylus brasiliensis. IL-13Ralpha1 was essential for allergen-induced airway hyperreactivity and mucus hypersecretion but not for fibroblast or alternative macrophage activation. Thus, type I and II IL-4 receptors exert distinct effects on immune responses.


Subject(s)
Interleukin-13 Receptor alpha1 Subunit/physiology , Receptors, Interleukin-4, Type II/physiology , Th2 Cells/immunology , Allergens/immunology , Animals , Antigens, Helminth/immunology , Bronchial Hyperreactivity/immunology , Cells, Cultured , Disease Susceptibility , Fibroblasts/immunology , Interleukin-13 Receptor alpha1 Subunit/genetics , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mucus/metabolism , Nippostrongylus/physiology , Schistosoma mansoni/immunology , Schistosoma mansoni/physiology , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/mortality , Strongylida Infections/immunology
20.
Cytokine ; 127: 154931, 2020 03.
Article in English | MEDLINE | ID: mdl-31783260

ABSTRACT

Human co-infection by helminth species is frequent, but their consequences are mostly unknown. Here, we investigate the impact of Strongyloides venezuelensis co-infection on the immune response, schistosome burden, and the associated pathology of schistosomiasis in mice. Co-infection did not alter the schistosome parasite burden, but reduced the IL-4/IL-10 ratio during acute schistosomiasis, indicating induction of modulatory mechanisms. Simultaneous infection with S. venezuelensis and S. mansoni increased the liver concentration of IFN-γ and altered the Th2/Th1 balance, leading to great infiltration of neutrophils and macrophages, which resulted in larger liver inflammation and increased serum transaminase activity in comparison with mono-infected mice. Mice infected with S. venezuelensis at two and four weeks after S. mansoni infection showed significant increase of Th1/Th2/Th17/Treg cytokines and strong cellular infiltration in the liver in comparison with mono-infected mice. However, only in mice co-infected after two weeks of schistosomiasis, the liver immune response leads to more intense Th2 polarization, increased liver inflammation, and transaminase serum activity. S. venezuelensis co-infection during chronic schistosomiasis did not significantly alter liver inflammation. Therefore, S. venezuelensis co-infection affects the host immune responses and morbidity of schistosomiasis, but the effects largely depend on the stage of the S. mansoni infection.


Subject(s)
Coinfection/immunology , Cytokines/immunology , Inflammation/immunology , Liver/immunology , Schistosomiasis mansoni/immunology , Strongyloidiasis/immunology , Animals , Coinfection/metabolism , Coinfection/parasitology , Cytokines/blood , Cytokines/metabolism , Female , Host-Parasite Interactions/immunology , Inflammation/metabolism , Liver/metabolism , Liver/pathology , Mice , Schistosoma mansoni/immunology , Schistosoma mansoni/physiology , Schistosomiasis mansoni/metabolism , Schistosomiasis mansoni/parasitology , Strongyloides/immunology , Strongyloides/physiology , Strongyloidiasis/metabolism , Strongyloidiasis/parasitology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL