Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Immunity ; 47(4): 739-751.e5, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045903

RESUMEN

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.


Asunto(s)
Proteínas del Helminto/inmunología , Interleucina-33/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Alérgenos/inmunología , Alternaria/inmunología , Secuencia de Aminoácidos , Animales , Western Blotting , Eosinófilos/inmunología , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunidad Innata/inmunología , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33/genética , Interleucina-33/metabolismo , Linfocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/genética , Nematospiroides dubius/metabolismo , Unión Proteica/inmunología , Receptores de Interleucina/inmunología , Receptores de Interleucina/metabolismo , Homología de Secuencia de Aminoácido , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología
2.
Eur J Immunol ; 50(11): 1663-1675, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32447774

RESUMEN

IL-1 is a powerful cytokine that drives inflammation and modulates adaptive immunity. Both IL-1α and IL-1ß are translated as proforms that require cleavage for full cytokine activity and release, while IL-1α is reported to occur as an alternative plasma membrane-associated form on many cell types. However, the existence of cell surface IL-1α (csIL-1α) is contested, how IL-1α tethers to the membrane is unknown, and signaling pathways controlling trafficking are not specified. Using a robust and fully validated system, we show that macrophages present bona fide csIL-1α after ligation of TLRs. Pro-IL-1α tethers to the plasma membrane in part through IL-1R2 or via association with a glycosylphosphatidylinositol-anchored protein, and can be cleaved, activated, and released by proteases. csIL-1α requires de novo protein synthesis and its trafficking to the plasma membrane is exquisitely sensitive to inhibition by IFN-γ, independent of expression level. We also reveal how prior csIL-1α detection could occur through inadvertent cell permeabilisation, and that senescent cells do not drive the senescent-associated secretory phenotype via csIL-1α, but rather via soluble IL-1α. We believe these data are important for determining the local or systemic context in which IL-1α can contribute to disease and/or physiological processes.


Asunto(s)
Membrana Celular/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Interferón gamma/metabolismo , Interleucina-1alfa/metabolismo , Receptores Tipo II de Interleucina-1/metabolismo , Animales , Humanos , Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica/fisiología , Transporte de Proteínas/fisiología
3.
PLoS Pathog ; 11(3): e1004676, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25816012

RESUMEN

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.


Asunto(s)
Anticuerpos Antihelmínticos/inmunología , Antígenos Helmínticos/inmunología , Inmunoglobulina G/inmunología , Interleucina-4/inmunología , Interleucinas/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Vacunación , Animales , Anticuerpos Antihelmínticos/genética , Humanos , Inmunoglobulina G/genética , Interleucina-4/genética , Interleucinas/genética , Larva/inmunología , Ratones , Ratones Noqueados , Infecciones por Strongylida/genética , Infecciones por Strongylida/prevención & control
4.
PLoS Pathog ; 9(8): e1003492, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23966853

RESUMEN

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.


Asunto(s)
Antígenos Helmínticos/metabolismo , Proteínas del Helminto/metabolismo , Larva/metabolismo , Infecciones por Nematodos/inmunología , Nematospiroides dubius/inmunología , Proteómica , Animales , Anticuerpos Antihelmínticos/análisis , Anticuerpos Antihelmínticos/inmunología , Antígenos Helmínticos/inmunología , Western Blotting , Cromatografía Liquida , Biología Computacional , Electroforesis en Gel Bidimensional , Ensayo de Inmunoadsorción Enzimática , Femenino , Perfilación de la Expresión Génica , Proteínas del Helminto/inmunología , Interacciones Huésped-Parásitos , Inmunización , Inmunoprecipitación , Larva/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Infecciones por Nematodos/parasitología , Nematospiroides dubius/crecimiento & desarrollo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Vacunación
5.
Immunol Cell Biol ; 92(5): 436-48, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24492801

RESUMEN

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.


Asunto(s)
Inmunidad Adaptativa , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Helmintiasis/genética , Helmintiasis/inmunología , Helmintos/inmunología , Inmunidad Innata , Enfermedades Intestinales/genética , Enfermedades Intestinales/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Anticuerpos Antihelmínticos/inmunología , Ácido Clodrónico/farmacología , Femenino , Granuloma , Helmintiasis/tratamiento farmacológico , Helmintiasis/metabolismo , Helmintiasis/patología , Helmintiasis Animal , Inmunidad Celular , Inmunidad Innata/efectos de los fármacos , Interferón gamma/metabolismo , Enfermedades Intestinales/tratamiento farmacológico , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/patología , Parasitosis Intestinales , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Receptores de Interleucina-4/metabolismo , Transducción de Señal
6.
Eur J Immunol ; 42(10): 2667-82, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22706967

RESUMEN

Allergic asthma is less prevalent in countries with parasitic helminth infections, and mice infected with parasites such as Heligmosomoides polygyrus are protected from allergic airway inflammation. To establish whether suppression of allergy could be mediated by soluble products of this helminth, we tested H. polygyrus excretory-secretory (HES) material for its ability to impair allergic inflammation. When HES was added to sensitising doses of ovalbumin, the subsequent allergic airway response was suppressed, with ablated cell infiltration, a lower ratio of effector (CD4(+) CD25(+) Foxp3(-) ) to regulatory (CD4(+) Foxp3(+) ) T (Treg) cells, and reduced Th1, Th2 and Th17 cytokine production. HES exposure reduced IL-5 responses and eosinophilia, abolished IgE production and inhibited the type 2 innate molecules arginase-1 and RELM-α (resistin-like molecule-α). Although HES contains a TGF-ß-like activity, similar effects in modulating allergy were not observed when administering mammalian TGF-ß alone. HES also protected previously sensitised mice, suppressing recruitment of eosinophils to the airways when given at challenge, but no change in Th or Treg cell populations was apparent. Because heat-treatment of HES did not impair suppression at sensitisation, but compromised its ability to suppress at challenge, we propose that HES contains distinct heat-stable and heat-labile immunomodulatory molecules, which modulate pro-allergic adaptive and innate cell populations.


Asunto(s)
Antígenos Helmínticos/administración & dosificación , Asma/inmunología , Citocinas/metabolismo , Eosinófilos/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Terapia con Helmintos , Animales , Arginasa/metabolismo , Asma/terapia , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Eosinófilos/efectos de los fármacos , Humanos , Inmunoglobulina E/sangre , Terapia de Inmunosupresión , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Linfocitos T Reguladores/efectos de los fármacos , Células Th2/efectos de los fármacos
7.
J Immunol ; 187(9): 4764-77, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21964031

RESUMEN

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.


Asunto(s)
Anticuerpos Antihelmínticos/biosíntesis , Antígenos Helmínticos/inmunología , Nematospiroides dubius/inmunología , Péptidos/inmunología , Polisacáridos/inmunología , Animales , Especificidad de Anticuerpos/inmunología , Antígenos Helmínticos/biosíntesis , Antígenos Helmínticos/genética , Epítopos/inmunología , Epítopos/metabolismo , Femenino , Sueros Inmunes/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Péptidos/metabolismo , Polisacáridos/metabolismo , Conformación Proteica , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología
8.
Exp Parasitol ; 132(1): 76-89, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21875581

RESUMEN

The intestinal nematode parasite Heligmosomoides polygyrus bakeri exerts widespread immunomodulatory effects on both the innate and adaptive immune system of the host. Infected mice adopt an immunoregulated phenotype, with abated allergic and autoimmune reactions. At the cellular level, infection is accompanied by expanded regulatory T cell populations, skewed dendritic cell and macrophage phenotypes, B cell hyperstimulation and multiple localised changes within the intestinal environment. In most mouse strains, these act to block protective Th2 immunity. The molecular basis of parasite interactions with the host immune system centres upon secreted products termed HES (H. polygyrus excretory-secretory antigen), which include a TGF-ß-like ligand that induces de novo regulatory T cells, factors that modify innate inflammatory responses, and molecules that block allergy in vivo. Proteomic and transcriptomic definition of parasite proteins, combined with biochemical identification of immunogenic molecules in resistant mice, will provide new candidate immunomodulators and vaccine antigens for future research.


Asunto(s)
Inmunomodulación , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Inmunidad Adaptativa/inmunología , Animales , Anticuerpos Antihelmínticos/inmunología , Especificidad de Anticuerpos , Antígenos Helmínticos/inmunología , Linfocitos B/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Humanos , Inmunidad Innata/inmunología , Ratones , Fenotipo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
9.
Mucosal Immunol ; 15(6): 1243-1256, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35288645

RESUMEN

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 RELMß, 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-ß, 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.


Asunto(s)
Factores Inhibidores de la Migración de Macrófagos , Infecciones por Strongylida , Ratones , Animales , Factores Inhibidores de la Migración de Macrófagos/genética , Inmunidad Innata , Linfocitos , Nippostrongylus
10.
Front Immunol ; 11: 950, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32508831

RESUMEN

Helminths regulate host immune responses to ensure their own long-term survival. Numerous studies have demonstrated that these helminth-induced regulatory mechanisms can also limit host inflammatory responses in several disease models. We used the Heligmosomoides bakeri (Hb) infection model (also known as H. polygyrus or H. polygyrus bakeri in the literature) to test whether such immune regulation affects skin inflammatory responses induced by the model contact sensitiser dibutyl phthalate fluorescein isothiocynate (DBP-FITC). Skin lysates from DBP-FITC-sensitized, Hb-infected mice produced less neutrophil specific chemokines and had significantly reduced levels of skin thickening and cellular inflammatory responses in tissue and draining lymph nodes (LNs) compared to uninfected mice. Hb-induced suppression did not appear to be mediated by regulatory T cells, nor was it due to impaired dendritic cell (DC) activity. Mice cleared of infection remained unresponsive to DBP-FITC sensitization indicating that suppression was not via the secretion of Hb-derived short-lived regulatory molecules, although long-term effects on cells cannot be ruled out. Importantly, similar helminth-induced suppression of inflammation was also seen in the draining LN after intradermal injection of the ubiquitous allergen house dust mite (HDM). These findings demonstrate that Hb infection attenuates skin inflammatory responses by suppressing chemokine production and recruitment of innate cells. These findings further contribute to the growing body of evidence that helminth infection can modulate inflammatory and allergic responses via a number of mechanisms with potential to be exploited in therapeutic and preventative strategies in the future.


Asunto(s)
Dermatitis por Contacto/inmunología , Tracto Gastrointestinal/parasitología , Heligmosomatoidea/inmunología , Inflamación/parasitología , Infecciones por Strongylida/inmunología , Animales , Quimiocinas/inmunología , Células Dendríticas/inmunología , Dermatitis por Contacto/parasitología , Dermatitis por Contacto/prevención & control , Modelos Animales de Enfermedad , Femenino , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Piel/inmunología , Piel/parasitología , Piel/patología
11.
Mucosal Immunol ; 12(2): 352-362, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30401814

RESUMEN

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


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Coinfección , Eosinófilos/inmunología , Interleucina-5/metabolismo , Pulmón/inmunología , Nematospiroides dubius/fisiología , Nippostrongylus/fisiología , Infecciones por Strongylida/inmunología , Tricuriasis/inmunología , Trichuris/fisiología , Animales , Antígenos Helmínticos/inmunología , Movimiento Celular , Células Cultivadas , Citotoxicidad Inmunológica , Femenino , Interacciones Huésped-Parásitos , Inmunidad , Interleucina-33/metabolismo , Pulmón/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Front Immunol ; 10: 2375, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708913

RESUMEN

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


Asunto(s)
Inmunidad Celular , Oxidorreductasas Intramoleculares/inmunología , Activación de Macrófagos , Factores Inhibidores de la Migración de Macrófagos/inmunología , Macrófagos/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Animales , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/genética , Macrófagos/patología , Ratones Endogámicos BALB C , Ratones Mutantes , Infecciones por Strongylida/genética , Infecciones por Strongylida/patología , Linfocitos T Reguladores/patología
13.
Nat Commun ; 8(1): 1741, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29170498

RESUMEN

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-ß signalling, but the identity of the active molecule is unknown. Here we identify an H. polygyrus TGF-ß mimic (Hp-TGM) that replicates the biological and functional properties of TGF-ß, including binding to mammalian TGF-ß receptors and inducing mouse and human Foxp3+ Treg cells. Hp-TGM has no homology with mammalian TGF-ß or other members of the TGF-ß 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.


Asunto(s)
Imitación Molecular/inmunología , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Secuencia de Aminoácidos , Animales , Antígenos Helmínticos/química , Antígenos Helmínticos/genética , Antígenos Helmínticos/inmunología , Femenino , Proteínas del Helminto/química , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Imitación Molecular/genética , Nematospiroides dubius/genética , Unión Proteica , Dominios Proteicos , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología
15.
Nat Commun ; 6: 5862, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25575242

RESUMEN

Presentation of peptide:MHCII by RORγ-expressing group 3 innate lymphoid cells (ILC3s), which are enriched within gut tissue, is required for control of CD4 T-cell responses to commensal bacteria. It is not known whether ILC populations migrate from their mucosal and peripheral sites to local draining secondary lymphoid tissues. Here we demonstrate that ILC3s reside within the interfollicular areas of mucosal draining lymph nodes, forming a distinct microenvironment not observed in peripheral lymph nodes. By photoconverting intestinal cells in Kaede mice we reveal constitutive trafficking of ILCs from the intestine to the draining mesenteric lymph nodes, which specifically for the LTi-like ILC3s was CCR7-dependent. Thus, ILC populations traffic to draining lymph nodes using different mechanisms.


Asunto(s)
Ganglios Linfáticos/citología , Linfocitos/citología , Membrana Mucosa/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores CCR7/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Movimiento Celular , Femenino , Inmunidad Innata , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Luz , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Membrana Mucosa/patología
16.
Gut Microbes ; 5(4): 522-32, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25144609

RESUMEN

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.


Asunto(s)
Tracto Gastrointestinal/microbiología , Lactobacillus/fisiología , Interacciones Microbianas , Nematospiroides dubius/fisiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Tracto Gastrointestinal/inmunología , Interacciones Huésped-Patógeno , Lactobacillus/crecimiento & desarrollo , Lactobacillus/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nematospiroides dubius/crecimiento & desarrollo , Nematospiroides dubius/inmunología
17.
Semin Immunopathol ; 34(6): 829-46, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23053394

RESUMEN

Heligmosomoides polygyrus is a natural intestinal parasite of mice, which offers an excellent model of the immunology of gastrointestinal helminth infections of humans and livestock. It is able to establish long-term chronic infections in many strains of mice, exerting potent immunomodulatory effects that dampen both protective immunity and bystander reactions to allergens and autoantigens. Immunity to the parasite develops naturally in some mouse strains and can be induced in others through immunization; while the mechanisms of protective immunity are not yet fully defined, both antibodies and a host cellular component are required, with strongest evidence for a role of alternatively activated macrophages. We discuss the balance between resistance and susceptibility in this model system and highlight new themes in innate and adaptive immunity, immunomodulation, and regulation of responsiveness in helminth infection.


Asunto(s)
Parasitosis Intestinales/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Inmunidad Adaptativa , Animales , Enfermedad Crónica , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Predisposición Genética a la Enfermedad , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunidad Innata , Parasitosis Intestinales/genética , Parasitosis Intestinales/microbiología , Metagenoma , Vacunas Antiprotozoos/inmunología , Infecciones por Strongylida/genética
19.
J Proteomics ; 74(9): 1573-94, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21722761

RESUMEN

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.


Asunto(s)
Enfermedades Gastrointestinales/parasitología , Proteínas del Helminto/análisis , Nematospiroides dubius/química , Proteómica , Animales , Antígenos Helmínticos , Modelos Animales de Enfermedad , Proteínas del Helminto/metabolismo , Proteómica/métodos
20.
J Exp Med ; 207(11): 2331-41, 2010 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-20876311

RESUMEN

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.


Asunto(s)
Antígenos Helmínticos/inmunología , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Nematospiroides dubius/inmunología , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Antígenos Helmínticos/metabolismo , Benzamidas/farmacología , Proliferación Celular/efectos de los fármacos , Enfermedad Crónica , Dioxoles/farmacología , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Interacciones Huésped-Parásitos/efectos de los fármacos , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Nematospiroides dubius/metabolismo , Fosforilación/genética , Fosforilación/inmunología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Smad2/genética , Proteína Smad2/inmunología , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/inmunología , Proteína smad3/metabolismo , Infecciones por Strongylida/genética , Infecciones por Strongylida/metabolismo , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA