Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Nat Commun ; 15(1): 5226, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38890291

ABSTRACT

IL-33 plays a significant role in inflammation, allergy, and host defence against parasitic helminths. The model gastrointestinal nematode Heligmosomoides polygyrus bakeri secretes the Alarmin Release Inhibitor HpARI2, an effector protein that suppresses protective immune responses and asthma in its host by inhibiting IL-33 signalling. Here we reveal the structure of HpARI2 bound to mouse IL-33. HpARI2 contains three CCP-like domains, and we show that it contacts IL-33 primarily through the second and third of these. A large loop which emerges from CCP3 directly contacts IL-33 and structural comparison shows that this overlaps with the binding site on IL-33 for its receptor, ST2, preventing formation of a signalling complex. Truncations of HpARI2 which lack the large loop from CCP3 are not able to block IL-33-mediated signalling in a cell-based assay and in an in vivo female mouse model of asthma. This shows that direct competition between HpARI2 and ST2 is responsible for suppression of IL-33-dependent responses.


Subject(s)
Asthma , Helminth Proteins , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Nematospiroides dubius , Animals , Interleukin-33/metabolism , Interleukin-33/chemistry , Nematospiroides dubius/immunology , Helminth Proteins/metabolism , Helminth Proteins/chemistry , Helminth Proteins/immunology , Mice , Female , Interleukin-1 Receptor-Like 1 Protein/metabolism , Asthma/immunology , Asthma/metabolism , Humans , Signal Transduction , Strongylida Infections/immunology , Strongylida Infections/parasitology , Strongylida Infections/metabolism , Protein Binding , Disease Models, Animal , Binding Sites , Mice, Inbred BALB C , Mice, Inbred C57BL
2.
Immunol Cell Biol ; 100(5): 301-303, 2022 05.
Article in English | MEDLINE | ID: mdl-35302670

ABSTRACT

A new study by Varyani et al. identifies that macrophage migration inhibitory factor (MIF) is required to mount a strong type 2 immune response in the gut. Such immune response is required to properly expel the helminth Nippostrongylus brasiliensis, for example by activating goblet cells to secrete RELM-ß.


Subject(s)
Helminths , Strongylida Infections , Animals , Goblet Cells/metabolism , Immunity, Innate , Lymphocytes , Nippostrongylus/metabolism , Strongylida Infections/metabolism , Strongylida Infections/parasitology
3.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34779829

ABSTRACT

Helminth parasites are adept manipulators of the immune system, using multiple strategies to evade the host type 2 response. In the intestinal niche, the epithelium is crucial for initiating type 2 immunity via tuft cells, which together with goblet cells expand dramatically in response to the type 2 cytokines IL-4 and IL-13. However, it is not known whether helminths modulate these epithelial cell populations. In vitro, using small intestinal organoids, we found that excretory/secretory products (HpES) from Heligmosomoides polygyrus blocked the effects of IL-4/13, inhibiting tuft and goblet cell gene expression and expansion, and inducing spheroid growth characteristic of fetal epithelium and homeostatic repair. Similar outcomes were seen in organoids exposed to parasite larvae. In vivo, H. polygyrus infection inhibited tuft cell responses to heterologous Nippostrongylus brasiliensis infection or succinate, and HpES also reduced succinate-stimulated tuft cell expansion. Our results demonstrate that helminth parasites reshape their intestinal environment in a novel strategy for undermining the host protective response.


Subject(s)
Epithelial Cells/metabolism , Goblet Cells/metabolism , Intestine, Small/cytology , Organoids/metabolism , Strongylida Infections/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Epithelial Cells/parasitology , Female , Gene Expression Regulation/drug effects , Goblet Cells/parasitology , Helminth Proteins/metabolism , Helminth Proteins/pharmacology , Host-Parasite Interactions , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Intestine, Small/parasitology , Mice, Inbred C57BL , Nematospiroides dubius/metabolism , Nematospiroides dubius/physiology , Nippostrongylus/metabolism , Nippostrongylus/physiology , Organoids/cytology , Organoids/parasitology , Strongylida Infections/parasitology , Succinic Acid/pharmacology , Transcriptome/drug effects
4.
Science ; 374(6568): eabe6723, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34735226

ABSTRACT

A diverse group of antimicrobial proteins (AMPs) helps protect the mammalian intestine from varied microbial challenges. We show that small proline-rich protein 2A (SPRR2A) is an intestinal antibacterial protein that is phylogenetically unrelated to previously discovered mammalian AMPs. In this study, SPRR2A was expressed in Paneth cells and goblet cells and selectively killed Gram-positive bacteria by disrupting their membranes. SPRR2A shaped intestinal microbiota composition, restricted bacterial association with the intestinal surface, and protected against Listeria monocytogenes infection. SPRR2A differed from other intestinal AMPs in that it was induced by type 2 cytokines produced during helminth infection. Moreover, SPRR2A protected against helminth-induced bacterial invasion of intestinal tissue. Thus, SPRR2A is a distinctive AMP triggered by type 2 immunity that protects the intestinal barrier during helminth infection.


Subject(s)
Cornified Envelope Proline-Rich Proteins/metabolism , Gastrointestinal Microbiome , Gram-Positive Bacteria/physiology , Intestinal Mucosa/metabolism , Intestines/microbiology , Nematospiroides dubius , Strongylida Infections/immunology , Animals , Bacterial Load , Cell Membrane/metabolism , Cell Membrane Permeability , Cornified Envelope Proline-Rich Proteins/genetics , Cytokines/metabolism , Disease Susceptibility , Goblet Cells/metabolism , Humans , Immunity, Innate , Intestinal Mucosa/microbiology , Listeria monocytogenes/physiology , Listeriosis/microbiology , Mice , Microbial Viability , Paneth Cells/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Strongylida Infections/metabolism , Strongylida Infections/microbiology
5.
Infect Immun ; 89(12): e0022521, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34460289

ABSTRACT

Heligmosomoides polygyrus is a helminth which naturally infects mice and is widely used as a laboratory model of chronic small intestinal helminth infection. While it is known that infection with H. polygyrus alters the composition of the host's bacterial microbiota, the functional implications of this alteration are unclear. We investigated the impact of H. polygyrus infection on short-chain fatty acid (SCFA) levels in the mouse intestine and sera. We found that helminth infection resulted in significantly upregulated levels of the branched SCFA isovaleric acid, exclusively in the proximal small intestine, which is the site of H. polygyrus colonization. We next set out to test the hypothesis that elevating local levels of isovaleric acid was a strategy used by H. polygyrus to promote its own fitness within the mammalian host. To test this, we supplemented the drinking water of mice with isovalerate during H. polygyrus infection and examined whether this affected helminth fecundity or chronicity. We did not find that isovaleric acid supplementation affected helminth chronicity; however, we found that it did promote helminth fecundity, as measured by helminth egg output in the feces of mice. Through antibiotic treatment of helminth-infected mice, we found that the bacterial microbiota was required in order to support elevated levels of isovaleric acid in the proximal small intestine during helminth infection. Overall, our data reveal that during H. polygyrus infection there is a microbiota-dependent localized increase in the production of isovaleric acid in the proximal small intestine and that this supports helminth fecundity in the murine host.


Subject(s)
Fatty Acids, Volatile/metabolism , Host-Parasite Interactions , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Nematospiroides dubius/physiology , Strongylida Infections/metabolism , Strongylida Infections/parasitology , Animals , Disease Models, Animal , Lipid Metabolism , Mice
6.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34290141

ABSTRACT

"Taste-like" tuft cells in the intestine trigger type 2 immunity in response to worm infection. The secretion of interleukin-13 (IL-13) from type 2 innate lymphoid cells (ILC2) represents a key step in the tuft cell-ILC2 cell-intestinal epithelial cell circuit that drives the clearance of worms from the gut via type 2 immune responses. Hallmark features of type 2 responses include tissue remodeling, such as tuft and goblet cell expansion, and villus atrophy, yet it remains unclear if additional molecular changes in the gut epithelium facilitate the clearance of worms from the gut. Using gut organoids, we demonstrated that IL-4 and IL-13, two type 2 cytokines with similar functions, not only induced the classical type 2 responses (e.g., tuft cell expansion) but also drastically up-regulated the expression of gasdermin C genes (Gsdmcs). Using an in vivo worm-induced type 2 immunity model, we confirmed the up-regulation of Gsdmcs in Nippostrongylus brasiliensis-infected wild-type C57BL/6 mice. Consistent with gasdermin family members being principal effectors of pyroptosis, overexpression of Gsdmc2 in human embryonic kidney 293 (HEK293) cells triggered pyroptosis and lytic cell death. Moreover, in intestinal organoids treated with IL-4 or IL-13, or in wild-type mice infected with N. brasiliensis, lytic cell death increased, which may account for villus atrophy observed in worm-infected mice. Thus, we propose that the up-regulated Gsdmc family may be major effectors for type 2 responses in the gut and that Gsdmc-mediated pyroptosis may provide a conduit for the release of antiparasitic factors from enterocytes to facilitate the clearance of worms.


Subject(s)
Cell Death , DNA-Binding Proteins/metabolism , Enterocytes/pathology , Immunity, Innate/immunology , Intestine, Small/pathology , Strongylida Infections/complications , Th2 Cells/immunology , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , Enterocytes/immunology , Enterocytes/metabolism , Enterocytes/parasitology , Female , Interleukin-13/metabolism , Interleukin-4/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/parasitology , Male , Mice , Mice, Inbred C57BL , Nippostrongylus/physiology , Signal Transduction , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology
7.
Life Sci Alliance ; 4(8)2021 08.
Article in English | MEDLINE | ID: mdl-34127548

ABSTRACT

IL-13 is implicated in effective repair after acute lung injury and the pathogenesis of chronic diseases such as allergic asthma. Both these processes involve matrix remodelling, but understanding the specific contribution of IL-13 has been challenging because IL-13 shares receptors and signalling pathways with IL-4. Here, we used Nippostrongylus brasiliensis infection as a model of acute lung damage comparing responses between WT and IL-13-deficient mice, in which IL-4 signalling is intact. We found that IL-13 played a critical role in limiting tissue injury and haemorrhaging in the lung, and through proteomic and transcriptomic profiling, identified IL-13-dependent changes in matrix and associated regulators. We further showed a requirement for IL-13 in the induction of epithelial-derived type 2 effector molecules such as RELM-α and surfactant protein D. Pathway analyses predicted that IL-13 induced cellular stress responses and regulated lung epithelial cell differentiation by suppression of Foxa2 pathways. Thus, in the context of acute lung damage, IL-13 has tissue-protective functions and regulates epithelial cell responses during type 2 immunity.


Subject(s)
Acute Lung Injury/parasitology , Interleukin-13/deficiency , Nippostrongylus/pathogenicity , Strongylida Infections/genetics , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Female , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Proteomics , Strongylida Infections/metabolism , Up-Regulation
8.
Front Immunol ; 12: 627638, 2021.
Article in English | MEDLINE | ID: mdl-33936040

ABSTRACT

Background: Tuberculosis (TB) is still a major challenge for humankind. Because regions with the highest incidence also have a high prevalence of helminthiasis and nutritional scarcity, we wanted to understand the impact of these on TB progression. Methods: We have developed an experimental murine model for active TB in C3HeB/FeJ, coinfected with Trichuris muris and Heligmosomoides polygyrus nematodes, and exposed to an environmental mycobacterium (M. manresensis) and intermittent fasting. Cause-effect relationships among these factors were explored with Partial Least Squares Path modelling (PLSPM). Results: Previous parasitization had a major anti-inflammatory effect and reduced systemic levels of ADA, haptoglobin, local pulmonary levels of IL-1ß, IL-6, TNF-α, CXCL-1, CXCL-5 and IL-10. Oral administration of heat-killed M. manresensis resulted in a similar outcome. Both interventions diminished pulmonary pathology and bacillary load, but intermittent food deprivation reduced this protective effect increasing stress and inflammation. The PLSPM revealed nematodes might have protective effects against TB progression. Conclusions: Significantly higher cortisol levels in food-deprivation groups showed it is a stressful condition, which might explain its deleterious effect. This highlights the impact of food security on TB eradication policies and the need to prioritize food supply over deworming activities.


Subject(s)
Coinfection , Food Deprivation , Helminthiasis/parasitology , Intestinal Diseases, Parasitic/parasitology , Lung/microbiology , Mycobacterium tuberculosis/pathogenicity , Nematospiroides dubius/pathogenicity , Strongylida Infections/parasitology , Trichuriasis/parasitology , Trichuris/pathogenicity , Tuberculosis, Pulmonary/microbiology , Animal Nutritional Physiological Phenomena , Animals , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Helminthiasis/immunology , Helminthiasis/metabolism , Host-Parasite Interactions , Inflammation Mediators/metabolism , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/metabolism , Lung/immunology , Lung/metabolism , Male , Mice, Inbred C3H , Mycobacterium tuberculosis/immunology , Nematospiroides dubius/immunology , Nutritional Status , Strongylida Infections/immunology , Strongylida Infections/metabolism , Trichuriasis/immunology , Trichuriasis/metabolism , Trichuris/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism
9.
Mol Neurobiol ; 58(8): 3968-3982, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33904019

ABSTRACT

Angiostrongylus cantonensis infection is a typical cause of eosinophilic encephalitis (EM), which has been reported to induce serious damage in the central nervous system. Both parasite and host factors contribute to the onset of EM, but the related immune-inflammation pathogenesis remains poorly characterised. An A. cantonensis infection model was generated through the infection of mice by gavage. Transmission electron microscopy and immunohistochemistry were used to assess the pathologic changes in the brain. The mRNA expression of inflammatory factors was tested using qRT-PCR. A combination of flow cytometry and western blotting was used to evaluate the alteration of leukocytes and related cytokines. A critical role of IL-17 was found by injecting IL-17A monoclonal antibody into naïve and A. cantonensis-infected mice. A. cantonensis larvae altered the immune homeostasis in the brain, leading to the destruction of myelin sheaths and activation of microglia and macrophage. During this process, IL-17A accumulation was observed, and IL-17RA was expressed in oligodendrocytes and microglia during the infection. Notably, γδ T cell was the major origin of IL-17A production induced by the parasite. After an IL-17A-neutralising antibody was applied, alterations in myelination and the state of the microglia/macrophage were discovered; the neurobehavioural scores of the mice also improved. Our study reveals one unrecognised impact of the γδ T cells in parasitic encephalopathy and emphasises that blocking IL-17A signalling can attenuate microglia and macrophage activation, thus reducing CNS demyelination and ameliorating the neurobehavioural deficit in A. cantonensis-infected mice.


Subject(s)
Brain/metabolism , Demyelinating Diseases/metabolism , Interleukin-17/biosynthesis , Intraepithelial Lymphocytes/metabolism , Strongylida Infections/metabolism , Animals , Brain/immunology , Demyelinating Diseases/immunology , Intraepithelial Lymphocytes/immunology , Male , Mice , Mice, Inbred BALB C , Microglia/immunology , Microglia/metabolism , Strongylida Infections/immunology
10.
J Parasitol ; 107(2): 205-213, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33684197

ABSTRACT

Peroxisome-proliferator activator receptor γ (PPARγ) has an anti-inflammatory role that inhibits the nuclear factor-κB (NF-κB) pathway and regulates the expressions of pro-inflammatory proteins, whereas its role in parasitic meningoencephalitis remains unknown. In this study we investigated the role of PPARγ and related mechanisms in eosinophilic meningoencephalitis caused by the rat lungworm Angiostrongylus cantonensis. We observed increased protein NF-κB expression in mouse brain tissue using GW9662, which is the specific antagonist of PPARγ, in a mouse model of angiostrongyliasis. Then we investigated NF-κB-related downstream proteins, such as COX-2, NOSs, and IL-1ß, with Western blot or enzyme-linked immunosorbent assay and found that the protein expression was upregulated. The results of gelatin zymography also showed that the MMP-9 activities were upregulated. Treatment with GW9662 increased the permeability of the blood-brain barrier and the number of eosinophils in cerebrospinal fluid. These results suggested that in angiostrongyliasis, PPARγ may play an anti-inflammation role in many inflammatory mediators, including NOS-related oxidative stress, cytokines, and matrix metalloproteinase cascade by decreasing the NF-κB action.


Subject(s)
Angiostrongylus cantonensis/pathogenicity , Meningoencephalitis/metabolism , Meningoencephalitis/parasitology , PPAR gamma/metabolism , Strongylida Infections/metabolism , Animals , Blood-Brain Barrier/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Male , Matrix Metalloproteinase 2/cerebrospinal fluid , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/cerebrospinal fluid , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Random Allocation , Strongylida Infections/parasitology , Taiwan
11.
Parasitology ; 148(2): 187-197, 2021 02.
Article in English | MEDLINE | ID: mdl-33004090

ABSTRACT

Steroids have been shown to be beneficial in patients and mice with eosinophilic meningitis caused by Angiostrongylus cantonensis infection; however, the mechanism for this beneficial effect is unknown. We speculated that the effect of steroids in eosinophilic meningitis caused by A. cantonensis infection may be mediated by the downregulation of matrix metallopeptidase-9 (MMP-9) and oxidative stress pathways via glucocorticoid receptors (GRs). We found blood-brain barrier (BBB) dysfunction in mice with eosinophilic meningitis 2-3 weeks after infection as evidenced by increased extravasation of Evans blue and cerebrospinal fluid (CSF) albumin levels. The administration of dexamethasone significantly decreased the amount of Evans blue and CSF albumin. The effect of dexamethasone was mediated by GRs and heat shock protein 70, resulting in subsequent decreases in the expressions of nuclear factor kappa B (NF-κB), c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) in the CSF and brain parenchymal after 2 weeks of steroid administration. Steroid treatment also decreased CSF/brain homogenate MMP-9 concentrations, but had no effect on CSF MMP-2 levels, indicating that MMP-9 rather than MMP-2 played a major role in BBB dysfunction in mice with eosinophilic meningitis. The concentration of 8-hydroxy-2'-deoxyguanosine (8-OHdG) gradually increased after 1-3 weeks of infection, and the administration of dexamethasone significantly downregulated the concentration of oxidized derivative 8-OHdG in CSF. In conclusion, increased 8-OHdG and MMP-9 concentrations were found in mice with eosinophilic meningitis caused by A. cantonensis infection. The effect of dexamethasone was mediated by GRs and significantly decreased not only the levels of 8-OHdG and MMP-9 but also NF-κB, JNK and ERK.


Subject(s)
Dexamethasone/administration & dosage , Down-Regulation/drug effects , Glucocorticoids/administration & dosage , Matrix Metalloproteinase 9/genetics , Oxidative Stress/drug effects , Rodent Diseases/parasitology , Strongylida Infections/parasitology , Angiostrongylus cantonensis/physiology , Animals , Eosinophilia/metabolism , Eosinophilia/parasitology , Gene Expression Regulation/drug effects , Injections, Intraperitoneal , Matrix Metalloproteinase 9/metabolism , Meningitis/metabolism , Meningitis/parasitology , Mice , Mice, Inbred BALB C , Rodent Diseases/metabolism , Strongylida Infections/metabolism
12.
Acta Trop ; 212: 105684, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32931750

ABSTRACT

Angiostrongylus cantonensis is the main cause of human eosinophilic meningitis. Humans are accidental hosts, becoming infected due to ingestion of raw intermediate (snails and slugs) or paratenic hosts. Once ingested, the larvae migrate towards the brain where they die, causing the disease. To develop better mollusk control strategies, it is important to first understand what happens in the snail during infection, therefore our purpose was to characterize proteomic, metabolic and immunologic changes in Biomphalaria glabrata 24 h after infection with A. cantonensis. For this purpose, proteins were extracted from infected and uninfected snails and analyzed through mass spectrometry. Hemolymph was also collected, the number of hemocytes was counted and urea, nitric oxide, calcium, glycogen levels as well as alanine and aspartate aminotransferases activities were assessed. The cephalopodal region and gonad-digestive gland complex were dissected and their glycogen content was measured. After infection with A. cantonensis, we observed an increase of hemocytes and granulocytes as well as an increase in hemoglobin type 2 proteins. Temptin-like protein was also found up-regulated in infected snails. Several proteins with structural function (such as myosin heavy chain - striated muscle - like and protein LOC106059779 with ADAM/reprosolin domain) were also differentially expressed, suggesting loss/damage of internal tissues. Increase in phosphoglycerate mutase indicates an increase in glycolysis, possible to compensate the increase in energetic needs. Consequently, there is a decrease in glycogen reserves, particularly in the gonad - digestive gland complex.


Subject(s)
Biomphalaria/parasitology , Proteomics/methods , Strongylida Infections/metabolism , Animals , Hemolymph/chemistry , Host-Parasite Interactions , Humans , Rats , Rats, Wistar
13.
Parasitol Res ; 119(10): 3433-3441, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32789733

ABSTRACT

Angiostrongylus cantonensis causes a human central nervous system (CNS) infection characterized by eosinophilic meningitis or meningoencephalitis. Individuals infected with A. cantonensis exhibit unbalanced walking. The mechanism of extensive neurological impairments of hosts caused by A. cantonensis larvae remains unclear. Tight junction proteins (e.g., claudin-5 and zonula occludens-1) are the most important regulators of paracellular permeability and cellular adhesion. In a previous study, we found that increased matrix metalloproteinase-9 (MMP-9) activity may be associated with blood-CNS barrier disruption and/or the degeneration of Purkinje cells in eosinophilic meningitis caused by A. cantonensis. In the present study, the co-localization of MMP-9 and tight junction proteins on the degeneration of Purkinje cells was measured via confocal laser scanning immunofluorescence microscopy. The statistical evidence indicated that MMP-9 correlated between tight junction protein disruption and Purkinje cell degeneration at 20 days post-infection with A. cantonensis. In conclusion, Purkinje cell degeneration is highly correlated with tight junction protein disruption via the MMP-9 activation pathway.


Subject(s)
Angiostrongylus cantonensis/physiology , Matrix Metalloproteinase 9/metabolism , Purkinje Cells/pathology , Strongylida Infections/parasitology , Tight Junction Proteins/metabolism , Animals , Disease Models, Animal , Larva/physiology , Mice , Purkinje Cells/metabolism , Purkinje Cells/parasitology , Strongylida Infections/metabolism , Strongylida Infections/pathology
14.
Am J Trop Med Hyg ; 103(2): 723-726, 2020 08.
Article in English | MEDLINE | ID: mdl-32431278

ABSTRACT

Meningoencephalitis is not a rare disease in small children. However, eosinophilic meningitis due to Angiostrongylus cantonensis is unusual in a baby. We describe the case of a 9-month-old baby from North Vietnam with eosinophilic meningoencephalitis. The baby lived in a rural area, where farming is widespread, and presented with fever and seizures. Laboratory results showed peripheral eosinophilia (16.1%), cerebrospinal fluid (CSF) white blood cell count 220/mm3 (26% eosinophils), CSF antibody test positive for Ascaris, CSF ELISA positive for Angiostrongylus cantonensis, and blood ELISA positive for A. cantonensis. A mobile worm was identified in the CSF. The presentation was consistent with a diagnosis of A. cantonensis eosinophilic meningitis. The baby recovered fully after administering albendazole (200 mg/day for 2 weeks), and intravenous dexamethasone (0.6 mg/kg/day every 8 hours) and mannitol (1.5 g/kg/day every 8 hours) for the first 3 days, followed by 5 days of oral prednisolone (2 mg/kg/day).


Subject(s)
Angiostrongylus cantonensis/isolation & purification , Eosinophilia/blood , Meningoencephalitis/physiopathology , Strongylida Infections/physiopathology , Albendazole/therapeutic use , Animals , Anthelmintics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antibodies, Helminth/blood , Antibodies, Helminth/cerebrospinal fluid , Dexamethasone/therapeutic use , Diuretics, Osmotic/therapeutic use , Eosinophilia/etiology , Humans , Infant , Intracranial Hypertension/drug therapy , Intracranial Hypertension/etiology , Magnetic Resonance Imaging , Male , Mannitol/therapeutic use , Meningoencephalitis/complications , Meningoencephalitis/drug therapy , Meningoencephalitis/metabolism , Prednisolone/therapeutic use , Seizures/etiology , Seizures/physiopathology , Strongylida Infections/complications , Strongylida Infections/drug therapy , Strongylida Infections/metabolism , Tomography, X-Ray Computed , Vietnam
16.
Int J Biol Sci ; 15(10): 2211-2223, 2019.
Article in English | MEDLINE | ID: mdl-31592236

ABSTRACT

BACKGROUND: Angiostrongylus cantonensis infection can cause demyelination in the central nervous system, and there is no effective treatment. METHODS: We used dexamethasone, Tanshinone IIA (TSIIA) and Cryptotanshinone(Two traditional Chinese medicine monomers) in combination with albendazole (AB, a standard anti-helminthic compound) to observe their therapeutic effect on demyelination in A. cantonensis-infected mice. Luxol fast blue staining and electron microscope of myelin sheath, Oligodendrocyte (OL) number and myelin basic protein (MBP) expression in brain was detected in above groups. RESULTS: TSIIA+AB facilitated OL proliferation and significantly increased both myelin sheath thickness and the population of small-diameter axons. In addition, TSIIA treatment inhibited the expression of inflammation-related factors (interleukin [IL]-6, IL-1ß, tumor necrosis factor [TNF]-α, inducible nitric oxide synthase [iNOS]) rather than inhibiting eosinophil infiltration in brain. TSIIA also decreased microglial activation and shifted their phenotype from M1 to M2. CONCLUSIONS: Taken together, these results provide evidence that TSIIA combined with AB may be an effective treatment for demyelination caused by A. cantonensis infection and other demyelinating diseases.


Subject(s)
Abietanes/therapeutic use , Angiostrongylus cantonensis/drug effects , Angiostrongylus cantonensis/pathogenicity , Strongylida Infections/drug therapy , Albendazole/pharmacology , Animals , Blotting, Western , Demyelinating Diseases/drug therapy , Fluorescent Antibody Technique , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Myelin Sheath/drug effects , Nitric Oxide Synthase Type II/metabolism , Remyelination/drug effects , Strongylida Infections/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
PLoS One ; 14(8): e0220503, 2019.
Article in English | MEDLINE | ID: mdl-31415587

ABSTRACT

Proteasome primarily degrades the unneeded or damaged proteins by proteolysis. Disruption of the brain barrier and its resulting meningoencephalitis caused by Angiostrongylus cantonensis are important pathological events in non-permissive hosts. In this study, the results showed upregulated proteasome during A. cantonensis infection. Occludin degradation and matrix metalloproteinase-9 (MMP-9) activity were significantly increased in infected mice than in uninfected mice. Moreover, confocal immunoflourescence microscopy showed that occludin was co-localized with MMP-9. The infected-mice were treated with proteasomal activity inhibitor MG132 by 1.5 and 3.0 mg/kg/day, which resulted in significantly reduced protein levels of phosphorylated IκBα (P<0.05) compared with the untreated control. The phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) showed similar result. In addition, MMP-9 activity and occludin degradation were reduced because of MG132 treatment. These results suggested that the proteasome in A. cantonensis infection degraded phosphorylated IκBα, modulated phosphorylated NF-κB, and then regulated the activation of MMP-9 and occludin degradation. Proteasome alterations were presented in eosinophilic meningitis of BALB/c mice and may contribute to the pathophysiology of eosinophilic meningitis by increasing occludin degradation. This molecule would serve as pivotal regulator in A. cantonensis-induced eosinophilic meningoencephalitis.


Subject(s)
Angiostrongylus cantonensis , Brain/metabolism , Meningoencephalitis/parasitology , Proteasome Endopeptidase Complex/metabolism , Strongylida Infections/metabolism , Animals , Brain/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Disease Models, Animal , Leupeptins/pharmacology , Male , Matrix Metalloproteinase 9/metabolism , Meningoencephalitis/metabolism , Mice , NF-kappa B/metabolism , Occludin/metabolism , Phosphorylation , Up-Regulation
18.
J Neuroinflammation ; 16(1): 159, 2019 Jul 29.
Article in English | MEDLINE | ID: mdl-31352901

ABSTRACT

BACKGROUND: It has become increasingly evident that the immune and nervous systems are closely intertwined, relying on one another during regular homeostatic conditions. Prolonged states of imbalance between neural and immune homeostasis, such as chronic neuroinflammation, are associated with a higher risk for neural damage. Toxoplasma gondii is a highly successful neurotropic parasite causing persistent subclinical neuroinflammation, which is associated with psychiatric and neurodegenerative disorders. Little is known, however, by what means neuroinflammation and the associated neural impairment can be modulated by peripheral inflammatory processes. METHODS: Expression of immune and synapse-associated genes was assessed via quantitative real-time PCR to investigate how T. gondii infection-induced chronic neuroinflammation and associated neuronal alterations can be reshaped by a subsequent acute intestinal nematode co-infection. Immune cell subsets were characterized via flow cytometry in the brain of infected mice. Sulfadiazine and interferon-γ-neutralizing antibody were applied to subdue neuroinflammation. RESULTS: Neuroinflammation induced by T. gondii infection of mice was associated with increased microglia activation, recruitment of immune cells into the brain exhibiting Th1 effector functions, and enhanced production of Th1 and pro-inflammatory molecules (IFN-γ, iNOS, IL-12, TNF, IL-6, and IL-1ß) following co-infection with Heligmosomoides polygyrus. The accelerated cerebral Th1 immune response resulted in enhanced T. gondii removal but exacerbated the inflammation-related decrease of synapse-associated gene expression. Synaptic proteins EAAT2 and GABAAα1, which are involved in the excitation/inhibition balance in the CNS, were affected in particular. These synaptic alterations were partially recovered by reducing neuroinflammation indirectly via antiparasitic treatment and especially by application of IFN-γ-neutralizing antibody. Impaired iNOS expression following IFN-γ neutralization directly affected EAAT2 and GABAAα1 signaling, thus contributing to the microglial regulation of neurons. Besides, reduced CD36, TREM2, and C1qa gene expression points toward inflammation induced synaptic pruning as a fundamental mechanism. CONCLUSION: Our results suggest that neuroimmune responses following chronic T. gondii infection can be modulated by acute enteric nematode co-infection. While consecutive co-infection promotes parasite elimination in the CNS, it also adversely affects gene expression of synaptic proteins, via an IFN-γ-dependent manner.


Subject(s)
Brain/metabolism , Interferon-gamma/metabolism , Microglia/metabolism , Neurons/metabolism , Strongylida Infections/metabolism , Toxoplasmosis/metabolism , Animals , Brain/parasitology , Coinfection , Macrophage Activation/physiology , Mice , Microglia/parasitology , Nematospiroides dubius , Neurons/parasitology , Synapses/metabolism , Synapses/parasitology , Toxoplasma
19.
PLoS One ; 14(3): e0213244, 2019.
Article in English | MEDLINE | ID: mdl-30845271

ABSTRACT

The 14-3-3 proteins are cerebrospinal fluid (CSF) markers of neuronal damage during infectious meningitis and Creutzfeldt-Jakob disease. Little is known about dynamic changes in the individual isoforms in response to parasitic eosinophilic meningitis. The purposes of this study were to determine the 14-3-3 protein isoform patterns, examine the kinetics and correlate the severity of blood brain barrier (BBB) damage with the expressions of these markers in mice with eosinophilic meningitis. Mice were orally infected with 50 A. cantonensis L3 via an oro-gastric tube and sacrificed every week for 3 consecutive weeks after infection. The Evans blue method and BBB junctional protein expressions were used to measure changes in the BBB. Hematoxylin and eosin staining was used to analyze pathological changes in the mice brains following 1-3 weeks of infection with A. cantonensis. The levels of 14-3-3 protein isoforms in serum/CSF and brain homogenates were analyzed by Western blot, and immunohistochemistry (IHC) was used to explore the different isoform distributions of 14-3-3 proteins and changes in BBB junctional proteins in the mice brain meninges. Dexamethasone was injected intraperitoneally from the seventh day post infection (dpi) until the end of the study (21 dpi) to study the changes in BBB junctional proteins. The amounts of Evans blue, tight junction and 14-3-3 protein isoforms in the different groups of mice were compared using the nonparametric Kruskal-Wallis test. There were significant increases in 14-3-3 protein isoforms ß and γ in the CSF in the second and third weeks after infection compared to the controls and first week of infection, which were correlated with the severity of BBB damage in brain histology, and Evans blue extravasation. Using IHC to assess the distribution of 14-3-3 protein isoforms and changes in BBB junctional proteins in the mice brain meninges, the expressions of isoforms ß, γ, ε, and θ and junctional proteins occludin and claudin-5 in the brain meninges increased over a 3-week period after infection compared to the controls and 1 week after infection. The administration of dexamethasone decreased the expressions of BBB junctional proteins occludin and claudin-5 in the mice brain meninges. Our findings support that 14-3-3 proteins ß and γ can potentially be used as a CSF marker of neuronal damage in parasitic eosinophilic meningitis caused by A. cantonensis.


Subject(s)
14-3-3 Proteins/metabolism , Angiostrongylus cantonensis/pathogenicity , Blood-Brain Barrier/metabolism , Brain/metabolism , Eosinophilia/metabolism , Meningitis/metabolism , Strongylida Infections/metabolism , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Blood-Brain Barrier/parasitology , Brain/parasitology , Disease Models, Animal , Eosinophilia/epidemiology , Eosinophilia/parasitology , Female , Incidence , Male , Meningitis/epidemiology , Meningitis/parasitology , Mice , Mice, Inbred BALB C , Protein Isoforms , Strongylida Infections/complications , Strongylida Infections/parasitology
20.
PLoS Pathog ; 14(11): e1007423, 2018 11.
Article in English | MEDLINE | ID: mdl-30500858

ABSTRACT

Ym1 and RELMα are established effector molecules closely synonymous with Th2-type inflammation and associated pathology. Here, we show that whilst largely dependent on IL-4Rα signaling during a type 2 response, Ym1 and RELMα also have IL-4Rα-independent expression patterns in the lung. Notably, we found that Ym1 has opposing effects on type 2 immunity during nematode infection depending on whether it is expressed at the time of innate or adaptive responses. During the lung migratory stage of Nippostrongylus brasiliensis, Ym1 promoted the subsequent reparative type 2 response but once that response was established, IL-4Rα-dependent Ym1 was important for limiting the magnitude of type 2 cytokine production from both CD4+ T cells and innate lymphoid cells in the lung. Importantly, our study demonstrates that delivery of Ym1 to IL-4Rα deficient animals drives RELMα production and overcomes lung repair deficits in mice deficient in type 2 immunity. Together, Ym1 and RELMα, exhibit time and dose-dependent interactions that determines the outcome of lung repair during nematode infection.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Lectins/metabolism , Nematode Infections/metabolism , Receptors, Cell Surface/deficiency , beta-N-Acetylhexosaminidases/metabolism , Animals , Lung/immunology , Lung/metabolism , Lung/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nematode Infections/immunology , Nippostrongylus/immunology , Receptors, Cell Surface/metabolism , Signal Transduction , Strongylida Infections/immunology , Strongylida Infections/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...