Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
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
2.
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
3.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33636133

ABSTRACT

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Subject(s)
Coinfection , Nematospiroides dubius/physiology , Signal Transduction , Strongylida Infections/pathology , West Nile virus/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Disease Susceptibility , Intestinal Mucosa/parasitology , Intestinal Mucosa/virology , Mice , Mice, Inbred C57BL , Neurons/parasitology , Neurons/virology , Receptors, Interleukin-4/metabolism , STAT6 Transcription Factor/genetics , Severity of Illness Index , Strongylida Infections/parasitology
4.
Parasit Vectors ; 14(1): 106, 2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33557937

ABSTRACT

BACKGROUND: Excretory-secretory (ES) products are crucial in maintaining helminths in the host. Consequently, the proteins of ES are potential vaccine molecules and potential therapeutic agents for autoimmune diseases. Heligmosomoides polygyrus bakeri, a gastrointestinal parasite of mice, is a model of hookworm infection in humans. ES produced by both sexes of H. polygyrus bakeri L4 stage cultured separately shows different immunomodulatory properties than ES obtained when both sexes are cultured together. Accordingly, the objective of this study was to identify and compare the excretory-secretory molecules from single-sex and mixed cultures. METHODS: The composition of ES of male and female L4 stage nematodes in the presence (cultured together) or absence (cultured alone) of the opposite sex was examined. Proteins were identified using mass spectrometry. The functions of identified proteins were explored with Blast2GO. RESULTS: A total of 258 proteins derived from mixed larval culture in the presence of sex pheromones were identified, 160 proteins from pure female cultures and 172 from pure male cultures. Exposure of nematodes to the sex pheromones results in abundant production of proteins with immunomodulatory properties such as Val proteins, acetylcholinesterases, TGF-ß mimic 9 and HpARI. Proteins found only in ES from mixed larval cultures were TGF-ß mimics 6 and 7 as well as galectin. CONCLUSIONS: The presence of the opposite sex strongly influences the composition of ES products, probably by chemical (pheromone) communication between individuals. However, examination of the composition of ES from various conditions gives an opportunity for searching for new potentially therapeutic compounds and anthelminthics as well as components of vaccines. Manipulation of the nematode environment might be important for the studies on the immunomodulatory potential of nematodes.


Subject(s)
Helminth Proteins/analysis , Helminth Proteins/metabolism , Larva/physiology , Nematospiroides dubius/physiology , Animals , Computational Biology , Female , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Nematospiroides dubius/growth & development , Specific Pathogen-Free Organisms
5.
PLoS Negl Trop Dis ; 15(1): e0009052, 2021 01.
Article in English | MEDLINE | ID: mdl-33471793

ABSTRACT

Intestinal helminth infection can impair host resistance to co-infection with enteric bacterial pathogens. However, it is not known whether helminth drug-clearance can restore host resistance to bacterial infection. Using a mouse helminth-Salmonella co-infection system, we show that anthelmintic treatment prior to Salmonella challenge is sufficient to restore host resistance to Salmonella. The presence of the small intestine-dwelling helminth Heligmosomoides polygyrus at the point of Salmonella infection supports the initial establishment of Salmonella in the small intestinal lumen. Interestingly, if helminth drug-clearance is delayed until Salmonella has already established in the small intestinal lumen, anthelmintic treatment does not result in complete clearance of Salmonella. This suggests that while the presence of helminths supports initial Salmonella colonization, helminths are dispensable for Salmonella persistence in the host small intestine. These data contribute to the mechanistic understanding of how an ongoing or prior helminth infection can affect pathogenic bacterial colonization and persistence in the mammalian intestine.


Subject(s)
Coinfection/microbiology , Coinfection/parasitology , Disease Resistance/physiology , Helminthiasis/complications , Intestinal Diseases, Parasitic/complications , Nematospiroides dubius/physiology , Salmonella/physiology , Animals , Disease Models, Animal , Female , Gastrointestinal Microbiome , Intestines/microbiology , Intestines/parasitology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Salmonella Infections/complications , Salmonella typhi
6.
J Exp Med ; 218(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33237304

ABSTRACT

Expression of the signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) is critical for the germinal center (GC) reaction and T cell-dependent antibody production. However, when SAP is expressed normally, the role of the associated SLAM family receptors (SFRs) in these processes is nebulous. Herein, we established that in the presence of SAP, SFRs suppressed the expansion of the GC reaction but facilitated the generation of antigen-specific B cells and antibodies. SFRs favored the generation of antigen-reactive B cells and antibodies by boosting expression of pro-survival effectors, such as the B cell antigen receptor (BCR) and Bcl-2, in activated GC B cells. The effects of SFRs on the GC reaction and T cell-dependent antibody production necessitated expression of multiple SFRs, both in T cells and in B cells. Hence, while in the presence of SAP, SFRs inhibit the GC reaction, they are critical for the induction of T cell-mediated humoral immunity by enhancing expression of pro-survival effectors in GC B cells.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Germinal Center/cytology , Immunity, Humoral , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Antibodies/metabolism , Antibody Formation/immunology , Antigens, Helminth/metabolism , Apoptosis , Bone Marrow/metabolism , Cell Count , Cell Cycle , Cell Proliferation , Cell Survival , Dose-Response Relationship, Immunologic , Immunization , Immunoglobulin Class Switching , Immunologic Memory , Mice, Knockout , Nematospiroides dubius/physiology , Plasma Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Signaling Lymphocytic Activation Molecule Family/deficiency , Somatic Hypermutation, Immunoglobulin , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Vaccination
7.
Parasitology ; 147(13): 1480-1487, 2020 11.
Article in English | MEDLINE | ID: mdl-32729450

ABSTRACT

The gastrointestinal nematode Heligmosomoides polygyrus bakeri shows enhanced survival in mice with colitis. As the antibody response plays an important role in antiparasitic immunity, antibodies against male and female L4 H. polygyrus were examined in mice with and without colitis. Levels of specific antibodies in the mucosa and serum were determined by enzyme-linked immunosorbent assay and immunogenic proteins of male and female parasites were identified using 2D electrophoresis and mass spectrometry. The function of identified proteins was explored with Blast2Go. Nematodes in mice with colitis induced higher levels of specific immunoglobulin G (IgG1) and IgA, a lower level of IgE in the small intestine and a higher level of IgE in serum against female L4. Infected mice with colitis recognized 12 proteins in male L4 and 10 in female L4. Most of the recognized proteins from male L4 were intermediate filament proteins, whereas the proteins from female L4 were primarily actins and galectins. Nematodes from mice with colitis were immunogenically different from nematodes from control mice. This phenomenon gives new insights into helminth therapy as well as host-parasite interactions.


Subject(s)
Antibodies, Helminth/immunology , Colitis/immunology , Helminth Proteins/immunology , Nematospiroides dubius/physiology , Proteome/immunology , Strongylida Infections/immunology , Animals , Colitis/parasitology , Female , Intestines/parasitology , Male , Mice , Mice, Inbred BALB C , Strongylida Infections/parasitology
8.
Mucosal Immunol ; 13(2): 357-370, 2020 03.
Article in English | MEDLINE | ID: mdl-31776431

ABSTRACT

Parasitic helminths cause significant damage as they migrate through host tissues to complete their life cycle. While chronic helminth infections are characterized by a well-described Type 2 immune response, the early, tissue-invasive stages are not well understood. Here we investigate the immune pathways activated during the early stages of Heligmosomoides polygyrus bakeri (Hpb), a natural parasitic roundworm of mice. In contrast to the Type 2 immune response present at later stages of infection, a robust Type 1 immune signature including IFNg production was dominant at the time of parasite invasion and granuloma formation. This early response was associated with an accumulation of activated Natural Killer (NK) cells, with no increase of other innate lymphoid cell populations. Parabiosis and confocal microscopy studies indicated that NK cells were recruited from circulation to the small intestine, where they surrounded parasitic larvae. NK cell recruitment required IFNγ receptor signaling, but was independent of CXCR3 expression. The depletion of tissue-infiltrating NK cells altered neither worm burden nor parasite fitness, but increased vascular injury, suggesting a role for NK cells in mediating tissue protection. Together, these data identify an unexpected role for NK cells in promoting disease tolerance during the invasive stage of an enteric helminth infection.


Subject(s)
Gastrointestinal Tract/immunology , Immunologic Surveillance , Intestines/immunology , Killer Cells, Natural/immunology , Nematospiroides dubius/physiology , Strongylida Infections/immunology , Th1 Cells/metabolism , Vascular System Injuries/immunology , Animals , Cell Movement , Female , Immunity, Innate , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Parabiosis , Receptors, CXCR/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Interferon gamma Receptor
9.
Integr Comp Biol ; 59(5): 1190-1202, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31368489

ABSTRACT

The immune system represents a host's main defense against infection to parasites and pathogens. In the wild, a host's response to immune challenges can vary due to physiological condition, demography (age, sex), and coinfection by other parasites or pathogens. These sources of variation, which are intrinsic to natural populations, can significantly impact the strength and type of immune responses elicited after parasite exposure and infection. Importantly, but often neglected, a host's immune response can also vary within the individual, across tissues and between local and systemic scales. Consequently, how a host responds at each scale may impact its susceptibility to concurrent and subsequent infections. Here we analyzed how characteristics of hosts and their parasite infections drive variation in the pro-inflammatory immune response in wild wood mice (Apodemus sylvaticus) at both the local and systemic scale by experimentally manipulating within-host parasite communities through anthelmintic drug treatment. We measured concentrations of the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α) produced in vitro in response to a panel of toll-like receptor agonists at the local (mesenteric lymph nodes [MLNs]) and systemic (spleen) scales of individuals naturally infected with two gastrointestinal parasites, the nematode Heligmosomoides polygyrus and the protozoan Eimeria hungaryensis. Anthelmintic-treated mice had a 20-fold lower worm burden compared to control mice, as well as a four-fold higher intensity of the non-drug targeted parasite E. hungaryensis. Anthelmintic treatment differentially impacted levels of TNF-α expression in males and females at the systemic and local scales, with treated males producing higher, and treated females lower, levels of TNF-α, compared to control mice. Also, TNF-α was affected by host age, at the local scale, with MLN cells of young, treated mice producing higher levels of TNF-α than those of old, treated mice. Using complementary, but distinct, measures of inflammation measured across within-host scales allowed us to better assess the wood mouse immune response to changes in parasite infection dynamics after anthelmintic treatment. This same approach could be used to understand helminth infections and responses to parasite control measures in other systems in order to gain a broader view of how variation impacts the immune response.


Subject(s)
Anthelmintics/pharmacology , Coccidiosis/veterinary , Eimeria/physiology , Murinae , Nematospiroides dubius/physiology , Rodent Diseases/immunology , Strongylida Infections/veterinary , Animals , Biomarkers , Coccidiosis/immunology , Coccidiosis/parasitology , Host-Parasite Interactions , Ivermectin/pharmacology , Pyrantel Pamoate/pharmacology , Rodent Diseases/parasitology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Tumor Necrosis Factor-alpha/metabolism
10.
Clin Exp Immunol ; 198(3): 390-402, 2019 12.
Article in English | MEDLINE | ID: mdl-31397879

ABSTRACT

The prevalence of allergic asthma and incidences of helminth infections in humans are inversely correlated. Although experimental studies have established the causal relation between parasite infection and allergic asthma, the mechanism of the parasite-associated immunomodulation is not fully elucidated. Using a murine model of asthma and nematode parasite Heligmosomoides polygyrus, we investigated the roles of regulatory B cells (Breg ) and T cells (Treg ) in mediation of the protection against allergic asthma by parasite. H. polygyrus infection significantly suppressed ovalbumin (OVA)-induced allergic airway inflammation (AAI) evidenced by alleviated lung histopathology and reduced numbers of bronchoalveolar inflammatory cell infiltration, and induced significant responses of interleukin (IL)-10+ Breg , IL-10+ Treg and forkhead box protein 3 (FoxP3)+ Treg in mesenteric lymph node and spleen of the mice. Adoptive transfer of IL-10+ Breg and IL-10+ Treg cell prevented the lung immunopathology in AAI mice. Depletion of FoxP3+ Treg cells in FoxP3-diphtheria toxin (DT) receptor transgenic mice by diphtheria toxin (DT) treatment exacerbated airway inflammation in parasite-free AAI mice and partially abrogated the parasite-induced protection against AAI. IL-10+ Breg cells were able to promote IL-10+ Treg expansion and maintain FoxP3+ Treg cell population. These two types of Tregs failed to induce CD19+ B cells to transform into IL-10+ Breg cells. These results demonstrate that Breg , IL-10+ Treg and FoxP3+ Treg cells contribute in A discrepant manner to the protection against allergic airway immunopathology by parasiteS. Breg cell might be a key upstream regulatory cell that induces IL-10+ Treg response and supports FoxP3+ Treg cell population which, in turn, mediate the parasite-imposed immunosuppression of allergic airway inflammation. These results provide insight into the immunological relationship between parasite infection and allergic asthma.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Hypersensitivity/immunology , Nematospiroides dubius/immunology , Pneumonia/immunology , Strongylida Infections/immunology , Animals , Asthma/immunology , Asthma/metabolism , Asthma/parasitology , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Hypersensitivity/metabolism , Hypersensitivity/parasitology , Immune Tolerance/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Nematospiroides dubius/physiology , Pneumonia/metabolism , Pneumonia/parasitology , Strongylida Infections/parasitology , T-Lymphocytes, Regulatory/immunology
11.
Infect Immun ; 87(6)2019 06.
Article in English | MEDLINE | ID: mdl-30962398

ABSTRACT

Obesity is increasingly causing lifestyle diseases in developed countries where helminthic infections are rarely seen. Here, we investigated whether an intestinal nematode, Heligmosomoides polygyrus, has a suppressive role in diet-induced obesity in mice. Infection with H. polygyrus suppressed weight gain in obese mice, which was associated with increased uncoupling protein 1 (UCP1) expression in adipocytes and a higher serum norepinephrine (NE) concentration. Blocking interactions of NE with its receptor on adipocytes resulted in the failure to prevent weight gain and to enhance UCP1 expression in obese mice infected with H. polygyrus, indicating that NE is responsible for the protective effects of H. polygyrus on obesity. In addition to sympathetic nerve-derived NE, the intestinal microbiota was involved in the increase in NE. Infection with H. polygyrus altered the composition of intestinal bacteria, and antibiotic treatment to reduce intestinal bacteria reversed the higher NE concentration, UCP1 expression, and prevention of the weight gain observed after H. polygyrus infection. Our data indicate that H. polygyrus exerts suppressive roles on obesity through modulation of microbiota that produce NE.


Subject(s)
Biological Therapy , Gastrointestinal Microbiome , Nematospiroides dubius/physiology , Obesity/microbiology , Obesity/therapy , Adipocytes/metabolism , Animals , Humans , Intestines/microbiology , Intestines/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Norepinephrine/metabolism , Obesity/metabolism , Obesity/parasitology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
12.
Parasitology ; 146(8): 1096-1106, 2019 07.
Article in English | MEDLINE | ID: mdl-30915927

ABSTRACT

Within-host interactions among coinfecting parasites are common and have important consequences for host health and disease dynamics. However, these within-host interactions have traditionally been studied in laboratory mouse models, which often exclude important variation and use unnatural host-parasite combinations. Conversely, the few wild studies of within-host interactions often lack knowledge of parasite exposure and infection history. Here we exposed laboratory-reared wood mice (Apodemus sylvaticus) that were derived from wild-caught animals to two naturally-occurring parasites (nematode: Heligmosomoides polygyrus, coccidia: Eimeria hungaryensis) to investigate the impact of coinfection on parasite infection dynamics, and to determine if the host immune response mediates this interaction. Coinfection led to delayed worm expulsion and prolonged egg shedding in H. polygyrus infections and lower peak E. hungaryensis oocyst burdens. By comparing antibody levels between wild and colony-housed mice, we also found that wild mice had elevated H. polygyrus-IgG1 titres even if currently uninfected with H. polygyrus. Using this unique wild-laboratory system, we demonstrate, for the first time, clear evidence for a reciprocal interaction between these intestinal parasites, and that there is a great discrepancy between antibody levels measured in the wild vs those measured under controlled laboratory conditions in relation to parasite infection and coinfection.


Subject(s)
Coccidiosis/veterinary , Coinfection/veterinary , Eimeria/physiology , Murinae , Nematospiroides dubius/physiology , Rodent Diseases/parasitology , Strongylida Infections/veterinary , Animals , Coccidiosis/parasitology , Coinfection/parasitology , Female , Intestinal Diseases, Parasitic/parasitology , Intestinal Diseases, Parasitic/veterinary , Male , Strongylida Infections/parasitology
13.
Exp Parasitol ; 196: 12-21, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30465732

ABSTRACT

Helminths and their products are strong candidates for the treatment of autoimmunological disorders and allergies. Being a key population of antigen-presenting cells, dendritic cells play a crucial role in the therapeutic potential of worms. The study compares the effects of live pre-male and pre-female L4 stage Heligmosomoides polygyrus administration on the maturation and activation of the JAWS II line of immature dendritic cells. On stimulation with L4 stage H. polygyrus, JAWS II cells acquire semi-mature status and induce Th2 and regulatory responses in vitro. The strongest immunosuppressive effect on JAWS II cells was observed following stimulation with both sexes of nematodes together; this was manifested as immature dendritic cell morphology, proliferation inhibition, cell cycle change, decreased translocation of NF-κB into the nucleus, and lower expression of surface cellular costimulatory molecules CD80, CD86 and MHC I. However, greater production of proinflammatory (IL-12p70, TNF-α, IL-6) and Th2 response-promoting cytokines (IL-4) was observed by JAWS II following exposure to both sexes compared to male or female larvae alone. Sex had no influence on the viability, apoptosis process or endocytosis abilities of the JAWS II cell line. The findings indicate that the presence of only a single sex of the parasite influences a developed response, resulting in reduced proinflammatory and an antiparasitic reaction.


Subject(s)
Dendritic Cells/parasitology , Nematospiroides dubius/physiology , Animals , Apoptosis , Bone Marrow Cells/immunology , Bone Marrow Cells/parasitology , Bone Marrow Cells/physiology , Cell Cycle , Cell Line , Chemokines/analysis , Cytokines/analysis , Dendritic Cells/immunology , Dendritic Cells/physiology , Endocytosis , Female , Larva/physiology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/analysis , Nematospiroides dubius/growth & development , Sex Factors , Specific Pathogen-Free Organisms
14.
J Helminthol ; 93(5): 552-558, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30001752

ABSTRACT

The yellow-necked mouse, Apodemus flavicollis, can be considered as a model for genetic polymorphism produced by the frequent presence of supernumerary or B chromosomes (Bs). Host genetic background is rarely taken into account in studies of parasite sex ratio. The main aim of this study was to investigate the range of infrapopulation sex ratios for nematode parasites of the yellow-necked mouse and to determine which factors most influence variation in parasite sex ratios. Six nematode species found in the collected yellow-necked mice were analysed. We confirmed the predominant pattern of female-biased sex ratios in vertebrate parasite infrapopulations. The presence of B chromosomes in host genomes played an important role in infrapopulations of Heligmosomoides polygyrus, Syphacia stroma and Trichuris muris, as hosts with B chromosomes carried a higher proportion of males. The relative increase of males in infrapopulations could result from a shift in parasite life history strategy, induced by adaptation to the specific host genotypes (Bs present). In a meta-analysis with previously published data, the sex determination system was demonstrated to play a significant role in nematode sex ratio variation, as well as specific life history patterns, such as the place of egg hatching.


Subject(s)
Chromosomes , Host-Parasite Interactions/genetics , Murinae/genetics , Nematoda/physiology , Sex Ratio , Animals , Female , Genome , Male , Murinae/parasitology , Nematospiroides dubius/physiology , Serbia , Sex Determination Processes , Trichuris/physiology
15.
Mucosal Immunol ; 12(2): 352-362, 2019 03.
Article in English | MEDLINE | ID: mdl-30401814

ABSTRACT

The ability of helminths to manipulate the immune system of their hosts to ensure their own survival is often credited with affecting responses to other pathogens. We undertook co-infection experiments in mice to determine how infection with the intestinal helminth Heligmosomoides polygyrus affected the parasitological, immunological and physiological outcomes of a primary infection with a distinct species of helminth; the lung migratory parasite Nippostrongylus brasiliensis. We found that migrating N. brasiliensis larvae were killed in the lungs of H. polygyrus-infected mice by a process involving IL-33-activated CD4+ T cells that released IL-5 and recruited activated eosinophils. The lung pathology normally associated with N. brasiliensis larval migration was also reduced. Importantly, lung immunity remained intact in mice cleared of prior H. polygyrus infection and also occurred during infection with another entirely enteric helminth, Trichuris muris. This study identifies a cross-mucosal immune mechanism by which intestinal helminths may protect their hosts against co-infection by a different parasite at a distal site, via circulation of activated CD4+ T cells that can be triggered to release effector cytokines and mount inflammatory responses by tissue damage-associated alarmins, such as IL-33.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Coinfection , Eosinophils/immunology , Interleukin-5/metabolism , Lung/immunology , Nematospiroides dubius/physiology , Nippostrongylus/physiology , Strongylida Infections/immunology , Trichuriasis/immunology , Trichuris/physiology , Animals , Antigens, Helminth/immunology , Cell Movement , Cells, Cultured , Cytotoxicity, Immunologic , Female , Host-Parasite Interactions , Immunity , Interleukin-33/metabolism , Lung/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Mucosal Immunol ; 12(2): 413-424, 2019 03.
Article in English | MEDLINE | ID: mdl-30446727

ABSTRACT

Intestinal epithelial cells (IECs) constitute an important barrier between host and pathogen. Immune mechanisms that provide protection against gastrointestinal helminths often require IL-4Rα-induced activation of STAT6-regulated genes in IECs. However, it is not known whether STAT6 activation in IECs enhances protective immunity against helminths. Furthermore, the regulation of proliferation and differentiation processes of the intestinal epithelium by IEC-intrinsic STAT6 signaling remains unclear. To address these questions, we generated mice with specific expression of a constitutively active version of STAT6 in IECs. These VillinCre_STAT6vt mice show accumulation of secretory IECs, increased proliferation of IECs and lengthening of the small intestine. They rapidly expelled Nippostrongylus brasiliensis worms even in the absence of T cells. Furthermore, primary infection with Heligmosomoides polygyrus resulted in larval trapping in the submucosa and the fecundity of adult worms was severely impaired. Our results reveal an important IEC-intrinsic role of STAT6-regulated genes for intestinal homeostasis and protective immunity against helminths.


Subject(s)
Epithelial Cells/physiology , Intestines/pathology , Nematospiroides dubius/physiology , Nippostrongylus/physiology , STAT6 Transcription Factor/metabolism , Strongylida Infections/immunology , Animals , Bodily Secretions , Cell Differentiation , Female , Homeostasis , Host-Pathogen Interactions , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , STAT6 Transcription Factor/genetics , Signal Transduction
17.
Front Immunol ; 9: 2282, 2018.
Article in English | MEDLINE | ID: mdl-30349532

ABSTRACT

Intestinal parasitic nematodes live in intimate contact with the host microbiota. Changes in the microbiome composition during nematode infection affect immune control of the parasites and shifts in the abundance of bacterial groups have been linked to the immunoregulatory potential of nematodes. Here we asked if the small intestinal parasite Heligmosomoides polygyrus produces factors with antimicrobial activity, senses its microbial environment and if the anti-nematode immune and regulatory responses are altered in mice devoid of gut microbes. We found that H. polygyrus excretory/secretory products exhibited antimicrobial activity against gram+/- bacteria. Parasites from germ-free mice displayed alterations in gene expression, comprising factors with putative antimicrobial functions such as chitinase and lysozyme. Infected germ-free mice developed increased small intestinal Th2 responses coinciding with a reduction in local Foxp3+RORγt+ regulatory T cells and decreased parasite fecundity. Our data suggest that nematodes sense their microbial surrounding and have evolved factors that limit the outgrowth of certain microbes. Moreover, the parasites benefit from microbiota-driven immune regulatory circuits, as an increased ratio of intestinal Th2 effector to regulatory T cells coincides with reduced parasite fitness in germ-free mice.


Subject(s)
Gastrointestinal Microbiome/immunology , Intestinal Diseases, Parasitic/immunology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , Cluster Analysis , Genes, Helminth/genetics , Host Microbial Interactions/immunology , Intestinal Diseases, Parasitic/parasitology , Intestine, Small/microbiology , Intestine, Small/parasitology , Mice, Inbred C57BL , Nematospiroides dubius/genetics , Nematospiroides dubius/physiology , Specific Pathogen-Free Organisms , Strongylida Infections/parasitology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transcriptome/immunology
18.
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
19.
Eur J Immunol ; 48(12): 1958-1964, 2018 12.
Article in English | MEDLINE | ID: mdl-30267404

ABSTRACT

T-helper type 2 (Th2) responses are central to the control of helminth infections, but sensitive to opposing cytokine signals favoring Th1 priming. We previously reported on GATA-3+ T-bet+ Th2/1 hybrid cell differentiation in helminth mono-infections, resulting in a substantial proportion of cells coproducing IFN-γ next to Th2 cytokines. Here, we demonstrate Th2/1 cells as the major source of parasite-specific IFN-γ production in acute and chronic infections with the enteric nematode Heligmosomoides polygyrus. Th2/1 cells differentiated from naive precursors and accumulated in spleen and intestine of infected mice, resulting in increased systemic and mucosal IFN-γ production. IFN-γ supplementation early during infection supported Th2/1 differentiation, associated with elevated parasite fecundity and the maintenance of high worm burdens in the chronic stage of infection, whereas mice lacking IFN-γ signals generated poor Th2/1 responses and restricted parasite fecundity more efficiently. These findings suggest that Th2/1 hybrid responses take part in immune regulation during helminth infection and restrain effective anti-helminth immunity.


Subject(s)
Interferon-gamma/metabolism , Intestines/immunology , Nematospiroides dubius/physiology , Spleen/immunology , Strongylida Infections/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Interferon-gamma/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/parasitology
20.
Nature ; 559(7712): 109-113, 2018 07.
Article in English | MEDLINE | ID: mdl-29950724

ABSTRACT

Epithelial surfaces form critical barriers to the outside world and are continuously renewed by adult stem cells1. Whereas dynamics of epithelial stem cells during homeostasis are increasingly well understood, how stem cells are redirected from a tissue-maintenance program to initiate repair after injury remains unclear. Here we examined infection by Heligmosomoides polygyrus, a co-evolved pathosymbiont of mice, to assess the epithelial response to disruption of the mucosal barrier. H. polygyrus disrupts tissue integrity by penetrating the duodenal mucosa, where it develops while surrounded by a multicellular granulomatous infiltrate2. Crypts overlying larvae-associated granulomas did not express intestinal stem cell markers, including Lgr53, in spite of continued epithelial proliferation. Granuloma-associated Lgr5- crypt epithelium activated an interferon-gamma (IFN-γ)-dependent transcriptional program, highlighted by Sca-1 expression, and IFN-γ-producing immune cells were found in granulomas. A similar epithelial response accompanied systemic activation of immune cells, intestinal irradiation, or ablation of Lgr5+ intestinal stem cells. When cultured in vitro, granuloma-associated crypt cells formed spheroids similar to those formed by fetal epithelium, and a sub-population of H. polygyrus-induced cells activated a fetal-like transcriptional program, demonstrating that adult intestinal tissues can repurpose aspects of fetal development. Therefore, re-initiation of the developmental program represents a fundamental mechanism by which the intestinal crypt can remodel itself to sustain function after injury.


Subject(s)
Fetus/cytology , Helminths/physiology , Intestines/cytology , Parasites/physiology , Stem Cell Niche , Stem Cells/cytology , Animals , Antigens, Ly/biosynthesis , Epithelial Cells/cytology , Female , Fetus/metabolism , Interferon-gamma/immunology , Male , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , Nematospiroides dubius/physiology , Receptors, G-Protein-Coupled/metabolism , Strongylida Infections/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...