Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Parasit Vectors ; 17(1): 205, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715092

ABSTRACT

BACKGROUND: Angiostrongyliasis is a highly dangerous infectious disease. Angiostrongylus cantonensis larvae migrate to the mouse brain and cause symptoms, such as brain swelling and bleeding. Noncoding RNAs (ncRNAs) are novel targets for the control of parasitic infections. However, the role of these molecules in A. cantonensis infection has not been fully clarified. METHODS: In total, 32 BALB/c mice were randomly divided into four groups, and the infection groups were inoculated with 40 A. cantonensis larvae by gavage. Hematoxylin and eosin (H&E) staining and RNA library construction were performed on brain tissues from infected mice. Differential expression of long noncoding RNAs (lncRNAs) and mRNAs in brain tissues was identified by high-throughput sequencing. The pathways and functions of the differentially expressed lncRNAs were determined by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. The functions of the differentially expressed lncRNAs were further characterized by lncRNA‒microRNA (miRNA) target interactions. The potential host lncRNAs involved in larval infection of the brain were validated by quantitative real-time polymerase chain reaction (qRT‒PCR). RESULTS: The pathological results showed that the degree of brain tissue damage increased with the duration of infection. The transcriptome results showed that 859 lncRNAs and 1895 mRNAs were differentially expressed compared with those in the control group, and several lncRNAs were highly expressed in the middle-late stages of mouse infection. GO and KEGG pathway analyses revealed that the differentially expressed target genes were enriched mainly in immune system processes and inflammatory response, among others, and several potential regulatory networks were constructed. CONCLUSIONS: This study revealed the expression profiles of lncRNAs in the brains of mice after infection with A. cantonensis. The lncRNAs H19, F630028O10Rik, Lockd, AI662270, AU020206, and Mexis were shown to play important roles in the infection of mice with A. cantonensis infection.


Subject(s)
Angiostrongylus cantonensis , Brain , Mice, Inbred BALB C , RNA, Long Noncoding , Strongylida Infections , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Angiostrongylus cantonensis/genetics , Strongylida Infections/parasitology , Strongylida Infections/genetics , Brain/parasitology , Brain/metabolism , Brain/pathology , Mice , Larva/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Expression Profiling , Female , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
PLoS Pathog ; 19(4): e1011296, 2023 04.
Article in English | MEDLINE | ID: mdl-37018382

ABSTRACT

Gastrointestinal helminths are a major health threat worldwide. Alternatively activated macrophages (AAMs) have been shown to contribute to host protection during secondary helminth infections. AAMs express effector molecules that depend on activation of the IL-4- or IL-13-induced transcription factor signal transducer and activator of transcription 6 (STAT6). However, the specific role of STAT6-regulated genes like Arginase-1 (Arg1) from AAMs or STAT6-regulated genes in other cell types for host protection remains unclear. To address this point, we generated mice expressing STAT6 only in macrophages (Mac-STAT6 mouse). In the model of Heligmosomoides polygyrus bakeri (Hpb) infection, Mac-STAT6 mice could not trap larvae in the submucosa of the small intestine after secondary infection. Further, mice lacking Arg1 in hematopoietic and endothelial cells were still protected from secondary Hpb infection. On the other hand, specific deletion of IL-4/IL-13 in T cells blunted AAM polarization, activation of intestinal epithelial cells (IECs) and protective immunity. Deletion of IL-4Rα on IEC also caused loss of larval trapping while AAM polarization remained intact. These results show that Th2-dependent and STAT6-regulated genes in IECs are required and AAMs are not sufficient for protection against secondary Hpb infection by mechanisms that remain to be investigated.


Subject(s)
Coinfection , Nematospiroides dubius , Strongylida Infections , Mice , Animals , Nematospiroides dubius/metabolism , Mice, Knockout , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukin-13/metabolism , Larva/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Strongylida Infections/genetics
3.
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
4.
Biomolecules ; 10(7)2020 07 05.
Article in English | MEDLINE | ID: mdl-32635653

ABSTRACT

Abstract: Currently, Angiostrongylus cantonensis infections are predominantly treated with albendazole. However, the use of albendazole can provoke certain neurological symptoms as a result of the immune response triggered by the dead worms. Therefore, treatment usually involves co-administration of corticosteroids to limit the inflammatory reaction. Corticosteroids play a useful role in suppressing inflammation in the brain; however, long-term usage or high dosage may make it problematic.Schisandrin B, an active ingredient from Schisandra chinensis, has been shown to have anti-inflammatory effects on the brain. This study aimed to investigate the effects and potential of schisandrin B in combination with albendazole to treat Angiostrongylus-induced meningoencephalitis. Here, we show that albendazole-schisandrin B co-treatment suppressed neuroinflammation in Angiostrongylus-infected mice and increased the survival of the mice. Accordingly, albendazole-schisandrin B co-treatment significantly inhibited inflammasome activation, pyroptosis, and apoptosis. The sensorimotor functions of the mice were also repaired after albendazole-schisandrin B treatment. Immune response was shown to shift from Th2 to Th1, which reduces inflammation and enhances immunity against A. cantonensis. Collectively, our study showed that albendazole-schisandrin B co-therapy may be used as an encouraging treatment for Angiostrongylus-induced meningoencephalitis.


Subject(s)
Albendazole/administration & dosage , Angiostrongylus cantonensis/parasitology , Lignans/administration & dosage , Meningoencephalitis/drug therapy , Polycyclic Compounds/administration & dosage , Strongylida Infections/drug therapy , Albendazole/pharmacology , Angiostrongylus cantonensis/drug effects , Animals , Apoptosis , Cyclooctanes/administration & dosage , Cyclooctanes/pharmacology , Disease Models, Animal , Drug Synergism , Gene Expression Regulation/drug effects , Inflammasomes/drug effects , Lignans/pharmacology , Meningoencephalitis/genetics , Meningoencephalitis/parasitology , Mice , Mice, Inbred BALB C , Polycyclic Compounds/pharmacology , Pyroptosis , Strongylida Infections/genetics , Survival Analysis , Th1 Cells/metabolism , Th2 Cells/metabolism
5.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32160524

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) regulate immunity, inflammation, and tissue homeostasis. Two distinct subsets of ILC2s have been described: steady-state natural ILC2s and inflammatory ILC2s, which are elicited following helminth infection. However, how tissue-specific cues regulate these two subsets of ILC2s and their effector functions remains elusive. Here, we report that interleukin-33 (IL-33) promotes the generation of inflammatory ILC2s (ILC2INFLAM) via induction of the enzyme tryptophan hydroxylase 1 (Tph1). Tph1 expression was upregulated in ILC2s upon activation with IL-33 or following helminth infection in an IL-33-dependent manner. Conditional deletion of Tph1 in lymphocytes resulted in selective impairment of ILC2INFLAM responses and increased susceptibility to helminth infection. Further, RNA sequencing analysis revealed altered gene expression in Tph1 deficient ILC2s including inducible T cell co-stimulator (Icos). Collectively, these data reveal a previously unrecognized function for IL-33, Tph1, and ICOS in promoting inflammatory ILC2 responses and type 2 immunity at mucosal barriers.


Subject(s)
Immunity, Cellular , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-33/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , Tryptophan Hydroxylase/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Disease Susceptibility , Gene Expression Regulation/immunology , Immunity, Innate , Immunity, Mucosal , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukin-33/genetics , Larva/growth & development , Larva/immunology , Larva/pathogenicity , Lymph Nodes/immunology , Lymph Nodes/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/growth & development , Nippostrongylus/pathogenicity , Primary Cell Culture , Signal Transduction , Strongylida Infections/genetics , Strongylida Infections/parasitology , Strongylida Infections/pathology , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/parasitology , Tryptophan Hydroxylase/genetics
6.
Parasite Immunol ; 42(5): e12704, 2020 05.
Article in English | MEDLINE | ID: mdl-32049381

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are heterogeneous population of monocyte and granulocyte progenitors that are highly suppressive against T cells. In BALB/c mice infected with a nematode Heligmosomoides polygyrus bakeri, we studied the dynamics of MDSCs, identified as CD11b+Gr-1+, induction in different tissues along with the development of parasite infection. We observed that MDSC-like cells are induced both by larvae and adult stages of H polygyrus bakeri. Gr-1+ cells of suppressive phenotype are recruited in the bone marrow, peripheral blood and peritoneal cavity during histotropic phase of infection and are present at that time in the intestine wall, where worms reside. Later, during intestinal phase, suppressive Gr-1+ cells increased in mesenteric lymph nodes and the spleen. l-arginine metabolism was important for the protective immunity, and parasite-induced Gr-1+ cells showed elevated arginase-1 and iNOS expression. Inhibition of arginase-1 and l-arginine administration caused reduced level of infection that coincided with weaker suppressive phenotype of Gr-1+ cells. We identified that l-arginine pathway activation and induction of MDSC-like cells characterize immunosuppressive state during H polygyrus bakeri infection in mice. Our findings confirm the role of MDSCs in parasitic infections and point l-arginine pathway as a potential target for immunomodulation during nematode infections.


Subject(s)
Arginine/immunology , CD11b Antigen/immunology , Monocytes/immunology , Nematospiroides dubius/immunology , Receptors, Chemokine/immunology , Strongylida Infections/immunology , Animals , CD11b Antigen/genetics , Female , Humans , Immune Tolerance , Mice , Mice, Inbred BALB C , Monocytes/parasitology , Nematospiroides dubius/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Receptors, Chemokine/genetics , Spleen/immunology , Strongylida Infections/genetics , Strongylida Infections/parasitology
7.
Front Immunol ; 10: 2375, 2019.
Article in English | MEDLINE | ID: mdl-31708913

ABSTRACT

Immunity to intestinal helminths is known to require both innate and adaptive components of the immune system activated along the Type 2 IL-4R/STAT6-dependent pathway. We have found that macrophage migration inhibitory factor (MIF) is essential for the development of effective immunity to the intestinal helminth Heligmosomoides polygyrus, even following vaccination which induces sterile immunity in wild-type mice. A chemical inhibitor of MIF, 4-IPP, was similarly found to compromise anti-parasite immunity. Cellular analyses found that the adaptive arm of the immune response, including IgG1 antibody responses and Th2-derived cytokines, was intact and that Foxp3+ T regulatory cell responses were unaltered in the absence of MIF. However, MIF was found to be an essential cytokine for innate cells, with ablated eosinophilia and ILC2 responses, and delayed recruitment and activation of macrophages to the M2 phenotype (expressing Arginase 1, Chil3, and RELM-α) upon infection of MIF-deficient mice; a macrophage deficit was also seen in wild-type BALB/c mice exposed to 4-IPP. Gene expression analysis of intestinal and lymph node tissues from MIF-deficient and -sufficient infected mice indicated significantly reduced levels of Arl2bp, encoding a factor involved in nuclear localization of STAT3. We further found that STAT3-deficient macrophages expressed less Arginase-1, and that mice lacking STAT3 in the myeloid compartment (LysMCrexSTAT3fl/fl) were unable to reject a secondary infection with H. polygyrus. We thus conclude that in the context of a Type 2 infection, MIF plays a critical role in polarizing macrophages into the protective alternatively-activated phenotype, and that STAT3 signaling may make a previously unrecognized contribution to immunity to helminths.


Subject(s)
Immunity, Cellular , Intramolecular Oxidoreductases/immunology , Macrophage Activation , Macrophage Migration-Inhibitory Factors/immunology , Macrophages/immunology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Macrophages/pathology , Mice, Inbred BALB C , Mice, Mutant Strains , Strongylida Infections/genetics , Strongylida Infections/pathology , T-Lymphocytes, Regulatory/pathology
8.
Korean J Parasitol ; 57(3): 249-256, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31284347

ABSTRACT

Steroids are commonly used in patients with eosinophilic meningitis caused by A. cantonensis infections. The mechanism steroids act on eosinophilic meningitis remains unclear. In this mouse experiments, expressions of 14-3-3 isoform ß and γ proteins significantly increased in the CSF 2-3 weeks after the infection, but not increasedin the dexamethasone-treated group. Expression of 14-3-3 ß, γ, ε, and θ isoforms increased in brain meninges over the 3-week period after infection and decreased due to dexamethasone treatment. In conclusion, administration of dexamethasone in mice with eosinophilic meningitis decreased expressions of 14-3-3 isoform proteins in the CSF and in brain meninges.


Subject(s)
14-3-3 Proteins/genetics , Angiostrongylus cantonensis/drug effects , Dexamethasone/administration & dosage , Eosinophilia/drug therapy , Meningitis/genetics , Strongylida Infections/genetics , 14-3-3 Proteins/cerebrospinal fluid , Angiostrongylus cantonensis/physiology , Animals , Down-Regulation/drug effects , Eosinophilia/cerebrospinal fluid , Eosinophilia/genetics , Female , Humans , Male , Meningitis/cerebrospinal fluid , Meningitis/parasitology , Mice , Mice, Inbred BALB C , Protein Isoforms/genetics , Protein Isoforms/metabolism , Strongylida Infections/cerebrospinal fluid , Strongylida Infections/drug therapy , Strongylida Infections/parasitology
9.
Nat Immunol ; 20(5): 571-580, 2019 05.
Article in English | MEDLINE | ID: mdl-30936493

ABSTRACT

Fine control of macrophage activation is needed to prevent inflammatory disease, particularly at barrier sites such as the lungs. However, the dominant mechanisms that regulate the activation of pulmonary macrophages during inflammation are poorly understood. We found that alveolar macrophages (AlvMs) were much less able to respond to the canonical type 2 cytokine IL-4, which underpins allergic disease and parasitic worm infections, than macrophages from lung tissue or the peritoneal cavity. We found that the hyporesponsiveness of AlvMs to IL-4 depended upon the lung environment but was independent of the host microbiota or the lung extracellular matrix components surfactant protein D (SP-D) and mucin 5b (Muc5b). AlvMs showed severely dysregulated metabolism relative to that of cavity macrophages. After removal from the lungs, AlvMs regained responsiveness to IL-4 in a glycolysis-dependent manner. Thus, impaired glycolysis in the pulmonary niche regulates AlvM responsiveness during type 2 inflammation.


Subject(s)
Inflammation/immunology , Lung/immunology , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Animals , Inflammation/genetics , Inflammation/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Larva/immunology , Larva/physiology , Lung/metabolism , Lung/pathology , Macrophage Activation/genetics , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/parasitology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucin-5B/genetics , Mucin-5B/immunology , Mucin-5B/metabolism , Nippostrongylus/immunology , Nippostrongylus/physiology , Pulmonary Surfactant-Associated Protein D/genetics , Pulmonary Surfactant-Associated Protein D/immunology , Pulmonary Surfactant-Associated Protein D/metabolism , Strongylida Infections/genetics , Strongylida Infections/immunology , Strongylida Infections/parasitology
10.
J Immunol ; 202(8): 2473-2481, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30850474

ABSTRACT

Helminthic infections modulate host immunity and may protect their hosts from developing immunological diseases like inflammatory bowel disease. Induction of regulatory T cells (Tregs) may be an important part of this protective process. Heligmosomoides polygyrus bakeri infection also promotes the production of the regulatory cytokines TGF-ß and IL-10 in the gut. In the intestines, TGF-ß helps induce regulatory T cells. This study used Foxp3/IL-10 double reporter mice to investigate the effect of TGF-ß on the differentiation of colon and mesenteric lymph node-derived murine Foxp3- IL-10- CD4+ T cells into their regulatory phenotypes. Foxp3- IL-10- CD4+ T cells from H. polygyrus bakeri-infected mice, as opposed to T cells from uninfected animals, cultured in vitro with TGF-ß and anti-CD3/CD28 mAb differentiated into Foxp3+ and/or IL-10+ T cells. The IL-10-producing T cells nearly all displayed CD25. Smad7 is a natural inhibitor of TGF-ß signaling. In contrast to gut T cells from uninfected mice, Foxp3- IL10- CD4+ T cells from H. polygyrus bakeri-infected mice displayed reduced Smad7 expression and responded to TGF-ß with Smad2/3 phosphorylation. The TGF-ß-induced Tregs that express IL-10 blocked colitis when transferred into the Rag/CD25- CD4+ T cell transfer model of inflammatory bowel disease. TGF-ß had a greatly diminished capacity to induce Tregs in H. polygyrus bakeri-infected transgenic mice with constitutively high T cell-specific Smad7 expression. Thus, infection with H. polygyrus bakeri causes down-modulation in Smad7 expression in intestinal CD4+ T cells, which allows the TGF-ß produced in response to the infection to induce the Tregs that prevent colitis.


Subject(s)
Colitis/immunology , Interleukin-10/immunology , Nematospiroides dubius/immunology , Smad7 Protein/immunology , Strongylida Infections/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Colitis/pathology , Colitis/prevention & control , Interleukin-10/genetics , Mice , Mice, Transgenic , Smad7 Protein/genetics , Strongylida Infections/genetics , Strongylida Infections/pathology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/genetics
11.
Biochim Biophys Acta Gene Regul Mech ; 1862(5): 557-566, 2019 05.
Article in English | MEDLINE | ID: mdl-30763737

ABSTRACT

MicroRNA plays a vital role in the regulation of host-parasite interaction. In recent years, genomic and transcriptomic resources have become increasingly available for many helminths, but only a limited number of reports in this area are on the regulatory effects of host microRNAs on parasitic nematodes. In this work, we screened increased expression of host microRNAs after nematode infection from miRNA-seq data and predicted target genes by combined bioinformatics analysis and transcriptional profiling. We elucidated regulatory effects of one host miRNA on nematode infection using miRNA inhibitor and adeno-associated virus (AAV)-based TuD miRNA inhibitor. Using AAV-based TuD miRNA inhibitor, we showed that stable blockade of mmu-miR-101b-3p could alleviate the pathological damages of Angiostrongylus cantonensis, a parasitic nematode. Data from a luciferase report assay showed that mmu-miR-101b-3p targeted the extracellular superoxide dismutase 3 (Acsod3). Increased Acsod3 expression in larvae and alleviated oxidative damages were seen in the groups receiving mmu-miR-101b-3p inhibitor treatment in vitro and AAV-based TuD miRNA inhibitor injection in vivo. Results of this study demonstrate that murine miR-101b-3p inhibits the expression of antioxidant enzyme in A. cantonensis to strengthen host oxidative responses to nematodes. This work expands our knowledge of interspecies regulation of nematode gene expression by of host miRNAs.


Subject(s)
Angiostrongylus cantonensis/enzymology , MicroRNAs/physiology , Strongylida Infections/genetics , Superoxide Dismutase/genetics , Angiostrongylus cantonensis/genetics , Angiostrongylus cantonensis/growth & development , Angiostrongylus cantonensis/ultrastructure , Animals , Female , Larva/enzymology , Larva/ultrastructure , Mice , MicroRNAs/metabolism , Oxidative Stress , Rats, Sprague-Dawley , Strongylida Infections/parasitology , Superoxide Dismutase/metabolism
12.
Elife ; 72018 09 21.
Article in English | MEDLINE | ID: mdl-30238872

ABSTRACT

Interleukin 25 (IL-25) is a major 'alarmin' cytokine, capable of initiating and amplifying the type immune response to helminth parasites. However, its role in the later effector phase of clearing chronic infection remains unclear. The helminth Heligmosomoides polygyrus establishes long-term infections in susceptible C57BL/6 mice, but is slowly expelled in BALB/c mice from day 14 onwards. We noted that IL-25R (Il17rb)-deficient BALB/c mice were unable to expel parasites despite type 2 immune activation comparable to the wild-type. We then established that in C57BL/6 mice, IL-25 adminstered late in infection (days 14-17) drove immunity. Moreover, when IL-25 and IL-4 were delivered to Rag1-deficient mice, the combination resulted in near complete expulsion of the parasite, even following administration of an anti-CD90 antibody to deplete innate lymphoid cells (ILCs). Hence, effective anti-helminth immunity during chronic infection requires an innate effector cell population that is synergistically activated by the combination of IL-4Rα and IL-25R signaling.


Subject(s)
Immunity, Innate/immunology , Nematospiroides dubius/immunology , Receptors, Cell Surface/immunology , Receptors, Interleukin-17/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Host-Parasite Interactions/immunology , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interleukin-17/immunology , Interleukin-17/pharmacology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/physiology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Strongylida Infections/genetics , Strongylida Infections/parasitology , Th2 Cells/metabolism
13.
Sci Rep ; 8(1): 5933, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29651122

ABSTRACT

Strongylid nematodes in large terrestrial herbivores such as great apes, equids, elephants, and humans tend to occur in complex communities. However, identification of all species within strongylid communities using traditional methods based on coproscopy or single nematode amplification and sequencing is virtually impossible. High-throughput sequencing (HTS) technologies provide opportunities to generate large amounts of sequence data and enable analyses of samples containing a mixture of DNA from multiple species/genotypes. We designed and tested an HTS approach for strain-level identification of gastrointestinal strongylids using ITS-2 metabarcoding at the MiSeq Illumina platform in samples from two free-ranging non-human primate species inhabiting the same environment, but differing significantly in their host traits and ecology. Although we observed overlapping of particular haplotypes, overall the studied primate species differed in their strongylid nematode community composition. Using HTS, we revealed hidden diversity in the strongylid nematode communities in non-human primates, more than one haplotype was found in more than 90% of samples and coinfections of more than one putative species occurred in 80% of samples. In conclusion, the HTS approach on strongylid nematodes, preferably using fecal samples, represents a time and cost-efficient way of studying strongylid communities and provides a resolution superior to traditional approaches.


Subject(s)
DNA Barcoding, Taxonomic , Horse Diseases/genetics , Strongylida Infections/genetics , Strongylida/genetics , Animals , Feces/parasitology , Genetic Variation , High-Throughput Nucleotide Sequencing , Horse Diseases/parasitology , Horses/genetics , Horses/parasitology , Interspersed Repetitive Sequences/genetics , Strongylida/classification , Strongylida Infections/parasitology , Sympatry
14.
Parasit Vectors ; 10(1): 445, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28950910

ABSTRACT

BACKGROUND: Angiostrongylus cantonensis is an important causative agent of eosinophilic meningitis and eosinophilic meningoencephalitis in humans. Previous studies have shown that the Sonic hedgehog (Shh) signaling pathway may reduce cell apoptosis by inhibiting oxidative stress in A. cantonensis infection. In this study, we investigated the relationship between cytokine secretion and Shh pathway activation after treatment with excretory/secretory products (ESP) of fifth-stage larval A. cantonensis (L5). RESULTS: The results showed that IL-1ß and IL-6 levels in mouse astrocytes were increased. Moreover, ESP stimulated the protein expression of Shh pathway molecules, including Shh, Ptch, Smo and Gli-1, and induced IL-1ß and IL-6 secretion. The transcription factor nuclear factor-κB (NF-κB) plays an important role in inflammation, and it regulates the expression of proinflammatory genes, including cytokines and chemokines, such as IL-1ß and TNF-α. After ESP treatment, NF-κB induced IL-1ß and IL-6 secretion in astrocytes by activating the Shh signaling pathway. CONCLUSIONS: Overall, the data presented in this study showed that ESP of fifth-stage larval A. cantonensis stimulates astrocyte activation and cytokine generation through NF-κB and the Shh signaling pathway.


Subject(s)
Angiostrongylus cantonensis/metabolism , Astrocytes/metabolism , Hedgehog Proteins/metabolism , Helminth Proteins/metabolism , Interleukin-18/metabolism , Interleukin-6/metabolism , NF-kappa B/metabolism , Strongylida Infections/parasitology , Angiostrongylus cantonensis/chemistry , Angiostrongylus cantonensis/growth & development , Animals , Astrocytes/parasitology , Female , Hedgehog Proteins/genetics , Host-Parasite Interactions , Humans , Interleukin-18/genetics , Interleukin-6/genetics , Larva/chemistry , Larva/growth & development , Larva/metabolism , Male , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction , Strongylida Infections/genetics , Strongylida Infections/metabolism
15.
PLoS Pathog ; 13(7): e1006536, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28759611

ABSTRACT

TPL-2 (COT, MAP3K8) kinase activates the MEK1/2-ERK1/2 MAPK signaling pathway in innate immune responses following TLR, TNFR1 and IL-1R stimulation. TPL-2 contributes to type-1/Th17-mediated autoimmunity and control of intracellular pathogens. We recently demonstrated TPL-2 reduces severe airway allergy to house dust mite by negatively regulating type-2 responses. In the present study, we found that TPL-2 deficiency resulted in resistance to Heligmosomoides polygyrus infection, with accelerated worm expulsion, reduced fecal egg burden and reduced worm fitness. Using co-housing experiments, we found resistance to infection in TPL-2 deficient mice (Map3k8-/-) was independent of microbiota alterations in H. polygyrus infected WT and Map3k8-/-mice. Additionally, our data demonstrated immunity to H. polygyrus infection in TPL-2 deficient mice was not due to dysregulated type-2 immune responses. Genome-wide analysis of intestinal tissue from infected TPL-2-deficient mice identified elevated expression of genes involved in chemotaxis and homing of leukocytes and cells, including Ccl24 and alternatively activated genes. Indeed, Map3k8-/-mice had a significant influx of eosinophils, neutrophils, monocytes and Il4GFP+ T cells. Conditional knockout experiments demonstrated that specific deletion of TPL-2 in CD11c+ cells, but not Villin+ epithelial cells, LysM+ myeloid cells or CD4+ T cells, led to accelerated resistance to H. polygyrus. In line with a central role of CD11c+ cells, CD11c+ CD11b+ cells isolated from TPL-2-deficient mice had elevated Ccl24. Finally, Ccl24 neutralization in TPL-2 deficient mice significantly decreased the expression of Arg1, Retnla, Chil3 and Ear11 correlating with a loss of resistance to H. polygyrus. These observations suggest that TPL-2-regulated Ccl24 in CD11c+CD11b+ cells prevents accelerated type-2 mediated immunity to H. polygyrus. Collectively, this study identifies a previously unappreciated role for TPL-2 controlling immune responses to H. polygyrus infection by restricting Ccl24 production.


Subject(s)
Chemokine CCL24/immunology , MAP Kinase Kinase Kinases/immunology , Nematospiroides dubius/immunology , Proto-Oncogene Proteins/immunology , Strongylida Infections/immunology , Animals , Chemokine CCL24/genetics , Female , Humans , Immunity, Innate , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/genetics , Nematospiroides dubius/physiology , Proto-Oncogene Proteins/genetics , Strongylida Infections/enzymology , Strongylida Infections/genetics , Strongylida Infections/parasitology , Th2 Cells/immunology
17.
J Immunol ; 197(11): 4371-4381, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27798167

ABSTRACT

The AP-1 factor basic leucine zipper transcription factor, ATF-like (BATF) is important for CD4+ Th17, Th9, and follicular Th cell development. However, its precise role in Th2 differentiation and function remains unclear, and the requirement for BATF in nonallergic settings of type-2 immunity has not been explored. In this article, we show that, in response to parasitic helminths, Batf-/- mice are unable to generate follicular Th and Th2 cells. As a consequence, they fail to establish productive type-2 immunity during primary and secondary infection. Batf-/- CD4+ T cells do not achieve type-2 cytokine competency, which implies that BATF plays a key role in the regulation of IL-4 and IL-13. In contrast to Th17 and Th9 cell subsets in which BATF binds directly to promoter and enhancer regions to regulate cytokine expression, our results show that BATF is significantly enriched at Rad50 hypersensitivity site (RHS)6 and RHS7 of the locus control region relative to AP-1 sites surrounding type-2 cytokine loci in Th2 cells. Indeed, Batf-/- CD4+ T cells do not obtain permissive epigenetic modifications within the Th2 locus, which were linked to RHS6 and RHS7 function. In sum, these findings reveal BATF as a central modulator of peripheral and humoral hallmarks of type-2 immunity and begin to elucidate a novel mechanism by which it regulates type-2 cytokine production through its modification of the Th2 locus control region.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Epigenesis, Genetic/immunology , Locus Control Region/immunology , Strongylida Infections/immunology , Th2 Cells/immunology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Acid Anhydride Hydrolases , Animals , Basic-Leucine Zipper Transcription Factors/genetics , DNA-Binding Proteins , Mice , Mice, Knockout , Strongylida Infections/genetics , Strongylida Infections/pathology , Th2 Cells/pathology
18.
Proc Natl Acad Sci U S A ; 113(36): 10139-44, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27551096

ABSTRACT

Natural killer (NK) cells are known to be activated by Th1-type cytokines, such as IL-2, -12, or -18, and they secrete a large amount of IFN-γ that accelerates Th1-type responses. However, the roles of NK cells in Th2-type responses have remained unclear. Because IL-4 acts as an initiator of Th2-type responses, we examined the characteristics of NK cells in mice overexpressing IL-4. In this study, we report that IL-4 overexpression induces distinctive characteristics of NK cells (B220(high)/CD11b(low)/IL-18Rα(low)), which are different from mature conventional NK (cNK) cells (B220(low)/CD11b(high)/IL-18Rα(high)). IL-4 overexpression induces proliferation of tissue-resident macrophages, which contributes to NK cell proliferation via production of IL-15. These IL-4-induced NK cells (IL4-NK cells) produce higher levels of IFN-γ, IL-10, and GM-CSF, and exhibit high cytotoxicity compared with cNK cells. Furthermore, incubation of cNK cells with IL-15 and IL-4 alters their phenotype to that similar to IL4-NK cells. Finally, parasitic infection, which typically causes strong Th2-type responses, induces the development of NK cells with characteristics similar to IL4-NK cells. These IL4-NK-like cells do not develop in IL-4Rα KO mice by parasitic infection. Collectively, these results suggest a novel role of IL-4 in immune responses through the induction of the unique NK cells.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-15/immunology , Interleukin-4/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Strongylida Infections/immunology , Animals , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Proliferation , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-15/genetics , Interleukin-15/pharmacology , Interleukin-4/genetics , Interleukin-4/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/parasitology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Macrophages/immunology , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/immunology , Nippostrongylus/pathogenicity , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/immunology , Receptors, Interleukin-4/deficiency , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/immunology , Signal Transduction , Strongylida Infections/genetics , Strongylida Infections/parasitology
19.
Parasitol Res ; 115(10): 3737-46, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27256220

ABSTRACT

Angiostrongyliasis caused by Angiostrongylus cantonensis (A. cantonensis) is an emerging food-borne parasitic disease, which refers basically to eosinophilic meningitis. Chitinase-like protein 3 (Chil3), a member of chitinase-like protein family which has chemotactic activity for eosinophils, is reported to be highly upregulated in brain of mouse infected with A. cantonensis. The mechanisms of high expression of Chil3 and the association between A. cantonensis and Chil3 are rarely reported. In order to understand the mechanism of high expression of Chil3 in A. cantonensis-infected mouse, we measured the level of Chil3 in RAW 264.7 and BV2 cell lines stimulated with soluble antigen of A. cantonensis by qPCR and ELISA. To explore the role of Chil3 in inflammation caused by A. cantonensis, we extracted and cultured brain mononuclear cells (BMNCs) and detected the eosinophil chemotactic activity of Chil3 using transwell assay and flow cytometer. Furthermore, we treated the infected mice by injection with rmChil3 and then counted the number of larvae in brains of infected mice and treated mice to examine the association between the worm and Chil3. Our results showed the soluble antigen from A. cantonensis could promote the Chil3 expression in macrophage and microglial cell lines induced by interleukin-13. In conclusion, we supposed that high expression of Chil3 enhanced by soluble antigens from A. cantonensis might be the reason of serious eosinophil infiltration in mouse brain after A. cantonensis infection.


Subject(s)
Angiostrongylus cantonensis/metabolism , Antigens, Helminth/metabolism , Chitinases/genetics , Interleukin-13/metabolism , Larva/metabolism , Strongylida Infections/metabolism , Strongylida Infections/parasitology , Angiostrongylus cantonensis/genetics , Angiostrongylus cantonensis/growth & development , Animals , Antigens, Helminth/genetics , Brain/enzymology , Brain/parasitology , Chitinases/metabolism , Female , Gene Expression Profiling , Humans , Interleukin-13/genetics , Larva/genetics , Larva/growth & development , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Snails , Strongylida Infections/enzymology , Strongylida Infections/genetics
20.
Am J Physiol Gastrointest Liver Physiol ; 311(1): G130-41, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27173511

ABSTRACT

Despite increased appreciation for the role of nicotinic receptors in the modulation of and response to inflammation, the contribution of muscarinic receptors to mucosal homeostasis, clearance of enteric pathogens, and modulation of immune cell function remains relatively undefined. Uninfected and Nippostrongylus brasiliensis-infected wild-type and type 3 muscarinic receptor (M3R)-deficient (Chrm3(-/-)) mice were studied to determine the contribution of M3R to mucosal homeostasis as well as host defense against the TH2-eliciting enteric nematode N. brasiliensis Intestinal permeability and expression of TH1/TH17 cytokines were increased in uninfected Chrm3(-/-) small intestine. Notably, in Chrm3(-/-) mice infected with N. brasiliensis, small intestinal upregulation of TH2 cytokines was attenuated and nematode clearance was delayed. In Chrm3(-/-) mice, TH2-dependent changes in small intestinal function including smooth muscle hypercontractility, increased epithelial permeability, decreased epithelial secretion and absorption, and goblet cell expansion were absent despite N. brasiliensis infection. These findings identify an important role for M3R in host defense and clearance of N. brasiliensis, and support the expanding role of cholinergic muscarinic receptors in maintaining mucosal homeostasis.


Subject(s)
Cytokines/metabolism , Immunity, Mucosal , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Nippostrongylus/pathogenicity , Receptor, Muscarinic M3/metabolism , Strongylida Infections/metabolism , Th2 Cells/metabolism , Animals , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Genetic Predisposition to Disease , Homeostasis , Host-Pathogen Interactions , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestine, Small/immunology , Intestine, Small/parasitology , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/parasitology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/immunology , Phenotype , Receptor, Muscarinic M3/deficiency , Receptor, Muscarinic M3/genetics , Strongylida Infections/genetics , Strongylida Infections/immunology , Strongylida Infections/parasitology , Th2 Cells/immunology , Th2 Cells/parasitology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...