44 results on '"Harcus Y"'
Search Results
2. Low-level regulatory T-cell activity is essential for functional type-2 effector immunity to expel gastrointestinal helminths
- Author
-
Smith, K A, Filbey, K J, Reynolds, L A, Hewitson, J P, Harcus, Y, Boon, L, Sparwasser, T, Hämmerling, G, and Maizels, R M
- Published
- 2016
- Full Text
- View/download PDF
3. Differential recognition patterns of Schistosoma haematobium adult worm antigens by the human antibodies IgA, IgE, IgG1 and IgG4
- Author
-
MUTAPI, F., BOURKE, C., HARCUS, Y., MIDZI, N., MDULUZA, T., TURNER, C. M., BURCHMORE, R., and MAIZELS, R. M.
- Published
- 2011
- Full Text
- View/download PDF
4. daf-7-related TGF-β homologues from Trichostrongyloid nematodes show contrasting life-cycle expression patterns
- Author
-
McSORLEY, H. J., GRAINGER, J. R., HARCUS, Y., MURRAY, J., NISBET, A. J., KNOX, D. P., and MAIZELS, R. M.
- Published
- 2010
5. Development of type 2 immune responses in the gastrointestinal environment: or why do GI nematodes drive Th2 responses?: 7.26
- Author
-
Balic, A., Harcus, Y., and Maizels, R. M.
- Published
- 2005
6. Early Th2 differentiation and development induced by excretory/secretory products of Nippostrongylus brasiliensis: 12.31
- Author
-
Balic, A., Harcus, Y. M., and Maizels, R. M.
- Published
- 2004
7. Inhibition of neutrophil recruitment by ES of Nippostrongylus brasiliensis
- Author
-
KEIR, P. A., BROWN, D. M., CLOUTER-BAKER, A., HARCUS, Y. M., and PROUDFOOT, L.
- Published
- 2004
8. SECRETORY/EXCRETORY ANTIGENS OF NIPPOSTRONGYLUS BRASILIENSIS DRIVE TH2 RESPONSES AND DEMONSTRATE SELF ADJUVANTICITY.
- Author
-
Holland, M. J., Harcus, Y. M., and Maizels, R. M.
- Published
- 1999
9. T-HELPER CELL CYTOKINE BIAS INDUCED BY EXCRETORY/SECRETORY (E/S) ANTIGENS OF NIPPOSTRONGYLUS BRASILIENSIS
- Author
-
Holland, M. J., Harcus, Y. M., and Maizels, R. M.
- Published
- 1998
10. A structurally distinct TGF-β mimic from an intestinal helminth parasite potently induces regulatory T cells
- Author
-
Johnston, CJC, Smyth, DJ, Kodali, RB, White, MPJ, Harcus, Y, Filbey, KJ, Hewitson, JP, Hinck, CS, Ivens, A, Kemter, AM, Kildemoes, AO, Le, Bihan T, Soares, DC, and Maizels, RM
- Abstract
Helminth parasites defy immune exclusion through sophisticated evasion mechanisms, including activation of host immunosuppressive regulatory T (Treg) cells. The mouse parasite Heligmosomoides polygyrus can expand the host Treg population by secreting products that activate TGF-β signalling, but the identity of the active molecule is unknown. Here we identify an H. polygyrus TGF-β mimic (Hp-TGM) that replicates the biological and functional properties of TGF-β, including binding to mammalian TGF-β receptors and inducing mouse and human Foxp3+ Treg cells. Hp-TGM has no homology with mammalian TGF-β or other members of the TGF-β family, but is a member of the complement control protein superfamily. Thus, our data indicate that through convergent evolution, the parasite has acquired a protein with cytokine-like function that is able to exploit an endogenous pathway of immunoregulation in the host.
- Published
- 2017
11. Innate and adaptive type 2 immune cell responses in genetically controlled resistance to intestinal helminth infection
- Author
-
Filbey, K.J., Grainger, J.R., Smith, K.A., Boon, L., van Rooijen, N., Harcus, Y., Jenkins, S., Hewitson, J.P., Maizels, R.M., Molecular cell biology and Immunology, and CCA - Immuno-pathogenesis
- Published
- 2014
12. daf-7-related TGF-β homologues from Trichostrongyloid nematodes show contrasting life-cycle expression patterns
- Author
-
McSORLEY, H. J., primary, GRAINGER, J. R., additional, HARCUS, Y., additional, MURRAY, J., additional, NISBET, A. J., additional, KNOX, D. P., additional, and MAIZELS, R. M., additional
- Published
- 2009
- Full Text
- View/download PDF
13. Signal sequence analysis of expressed sequence tags from the nematode Nippostrongylus brasiliensis and the evolution of secreted proteins in parasites
- Author
-
Harcus, Y. M., Parkinson, J., Fernández, C., Jennifer Daub, Selkirk, M. E., Blaxter, M. L., and Maizels, R. M.
- Subjects
GENES ,ANTIBODY-RESPONSE ,Bioinformatics ,05 Environmental Sciences ,Protein Sorting Signals ,ACETYLCHOLINESTERASE ,Trans-Splicing ,Evolution, Molecular ,Sequence Analysis, Protein ,Sequence Homology, Nucleic Acid ,Animals ,Parasites ,Selection, Genetic ,GENETICS & HEREDITY ,Caenorhabditis elegans Proteins ,Conserved Sequence ,Expressed Sequence Tags ,08 Information And Computing Sciences ,Science & Technology ,CDNA LIBRARIES ,INFECTIVE LARVAL STAGE ,Research ,BIOTECHNOLOGY & APPLIED MICROBIOLOGY ,HELMINTH-PARASITES ,CYSTEINE PROTEASE ,Helminth Proteins ,06 Biological Sciences ,MIGRATION INHIBITORY FACTOR ,PLATELET-ACTIVATING-FACTOR ,CLEAVAGE SITES ,Nippostrongylus ,Life Sciences & Biomedicine - Abstract
Analysis of secreted proteins indicate that they may be undergoing accelerated evolution, either because of relaxed functional constraints, or in response to stronger selective pressure from host immunity., Background Parasitism is a highly successful mode of life and one that requires suites of gene adaptations to permit survival within a potentially hostile host. Among such adaptations is the secretion of proteins capable of modifying or manipulating the host environment. Nippostrongylus brasiliensis is a well-studied model nematode parasite of rodents, which secretes products known to modulate host immunity. Results Taking a genomic approach to characterize potential secreted products, we analyzed expressed sequence tag (EST) sequences for putative amino-terminal secretory signals. We sequenced ESTs from a cDNA library constructed by oligo-capping to select full-length cDNAs, as well as from conventional cDNA libraries. SignalP analysis was applied to predicted open reading frames, to identify potential signal peptides and anchors. Among 1,234 ESTs, 197 (~16%) contain predicted 5' signal sequences, with 176 classified as conventional signal peptides and 21 as signal anchors. ESTs cluster into 742 distinct genes, of which 135 (18%) bear predicted signal-sequence coding regions. Comparisons of clusters with homologs from Caenorhabditis elegans and more distantly related organisms reveal that the majority (65% at P < e-10) of signal peptide-bearing sequences from N. brasiliensis show no similarity to previously reported genes, and less than 10% align to conserved genes recorded outside the phylum Nematoda. Of all novel sequences identified, 32% contained predicted signal peptides, whereas this was the case for only 3.4% of conserved genes with sequence homologies beyond the Nematoda. Conclusions These results indicate that secreted proteins may be undergoing accelerated evolution, either because of relaxed functional constraints, or in response to stronger selective pressure from host immunity.
- Published
- 2004
14. Praziquantrel treatment of individuals exposed to Schistosoma haematobium enhances serological recognition of defined antigens.
- Author
-
Mutapi F, Burchmore R, Mduluza T, Foucher A, Harcus Y, Nicoll G, Midzi N, Turner CM, and Maizels RM
- Abstract
BACKGROUND: Schistosomiasis is a major parasitic disease affecting >200 million people in the developing world, and 400 million people are at risk for infection. This study aimed to identify and compare proteins recognized by serum samples from schistosome-exposed individuals before and after curative praziquantel treatment. METHODS: Proteins recognized by pooled serum samples from Schistosoma haematobium-exposed Zimbabweans were determined by 2-dimensional Western blotting and identified by mass spectrometry. RESULTS: Serum samples recognized 71 spots, which resolved to 26 different characterized proteins. Eleven of these proteins have not previously been shown to be immunogenic in natural human infection or in experimental models of schistosomiasis, making them novel antigens in the parasite. Pretreatment serum samples recognized 59 spots, which resolved to 21 different identified proteins. Posttreatment serum samples recognized an additional 12 spots, which resolved to 8 different identified proteins. Of these 8 proteins, 3 had putative isoforms recognized before treatment, and 5 (calreticulin, tropomyosin 1, tropomyosin 2, paramyosin, and triose phosphate isomerase) did not. CONCLUSIONS: This study is the most comprehensive characterization of S. haematobium antigens to date and describes novel antigens in all schistosome species. Posttreatment results are consistent with praziquantel treatment inducing quantitative and qualitative changes in schistosome-specific antibody responses. Copyright © 2005 Infectious Diseases Society of America [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
15. Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway
- Author
-
Grainger, Smith, KA, Hewitson, JP, McSorley, HJ, Harcus, Y, Filbey, KJ, Finney, CAM, Greenwood, EJD, Knox, DP, Wilson, MS, Belkaid, Y, Rudensky, AY, and Maizels, RM
- Subjects
chemical and pharmacologic phenomena ,Mice, Transgenic ,Dioxoles ,Smad2 Protein ,Protein Serine-Threonine Kinases ,T-Lymphocytes, Regulatory ,Host-Parasite Interactions ,Mice ,Th2 Cells ,Transforming Growth Factor beta ,parasitic diseases ,Animals ,Smad3 Protein ,Phosphorylation ,Cell Proliferation ,Strongylida Infections ,Mice, Inbred BALB C ,Nematospiroides dubius ,Receptor, Transforming Growth Factor-beta Type II ,hemic and immune systems ,Forkhead Transcription Factors ,Th1 Cells ,3. Good health ,Gene Expression Regulation ,Antigens, Helminth ,Benzamides ,Chronic Disease ,biological phenomena, cell phenomena, and immunity ,Receptors, Transforming Growth Factor beta ,Signal Transduction - Abstract
Foxp3-expressing regulatory T (T reg) cells have been implicated in parasite-driven inhibition of host immunity during chronic infection. We addressed whether parasites can directly induce T reg cells. Foxp3 expression was stimulated in naive Foxp3⁻ T cells in mice infected with the intestinal helminth Heligmosomoides polygyrus. In vitro, parasite-secreted proteins (termed H. polygyrus excretory-secretory antigen [HES]) induced de novo Foxp3 expression in fluorescence-sorted Foxp3⁻ splenocytes from Foxp3-green fluorescent protein reporter mice. HES-induced T reg cells suppressed both in vitro effector cell proliferation and in vivo allergic airway inflammation. HES ligated the transforming growth factor (TGF) β receptor and promoted Smad2/3 phosphorylation. Foxp3 induction by HES was lost in dominant-negative TGF-βRII cells and was abolished by the TGF-β signaling inhibitor SB431542. This inhibitor also reduced worm burdens in H. polygyrus-infected mice. HES induced IL-17 in the presence of IL-6 but did not promote Th1 or Th2 development under any conditions. Importantly, antibody to mammalian TGF-β did not recognize HES, whereas antisera that inhibited HES did not affect TGF-β. Foxp3 was also induced by secreted products of Teladorsagia circumcincta, a related nematode which is widespread in ruminant animals. We have therefore identified a novel pathway through which helminth parasites may stimulate T reg cells, which is likely to be a key part of the parasite's immunological relationship with the host.
16. The IL-25-dependent tuft cell circuit driven by intestinal helminths requires macrophage migration inhibitory factor (MIF).
- Author
-
Varyani F, Löser S, Filbey KJ, Harcus Y, Drurey C, Poveda MC, Rasid O, White MPJ, Smyth DJ, Gerbe F, Jay P, and Maizels RM
- Subjects
- Mice, Animals, Immunity, Innate, Lymphocytes, Nippostrongylus, Macrophage Migration-Inhibitory Factors genetics, Strongylida Infections
- Abstract
Macrophage migration inhibitory factor (MIF) is a key innate immune mediator with chemokine- and cytokine-like properties in the inflammatory pathway. While its actions on macrophages are well-studied, its effects on other cell types are less understood. Here we report that MIF is required for expansion of intestinal tuft cells during infection with the helminth Nippostrongylus brasiliensis. MIF-deficient mice show defective innate responses following infection, lacking intestinal epithelial tuft cell hyperplasia or upregulation of goblet cell RELMβ, and fail to expand eosinophil, type 2 innate lymphoid cell (ILC2) and macrophage (M2) populations. Similar effects were observed in MIF-sufficient wild-type mice given the MIF inhibitor 4-IPP. MIF had no direct effect on epithelial cells in organoid cultures, and MIF-deficient intestinal stem cells could generate tuft cells in vitro in the presence of type 2 cytokines. In vivo the lack of MIF could be fully compensated by administration of IL-25, restoring tuft cell differentiation and goblet cell expression of RELM-β, demonstrating its requirement upstream of the ILC2-tuft cell circuit. Both ILC2s and macrophages expressed the MIF receptor CXCR4, indicating that MIF may act as an essential co-factor on both cell types to activate responses to IL-25 in helminth infection., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
17. Characterisation of the secreted apyrase family of Heligmosomoides polygyrus.
- Author
-
Berkachy R, Smyth DJ, Schnoeller C, Harcus Y, Maizels RM, Selkirk ME, and Gounaris K
- Subjects
- Animals, Apyrase genetics, Mice, Saccharomycetales, Intestinal Diseases, Parasitic, Nematospiroides dubius
- Abstract
Apyrases are a recurrent feature of secretomes from numerous species of parasitic nematodes. Here we characterise the five apyrases secreted by Heligmosomoides polygyrus, a natural parasite of mice and a widely used laboratory model for intestinal nematode infection. All five enzymes are closely related to soluble calcium-activated nucleotidases described in a variety of organisms, and distinct from the CD39 family of ecto-nucleotidases. Expression is maximal in adult worms and restricted to adults and L4s. Recombinant apyrases were produced and purified from Pichia pastoris. The five enzymes showed very similar biochemical properties, with strict calcium dependence and a broad substrate specificity, catalysing the hydrolysis of all nucleoside tri- and diphosphates, with no activity against nucleoside monophosphates. Natural infection of mice provoked very low antibodies to any enzyme, but immunisation with an apyrase cocktail showed partial protection against reinfection, with reduced egg output and parasite recovery. The most likely role for nematode secreted apyrases is hydrolysis of extracellular ATP, which acts as an alarmin for cellular release of IL-33 and initiation of type 2 immunity., (Copyright © 2020 Australian Society for Parasitology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. Macrophage Migration Inhibitory Factor (MIF) Is Essential for Type 2 Effector Cell Immunity to an Intestinal Helminth Parasite.
- Author
-
Filbey KJ, Varyani F, Harcus Y, Hewitson JP, Smyth DJ, McSorley HJ, Ivens A, Nylén S, Rottenberg M, Löser S, and Maizels RM
- Subjects
- 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, Immunity, Cellular, Intramolecular Oxidoreductases immunology, Macrophage Activation, Macrophage Migration-Inhibitory Factors immunology, Macrophages immunology, Nematospiroides dubius immunology, Strongylida Infections immunology, T-Lymphocytes, Regulatory immunology
- 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 (LysMCre xSTAT3fl/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., (Copyright © 2019 Filbey, Varyani, Harcus, Hewitson, Smyth, McSorley, Ivens, Nylén, Rottenberg, Löser and Maizels.)- Published
- 2019
- Full Text
- View/download PDF
19. Concerted IL-25R and IL-4Rα signaling drive innate type 2 effector immunity for optimal helminth expulsion.
- Author
-
Smith KA, Löser S, Varyani F, Harcus Y, McSorley HJ, McKenzie AN, and Maizels RM
- Subjects
- 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, Immunity, Innate immunology, Nematospiroides dubius immunology, Receptors, Cell Surface immunology, Receptors, Interleukin-17 immunology, Strongylida Infections immunology, Th2 Cells immunology
- 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., Competing Interests: KS, SL, FV, YH, HM, AM, RM No competing interests declared, (© 2018, Smith et al.)
- Published
- 2018
- Full Text
- View/download PDF
20. TGF-β mimic proteins form an extended gene family in the murine parasite Heligmosomoides polygyrus.
- Author
-
Smyth DJ, Harcus Y, White MPJ, Gregory WF, Nahler J, Stephens I, Toke-Bjolgerud E, Hewitson JP, Ivens A, McSorley HJ, and Maizels RM
- Subjects
- Animals, Cloning, Molecular, Forkhead Transcription Factors metabolism, Helminth Proteins genetics, Mice, Spleen cytology, Transforming Growth Factor beta genetics, Gene Expression Regulation physiology, Helminth Proteins chemistry, Helminth Proteins metabolism, Nematospiroides dubius metabolism, Transforming Growth Factor beta metabolism
- Abstract
We recently reported the discovery of a new parasite-derived protein that functionally mimics the immunosuppressive cytokine transforming growth factor (TGF)-β. The Heligmosomoides polygyrus TGF-β Mimic (Hp-TGM) shares no homology to any TGF-β family member, however it binds the mammalian TGF-β receptor and induces expression of Foxp3, the canonical transcription factor of both mouse and human regulatory T cells. Hp-TGM consists of five atypical Complement Control Protein (CCP, Pfam 00084) domains, each lacking certain conserved residues and 12-15 amino acids longer than the 60-70 amino acids consensus domain, but with a recognizable 3-cysteine, tryptophan, cysteine motif. We now report on the identification of a family of nine related Hp-TGM homologues represented in the secreted proteome and transcriptome of H. polygyrus. Recombinant proteins from five of the nine new TGM members were tested for TGF-β activity, but only two were functionally active in an MFB-F11 reporter assay, and by the induction of T cell Foxp3 expression. Sequence comparisons reveal that proteins with functional activity are similar or identical to Hp-TGM across the first three CCP domains, but more variable in domains 4 and 5. Inactive proteins diverged in all domains, or lacked some domains entirely. Testing truncated versions of Hp-TGM confirmed that domains 1-3 are essential for full activity in vitro, while domains 4 and 5 are not required. Further studies will elucidate whether these latter domains fulfill other functions in promoting host immune regulation during infection and if the more divergent family members play other roles in immunomodulation., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
21. A structurally distinct TGF-β mimic from an intestinal helminth parasite potently induces regulatory T cells.
- Author
-
Johnston CJC, Smyth DJ, Kodali RB, White MPJ, Harcus Y, Filbey KJ, Hewitson JP, Hinck CS, Ivens A, Kemter AM, Kildemoes AO, Le Bihan T, Soares DC, Anderton SM, Brenn T, Wigmore SJ, Woodcock HV, Chambers RC, Hinck AP, McSorley HJ, and Maizels RM
- Subjects
- Amino Acid Sequence, Animals, Antigens, Helminth chemistry, Antigens, Helminth genetics, Antigens, Helminth immunology, Female, Helminth Proteins chemistry, Helminth Proteins genetics, Helminth Proteins immunology, Host-Pathogen Interactions genetics, Host-Pathogen Interactions immunology, Humans, Immune Evasion immunology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Molecular Mimicry genetics, Nematospiroides dubius genetics, Protein Binding, Protein Domains, Receptors, Transforming Growth Factor beta metabolism, Strongylida Infections immunology, Strongylida Infections parasitology, Molecular Mimicry immunology, Nematospiroides dubius immunology, Nematospiroides dubius pathogenicity, T-Lymphocytes, Regulatory immunology, Transforming Growth Factor beta metabolism
- Abstract
Helminth parasites defy immune exclusion through sophisticated evasion mechanisms, including activation of host immunosuppressive regulatory T (Treg) cells. The mouse parasite Heligmosomoides polygyrus can expand the host Treg population by secreting products that activate TGF-β signalling, but the identity of the active molecule is unknown. Here we identify an H. polygyrus TGF-β mimic (Hp-TGM) that replicates the biological and functional properties of TGF-β, including binding to mammalian TGF-β receptors and inducing mouse and human Foxp3
+ Treg cells. Hp-TGM has no homology with mammalian TGF-β or other members of the TGF-β family, but is a member of the complement control protein superfamily. Thus, our data indicate that through convergent evolution, the parasite has acquired a protein with cytokine-like function that is able to exploit an endogenous pathway of immunoregulation in the host.- Published
- 2017
- Full Text
- View/download PDF
22. Extracellular Vesicles from a Helminth Parasite Suppress Macrophage Activation and Constitute an Effective Vaccine for Protective Immunity.
- Author
-
Coakley G, McCaskill JL, Borger JG, Simbari F, Robertson E, Millar M, Harcus Y, McSorley HJ, Maizels RM, and Buck AH
- Subjects
- Animals, Antibodies, Helminth immunology, Antibody Formation drug effects, Bone Marrow Cells cytology, Cytochalasin D pharmacology, Extracellular Vesicles drug effects, Interleukin-1 Receptor-Like 1 Protein, Macrophages drug effects, Macrophages metabolism, Mice, Parasites drug effects, Receptors, Interleukin metabolism, Vaccination, Extracellular Vesicles metabolism, Immunity drug effects, Macrophage Activation drug effects, Nematospiroides dubius metabolism, Parasites metabolism, Vaccines immunology
- Abstract
Recent studies have demonstrated that many parasites release extracellular vesicles (EVs), yet little is known about the specific interactions of EVs with immune cells or their functions during infection. We show that EVs secreted by the gastrointestinal nematode Heligmosomoides polygyrus are internalized by macrophages and modulate their activation. EV internalization causes downregulation of type 1 and type 2 immune-response-associated molecules (IL-6 and TNF, and Ym1 and RELMα) and inhibits expression of the IL-33 receptor subunit ST2. Co-incubation with EV antibodies abrogated suppression of alternative activation and was associated with increased co-localization of the EVs with lysosomes. Furthermore, mice vaccinated with EV-alum generated protective immunity against larval challenge, highlighting an important role in vivo. In contrast, ST2-deficient mice are highly susceptible to infection, and they are unable to clear parasites following EV vaccination. Hence, macrophage activation and the IL-33 pathway are targeted by H. polygyrus EVs, while neutralization of EV function facilitates parasite expulsion., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
23. Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites.
- Author
-
Gerbe F, Sidot E, Smyth DJ, Ohmoto M, Matsumoto I, Dardalhon V, Cesses P, Garnier L, Pouzolles M, Brulin B, Bruschi M, Harcus Y, Zimmermann VS, Taylor N, Maizels RM, and Jay P
- Subjects
- Animals, Cell Lineage, Cell Proliferation, Feedback, Physiological, Female, Goblet Cells cytology, Goblet Cells immunology, Interleukin-13 immunology, Interleukin-17 immunology, Interleukin-17 metabolism, Intestinal Mucosa metabolism, Male, Mice, Octamer Transcription Factors deficiency, Receptors, Interleukin-4 immunology, Signal Transduction immunology, Stem Cells cytology, Stem Cells immunology, Strongylida Infections immunology, Th2 Cells cytology, Th2 Cells immunology, Immunity, Mucosal immunology, Intestinal Mucosa cytology, Intestinal Mucosa immunology, Nippostrongylus immunology, Parasites immunology
- Abstract
Helminth parasitic infections are a major global health and social burden. The host defence against helminths such as Nippostrongylus brasiliensis is orchestrated by type 2 cell-mediated immunity. Induction of type 2 cytokines, including interleukins (IL) IL-4 and IL-13, induce goblet cell hyperplasia with mucus production, ultimately resulting in worm expulsion. However, the mechanisms underlying the initiation of type 2 responses remain incompletely understood. Here we show that tuft cells, a rare epithelial cell type in the steady-state intestinal epithelium, are responsible for initiating type 2 responses to parasites by a cytokine-mediated cellular relay. Tuft cells have a Th2-related gene expression signature and we demonstrate that they undergo a rapid and extensive IL-4Rα-dependent amplification following infection with helminth parasites, owing to direct differentiation of epithelial crypt progenitor cells. We find that the Pou2f3 gene is essential for tuft cell specification. Pou2f3(-/-) mice lack intestinal tuft cells and have defective mucosal type 2 responses to helminth infection; goblet cell hyperplasia is abrogated and worm expulsion is compromised. Notably, IL-4Rα signalling is sufficient to induce expansion of the tuft cell lineage, and ectopic stimulation of this signalling cascade obviates the need for tuft cells in the epithelial cell remodelling of the intestine. Moreover, tuft cells secrete IL-25, thereby regulating type 2 immune responses. Our data reveal a novel function of intestinal epithelial tuft cells and demonstrate a cellular relay required for initiating mucosal type 2 immunity to helminth infection.
- Published
- 2016
- Full Text
- View/download PDF
24. Erratum: Exosomes secreted by nematode parasites transfer small RNAs to mammalian cells and modulate innate immunity.
- Author
-
Buck AH, Coakley G, Simbari F, McSorley HJ, Quintana JF, Le Bihan T, Kumar S, Abreu-Goodger C, Lear M, Harcus Y, Ceroni A, Babayan SA, Blaxter M, Ivens A, and Maizels RM
- Published
- 2015
- Full Text
- View/download PDF
25. Cultivation of Heligmosomoides polygyrus: an immunomodulatory nematode parasite and its secreted products.
- Author
-
Johnston CJ, Robertson E, Harcus Y, Grainger JR, Coakley G, Smyth DJ, McSorley HJ, and Maizels R
- Subjects
- Animals, Life Cycle Stages, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Nematospiroides dubius growth & development, Nematospiroides dubius metabolism
- Abstract
Heligmosomoides polygyrus (formerly known as Nematospiroides dubius, and also referred to by some as H. bakeri) is a gastrointestinal helminth that employs multiple immunomodulatory mechanisms to establish chronic infection in mice and closely resembles prevalent human helminth infections. H. polygyrus has been studied extensively in the field of helminth-derived immune regulation and has been found to potently suppress experimental models of allergy and autoimmunity (both with active infection and isolated secreted products). The protocol described in this paper outlines management of the H. polygyrus life cycle for consistent production of L3 larvae, recovery of adult parasites, and collection of their excretory-secretory products (HES).
- Published
- 2015
- Full Text
- View/download PDF
26. Concerted activity of IgG1 antibodies and IL-4/IL-25-dependent effector cells trap helminth larvae in the tissues following vaccination with defined secreted antigens, providing sterile immunity to challenge infection.
- Author
-
Hewitson JP, Filbey KJ, Esser-von Bieren J, Camberis M, Schwartz C, Murray J, Reynolds LA, Blair N, Robertson E, Harcus Y, Boon L, Huang SC, Yang L, Tu Y, Miller MJ, Voehringer D, Le Gros G, Harris N, and Maizels RM
- Subjects
- Animals, Antibodies, Helminth genetics, Humans, Immunoglobulin G genetics, Interleukin-4 genetics, Interleukins genetics, Larva immunology, Mice, Mice, Knockout, Strongylida Infections genetics, Strongylida Infections prevention & control, Antibodies, Helminth immunology, Antigens, Helminth immunology, Immunoglobulin G immunology, Interleukin-4 immunology, Interleukins immunology, Nematospiroides dubius immunology, Strongylida Infections immunology, Vaccination
- Abstract
Over 25% of the world's population are infected with helminth parasites, the majority of which colonise the gastrointestinal tract. However, no vaccine is yet available for human use, and mechanisms of protective immunity remain unclear. In the mouse model of Heligmosomoides polygyrus infection, vaccination with excretory-secretory (HES) antigens from adult parasites elicits sterilising immunity. Notably, three purified HES antigens (VAL-1, -2 and -3) are sufficient for effective vaccination. Protection is fully dependent upon specific IgG1 antibodies, but passive transfer confers only partial immunity to infection, indicating that cellular components are also required. Moreover, immune mice show greater cellular infiltration associated with trapping of larvae in the gut wall prior to their maturation. Intra-vital imaging of infected intestinal tissue revealed a four-fold increase in extravasation by LysM+GFP+ myeloid cells in vaccinated mice, and the massing of these cells around immature larvae. Mice deficient in FcRγ chain or C3 complement component remain fully immune, suggesting that in the presence of antibodies that directly neutralise parasite molecules, the myeloid compartment may attack larvae more quickly and effectively. Immunity to challenge infection was compromised in IL-4Rα- and IL-25-deficient mice, despite levels of specific antibody comparable to immune wild-type controls, while deficiencies in basophils, eosinophils or mast cells or CCR2-dependent inflammatory monocytes did not diminish immunity. Finally, we identify a suite of previously uncharacterised heat-labile vaccine antigens with homologs in human and veterinary parasites that together promote full immunity. Taken together, these data indicate that vaccine-induced immunity to intestinal helminths involves IgG1 antibodies directed against secreted proteins acting in concert with IL-25-dependent Type 2 myeloid effector populations.
- Published
- 2015
- Full Text
- View/download PDF
27. Exosomes secreted by nematode parasites transfer small RNAs to mammalian cells and modulate innate immunity.
- Author
-
Buck AH, Coakley G, Simbari F, McSorley HJ, Quintana JF, Le Bihan T, Kumar S, Abreu-Goodger C, Lear M, Harcus Y, Ceroni A, Babayan SA, Blaxter M, Ivens A, and Maizels RM
- Subjects
- Alternaria immunology, Alternaria physiology, Alternariosis genetics, Alternariosis immunology, Alternariosis microbiology, Animals, Dual Specificity Phosphatase 1 genetics, Dual Specificity Phosphatase 1 immunology, Exosomes genetics, Exosomes immunology, Humans, Interleukin-1 Receptor-Like 1 Protein, Mice, Mice, Inbred BALB C, MicroRNAs chemistry, MicroRNAs genetics, MicroRNAs immunology, Nematoda genetics, Nematoda metabolism, Nematode Infections genetics, Nematode Infections parasitology, RNA, Helminth chemistry, RNA, Helminth genetics, RNA, Helminth metabolism, Receptors, Interleukin genetics, Receptors, Interleukin immunology, Exosomes metabolism, Immunity, Innate, MicroRNAs metabolism, Nematoda immunology, Nematode Infections immunology, RNA, Helminth immunology
- Abstract
In mammalian systems RNA can move between cells via vesicles. Here we demonstrate that the gastrointestinal nematode Heligmosomoides polygyrus, which infects mice, secretes vesicles containing microRNAs (miRNAs) and Y RNAs as well as a nematode Argonaute protein. These vesicles are of intestinal origin and are enriched for homologues of mammalian exosome proteins. Administration of the nematode exosomes to mice suppresses Type 2 innate responses and eosinophilia induced by the allergen Alternaria. Microarray analysis of mouse cells incubated with nematode exosomes in vitro identifies Il33r and Dusp1 as suppressed genes, and Dusp1 can be repressed by nematode miRNAs based on a reporter assay. We further identify miRNAs from the filarial nematode Litomosoides sigmodontis in the serum of infected mice, suggesting that miRNA secretion into host tissues is conserved among parasitic nematodes. These results reveal exosomes as another mechanism by which helminths manipulate their hosts and provide a mechanistic framework for RNA transfer between animal species.
- Published
- 2014
- Full Text
- View/download PDF
28. Secreted proteomes of different developmental stages of the gastrointestinal nematode Nippostrongylus brasiliensis.
- Author
-
Sotillo J, Sanchez-Flores A, Cantacessi C, Harcus Y, Pickering D, Bouchery T, Camberis M, Tang SC, Giacomin P, Mulvenna J, Mitreva M, Berriman M, LeGros G, Maizels RM, and Loukas A
- Subjects
- Ancylostomatoidea metabolism, Animals, Base Sequence, Conserved Sequence, Gene Expression Profiling, Gene Expression Regulation, Developmental, Phylogeny, Proteome metabolism, Proteomics methods, Rats, Rats, Sprague-Dawley, Sequence Analysis, RNA, Ancylostomatoidea growth & development, Gastrointestinal Tract parasitology, Helminth Proteins metabolism, Life Cycle Stages, Proteome analysis
- Abstract
Hookworms infect more than 700 million people worldwide and cause more morbidity than most other human parasitic infections. Nippostrongylus brasiliensis (the rat hookworm) has been used as an experimental model for human hookworm because of its similar life cycle and ease of maintenance in laboratory rodents. Adult N. brasiliensis, like the human hookworm, lives in the intestine of the host and releases excretory/secretory products (ESP), which represent the major host-parasite interface. We performed a comparative proteomic analysis of infective larval (L3) and adult worm stages of N. brasiliensis to gain insights into the molecular bases of host-parasite relationships and determine whether N. brasiliensis could indeed serve as an appropriate model for studying human hookworm infections. Proteomic data were matched to a transcriptomic database assembled from 245,874,892 Illumina reads from different developmental stages (eggs, L3, L4, and adult) of N. brasiliensis yielding∼18,426 unigenes with 39,063 possible isoform transcripts. From this analysis, 313 proteins were identified from ESPs by LC-MS/MS-52 in the L3 and 261 in the adult worm. Most of the proteins identified in the study were stage-specific (only 13 proteins were shared by both stages); in particular, two families of proteins-astacin metalloproteases and CAP-domain containing SCP/TAPS-were highly represented in both L3 and adult ESP. These protein families are present in most nematode groups, and where studied, appear to play roles in larval migration and evasion of the host's immune response. Phylogenetic analyses of defined protein families and global gene similarity analyses showed that N. brasiliensis has a greater degree of conservation with human hookworm than other model nematodes examined. These findings validate the use of N. brasiliensis as a suitable parasite for the study of human hookworm infections in a tractable animal model., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
29. MyD88 signaling inhibits protective immunity to the gastrointestinal helminth parasite Heligmosomoides polygyrus.
- Author
-
Reynolds LA, Harcus Y, Smith KA, Webb LM, Hewitson JP, Ross EA, Brown S, Uematsu S, Akira S, Gray D, Gray M, MacDonald AS, Cunningham AF, and Maizels RM
- Subjects
- Adaptor Proteins, Vesicular Transport genetics, Animals, CD4-Positive T-Lymphocytes immunology, Granuloma genetics, Granuloma immunology, Interleukin-17 biosynthesis, Interleukin-4 biosynthesis, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptor, Interferon alpha-beta genetics, Receptors, Interleukin-1 Type I genetics, Signal Transduction genetics, Signal Transduction immunology, Strongylida Infections parasitology, Toll-Like Receptor 2 genetics, Toll-Like Receptor 4 genetics, Toll-Like Receptor 5 genetics, Toll-Like Receptor 9 genetics, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 immunology, Nematospiroides dubius immunology, Strongylida Infections immunology
- Abstract
Helminth parasites remain one of the most common causes of infections worldwide, yet little is still known about the immune signaling pathways that control their expulsion. C57BL/6 mice are chronically susceptible to infection with the gastrointestinal helminth parasite Heligmosomoides polygyrus. In this article, we report that C57BL/6 mice lacking the adapter protein MyD88, which mediates signaling by TLRs and IL-1 family members, showed enhanced immunity to H. polygyrus infection. Alongside increased parasite expulsion, MyD88-deficient mice showed heightened IL-4 and IL-17A production from mesenteric lymph node CD4(+) cells. In addition, MyD88(-/-) mice developed substantial numbers of intestinal granulomas around the site of infection, which were not seen in MyD88-sufficient C57BL/6 mice, nor when signaling through the adapter protein TRIF (TIR domain-containing adapter-inducing IFN-β adapter protein) was also ablated. Mice deficient solely in TLR2, TLR4, TLR5, or TLR9 did not show enhanced parasite expulsion, suggesting that these TLRs signal redundantly to maintain H. polygyrus susceptibility in wild-type mice. To further investigate signaling pathways that are MyD88 dependent, we infected IL-1R1(-/-) mice with H. polygyrus. This genotype displayed heightened granuloma numbers compared with wild-type mice, but without increased parasite expulsion. Thus, the IL-1R-MyD88 pathway is implicated in inhibiting granuloma formation; however, protective immunity in MyD88-deficient mice appears to be granuloma independent. Like IL-1R1(-/-) and MyD88(-/-) mice, animals lacking signaling through the type 1 IFN receptor (i.e., IFNAR1(-/-)) also developed intestinal granulomas. Hence, IL-1R1, MyD88, and type 1 IFN receptor signaling may provide pathways to impede granuloma formation in vivo, but additional MyD88-mediated signals are associated with inhibition of protective immunity in susceptible C57BL/6 mice., (Copyright © 2014 The Authors.)
- Published
- 2014
- Full Text
- View/download PDF
30. Commensal-pathogen interactions in the intestinal tract: lactobacilli promote infection with, and are promoted by, helminth parasites.
- Author
-
Reynolds LA, Smith KA, Filbey KJ, Harcus Y, Hewitson JP, Redpath SA, Valdez Y, Yebra MJ, Finlay BB, and Maizels RM
- Subjects
- Animals, Gastrointestinal Tract immunology, Host-Pathogen Interactions, Lactobacillus growth & development, Lactobacillus immunology, Mice, Inbred BALB C, Mice, Inbred C57BL, Nematospiroides dubius growth & development, Nematospiroides dubius immunology, Gastrointestinal Tract microbiology, Lactobacillus physiology, Microbial Interactions, Nematospiroides dubius physiology, T-Lymphocytes, Regulatory immunology, Th17 Cells immunology
- Abstract
The intestinal microbiota are pivotal in determining the developmental, metabolic and immunological status of the mammalian host. However, the intestinal tract may also accommodate pathogenic organisms, including helminth parasites which are highly prevalent in most tropical countries. Both microbes and helminths must evade or manipulate the host immune system to reside in the intestinal environment, yet whether they influence each other's persistence in the host remains unknown. We now show that abundance of Lactobacillus bacteria correlates positively with infection with the mouse intestinal nematode parasite, Heligmosomoides polygyrus, as well as with heightened regulatory T cell (Treg) and Th17 responses. Moreover, H. polygyrus raises Lactobacillus species abundance in the duodenum of C57BL/6 mice, which are highly susceptible to H. polygyrus infection, but not in BALB/c mice, which are relatively resistant. Sequencing of samples at the bacterial gyrB locus identified the principal Lactobacillus species as L. taiwanensis, a previously characterized rodent commensal. Experimental administration of L. taiwanensis to BALB/c mice elevates regulatory T cell frequencies and results in greater helminth establishment, demonstrating a causal relationship in which commensal bacteria promote infection with an intestinal parasite and implicating a bacterially-induced expansion of Tregs as a mechanism of greater helminth susceptibility. The discovery of this tripartite interaction between host, bacteria and parasite has important implications for both antibiotic and anthelmintic use in endemic human populations.
- Published
- 2014
- Full Text
- View/download PDF
31. Innate and adaptive type 2 immune cell responses in genetically controlled resistance to intestinal helminth infection.
- Author
-
Filbey KJ, Grainger JR, Smith KA, Boon L, van Rooijen N, Harcus Y, Jenkins S, Hewitson JP, and Maizels RM
- Subjects
- Animals, Antibodies, Helminth immunology, Clodronic Acid pharmacology, Female, Granuloma, Helminthiasis drug therapy, Helminthiasis metabolism, Helminthiasis pathology, Helminthiasis, Animal, Immunity, Cellular, Interferon-gamma metabolism, Intestinal Diseases drug therapy, Intestinal Diseases metabolism, Intestinal Diseases pathology, Intestinal Diseases, Parasitic, Macrophage Activation immunology, Macrophages immunology, Mice, Receptors, Interleukin-4 metabolism, Signal Transduction, Adaptive Immunity drug effects, Disease Resistance genetics, Disease Resistance immunology, Helminthiasis genetics, Helminthiasis immunology, Helminths immunology, Immunity, Innate drug effects, Intestinal Diseases genetics, Intestinal Diseases immunology
- Abstract
The nematode Heligmosomoides polygyrus is an excellent model for intestinal helminth parasitism. Infection in mice persists for varying lengths of time in different inbred strains, with CBA and C57BL/6 mice being fully susceptible, BALB/c partially so and SJL able to expel worms within 2-3 weeks of infection. We find that resistance correlates not only with the adaptive Th2 response, including IL-10 but with activation of innate lymphoid cell and macrophage populations. In addition, the titer and specificity range of the serum antibody response is maximal in resistant mice. In susceptible strains, Th2 responses were found to be counterbalanced by IFN-γ-producing CD4(+) and CD8(+) cells, but these are not solely responsible for susceptibility as mice deficient in either CD8(+) T cells or IFN-γ remain unable to expel the parasites. Foxp3(+) Treg numbers were comparable in all strains, but in the most resistant SJL strain, this population does not upregulate CD103 in infection, and in the lamina propria the frequency of Foxp3(+)CD103(+) T cells is significantly lower than in susceptible mice. The more resistant SJL and BALB/c mice develop macrophage-rich IL-4Rα-dependent Type 2 granulomas around intestinal sites of larval invasion, and expression of alternative activation markers Arginase-1, Ch3L3 (Ym1) and RELM-α within the intestine and the peritoneal lavage was also strongly correlated with helminth elimination in these strains. Clodronate depletion of phagocytic cells compromises resistance of BALB/c mice and slows expulsion in the SJL strain. Thus, Type 2 immunity involves IL-4Rα-dependent innate cells including but not limited to a phagocyte population, the latter likely involving the action of specific antibodies.
- Published
- 2014
- Full Text
- View/download PDF
32. The secreted triose phosphate isomerase of Brugia malayi is required to sustain microfilaria production in vivo.
- Author
-
Hewitson JP, Rückerl D, Harcus Y, Murray J, Webb LM, Babayan SA, Allen JE, Kurniawan A, and Maizels RM
- Subjects
- Animals, Antibodies, Helminth immunology, Antibodies, Neutralizing immunology, Blotting, Western, Brugia malayi enzymology, Brugia malayi immunology, Elephantiasis, Filarial immunology, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Gerbillinae, Humans, Immunohistochemistry, Mice, Mice, Inbred BALB C, Brugia malayi pathogenicity, Elephantiasis, Filarial enzymology, Microfilariae, Triose-Phosphate Isomerase metabolism
- Abstract
Human lymphatic filariasis is a major tropical disease transmitted through mosquito vectors which take up microfilarial larvae from the blood of infected subjects. Microfilariae are produced by long-lived adult parasites, which also release a suite of excretory-secretory products that have recently been subject to in-depth proteomic analysis. Surprisingly, the most abundant secreted protein of adult Brugia malayi is triose phosphate isomerase (TPI), a glycolytic enzyme usually associated with the cytosol. We now show that while TPI is a prominent target of the antibody response to infection, there is little antibody-mediated inhibition of catalytic activity by polyclonal sera. We generated a panel of twenty-three anti-TPI monoclonal antibodies and found only two were able to block TPI enzymatic activity. Immunisation of jirds with B. malayi TPI, or mice with the homologous protein from the rodent filaria Litomosoides sigmodontis, failed to induce neutralising antibodies or protective immunity. In contrast, passive transfer of neutralising monoclonal antibody to mice prior to implantation with adult B. malayi resulted in 60-70% reductions in microfilarial levels in vivo and both oocyte and microfilarial production by individual adult females. The loss of fecundity was accompanied by reduced IFNγ expression by CD4⁺ T cells and a higher proportion of macrophages at the site of infection. Thus, enzymatically active TPI plays an important role in the transmission cycle of B. malayi filarial parasites and is identified as a potential target for immunological and pharmacological intervention against filarial infections.
- Published
- 2014
- Full Text
- View/download PDF
33. Secretion of protective antigens by tissue-stage nematode larvae revealed by proteomic analysis and vaccination-induced sterile immunity.
- Author
-
Hewitson JP, Ivens AC, Harcus Y, Filbey KJ, McSorley HJ, Murray J, Bridgett S, Ashford D, Dowle AA, and Maizels RM
- Subjects
- Animals, Antibodies, Helminth analysis, Antibodies, Helminth immunology, Antigens, Helminth immunology, Blotting, Western, Chromatography, Liquid, Computational Biology, Electrophoresis, Gel, Two-Dimensional, Enzyme-Linked Immunosorbent Assay, Female, Gene Expression Profiling, Helminth Proteins immunology, Host-Parasite Interactions, Immunization, Immunoprecipitation, Larva immunology, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Nematode Infections parasitology, Nematospiroides dubius growth & development, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Vaccination, Antigens, Helminth metabolism, Helminth Proteins metabolism, Larva metabolism, Nematode Infections immunology, Nematospiroides dubius immunology, Proteomics
- Abstract
Gastrointestinal nematode parasites infect over 1 billion humans, with little evidence for generation of sterilising immunity. These helminths are highly adapted to their mammalian host, following a developmental program through successive niches, while effectively down-modulating host immune responsiveness. Larvae of Heligmosomoides polygyrus, for example, encyst in the intestinal submucosa, before emerging as adult worms into the duodenal lumen. Adults release immunomodulatory excretory-secretory (ES) products, but mice immunised with adult H. polygyrus ES become fully immune to challenge infection. ES products of the intestinal wall 4th stage (L4) larvae are similarly important in host-parasite interactions, as they readily generate sterile immunity against infection, while released material from the egg stage is ineffective. Proteomic analyses of L4 ES identifies protective antigen targets as well as potential tissue-phase immunomodulatory molecules, using as comparators the adult ES proteome and a profile of H. polygyrus egg-released material. While 135 proteins are shared between L4 and adult ES, 72 are L4 ES-specific; L4-specific proteins correspond to those whose transcription is restricted to larval stages, while shared proteins are generally transcribed by all life cycle forms. Two protein families are more heavily represented in the L4 secretome, the Sushi domain, associated with complement regulation, and the ShK/SXC domain related to a toxin interfering with T cell signalling. Both adult and L4 ES contain extensive but distinct arrays of Venom allergen/Ancylostoma secreted protein-Like (VAL) members, with acetylcholinesterases (ACEs) and apyrase APY-3 particularly abundant in L4 ES. Serum antibodies from mice vaccinated with L4 and adult ES react strongly to the VAL-1 protein and to ACE-1, indicating that these two antigens represent major vaccine targets for this intestinal nematode. We have thus defined an extensive and novel repertoire of H. polygyrus proteins closely implicated in immune modulation and protective immunity.
- Published
- 2013
- Full Text
- View/download PDF
34. Type 2 innate immunity in helminth infection is induced redundantly and acts autonomously following CD11c(+) cell depletion.
- Author
-
Smith KA, Harcus Y, Garbi N, Hämmerling GJ, MacDonald AS, and Maizels RM
- Subjects
- Animals, Basophils immunology, CD11c Antigen genetics, Cytokines genetics, Cytokines metabolism, Flow Cytometry, Macrophages metabolism, Macrophages, Alveolar immunology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Reverse Transcriptase Polymerase Chain Reaction, CD11c Antigen metabolism, Dendritic Cells immunology, Immunity, Innate physiology, Nematospiroides dubius immunology, Nippostrongylus immunology, Strongylida Infections immunology
- Abstract
Infection with gastrointestinal helminths generates a dominant type 2 response among both adaptive (Th2) and innate (macrophage, eosinophil, and innate lymphoid) immune cell types. Two additional innate cell types, CD11c(high) dendritic cells (DCs) and basophils, have been implicated in the genesis of type 2 immunity. Investigating the type 2 response to intestinal nematode parasites, including Heligmosomoides polygyrus and Nippostrongylus brasiliensis, we first confirmed the requirement for DCs in stimulating Th2 adaptive immunity against these helminths through depletion of CD11c(high) cells by administration of diphtheria toxin to CD11c.DOG mice. In contrast, responsiveness was intact in mice depleted of basophils by antibody treatment. Th2 responses can be induced by adoptive transfer of DCs, but not basophils, exposed to soluble excretory-secretory products from these helminths. However, innate type 2 responses arose equally strongly in the presence or absence of CD11c(high) cells or basophils; thus, in CD11c.DOG mice, the alternative activation of macrophages, as measured by expression of arginase-1, RELM-α, and Ym-1 (Chi3L3) in the intestine following H. polygyrus infection or in the lung following N. brasiliensis infection, was unaltered by depletion of CD11c-expressing DCs and alveolar macrophages or by antibody-mediated basophil depletion. Similarly, goblet cell-associated RELM-β in lung and intestinal tissues, lung eosinophilia, and expansion of innate lymphoid ("nuocyte") populations all proceeded irrespective of depletion of CD11c(high) cells or basophils. Thus, while CD11c(high) DCs initiate helminth-specific adaptive immunity, innate type 2 cells are able to mount an autonomous response to the challenge of parasite infection.
- Published
- 2012
- Full Text
- View/download PDF
35. Heligmosomoides polygyrus elicits a dominant nonprotective antibody response directed against restricted glycan and peptide epitopes.
- Author
-
Hewitson JP, Filbey KJ, Grainger JR, Dowle AA, Pearson M, Murray J, Harcus Y, and Maizels RM
- Subjects
- Animals, Antibody Specificity immunology, Antigens, Helminth biosynthesis, Antigens, Helminth genetics, Epitopes immunology, Epitopes metabolism, Female, Immune Sera metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Peptides metabolism, Polysaccharides metabolism, Protein Conformation, Strongylida Infections immunology, Strongylida Infections parasitology, Antibodies, Helminth biosynthesis, Antigens, Helminth immunology, Nematospiroides dubius immunology, Peptides immunology, Polysaccharides immunology
- Abstract
Heligmosomoides polygyrus is a widely used gastrointestinal helminth model of long-term chronic infection in mice, which has not been well-characterized at the antigenic level. We now identify the major targets of the murine primary Ab response as a subset of the secreted products in H. polygyrus excretory-secretory (HES) Ag. An immunodominant epitope is an O-linked glycan (named glycan A) carried on three highly expressed HES glycoproteins (venom allergen Ancylostoma-secreted protein-like [VAL]-1, -2, and -5), which stimulates only IgM Abs, is exposed on the adult worm surface, and is poorly represented in somatic parasite extracts. A second carbohydrate epitope (glycan B), present on both a non-protein high molecular mass component and a 65-kDa molecule, is widely distributed in adult somatic tissues. Whereas the high molecular mass component and 65-kDa molecules bear phosphorylcholine, the glycan B epitope itself is not phosphorylcholine. Class-switched IgG1 Abs are found to glycan B, but the dominant primary IgG1 response is to the polypeptides of VAL proteins, including also VAL-3 and VAL-4. Secondary Ab responses include the same specificities while also recognizing VAL-7. Although vaccination with HES conferred complete protection against challenge H. polygyrus infection, mAbs raised against each of the glycan epitopes and against VAL-1, VAL-2, and VAL-4 proteins were unable to do so, even though these specificities (with the exception of VAL-2) are also secreted by tissue-phase L4 larvae. The primary immune response in susceptible mice is, therefore, dominated by nonprotective Abs against a small subset of antigenic epitopes, raising the possibility that these act as decoy specificities that generate ineffective humoral immunity.
- Published
- 2011
- Full Text
- View/download PDF
36. Proteomic analysis of secretory products from the model gastrointestinal nematode Heligmosomoides polygyrus reveals dominance of venom allergen-like (VAL) proteins.
- Author
-
Hewitson JP, Harcus Y, Murray J, van Agtmaal M, Filbey KJ, Grainger JR, Bridgett S, Blaxter ML, Ashton PD, Ashford DA, Curwen RS, Wilson RA, Dowle AA, and Maizels RM
- Subjects
- Animals, Antigens, Helminth, Disease Models, Animal, Helminth Proteins metabolism, Gastrointestinal Diseases parasitology, Helminth Proteins analysis, Nematospiroides dubius chemistry, Proteomics methods
- Abstract
The intestinal helminth parasite, Heligmosomoides polygyrus bakeri offers a tractable experimental model for human hookworm infections such as Ancylostoma duodenale and veterinary parasites such as Haemonchus contortus. Parasite excretory-secretory (ES) products represent the major focus for immunological and biochemical analyses, and contain immunomodulatory molecules responsible for nematode immune evasion. In a proteomic analysis of adult H. polygyrus secretions (termed HES) matched to an extensive transcriptomic dataset, we identified 374 HES proteins by LC-MS/MS, which were distinct from those in somatic extract HEx, comprising 446 identified proteins, confirming selective export of ES proteins. The predominant secreted protein families were proteases (astacins and other metalloproteases, aspartic, cysteine and serine-type proteases), lysozymes, apyrases and acetylcholinesterases. The most abundant products were members of the highly divergent venom allergen-like (VAL) family, related to Ancylostoma secreted protein (ASP); 25 homologues were identified, with VAL-1 and -2 also shown to be associated with the parasite surface. The dominance of VAL proteins is similar to profiles reported for Ancylostoma and Haemonchus ES products. Overall, this study shows that the secretions of H. polygyrus closely parallel those of clinically important GI nematodes, confirming the value of this parasite as a model of helminth infection., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
37. Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway.
- Author
-
Grainger JR, Smith KA, Hewitson JP, McSorley HJ, Harcus Y, Filbey KJ, Finney CA, Greenwood EJ, Knox DP, Wilson MS, Belkaid Y, Rudensky AY, and Maizels RM
- Subjects
- Animals, Antigens, Helminth metabolism, Benzamides pharmacology, Cell Proliferation drug effects, Chronic Disease, Dioxoles pharmacology, Forkhead Transcription Factors biosynthesis, Forkhead Transcription Factors genetics, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Host-Parasite Interactions drug effects, Host-Parasite Interactions genetics, Host-Parasite Interactions immunology, Mice, Mice, Inbred BALB C, Mice, Transgenic, Nematospiroides dubius metabolism, Phosphorylation genetics, Phosphorylation immunology, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases immunology, Protein Serine-Threonine Kinases metabolism, Receptor, Transforming Growth Factor-beta Type II, Receptors, Transforming Growth Factor beta antagonists & inhibitors, Receptors, Transforming Growth Factor beta genetics, Receptors, Transforming Growth Factor beta immunology, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction drug effects, Signal Transduction genetics, Smad2 Protein genetics, Smad2 Protein immunology, Smad2 Protein metabolism, Smad3 Protein genetics, Smad3 Protein immunology, Smad3 Protein metabolism, Strongylida Infections genetics, Strongylida Infections metabolism, T-Lymphocytes, Regulatory metabolism, Th1 Cells immunology, Th1 Cells metabolism, Th2 Cells immunology, Th2 Cells metabolism, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Antigens, Helminth immunology, Forkhead Transcription Factors immunology, Gene Expression Regulation immunology, Nematospiroides dubius immunology, Signal Transduction immunology, Strongylida Infections immunology, T-Lymphocytes, Regulatory immunology, Transforming Growth Factor beta immunology
- Abstract
Foxp3-expressing regulatory T (T reg) cells have been implicated in parasite-driven inhibition of host immunity during chronic infection. We addressed whether parasites can directly induce T reg cells. Foxp3 expression was stimulated in naive Foxp3⁻ T cells in mice infected with the intestinal helminth Heligmosomoides polygyrus. In vitro, parasite-secreted proteins (termed H. polygyrus excretory-secretory antigen [HES]) induced de novo Foxp3 expression in fluorescence-sorted Foxp3⁻ splenocytes from Foxp3-green fluorescent protein reporter mice. HES-induced T reg cells suppressed both in vitro effector cell proliferation and in vivo allergic airway inflammation. HES ligated the transforming growth factor (TGF) β receptor and promoted Smad2/3 phosphorylation. Foxp3 induction by HES was lost in dominant-negative TGF-βRII cells and was abolished by the TGF-β signaling inhibitor SB431542. This inhibitor also reduced worm burdens in H. polygyrus-infected mice. HES induced IL-17 in the presence of IL-6 but did not promote Th1 or Th2 development under any conditions. Importantly, antibody to mammalian TGF-β did not recognize HES, whereas antisera that inhibited HES did not affect TGF-β. Foxp3 was also induced by secreted products of Teladorsagia circumcincta, a related nematode which is widespread in ruminant animals. We have therefore identified a novel pathway through which helminth parasites may stimulate T reg cells, which is likely to be a key part of the parasite's immunological relationship with the host.
- Published
- 2010
- Full Text
- View/download PDF
38. daf-7-related TGF-beta homologues from Trichostrongyloid nematodes show contrasting life-cycle expression patterns.
- Author
-
McSorley HJ, Grainger JR, Harcus Y, Murray J, Nisbet AJ, Knox DP, and Maizels RM
- Subjects
- Amino Acid Sequence, Animals, Gene Expression Profiling, Helminth Proteins chemistry, Helminth Proteins genetics, Humans, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Nematospiroides dubius, Phylogeny, Sequence Alignment, Trichostrongyloidea classification, Trichostrongyloidea genetics, Trichostrongyloidea metabolism, Caenorhabditis elegans Proteins chemistry, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism, Gene Expression Regulation, Developmental, Helminth Proteins metabolism, Life Cycle Stages, Sequence Homology, Amino Acid, Transforming Growth Factor beta chemistry, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Trichostrongyloidea growth & development
- Abstract
The transforming growth factor-beta (TGF-beta) gene family regulates critical processes in animal development, and plays a crucial role in regulating the mammalian immune response. We aimed to identify TGF-beta homologues from 2 laboratory model nematodes (Heligmosomoides polygyrus and Nippostrongylus brasiliensis) and 2 major parasites of ruminant livestock (Haemonchus contortus and Teladorsagia circumcincta). Parasite cDNA was used as a template for gene-specific PCR and RACE. Homologues of the TGH-2 subfamily were isolated, and found to differ in length (301, 152, 349 and 305 amino acids respectively), with variably truncated N-terminal pre-proteins. All contained conserved C-terminal active domains (>85% identical over 115 amino acids) containing 9 cysteine residues, as in C. elegans DAF-7, Brugia malayi TGH-2 and mammalian TGF-beta. Surprisingly, only the H. contortus homologue retained a conventional signal sequence, absent from shorter proteins of other species. RT-PCR assays of transcription showed that in H. contortus and N. brasiliensis expression was maximal in the infective larval stage, and very low in adult worms. In contrast, in H. polygyrus and T. circumcincta, tgh-2 transcription is higher in adults than infective larvae. The molecular evolution of this gene family in parasitic nematodes has diversified the pre-protein and life-cycle expression patterns of TGF-beta homologues while conserving the structure of the active domain.
- Published
- 2010
- Full Text
- View/download PDF
39. Dynamics of CD11c(+) dendritic cell subsets in lymph nodes draining the site of intestinal nematode infection.
- Author
-
Balic A, Smith KA, Harcus Y, and Maizels RM
- Subjects
- Animals, Antigens, CD biosynthesis, Cell Differentiation, Cell Movement, Cells, Cultured, Cytokines genetics, Dendritic Cells immunology, Dendritic Cells parasitology, Dendritic Cells pathology, Intestinal Diseases, Parasitic pathology, Lymph Nodes metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Nematospiroides dubius pathogenicity, Nippostrongylus pathogenicity, Strongylida Infections pathology, Th2 Cells immunology, Cytokines biosynthesis, Dendritic Cells metabolism, Intestinal Diseases, Parasitic immunology, Lymph Nodes pathology, Nematospiroides dubius immunology, Nippostrongylus immunology, Strongylida Infections immunology
- Abstract
Helminth parasites drive dominant Th2 responses through an as yet unidentified pathway. We have previously shown that the rodent gastrointestinal nematode Nippostrongylus brasiliensis secretes products which selectively activate in vitro-derived dendritic cells to induce Th2 responses on in vivo transfer. We now show that, during active infection with this parasite, the draining mesenteric lymph node dendritic cell population is altered significantly. Although there is substantial expansion of DC numbers during infection, the CD86(hi)-CD8alpha(int)-CD11b(-) subset is markedly diminished, and expression levels of CD40, CD86 and CD103 are reduced. Notably, the reduced frequency of CD8alpha(int) DCs is evident only in those mesenteric lymph nodes draining the anterior site of infestation. In infections with the longer lived Heligmosomoides polygyrus, the proportion of CD8alpha(int) DCs in the MLNC falls to below 10% of total DC numbers by 35 days post-infection. Further, infection alters TLR responsiveness, as IL-12 production (as measured by ex vivo intracellular staining of CD11c(+) DCs) in response to LPS stimulation is reduced, while IL-6, TNF-alpha and in particular, IL-10 all increase following infection with either nematode parasite. These changes suggest the possibility that helminth parasites modulate gastrointestinal immunity both by inhibiting migration of CD8alpha(int) DCs to the draining lymph nodes, and modifying DC responsiveness in a manner which favours a Th2 outcome.
- Published
- 2009
- Full Text
- View/download PDF
40. C-type lectins from the nematode parasites Heligmosomoides polygyrus and Nippostrongylus brasiliensis.
- Author
-
Harcus Y, Nicoll G, Murray J, Filbey K, Gomez-Escobar N, and Maizels RM
- Subjects
- Animals, Gene Library, Host-Parasite Interactions, Larva metabolism, Lectins, C-Type genetics, Life Cycle Stages, Mice, Molecular Sequence Data, Nematospiroides dubius genetics, Nematospiroides dubius metabolism, Nippostrongylus genetics, Nippostrongylus metabolism, Reverse Transcriptase Polymerase Chain Reaction, Sequence Analysis, DNA, Duodenum parasitology, Lectins, C-Type metabolism, Nematospiroides dubius growth & development, Nippostrongylus growth & development
- Abstract
The C-type lectin superfamily is highly represented in all metazoan phyla so far studied. Many members of this superfamily are important in innate immune defences against infection, while others serve key developmental and structural roles. Within the superfamily, many proteins contain multiple canonical carbohydrate-recognition domains (CRDs), together with additional non-lectin domains. In this report, we have studied two gastrointestinal nematode parasites which are widely used in experimental rodent systems, Heligmosomoides polygyrus and Nippostrongylus brasiliensis. From cDNA libraries, we have isolated 3 new C-type lectins from these species; all are single-CRD proteins with short additional N-terminal domains. The predicted Hp-CTL-1 protein contains 156 aa, Nb-CTL-1 191 aa and Nb-CTL-2 183 aa; all encode predicted signal peptides, as well as key conserved sequence motifs characteristic of the CTL superfamily. These lectins are most similar to C. elegans CLEC-48, 49 and 50, as well as to the lectin domains of mammalian immune system proteins CD23 and CD206. RT-PCR showed that these H. polygyrus and N. brasiliensis genes are primarily expressed in the gut-dwelling adult stages, although Nb-CTL-2 transcripts are also prominent in the free-living infective larval (L3) stage. Polyclonal antibodies raised to Hp-CTL-1 and Nb-CTL-1 reacted to both proteins by ELISA, and in Western blot analysis recognised a 15-kDa band in secreted proteins of adult N. brasiliensis (NES) and a 19-kDa band in H. polygyrus ES (HES). Anti-CTL-1 antibody also bound strongly to the cuticle of adult H. polygyrus. Hence, live parasites release C-type lectins homologous to some key receptors of the mammalian host immune system, raising the possibility that these products interfere in some manner with immunological recognition or effector function.
- Published
- 2009
- Full Text
- View/download PDF
41. Praziquantel treatment of individuals exposed to Schistosoma haematobium enhances serological recognition of defined parasite antigens.
- Author
-
Mutapi F, Burchmore R, Mduluza T, Foucher A, Harcus Y, Nicoll G, Midzi N, Turner CM, and Maizels RM
- Subjects
- Adolescent, Adult, Animals, Antibodies, Helminth immunology, Antigens, Helminth blood, Antigens, Helminth isolation & purification, Blotting, Western, Child, Child, Preschool, Electrophoresis, Gel, Two-Dimensional, Helminth Proteins blood, Helminth Proteins isolation & purification, Humans, Mass Spectrometry, Praziquantel pharmacology, Schistosomiasis haematobia diagnosis, Schistosomiasis haematobia immunology, Schistosomiasis haematobia parasitology, Schistosomicides pharmacology, Zimbabwe, Antigens, Helminth immunology, Helminth Proteins immunology, Praziquantel therapeutic use, Schistosoma haematobium immunology, Schistosomiasis haematobia drug therapy, Schistosomicides therapeutic use
- Abstract
Background: Schistosomiasis is a major parasitic disease affecting >200 million people in the developing world, and 400 million people are at risk for infection. This study aimed to identify and compare proteins recognized by serum samples from schistosome-exposed individuals before and after curative praziquantel treatment., Methods: Proteins recognized by pooled serum samples from Schistosoma haematobium-exposed Zimbabweans were determined by 2-dimensional Western blotting and identified by mass spectrometry., Results: Serum samples recognized 71 spots, which resolved to 26 different characterized proteins. Eleven of these proteins have not previously been shown to be immunogenic in natural human infection or in experimental models of schistosomiasis, making them novel antigens in the parasite. Pretreatment serum samples recognized 59 spots, which resolved to 21 different identified proteins. Posttreatment serum samples recognized an additional 12 spots, which resolved to 8 different identified proteins. Of these 8 proteins, 3 had putative isoforms recognized before treatment, and 5 (calreticulin, tropomyosin 1, tropomyosin 2, paramyosin, and triose phosphate isomerase) did not., Conclusions: This study is the most comprehensive characterization of S. haematobium antigens to date and describes novel antigens in all schistosome species. Posttreatment results are consistent with praziquantel treatment inducing quantitative and qualitative changes in schistosome-specific antibody responses.
- Published
- 2005
- Full Text
- View/download PDF
42. Selective maturation of dendritic cells by Nippostrongylus brasiliensis-secreted proteins drives Th2 immune responses.
- Author
-
Balic A, Harcus Y, Holland MJ, and Maizels RM
- Subjects
- Animals, Antigens, Helminth metabolism, Cell Differentiation immunology, Cytokines genetics, Cytokines immunology, Dendritic Cells cytology, Dendritic Cells parasitology, Enzyme-Linked Immunosorbent Assay, Female, Flow Cytometry, Mice, Mice, Inbred BALB C, Nippostrongylus metabolism, Oligonucleotide Array Sequence Analysis, RNA chemistry, RNA genetics, Up-Regulation immunology, Antigens, Helminth immunology, Dendritic Cells immunology, Nippostrongylus immunology, Strongylida Infections immunology, Th2 Cells immunology
- Abstract
Helminth infections at mucosal and tissue sites strongly polarize towards Th2 immune responses, following pathways which have yet to be elucidated. We investigated whether dendritic cells (DC) exposed to gastrointestinal nematodes induce Th2 differentiation and, if so, whether this outcome reflects the absence of DC activation (the default hypothesis). We studied secreted proteins from the parasite Nippostrongylus brasiliensis, which induce Th2 development in vivo without live infection. Murine bone marrow-derived DC pulsed with N. brasiliensis excretory/secretory antigen (NES) can, on transfer to naive recipients, prime mice for Th2 responsiveness. Heat inactivation of NES abolishes both its ability to drive Th2 responses in vivo and its capacity to stimulate DC for Th2 induction. NES, but not heat-inactivated NES, up-regulates DC maturation markers associated with Th2 promotion (CD86 and OX40L), with little change to CD80 and MHC class II. Moreover, DC exposed to NES readily produce IL-6 and IL-12p40, but not IL-12p70. LPS induced high IL-12p70 levels, except in DC that had been pre-incubated with NES. These data contradict the default hypothesis, demonstrating that a helminth product (NES) actively matures DC, selectively up-regulating CD86 and OX40L together with IL-6 production, while blocking IL-12p70 responsiveness in a manner consistent with Th2 generation in vivo.
- Published
- 2004
- Full Text
- View/download PDF
43. IL-4 is required to prevent filarial nematode development in resistant but not susceptible strains of mice.
- Author
-
Le Goff L, Lamb TJ, Graham AL, Harcus Y, and Allen JE
- Subjects
- Animals, Cytokines biosynthesis, Filariasis blood, Filariasis parasitology, Filarioidea immunology, Filarioidea isolation & purification, Genetic Predisposition to Disease, Host-Parasite Interactions, Immunity, Innate, Interferon-gamma biosynthesis, Interleukin-4 genetics, Interleukin-4 metabolism, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Microfilariae growth & development, Microfilariae immunology, Species Specificity, Th1 Cells immunology, Th2 Cells immunology, Filariasis immunology, Filarioidea growth & development, Interleukin-4 physiology
- Abstract
The murine Litomosoides sigmodontis model of filarial infection provides the opportunity to elucidate the immunological mechanisms that determine whether these nematode parasites can establish a successful infection or are rejected by the mammalian host. BALB/c mice are fully susceptible to L. sigmodontis infection and can develop patent infection, with the microfilarial stage circulating in the bloodstream. In contrast, mice on the C57BL background are largely resistant to the infection and never produce a patent infection. In this study, we used IL-4 deficient mice on the C57BL/6 background to address the role of IL-4 in the development of L. sigmodontis parasites in a resistant host. Two months after infection, adult worm recovery and the percentage of microfilaraemic mice in infected IL-4 deficient mice were comparable with those of the susceptible BALB/c mice while, as expected, healthy adults were not recovered from wild type C57BL/6 mice. The cytokine and antibody responses reveal that despite similar parasitology the two susceptible strains (BALB/c and IL-4 deficient C57BL/6) have markedly different immune responses: wild type BALB/c mice exhibit a strong Th2 immune response and the IL-4 deficient C57BL/6 mice exhibit a Th1 response. We also excluded a role for antibodies in resistance through infection of B-cell deficient C57BL/6 mice. Our data suggest that the mechanisms that determine parasite clearance in a resistant/non-permissive host are Th2 dependent but that in a susceptible/permissive host, the parasite can develop in the face of a Th2 dominated response.
- Published
- 2002
- Full Text
- View/download PDF
44. Proteins secreted by the parasitic nematode Nippostrongylus brasiliensis act as adjuvants for Th2 responses.
- Author
-
Holland MJ, Harcus YM, Riches PL, and Maizels RM
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, Female, Genotype, Helminth Proteins metabolism, Immunization methods, Immunoglobulin E biosynthesis, Interferon-gamma biosynthesis, Interleukin-4 biosynthesis, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred CBA, Rats, Rats, Sprague-Dawley, Th2 Cells parasitology, Adjuvants, Immunologic, Antigens, Helminth immunology, Helminth Proteins immunology, Nippostrongylus immunology, Th2 Cells immunology
- Abstract
Infections with parasitic helminths such as Nippostronglyus brasiliensis induce dominant type 2 responses from antigen-specific T helper cells. The potency of the Th2 bias can also drive Th2 responses to bystander antigens introduced at the same time as infection. We now report that the Th2-promoting effect of infection can be reproduced with soluble N. brasiliensis excretory-secretory proteins (NES) released by adult parasites in vitro. Immunization of BALB/c mice with NES results in the production of IL-4 with elevated total serum IgE and specific IgG1 antibodies. NES is also able to stimulate IL-4 and polyclonal IgE production in other mouse strains (C57BL/6, B10.D2, CBA). These features are seen whether NES is administered without adjuvant as soluble protein in phosphate-buffered saline or with complete Freund's adjuvant which normally favors Th1 responses. Thus, NES possesses intrinsic adjuvanticity. Moreover, co-administration of hen egg lysozyme (HEL) with NES in the absence of other adjuvants results in generation of HEL-specific lymphocyte proliferation, IL-4 release and IgG1 antibody responses, documenting that NES can act as an adjuvant for third-party antigens. Proteinase K digestion or heat treatment of NES before immunization abolished the IL-4-stimulating activity, indicating that the factors acting to promote Th2 induction are proteins secreted by the adult parasite.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.