Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
PLoS Pathog ; 19(4): e1011296, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37018382

RESUMO

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


Assuntos
Coinfecção , Nematospiroides dubius , Infecções por Strongylida , Camundongos , Animais , Nematospiroides dubius/metabolismo , Camundongos Knockout , Interleucina-4/genética , Interleucina-4/metabolismo , Interleucina-13/metabolismo , Larva/metabolismo , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo , Infecções por Strongylida/genética
2.
J Biol Chem ; 298(6): 101994, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35500648

RESUMO

The mouse intestinal helminth Heligmosomoides polygyrus modulates host immune responses by secreting a transforming growth factor (TGF)-ß mimic (TGM), to expand the population of Foxp3+ Tregs. TGM comprises five complement control protein (CCP)-like domains, designated D1-D5. Though lacking homology to TGF-ß, TGM binds directly to the TGF-ß receptors TßRI and TßRII and stimulates the differentiation of naïve T-cells into Tregs. However, the molecular determinants of binding are unclear. Here, we used surface plasmon resonance, isothermal calorimetry, NMR spectroscopy, and mutagenesis to investigate how TGM binds the TGF-ß receptors. We demonstrate that binding is modular, with D1-D2 binding to TßRI and D3 binding to TßRII. D1-D2 and D3 were further shown to compete with TGF-ß(TßRII)2 and TGF-ß for binding to TßRI and TßRII, respectively. The solution structure of TGM-D3 revealed that TGM adopts a CCP-like fold but is also modified to allow the C-terminal strand to diverge, leading to an expansion of the domain and opening potential interaction surfaces. TGM-D3 also incorporates a long structurally ordered hypervariable loop, adding further potential interaction sites. Through NMR shift perturbations and binding studies of TGM-D3 and TßRII variants, TGM-D3 was shown to occupy the same site of TßRII as bound by TGF-ß using both a novel interaction surface and the hypervariable loop. These results, together with the identification of other secreted CCP-like proteins with immunomodulatory activity in H. polygyrus, suggest that TGM is part of a larger family of evolutionarily plastic parasite effector molecules that mediate novel interactions with their host.


Assuntos
Proteínas de Helminto , Interações Hospedeiro-Patógeno , Nematospiroides dubius , Receptores de Fatores de Crescimento Transformadores beta , Fator de Crescimento Transformador beta , Animais , Evolução Biológica , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Nematospiroides dubius/classificação , Nematospiroides dubius/genética , Nematospiroides dubius/imunologia , Nematospiroides dubius/metabolismo , Ligação Proteica , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo
3.
J Exp Med ; 219(1)2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34779829

RESUMO

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


Assuntos
Células Epiteliais/metabolismo , Células Caliciformes/metabolismo , Intestino Delgado/citologia , Organoides/metabolismo , Infecções por Strongylida/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Epiteliais/parasitologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/parasitologia , Proteínas de Helminto/metabolismo , Proteínas de Helminto/farmacologia , Interações Hospedeiro-Parasita , Interleucina-13/farmacologia , Interleucina-4/farmacologia , Intestino Delgado/parasitologia , Camundongos Endogâmicos C57BL , Nematospiroides dubius/metabolismo , Nematospiroides dubius/fisiologia , Nippostrongylus/metabolismo , Nippostrongylus/fisiologia , Organoides/citologia , Organoides/parasitologia , Infecções por Strongylida/parasitologia , Ácido Succínico/farmacologia , Transcriptoma/efeitos dos fármacos
4.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33105843

RESUMO

As the global incidences of colorectal cancer rises, there is a growing importance in understanding the interaction between external factors, such as common infections, on the initiation and progression of this disease. While certain helminth infections have been shown to alter the severity and risk of developing colitis-associated colorectal cancer, whether these parasites can directly affect colorectal cancer progression is unknown. Here, we made use of murine and human colorectal cancer cell lines to demonstrate that exposure to antigens derived from the gastrointestinal nematode Heligmosomoides polygyrus significantly reduced colorectal cancer cell proliferation in vitro. Using a range of approaches, we demonstrate that antigen-dependent reductions in cancer cell proliferation and viability are associated with increased expression of the critical cell cycle regulators p53 and p21. Interestingly, H. polygyrus-derived antigens significantly increased murine colorectal cancer cell migration, which was associated with an increased expression of the adherens junction protein ß-catenin, whereas the opposite was true for human colorectal cancer cells. Together, these findings demonstrate that antigens derived from a gastrointestinal nematode can significantly alter colorectal cancer cell behavior. Further in-depth analysis may reveal novel candidates for targeting and treating late-stage cancer.


Assuntos
Antígenos de Helmintos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Nematospiroides dubius/imunologia , Animais , Antígenos de Helmintos/imunologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Células HCT116 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Nematospiroides dubius/metabolismo , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Especificidade da Espécie , Proteína Supressora de Tumor p53/metabolismo , beta Catenina/metabolismo
5.
Elife ; 92020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32420871

RESUMO

The IL-33-ST2 pathway is an important initiator of type 2 immune responses. We previously characterised the HpARI protein secreted by the model intestinal nematode Heligmosomoides polygyrus, which binds and blocks IL-33. Here, we identify H. polygyrus Binds Alarmin Receptor and Inhibits (HpBARI) and HpBARI_Hom2, both of which consist of complement control protein (CCP) domains, similarly to the immunomodulatory HpARI and Hp-TGM proteins. HpBARI binds murine ST2, inhibiting cell surface detection of ST2, preventing IL-33-ST2 interactions, and inhibiting IL-33 responses in vitro and in an in vivo mouse model of asthma. In H. polygyrus infection, ST2 detection is abrogated in the peritoneal cavity and lung, consistent with systemic effects of HpBARI. HpBARI_Hom2 also binds human ST2 with high affinity, and effectively blocks human PBMC responses to IL-33. Thus, we show that H. polygyrus blocks the IL-33 pathway via both HpARI which blocks the cytokine, and also HpBARI which blocks the receptor.


Assuntos
Alternaria/imunologia , Antígenos de Helmintos/metabolismo , Asma/patologia , Proteína 1 Semelhante a Receptor de Interleucina-1/antagonistas & inibidores , Interleucina-33/antagonistas & inibidores , Nematospiroides dubius/metabolismo , Animais , Linhagem Celular , Humanos , Fatores Imunológicos/metabolismo , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nematospiroides dubius/imunologia , Ovalbumina/imunologia
6.
Int J Parasitol ; 48(5): 379-385, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29510118

RESUMO

We recently reported the discovery of a new parasite-derived protein that functionally mimics the immunosuppressive cytokine transforming growth factor (TGF)-ß. The Heligmosomoides polygyrus TGF-ß Mimic (Hp-TGM) shares no homology to any TGF-ß family member, however it binds the mammalian TGF-ß 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-ß 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.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas de Helminto/química , Proteínas de Helminto/metabolismo , Nematospiroides dubius/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Clonagem Molecular , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Helminto/genética , Camundongos , Baço/citologia , Fator de Crescimento Transformador beta/genética
7.
Immunity ; 47(4): 739-751.e5, 2017 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-29045903

RESUMO

Infection by helminth parasites is associated with amelioration of allergic reactivity, but mechanistic insights into this association are lacking. Products secreted by the mouse parasite Heligmosomoides polygyrus suppress type 2 (allergic) immune responses through interference in the interleukin-33 (IL-33) pathway. Here, we identified H. polygyrus Alarmin Release Inhibitor (HpARI), an IL-33-suppressive 26-kDa protein, containing three predicted complement control protein (CCP) modules. In vivo, recombinant HpARI abrogated IL-33, group 2 innate lymphoid cell (ILC2) and eosinophilic responses to Alternaria allergen administration, and diminished eosinophilic responses to Nippostrongylus brasiliensis, increasing parasite burden. HpARI bound directly to both mouse and human IL-33 (in the cytokine's activated state) and also to nuclear DNA via its N-terminal CCP module pair (CCP1/2), tethering active IL-33 within necrotic cells, preventing its release, and forestalling initiation of type 2 allergic responses. Thus, HpARI employs a novel molecular strategy to suppress type 2 immunity in both infection and allergy.


Assuntos
Proteínas de Helminto/imunologia , Interleucina-33/imunologia , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Alérgenos/imunologia , Alternaria/imunologia , Sequência de Aminoácidos , Animais , Western Blotting , Eosinófilos/imunologia , Proteínas de Helminto/genética , Proteínas de Helminto/metabolismo , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunidade Inata/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33/genética , Interleucina-33/metabolismo , Linfócitos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nematospiroides dubius/genética , Nematospiroides dubius/metabolismo , Ligação Proteica/imunologia , Receptores de Interleucina/imunologia , Receptores de Interleucina/metabolismo , Homologia de Sequência de Aminoácidos , Infecções por Strongylida/metabolismo , Infecções por Strongylida/parasitologia
8.
Cell Rep ; 19(8): 1545-1557, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28538175

RESUMO

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.


Assuntos
Vesículas Extracelulares/metabolismo , Imunidade , Ativação de Macrófagos , Nematospiroides dubius/metabolismo , Parasitos/metabolismo , Vacinas/imunologia , Animais , Anticorpos Anti-Helmínticos/imunologia , Formação de Anticorpos/efeitos dos fármacos , Células da Medula Óssea/citologia , Citocalasina D/farmacologia , Vesículas Extracelulares/efeitos dos fármacos , Imunidade/efeitos dos fármacos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Parasitos/efeitos dos fármacos , Receptores de Interleucina/metabolismo , Vacinação
9.
J Vis Exp ; (98): e52412, 2015 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-25867600

RESUMO

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).


Assuntos
Nematospiroides dubius/crescimento & desenvolvimento , Nematospiroides dubius/metabolismo , Animais , Estágios do Ciclo de Vida , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA
10.
Exp Parasitol ; 135(3): 524-31, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24036322

RESUMO

Saponins of marigold (Calendula officinalis), in particular derivatives of 3-O-monoglucuronide of oleanolic acid, are able to reduce infectivity of Heligmosomoides polygyrus in mice. The purpose of this study was to understand the immune activation provoked by third-stage larvae exposed to marigold glucuronides. We also examined the pattern of glycosylation of larval antigens which appeared to be crucial for induction of cytokine production in BALB/c mice; higher concentrations of IL-6, IFN-γ, IL-10 and TNF-α were observed in serum or intestine one week post infection. Three weeks later, in the chronic phase of infection, cells in culture were able to produce IL-6, IFN-γ, TNF-α and IL-17. Restimulation of cells with H. polygyrus antigen resulted in reduced production of IL-6, and TNF-α. The pattern of cytokine production co-existed with reduced expression of terminal glucose, α-linked mannose, N-acetyl-galactosamine, ß-galactose, N-acetyl-glucosamine and α-fucose in several protein bands. Galactose, as a new terminal carbohydrate residue appeared in 20-24kDa protein bands. The number of immunogenic epitopes in parasitic antigens was reduced; only three protein bands of 56, 26 and 12kDa were recognized by IgG1. These studies provide a model system to find the glycosylated molecules expressed on nematodes that improve establishment and survival and characterize cytokine production in mice infected with larvae exposed to saponin. Identification of these molecules is the first step in the recognition of key antigenic epitopes able to induce protective or tolerogenic immune responses.


Assuntos
Glicoproteínas/química , Nematospiroides dubius/imunologia , Saponinas/farmacologia , Infecções por Strongylida/imunologia , Animais , Antígenos de Helmintos/análise , Antígenos de Helmintos/imunologia , Citocinas/metabolismo , Glucuronídeos/farmacologia , Glicoproteínas/efeitos dos fármacos , Glicoproteínas/imunologia , Glicosilação/efeitos dos fármacos , Soros Imunes/imunologia , Imunoglobulina G/imunologia , Larva/efeitos dos fármacos , Larva/imunologia , Larva/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nematospiroides dubius/efeitos dos fármacos , Nematospiroides dubius/metabolismo , Ácido Oleanólico/metabolismo , Ácido Oleanólico/farmacologia , Extratos Vegetais/farmacologia , Infecções por Strongylida/parasitologia , Tagetes/química
11.
J R Soc Interface ; 10(80): 20120588, 2013 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-23256186

RESUMO

Co-infection by multiple parasites is common within individuals. Interactions between co-infecting parasites include resource competition, direct competition and immune-mediated interactions and each are likely to alter the dynamics of single parasites. We posit that co-infection is a driver of variation in parasite establishment and growth, ultimately altering the production of parasite transmission stages. To test this hypothesis, three different treatment groups of laboratory mice were infected with the gastrointestinal helminth Heligmosomoides polygyrus, the respiratory bacterial pathogen Bordetella bronchiseptica lux(+) or co-infected with both parasites. To follow co-infection simultaneously, self-bioluminescent bacteria were used to quantify infection in vivo and in real-time, while helminth egg production was monitored in real-time using faecal samples. Co-infection resulted in high bacterial loads early in the infection (within the first 5 days) that could cause host mortality. Co-infection also produced helminth 'super-shedders'; individuals that chronically shed the helminth eggs in larger than average numbers. Our study shows that co-infection may be one of the underlying mechanisms for the often-observed high variance in parasite load and shedding rates, and should thus be taken into consideration for disease management and control. Further, using self-bioluminescent bacterial reporters allowed quantification of the progression of infection within the whole animal of the same individuals at a fine temporal scale (daily) and significantly reduced the number of animals used (by 85%) compared with experiments that do not use in vivo techniques. Thus, we present bioluminescent imaging as a novel, non-invasive tool offering great potential to be taken forward into other applications of infectious disease ecology.


Assuntos
Infecções por Bordetella/metabolismo , Bordetella bronchiseptica/metabolismo , Coinfecção/metabolismo , Nematospiroides dubius/metabolismo , Infecções por Strongylida/metabolismo , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Nematospiroides dubius/microbiologia , Óvulo/metabolismo
12.
Immunology ; 138(4): 370-81, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23240853

RESUMO

Modulation and suppression of the immune response of the host by nematode parasites have been reported extensively and the cysteine protease inhibitor (CPI or cystatin) is identified as one of the major immunomodulators. In the present study, we cloned and produced recombinant CPI protein from the murine nematode parasite Heligmosomoides polygyrus (rHp-CPI) and investigated its immunomodulatory effects on dendritic cell (DC) function and immune responses in mice. Bone-marrow-derived CD11c(+) DC (BMDC) that were exposed to rHp-CPI during the differentiation stage showed reduced MHC-II molecule expression compared with BMDC that were generated in normal culture conditions. The BMDC generated in the presence of rHp-CPI also exhibited reduced expression of CD40, CD86 and MHC-II molecules and reduced interleukin-6 and tumour necrosis factor-α cytokine production when stimulated with Toll-like receptor ligand CpG. Activation of BMDC generated in normal conditions induced by lipopolysaccharide and CpG was also suppressed by rHp-CPI, as shown by reduced co-stimulatory molecule expression and cytokine production. Furthermore, BMDC treated with rHp-CPI before ovalbumin (OVA) antigen pulsing induced a weaker proliferation response and less interferon-γ production of OVA-specific CD4(+) T cells compared with BMDC without rHp-CPI pre-treatment. Adoptive transfer of rHp-CPI-treated and OVA-loaded BMDC to mice induced significantly lower levels of antigen-specific antibody response than the BMDC loaded with antigen alone. These results demonstrated that the CPI from nematode parasites is able to modulate differentiation and activation stages of BMDC. It also interferes with antigen and MHC-II molecule processing and Toll-like receptor signalling pathway, resulting in functionally deficient DC that induce a suboptimum immune response.


Assuntos
Cistatinas/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Células Dendríticas/efeitos dos fármacos , Proteínas de Helminto/farmacologia , Nematospiroides dubius/imunologia , Infecções por Strongylida/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Antígenos CD/genética , Antígenos CD/imunologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cistatinas/genética , Cistatinas/imunologia , Cisteína Proteases/metabolismo , Inibidores de Cisteína Proteinase/genética , Inibidores de Cisteína Proteinase/imunologia , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Escherichia coli , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Interações Hospedeiro-Parasita , Camundongos , Camundongos Endogâmicos BALB C , Nematospiroides dubius/metabolismo , Ovalbumina/imunologia , Ovalbumina/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Infecções por Strongylida/parasitologia , Infecções por Strongylida/patologia
13.
J Exp Med ; 207(11): 2331-41, 2010 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-20876311

RESUMO

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) ß receptor and promoted Smad2/3 phosphorylation. Foxp3 induction by HES was lost in dominant-negative TGF-ßRII cells and was abolished by the TGF-ß 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-ß did not recognize HES, whereas antisera that inhibited HES did not affect TGF-ß. 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.


Assuntos
Antígenos de Helmintos/imunologia , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica/imunologia , Nematospiroides dubius/imunologia , Transdução de Sinais/imunologia , Infecções por Strongylida/imunologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Antígenos de Helmintos/metabolismo , Benzamidas/farmacologia , Proliferação de Células/efeitos dos fármacos , Doença Crônica , Dioxóis/farmacologia , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Interações Hospedeiro-Parasita/efeitos dos fármacos , Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Nematospiroides dubius/metabolismo , Fosforilação/genética , Fosforilação/imunologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/imunologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Smad2/genética , Proteína Smad2/imunologia , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/imunologia , Proteína Smad3/metabolismo , Infecções por Strongylida/genética , Infecções por Strongylida/metabolismo , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
14.
Parasitol Int ; 58(4): 461-70, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19751847

RESUMO

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.


Assuntos
Duodeno/parasitologia , Lectinas Tipo C/metabolismo , Nematospiroides dubius/crescimento & desenvolvimento , Nippostrongylus/crescimento & desenvolvimento , Animais , Biblioteca Gênica , Interações Hospedeiro-Parasita , Larva/metabolismo , Lectinas Tipo C/genética , Estágios do Ciclo de Vida , Camundongos , Dados de Sequência Molecular , Nematospiroides dubius/genética , Nematospiroides dubius/metabolismo , Nippostrongylus/genética , Nippostrongylus/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
15.
Proteomics ; 6(16): 4633-45, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16858733

RESUMO

The soluble global proteome of adult nematode Heligmosomoides polygyrus (H. p.) bakeri, a hookworm laboratory model was compared for the first time in the intestines of a slow-responder mouse host strain (C57/BL10) that is known to support a primary parasite infection for many months, and rapid-responder mouse host (SWR) that is known to eliminate the nematode infection by week 6 postinfection. At week 4 postinfection, major adult nematode proteins selectively produced following establishment of infection in C57/BL10 hosts include several globin forms, calreticulin and a phosphatidylethanolamine-binding protein. The increased synthesis of forms of myosin, actin and troponin in the nematode living in the rapid-responder SWR host may relate to the attempted reorganisation or repair of the cytoskeleton and/or muscle layer in the host immune initiated, increased mucus production and smooth muscle activity within intestinal environment. Initial evidence suggests weakly antigenic forms of globins dominant in the cytosol of H. p. bakeri adults in the intestinal environment compared to their low production in a related free-living nematode. The demonstration of considerable plasticity within a parasitic nematode proteome provides a molecular basis for the previously observed phenotypic plasticity within different host environments. Proteome plasticity has relevance to the efficiency of future vaccine and drug therapy, and the continued failure of defined antigen vaccines in mammalian populations.


Assuntos
Proteínas de Helminto/análise , Intestinos/parasitologia , Nematospiroides dubius/metabolismo , Proteoma/análise , Infecções por Strongylida/metabolismo , Sequência de Aminoácidos , Animais , Calreticulina/metabolismo , Proteínas do Citoesqueleto/análise , Eletroforese em Gel Bidimensional , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Especificidade da Espécie
16.
Vet Parasitol ; 116(1): 23-34, 2003 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-14519324

RESUMO

Albizia anthelmintica Brong., belongs to the plant family Mimosaceae. The plant is widely used in East Africa by poor smallholder farmers and pastoralists to treat their livestock against internal parasites. The anthelmintic effects of water extracts from the bark of A. anthelmintica, obtained from three different geographic areas in Kenya and using different methods of preparation, were tested at different doses in sheep and mice infected with the nematode parasites Haemonchus contortus and Heligmosomoides polygyrus, respectively. Lambs were infected with 3000 infective larvae of H. contortus and treated with the plant preparations 28 days later, while mice were infected with 200 infective larvae of H. polygyrus and treated 18 days later. Proximate analysis established high levels of crude proteins in A. anthelmintica bark. Two sheep out of the 45 treated with the plant preparations suffered from transient bloat, which was relieved by dosing with a surfactant. Significant reductions in faecal egg counts were observed in lambs treated with A. anthelmintica in two of the three experiments undertaken, but the efficacy levels achieved were well below the 70% reduction required. Similar values of packed red cell volume and live weight gain were observed for treated and control lambs. There was no overall significant effect of treatment with A. anthelmintica on faecal egg and total worm counts in mice. A dose rate of 1000 mg/kg bodyweight of A. anthelmintica preparation resulted in death of all mice. The results show that A. anthelmintica at the doses and preparations used is not efficacious against H. contortus in sheep or against H. polygyrus in mice.


Assuntos
Albizzia/química , Hemoncose/tratamento farmacológico , Hemoncose/veterinária , Haemonchus/crescimento & desenvolvimento , Fitoterapia/veterinária , Extratos Vegetais/uso terapêutico , Doenças dos Ovinos/tratamento farmacológico , Doenças dos Ovinos/parasitologia , Animais , Fezes/parasitologia , Hemoncose/parasitologia , Hematócrito/veterinária , Quênia , Masculino , Camundongos , Nematospiroides dubius/crescimento & desenvolvimento , Nematospiroides dubius/metabolismo , Contagem de Ovos de Parasitas/veterinária , Fitoterapia/métodos , Distribuição Aleatória , Ovinos
17.
Parasitology ; 114 ( Pt 1): 79-84, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9011077

RESUMO

A characteristic feature of infections with the nematode parasite of mice Heligmosomoides polygyrus, is a marked IgG1 hypergammaglobulinaemia. A possible source for this immunoglobulin has recently been demonstrated, through evidence that H. polygyrus adult worm homogenate (AWH) can induce the in vitro production of non-specific IgG1 from mouse lymphocytes. To determine the interactions between this immunoglobulin and the parasite, the ability of IgG1 to bind to AWH of H. polygyrus was investigated. Protein (Western) blotting indicated that mouse monoclonal antibodies are able to bind non-specifically to selected parasite antigens. Furthermore, by binding H. polygyrus adult worm homogenate to cyanogen bromide (CNBr)-activated Sepharose CL-4B, an affinity column was prepared which could be used to efficiently purify mouse IgG1 monoclonal antibodies. These antibodies were eluted from the affinity column and still retained their original specificity. These results indicate that H. polygyrus not only induces the production of non-specific IgG1 by the host, it can also bind this immunoglobulin to its own specific proteins. Thus, it is possible that IgG1 produced during a primary infection with H. polygyrus may not entirely benefit the host.


Assuntos
Anticorpos Monoclonais/isolamento & purificação , Imunoglobulina G/metabolismo , Nematospiroides dubius/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos , Western Blotting , Cromatografia de Afinidade , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Imunoglobulina G/imunologia , Imunoglobulina G/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Nematospiroides dubius/imunologia , Fator Tímico Circulante/imunologia
18.
Mol Reprod Dev ; 45(3): 332-41, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8916044

RESUMO

Nematode spermatozoa are amoeboid cells. In Caenorhabditis elegans and Ascaris suum, previous studies have reported that sperm motility does not involve actin, but, instead, requires a specific cytoskeletal protein, namely major-sperm-protein (MSP). In Heligmosomoides polygyrus, a species with large and elongate spermatids and spermatozoa, cell organelles are easily identified even with light microscopy. Electrophoresis of Heligmosomoides sperm proteins indicates that the main protein band has a molecular weight of about 15 kDa, as MSP in other nematodes, and is specifically labelled by an anti-MSP antibody raised against C. elegans MSP. A minor band at 43 kDa was specifically labelled by an anti-actin antibody. Reaction of anti-actin and anti-MSP antibodies is specific to, and restricted to, their respective targets. Actin and MSP localisation, studied by indirect immunofluorescence in male germ cells of Heligmosomoides polygyrus, are similar: spermatids show rows of dots, corresponding to the fibrous bodies, around an unlabelled central longitudinal core; spermatozoa are labelled strictly in an anterior crescent-shaped cap, at the opposite pole to the nucleus, which contains fibres of the MSP cytoskeleton. Phalloidin labelling shows that F-actin is present in spermatids, but absent in spermatozoa. Tropomyosin shows a distinct pattern in spermatids, but is located in the MSP and actin-containing cap in spermatozoa. It is hypothesized that actin plays a role in the shaping of the cell and in the arrangement of its organelles during nematode spermiogenesis, when MSP is present, in an inactive state, in the fibrous bodies. The concentration of actin and tropomyosin in the anterior cap is not compatible with previous theories about the MSP cytoskeleton, which is supposed to act in the absence of actin.


Assuntos
Actinas/metabolismo , Proteínas de Helminto/metabolismo , Nematospiroides dubius/metabolismo , Espermátides/metabolismo , Espermatozoides/metabolismo , Animais , Especificidade de Anticorpos , Técnica Indireta de Fluorescência para Anticorpo , Masculino , Camundongos , Microscopia Eletrônica , Microscopia Eletrônica de Varredura , Espermatozoides/ultraestrutura
19.
J Parasitol ; 81(2): 302-3, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7707211

RESUMO

A glutathione-affinity matrix was used to identify glutathione-dependent protein(s) in somatic extracts of the nematode Heligmosomoides polygyrus. Polypeptides of 70-80 kDa were retained by the affinity matrix following the elution of H. polygyrus glutathione S-transferases by 5 mM glutathione. The 70-80-kDa polypeptides were subsequently eluted from the matrix by the addition of 20 mM glutathione and these polypeptides did not show glutathione S-transferase activity. The high-affinity H. polygyrus glutathione-binding proteins may be related to the uncharacterized purification hindering factors previously demonstrated during the isolation of glutathione S-transferases in several other helminths.


Assuntos
Glutationa/metabolismo , Proteínas de Helminto/metabolismo , Nematospiroides dubius/química , Animais , Cromatografia de Afinidade , Eletroforese em Gel de Poliacrilamida , Glutationa Transferase/isolamento & purificação , Proteínas de Helminto/química , Ponto Isoelétrico , Nematospiroides dubius/metabolismo
20.
Parasitol Res ; 81(4): 302-4, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-7624287

RESUMO

The in vivo N-dealkylation of [13C-2]-labeled aminopyrine by the L1-L2 larvae of Heligmosomoides polygyrus was demonstrated by the use of a sensitive gas chromatography-mass spectrometry method. This is the first evidence for the possible existence of a cytochrome P-450-dependent activity in helminths.


Assuntos
Aminopirina/metabolismo , Nematospiroides dubius/metabolismo , Antagonistas Adrenérgicos alfa/farmacologia , Aminopirina/antagonistas & inibidores , Animais , Cromatografia Gasosa , Remoção de Radical Alquila/efeitos dos fármacos , Dimetil Sulfóxido/farmacologia , Imidazóis/farmacologia , Larva/metabolismo , Espectrometria de Massas , Camundongos , Nematospiroides dubius/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA