Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 850
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 259-288, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38277692

ABSTRACT

Gastrointestinal nematode (GIN) infection has applied significant evolutionary pressure to the mammalian immune system and remains a global economic and human health burden. Upon infection, type 2 immune sentinels activate a common antihelminth response that mobilizes and remodels the intestinal tissue for effector function; however, there is growing appreciation of the impact GIN infection also has on the distal tissue immune state. Indeed, this effect is observed even in tissues through which GINs never transit. This review highlights how GIN infection modulates systemic immunity through (a) induction of host resistance and tolerance responses, (b) secretion of immunomodulatory products, and (c) interaction with the intestinal microbiome. It also discusses the direct consequences that changes to distal tissue immunity can have for concurrent and subsequent infection, chronic noncommunicable diseases, and vaccination efficacy.


Subject(s)
Gastrointestinal Microbiome , Nematoda , Nematode Infections , Animals , Humans , Nematode Infections/immunology , Nematoda/immunology , Nematoda/physiology , Gastrointestinal Microbiome/immunology , Immunomodulation , Host-Parasite Interactions/immunology , Intestinal Diseases, Parasitic/immunology , Immune Tolerance , Gastrointestinal Tract/immunology , Gastrointestinal Tract/parasitology
2.
Nat Immunol ; 22(2): 216-228, 2021 02.
Article in English | MEDLINE | ID: mdl-33462454

ABSTRACT

CD4+ effector lymphocytes (Teff) are traditionally classified by the cytokines they produce. To determine the states that Teff cells actually adopt in frontline tissues in vivo, we applied single-cell transcriptome and chromatin analyses to colonic Teff cells in germ-free or conventional mice or in mice after challenge with a range of phenotypically biasing microbes. Unexpected subsets were marked by the expression of the interferon (IFN) signature or myeloid-specific transcripts, but transcriptome or chromatin structure could not resolve discrete clusters fitting classic helper T cell (TH) subsets. At baseline or at different times of infection, transcripts encoding cytokines or proteins commonly used as TH markers were distributed in a polarized continuum, which was functionally validated. Clones derived from single progenitors gave rise to both IFN-γ- and interleukin (IL)-17-producing cells. Most of the transcriptional variance was tied to the infecting agent, independent of the cytokines produced, and chromatin variance primarily reflected activities of activator protein (AP)-1 and IFN-regulatory factor (IRF) transcription factor (TF) families, not the canonical subset master regulators T-bet, GATA3 or RORγ.


Subject(s)
Bacteria/pathogenicity , Bacterial Infections/microbiology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/parasitology , Colon/microbiology , Colon/parasitology , Gastrointestinal Microbiome , Heligmosomatoidea/pathogenicity , Intestinal Diseases, Parasitic/parasitology , Animals , Bacteria/immunology , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chromatin/genetics , Chromatin/metabolism , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Colon/immunology , Colon/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Heligmosomatoidea/immunology , Host-Pathogen Interactions , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Intestinal Diseases, Parasitic/genetics , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Nematospiroides dubius/immunology , Nematospiroides dubius/pathogenicity , Nippostrongylus/immunology , Nippostrongylus/pathogenicity , Phenotype , Salmonella enterica/immunology , Salmonella enterica/pathogenicity , Single-Cell Analysis , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptome
3.
PLoS Pathog ; 20(8): e1011766, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39141685

ABSTRACT

Intestinal helminth infection triggers a type 2 immune response that promotes a 'weep-and sweep' response characterised by increased mucus secretion and intestinal hypermotility, which function to dislodge the worm from its intestinal habitat. Recent studies have discovered that several other pathogens cause intestinal dysmotility through major alterations to the immune and enteric nervous systems (ENS), and their interactions, within the gastrointestinal tract. However, the involvement of these systems has not been investigated for helminth infections. Eosinophils represent a key cell type recruited by the type 2 immune response and alter intestinal motility under steady-state conditions. Our study aimed to investigate whether altered intestinal motility driven by the murine hookworm, Nippostrongylus brasiliensis, infection involves eosinophils and how the ENS and smooth muscles of the gut are impacted. Eosinophil deficiency did not influence helminth-induced intestinal hypermotility and hypermotility did not involve gross structural or functional changes to the ENS. Hypermotility was instead associated with a dramatic increase in smooth muscle thickness and contractility, an observation that extended to another rodent nematode, Heligmosomoides polygyrus. In summary our data indicate that, in contrast to other pathogens, helminth-induced intestinal hypermotility is driven by largely by myogenic, rather than neurogenic, alterations with such changes occurring independently of eosinophils. (<300 words).


Subject(s)
Enteric Nervous System , Eosinophils , Gastrointestinal Motility , Muscle, Smooth , Nippostrongylus , Animals , Mice , Eosinophils/immunology , Muscle, Smooth/parasitology , Enteric Nervous System/parasitology , Enteric Nervous System/immunology , Gastrointestinal Motility/physiology , Nematospiroides dubius/physiology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Strongylida Infections/parasitology , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Helminthiasis/immunology , Helminthiasis/parasitology , Neurons/parasitology , Neurons/metabolism , Mice, Inbred C57BL
4.
Immunol Cell Biol ; 102(5): 396-406, 2024.
Article in English | MEDLINE | ID: mdl-38648862

ABSTRACT

Increased permeability of the intestinal epithelial layer is linked to the pathogenesis and perpetuation of a wide range of intestinal and extra-intestinal diseases. Infecting humans with controlled doses of helminths, such as human hookworm (termed hookworm therapy), is proposed as a treatment for many of the same diseases. Helminths induce immunoregulatory changes in their host which could decrease epithelial permeability, which is highlighted as a potential mechanism through which helminths treat disease. Despite this, the influence of a chronic helminth infection on epithelial permeability remains unclear. This study uses the chronically infecting intestinal helminth Heligmosomoides polygyrus to reveal alterations in the expression of intestinal tight junction proteins and epithelial permeability during the infection course. In the acute infection phase (1 week postinfection), an increase in intestinal epithelial permeability is observed. Consistent with this finding, jejunal claudin-2 is upregulated and tricellulin is downregulated. By contrast, in the chronic infection phase (6 weeks postinfection), colonic claudin-1 is upregulated and epithelial permeability decreases. Importantly, this study also investigates changes in epithelial permeability in a small human cohort experimentally challenged with the human hookworm, Necator americanus. It demonstrates a trend toward small intestinal permeability increasing in the acute infection phase (8 weeks postinfection), and colonic and whole gut permeability decreasing in the chronic infection phase (24 weeks postinfection), suggesting a conserved epithelial response between humans and mice. In summary, our findings demonstrate dynamic changes in epithelial permeability during a chronic helminth infection and provide another plausible mechanism by which chronic helminth infections could be utilized to treat disease.


Subject(s)
Intestinal Mucosa , Permeability , Animals , Humans , Intestinal Mucosa/parasitology , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Chronic Disease , Nematospiroides dubius/immunology , Mice , Necator americanus , Intestinal Diseases, Parasitic/immunology , Tight Junctions/metabolism , Tight Junction Proteins/metabolism , Intestine, Small/parasitology , Intestine, Small/immunology , Female , Mice, Inbred C57BL , Male , Helminthiasis/immunology , Helminthiasis/parasitology , Necatoriasis/immunology , MARVEL Domain Containing 2 Protein/metabolism
5.
BMC Vet Res ; 20(1): 335, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068442

ABSTRACT

Fish gut is a versatile organ serving as the primary pathway for invasion by pathogens, particularly parasites, playing a crucial role in modulating the intestinal adaptive immune response. This study aimed to investigate the cellular-mediated reaction, mucosal acidity, and the expression of proliferating cell nuclear antigen (PCNA), vascular endothelial growth factor (VEGF), and CD68 in the intestines of catfish, Clarias gariepinus, naturally infected with helminths. Forty catfish were collected from the Nile River and examined for intestinal parasites. The intestinal tissues of the control and infected fish were fixed for histochemical and immunohistochemical studies. Two groups of helminths were found: cestodes Tetracampos ciliotheca and Polyonchobothrium clarias, and nematodes Paracamallanus cyathopharynx, with a prevalence rate of 63.63%, 18.0%, and 18.0%, respectively. Our results showed that the infected fish had a statistically significant rise in the activity of immune cells, including mast cells, eosinophil granular cells, and dendritic cells. This correlated with upregulation in the expressions of PCNA, VEGF, and CD68. Histochemical analyses demonstrated a marked increase in acidic mucus production, Sudan black B, and bromophenol mercury blue. This study enriches our understanding of the evolution of vertebrate immunity in combating intestinal parasitic infections and the host's adaptive responses.


Subject(s)
Catfishes , Fish Diseases , Helminthiasis, Animal , Intestinal Mucosa , Animals , Catfishes/parasitology , Fish Diseases/parasitology , Fish Diseases/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/immunology , Helminthiasis, Animal/parasitology , Immunity, Cellular , Vascular Endothelial Growth Factor A/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Intestinal Diseases, Parasitic/veterinary , Intestinal Diseases, Parasitic/parasitology , Intestinal Diseases, Parasitic/immunology , Antigens, CD/metabolism
6.
PLoS Pathog ; 17(7): e1009768, 2021 07.
Article in English | MEDLINE | ID: mdl-34329367

ABSTRACT

The intestinal nematode parasite Trichuris muris dwells in the caecum and proximal colon driving an acute resolving intestinal inflammation dominated by the presence of macrophages. Notably, these macrophages are characterised by their expression of RELMα during the resolution phase of the infection. The RELMα+ macrophage phenotype associates with the presence of alternatively activated macrophages and work in other model systems has demonstrated that the balance of classically and alternatively activated macrophages is critically important in enabling the resolution of inflammation. Moreover, in the context of type 2 immunity, RELMα+ alternatively activated macrophages are associated with the activation of macrophages via the IL4Rα. Despite a breadth of inflammatory pathologies associated with the large intestine, including those that accompany parasitic infection, it is not known how colonic macrophages are activated towards an alternatively activated phenotype. Here, we address this important knowledge gap by using Trichuris muris infection, in combination with transgenic mice (IL4Rαfl/fl.CX3CR1Cre) and IL4Rα-deficient/wild-type mixed bone marrow chimaeras. We make the unexpected finding that education of colonic macrophages towards a RELMα+, alternatively activated macrophage phenotype during T. muris infection does not require IL4Rα expression on macrophages. Further, this independence is maintained even when the mice are treated with an anti-IFNγ antibody during infection to create a strongly polarised Th2 environment. In contrast to RELMα, PD-L2 expression on macrophages post infection was dependent on IL4Rα signalling in the macrophages. These novel data sets are important, revealing a surprising cell-intrinsic IL4R alpha independence of the colonic RELMα+ alternatively activated macrophage during Trichuris muris infection.


Subject(s)
Colon/immunology , Colon/parasitology , Intestinal Diseases, Parasitic/immunology , Macrophages/immunology , Trichuriasis/immunology , Animals , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-4 Receptor alpha Subunit/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Trichuris/immunology
7.
PLoS Pathog ; 16(12): e1009121, 2020 12.
Article in English | MEDLINE | ID: mdl-33351862

ABSTRACT

Parasitic helminths are sensed by the immune system via tissue-derived alarmins that promote the initiation of the appropriate type 2 immune responses. Here we establish the nuclear alarmin cytokine IL-33 as a non-redundant trigger of specifically IL-9-driven and mast cell-mediated immunity to the intestinal parasite Strongyloides ratti. Blockade of endogenous IL-33 using a helminth-derived IL-33 inhibitor elevated intestinal parasite burdens in the context of reduced mast cell activation while stabilization of endogenous IL-33 or application of recombinant IL-33 reciprocally reduced intestinal parasite burdens and increased mast cell activation. Using gene-deficient mice, we show that application of IL-33 triggered rapid mast cell-mediated expulsion of parasites directly in the intestine, independent of the adaptive immune system, basophils, eosinophils or Gr-1+ cells but dependent on functional IL-9 receptor and innate lymphoid cells (ILC). Thereby we connect the described axis of IL-33-mediated ILC2 expansion to the rapid initiation of IL-9-mediated and mast cell-driven intestinal anti-helminth immunity.


Subject(s)
Interleukin-33/immunology , Interleukin-9/immunology , Intestinal Diseases, Parasitic/immunology , Lymphocytes/immunology , Mast Cells/immunology , Strongyloidiasis/immunology , Animals , Immunity, Innate/immunology , Intestines/immunology , Intestines/parasitology , Mice , Strongyloides ratti/immunology
8.
Infect Immun ; 88(3)2020 02 20.
Article in English | MEDLINE | ID: mdl-31843966

ABSTRACT

Infections with Trichuris trichiura are among the most common causes of intestinal parasitism in children worldwide, and the diagnosis is based on microscopic egg identification in the chronic phase of the infection. During parasitism, the adult worm of the trichurid nematode maintains its anterior region inserted in the intestinal mucosa, which causes serious damage and which may open access for gut microorganisms through the intestinal tissue. The immune-regulatory processes taking place during the evolution of the chronic infection are still not completely understood. By use of the Swiss Webster outbred mouse model, mice were infected with 200 eggs, and tolerance to the establishment of a chronic Trichuris muris infection was induced by the administration of a short pulse of dexamethasone during nematode early larval development. The infected mice presented weight loss, anemia, an imbalance of the microbiota, and intense immunological cell infiltration in the large intestine. It was found that mice have a mixed Th1/Th2/Th17 response, with differences being found among the different anatomical locations. After 45 days of infection, the parasitism induced changes in the microbiota composition and bacterial invasion of the large intestine epithelium. In addition, we describe that the excretory-secretory products from the nematode have anti-inflammatory effects on mouse macrophages cultured in vitro, suggesting that T. muris may modulate the immune response at the site of insertion of the worm inside mouse tissue. The data presented in this study suggest that the host immune state at 45 days postinfection with T. muris during the chronic phase of infection is the result of factors derived from the worm as well as alterations to the microbiota and bacterial invasion. Taken together, these results provide new information about the parasite-host-microbiota relationship and open new treatment possibilities.


Subject(s)
Gastrointestinal Microbiome/physiology , Host Microbial Interactions/immunology , Immunity, Cellular/physiology , Intestinal Diseases, Parasitic/immunology , Trichuriasis/immunology , Animals , Host-Parasite Interactions/immunology , Mice , T-Lymphocytes, Helper-Inducer/immunology , Trichuris/immunology
9.
Parasite Immunol ; 42(9): e12700, 2020 09.
Article in English | MEDLINE | ID: mdl-32027755

ABSTRACT

Intestinal helminthes induce immunosuppressive responses as well as type 2 immunity. Their suppressive properties are intended to regulate inflammatory diseases such as allergies and autoimmune diseases. This study evaluated whether helminthic infections suppress obesity, a chronic inflammatory state, using an intestinal nematode, Heligmosomoides polygyrus (Hp). Infection with Hp at the same time as feeding a high-fat diet (HFD) prevented weight gain, dyslipidaemia and glucose intolerance observed in uninfected obese mice. Immunologically, Hp infection skewed M1 macrophages to M2 macrophages and induced type 2 innate lymphoid cells in adipose tissues. The expression of interleukin (IL)-33, a potent initiator of type 2 responses, was also increased in association with uncoupled protein 1 (UCP1). To further investigate the anti-obesity effects of IL-33 in mice infected with Hp, IL-33-deficient mice were fed the HFD and infected with Hp. These mutant mice rapidly gained weight compared with wild-type mice, indicating the anti-obesity effect of IL-33. In the absence of IL-33, the rapid increase in weight was not prevented, and type 2 responses and UCP1 expression were not observed even during Hp infection. These results suggested that the suppression of obesity by Hp is dependent on IL-33.


Subject(s)
Diet, High-Fat , Interleukin-33/physiology , Intestinal Diseases, Parasitic/immunology , Nematospiroides dubius , Obesity/prevention & control , Strongylida Infections/immunology , Adipose Tissue/immunology , Animals , Immunity, Innate , Intestinal Diseases, Parasitic/complications , Lymphocytes/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Nematospiroides dubius/immunology , Obesity/immunology , Therapy with Helminths
10.
Trop Anim Health Prod ; 53(1): 16, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33216227

ABSTRACT

The aim of this study was to determine the resistance to worm infection in Santa Inês sheep by combining different sets of gastrointestinal parasite resistance indicator traits, using the k-means algorithm. Records from 221 animals reared in the Mid-North sub-region of Brazil were used. The following phenotypes were used: hematocrit (HCT); white blood cell count; red blood cell count (RBC); hemoglobin (HGB); platelets; mean corpuscular hemoglobin; mean corpuscular volume; mean corpuscular hemoglobin concentration; fecal egg count (FEC); coloration of the ocular mucosa (FAMACHA score); body condition score (BCS); withers height; and rump height. Two files with phenotypic information of animals were edited: complete, including all traits, and reduced, in which only FAMACHA score, HCT, FEC, and BCS were used. For determination of worm resistance, three groups were formed using the k-means non-hierarchical clustering by combining the traits of the complete and reduced analyses. The animals of the group in which individuals had the lowest values for FEC and FAMACHA score, as well as the highest values for HCT, RBC, HGB, and BCS were classified as resistant. In the group with opposite values for the aforementioned traits, the animals were classified as sensitive. The animals of the group with values between the other two groups were classified as moderately resistant. The results obtained in complete and reduced analyses were equivalent. Thus, it is possible to identify animals of the Santa Inês sheep breed according to their status of resistance to worm infection based on a reduced trait set.


Subject(s)
Disease Resistance/immunology , Helminthiasis, Animal/immunology , Intestinal Diseases, Parasitic/veterinary , Sheep Diseases/immunology , Animals , Body Constitution/physiology , Brazil , Helminthiasis, Animal/parasitology , Hematologic Tests/veterinary , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Sheep , Sheep Diseases/parasitology , Sheep, Domestic
11.
PLoS Pathog ; 13(9): e1006649, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28938014

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract, strongly associated with an increased risk of colorectal cancer development. Parasitic infections caused by helminths have been shown to modulate the host's immune response by releasing immunomodulatory molecules and inducing regulatory T cells (Tregs). This immunosuppressive state provoked in the host has been considered as a novel and promising approach to treat IBD patients and alleviate acute intestinal inflammation. On the contrary, specific parasite infections are well known to be directly linked to carcinogenesis. Whether a helminth infection interferes with the development of colitis-associated colon cancer (CAC) is not yet known. In the present study, we demonstrate that the treatment of mice with the intestinal helminth Heligmosomoides polygyrus at the onset of tumor progression in a mouse model of CAC does not alter tumor growth and distribution. In contrast, H. polygyrus infection in the early inflammatory phase of CAC strengthens the inflammatory response and significantly boosts tumor development. Here, H. polygyrus infection was accompanied by long-lasting alterations in the colonic immune cell compartment, with reduced frequencies of colonic CD8+ effector T cells. Moreover, H. polygyrus infection in the course of dextran sulfate sodium (DSS) mediated colitis significantly exacerbates intestinal inflammation by amplifying the release of colonic IL-6 and CXCL1. Thus, our findings indicate that the therapeutic application of helminths during CAC might have tumor-promoting effects and therefore should be well-considered.


Subject(s)
Colitis/complications , Colonic Neoplasms/etiology , Helminthiasis/complications , Intestinal Diseases, Parasitic/complications , Strongylida Infections/complications , Animals , Carcinogenesis/immunology , Disease Models, Animal , Female , Flow Cytometry , Helminthiasis/immunology , Intestinal Diseases, Parasitic/immunology , Mice , Mice, Inbred BALB C , Nematospiroides dubius , Strongylida Infections/immunology
12.
Int Immunol ; 30(3): 93-102, 2018 03 10.
Article in English | MEDLINE | ID: mdl-29346656

ABSTRACT

Helminth infection remains common in developing countries, where residents who suffer from the consequences of such infections can develop serious physical and mental disorders and often persist in the face of serious economic problems. Intestinal nematode infection induces the development of Th2-type immune responses including the B-cell IgE response; additionally, this infection induces an increase in the numbers and activation of various types of effector cells, such as mast cells, eosinophils and basophils, as well as the induction of goblet cell hyperplasia, anti-microbial peptide production and smooth-muscle contraction, all of which contribute to expel nematodes. Innate immunity is important in efforts to eliminate helminth infection; cytokines, including IL-25, IL-33 and thymic stromal lymphopoietin, which are products of epithelial cells and mast cells, induce Th2 cells and group 2 innate lymphoid cells to proliferate and produce Th2 cytokines. Nematodes also facilitate chronic infection by suppression of immune reactions through an increased number of Treg cells. Immunosuppression by parasite infection may ultimately be beneficial for the host animals; indeed, a negative correlation has been found between parasite infection and the prevalence of inflammatory disease in humans.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Host-Pathogen Interactions/immunology , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Intestines/immunology , Intestines/parasitology , Nematoda/immunology , Animals , Humans , T-Lymphocytes/immunology
13.
Parasite Immunol ; 41(4): e12618, 2019 04.
Article in English | MEDLINE | ID: mdl-30742324

ABSTRACT

Leishmania donovani exposure often results in subclinical infection in immunocompetent individuals, and the factors dictating development of visceral leishmaniasis (VL) are not known. Infection with intestinal worms skew immunity towards type 2 and regulatory responses, thereby theoretically increases susceptibility to intracellular infections controlled by type 1 responses. Here we have tested how chronic infection with the intestinal nematode Heligmosomoides polygyrus affected immunity to a secondary infection with L donovani. We found that mice infected with H polygyrus displayed higher Leishmania burden in liver and spleen compared to worm-free animals. This increased infectious load was accompanied by reduced leucocyte infiltration and nos2 transcription in livers and increased il4 and il10 transcription in spleens. Collectively, these data show that chronic infection with intestinal nematodes skew immune responses in a way that may favour development of VL.


Subject(s)
Helminthiasis/immunology , Intestinal Diseases, Parasitic/immunology , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , Nematospiroides dubius/immunology , Strongylida Infections/immunology , Animals , Coinfection/immunology , Female , Helminthiasis/parasitology , Interleukin-10/metabolism , Interleukin-4/metabolism , Intestinal Diseases, Parasitic/parasitology , Mice , Nitric Oxide Synthase Type II/metabolism , Parasite Load , Spleen , Strongylida Infections/parasitology
14.
J Immunol ; 199(1): 244-252, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28533444

ABSTRACT

IgE production plays a crucial role in protective as well as pathogenic type 2 immune responses. Although the cytokine IL-4 is required for the development of IgE-producing plasma cells, the source of IL-4 and cellular requirements for optimal IgE responses remain unclear. Recent evidence suggests that T follicular helper (Tfh) cells are the primary producer of IL-4 in the reactive lymph node during type 2 immune responses. As Tfh cells are also required for the development of plasmablasts derived from germinal center and extrafollicular sources, we hypothesized that this cell subset is essential for the IgE plasmablast response. In this study, we show that during intestinal helminth infection, IL-4 derived from Tfh cells is required for IgE class switching and plasmablast formation. Notably, early IgE class switching did not require germinal center formation. Additionally, Tfh cell-derived IL-4 was required to maintain the Th2 response in the mesenteric lymph nodes of infected mice. Collectively, our results indicate that IL-4-producing Tfh cells are central orchestrators of the type 2 immune response in the reactive lymph nodes during parasitic helminth infection.


Subject(s)
Helminthiasis/immunology , Immunoglobulin E/biosynthesis , Interleukin-4/immunology , Intestinal Diseases, Parasitic/immunology , Nematospiroides dubius , Strongylida Infections/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , Cell Differentiation , Germinal Center/cytology , Germinal Center/immunology , Helminthiasis/parasitology , Immunoglobulin Class Switching , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Intestinal Diseases, Parasitic/parasitology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Plasma Cells/immunology , T-Lymphocyte Subsets
15.
J Immunol ; 199(12): 3952-3958, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29109124

ABSTRACT

The vitamin D receptor participates in the control of IgE class-switch recombination in B cells. The physiologic vitamin D receptor agonist, 1,25(OH)2D3 (calcitriol), is synthesized by the essential enzyme 25-hydroxyvitamin D3-1α-hydroxylase (CYP27B1), which can be expressed by activated immune cells. The role of endogenous calcitriol synthesis for the regulation of IgE has not been proven. In this study, we investigated IgE-responses in Cyp27b1-knockout (KO) mice following sensitization to OVA or intestinal infection with Heligmosomoides polygyrus Specific Igs and plasmablasts were determined by ELISA and ELISpot, Cyp27b1 expression was measured by quantitative PCR. The data show elevated specific IgE and IgG1 concentrations in the blood of OVA-sensitized Cyp27b1-KO mice compared with wild-type littermates (+898 and +219%). Accordingly, more OVA-specific IgG1-secreting cells are present in spleen and fewer in the bone marrow of Cyp27b1-KO mice. Ag-specific mechanisms are suggested as the leucopoiesis is in general unchanged and activated murine B and T lymphocytes express Cyp27b1 Accordingly, elevated specific IgE concentrations in the blood of sensitized T cell-specific Cyp27b1-KO mice support a lymphocyte-driven mechanism. In an independent IgE-inducing model, i.e., intestinal infection with H. polygyrus, we validated the increase of total and specific IgE concentrations of Cyp27b1-KO compared with wild-type mice, but not those of IgG1 or IgA. We conclude that endogenous calcitriol has an impact on the regulation of IgE in vivo. Our data provide genetic evidence supporting previous preclinical and clinical findings and suggest that vitamin D deficiency not only promotes bone diseases but also type I sensitization.


Subject(s)
25-Hydroxyvitamin D3 1-alpha-Hydroxylase/physiology , Calcitriol/immunology , Immunoglobulin Class Switching , Immunoglobulin E/blood , 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/deficiency , Animals , B-Lymphocytes/immunology , Bone Marrow/immunology , Calcitriol/biosynthesis , Calcitriol/deficiency , Female , Helminthiasis, Animal/immunology , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Intestinal Diseases, Parasitic/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , Organ Specificity , Ovalbumin/immunology , Receptors, Calcitriol/physiology , Spleen/immunology , T-Lymphocytes/immunology , Vitamin D Deficiency/immunology
16.
Parasitology ; 146(11): 1371-1378, 2019 09.
Article in English | MEDLINE | ID: mdl-31258097

ABSTRACT

The multifaceted interactions occurring between gastrointestinal (GI) parasitic helminths and the host gut microbiota are emerging as a key area of study within the broader research domain of host-pathogen relationships. Over the past few years, a wealth of investigations has demonstrated that GI helminths interact with the host gut flora, and that such interactions result in modifications of the host immune and metabolic statuses. Nevertheless, whilst selected changes in gut microbial composition are consistently observed in response to GI helminth infections across several host-parasite systems, research in this area to date is largely characterised by inconsistent findings. These discrepancies are particularly evident when data from studies of GI helminth-microbiota interactions conducted in humans from parasite-endemic regions are compared. In this review, we provide an overview of the main sources of variance that affect investigations on helminth-gut microbiota interactions in humans, and propose a series of methodological approaches that, whilst accounting for the inevitable constraints of fieldwork, are aimed at minimising confounding factors and draw biologically meaningful interpretations from highly variable datasets.


Subject(s)
Gastrointestinal Microbiome , Helminthiasis , Intestinal Diseases, Parasitic , Gastrointestinal Microbiome/immunology , Helminthiasis/immunology , Helminthiasis/metabolism , Humans , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/metabolism
17.
Parasitology ; 146(1): 89-96, 2019 01.
Article in English | MEDLINE | ID: mdl-30086804

ABSTRACT

Antibodies at gastrointestinal mucosal membranes play a vital role in immunological protection against a range of pathogens, including helminths. Gastrointestinal health is central to efficient livestock production, and such infections cause significant losses. Fecal samples were taken from 114 cattle, across three beef farms, with matched blood samples taken from 22 of those animals. To achieve fecal antibody detection, a novel fecal supernatant was extracted. Fecal supernatant and serum samples were then analysed, using adapted enzyme-linked immunosorbent assay protocols, for levels of total immunoglobulin (Ig)A, IgG, IgM, and Teladorsagia circumcincta-specific IgA, IgG, IgM and IgE (in the absence of reagents for cattle-specific nematode species). Fecal nematode egg counts were conducted on all fecal samples. Assays performed successfully and showed that IgA was the predominant antibody in fecal samples, whereas IgG was predominant in serum. Total IgA in feces and serum correlated within individuals (0.581, P = 0.005), but other Ig types did not. Results support the hypothesis that the tested protocols are an effective method for the non-invasive assessment of cattle immunology. The method could be used as part of animal health assessments, although further work is required to interpret the relationship between results and levels of infection and immunity.


Subject(s)
Antibodies, Helminth/analysis , Cattle Diseases/parasitology , Gastrointestinal Diseases/veterinary , Intestinal Diseases, Parasitic/veterinary , Nematode Infections/veterinary , Animals , Cattle , Cattle Diseases/immunology , Farms , Feces/parasitology , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/parasitology , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Nematode Infections/immunology , Nematode Infections/parasitology , Red Meat , United Kingdom
18.
Proc Natl Acad Sci U S A ; 113(11): 2970-5, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26884194

ABSTRACT

Global climate change is predicted to alter the distribution and dynamics of soil-transmitted helminth infections, and yet host immunity can also influence the impact of warming on host-parasite interactions and mitigate the long-term effects. We used time-series data from two helminth species of a natural herbivore and investigated the contribution of climate change and immunity on the long-term and seasonal dynamics of infection. We provide evidence that climate warming increases the availability of infective stages of both helminth species and the proportional increase in the intensity of infection for the helminth not regulated by immunity. In contrast, there is no significant long-term positive trend in the intensity for the immune-controlled helminth, as immunity reduces the net outcome of climate on parasite dynamics. Even so, hosts experienced higher infections of this helminth at an earlier age during critical months in the warmer years. Immunity can alleviate the expected long-term effect of climate on parasite infections but can also shift the seasonal peak of infection toward the younger individuals.


Subject(s)
Global Warming , Helminthiasis, Animal/immunology , Host-Parasite Interactions/immunology , Rabbits/parasitology , Aging/immunology , Animal Distribution , Animals , Helminthiasis, Animal/epidemiology , Helminthiasis, Animal/parasitology , Helminthiasis, Animal/transmission , Humidity , Intestinal Diseases, Parasitic/epidemiology , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Intestinal Diseases, Parasitic/veterinary , Intestine, Small/immunology , Intestine, Small/parasitology , Larva/physiology , Life Cycle Stages , Ovum/physiology , Population Dynamics , Rabbits/immunology , Scotland/epidemiology , Seasons , Soil/parasitology , Stomach/immunology , Stomach/parasitology , Stomach Diseases/epidemiology , Stomach Diseases/immunology , Stomach Diseases/parasitology , Stomach Diseases/veterinary , Temperature , Trichostrongyloidea/growth & development , Trichostrongyloidea/physiology , Trichostrongyloidiasis/epidemiology , Trichostrongyloidiasis/immunology , Trichostrongyloidiasis/parasitology , Trichostrongyloidiasis/transmission , Trichostrongyloidiasis/veterinary , Trichostrongylosis/epidemiology , Trichostrongylosis/immunology , Trichostrongylosis/parasitology , Trichostrongylosis/transmission , Trichostrongylosis/veterinary , Trichostrongylus/growth & development , Trichostrongylus/physiology
19.
Cytokine ; 108: 179-181, 2018 08.
Article in English | MEDLINE | ID: mdl-29684754

ABSTRACT

IL-21 is a much studied cytokine that has been implicated in the regulation of TH1, TH2, TH17 and regulatory immune responses; its signalling is a promising therapeutic target for autoimmune, inflammatory and infectious diseases. Despite its biological importance, measuring IL-21 reliably has proved difficult. ELISAs are commonly used to measure cytokines in various biological samples. However, results obtained are only as good as the quality of the sample. Here, we show that when using fresh samples, a significant increase in IL-21 was measured in the intestinal homogenate of mice infected with the intestinal worm Heligmosomoides polygyrus. This difference disappeared when samples were frozen in either liquid nitrogen for two days or at -80 °C for three weeks, with levels in both naïve and infected animals decreasing. This was not observed for the IL-13 cytokine, where freezing had no impact on levels measured. Our study highlights the importance of sample storage to measuring biomarkers. Since modulating IL-21 signalling is such an important potential therapeutic avenue, accurately measuring the levels of this cytokine is key to assessing its role in various research models and clinical settings.


Subject(s)
Freezing , Helminthiasis/immunology , Interleukins/analysis , Intestinal Diseases, Parasitic/immunology , Specimen Handling/methods , Tissue Extracts/analysis , Animals , Biomarkers/analysis , Female , Intestines/immunology , Intestines/parasitology , Mice , Mice, Inbred C57BL , Nematospiroides dubius
20.
Immunity ; 30(1): 12-20, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19144314

ABSTRACT

The role of basophils, the rarest of blood granulocytes, in host immunity has been a mystery. Long considered the poor relative of mast cells, basophils have received much recent attention because of the availability of new reagents and models that reveal unique properties of these cells. Basophils are known to have distinct roles in allergic hypersensitivity reactions and in the immune response to intestinal helminthes. In this review, we highlight these advances and summarize our current understanding of the repertoire of functions attributed to these cells. Despite these recent insights, we are likely only beginning to gain a full understanding of how and where these cells lend effector functions to vertebrate immunity. Advances are likely to come only with the development of specific reagents that enable the finer study of basophil lineage and function. Although many fundamental aspects of basophil biology remain unanswered, the prospects remain bright for unmasking new contributions by these unusual cells.


Subject(s)
Basophils/immunology , Host-Parasite Interactions/immunology , Immunity , Intestinal Diseases, Parasitic/immunology , Animals , Helminthiasis/immunology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL