Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 239
Filtrar
Más filtros

Intervalo de año de publicación
1.
PLoS Pathog ; 17(11): e1010067, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34784389

RESUMEN

Human ascariasis is the most prevalent but neglected tropical disease in the world, affecting approximately 450 million people. The initial phase of Ascaris infection is marked by larval migration from the host's organs, causing mechanical injuries followed by an intense local inflammatory response, which is characterized mainly by neutrophil and eosinophil infiltration, especially in the lungs. During the pulmonary phase, the lesions induced by larval migration and excessive immune responses contribute to tissue remodeling marked by fibrosis and lung dysfunction. In this study, we investigated the relationship between SIgA levels and eosinophils. We found that TLR2 and TLR4 signaling induces eosinophils and promotes SIgA production during Ascaris suum infection. Therefore, control of parasite burden during the pulmonary phase of ascariasis involves eosinophil influx and subsequent promotion of SIgA levels. In addition, we also demonstrate that eosinophils also participate in the process of tissue remodeling after lung injury caused by larval migration, contributing to pulmonary fibrosis and dysfunction in re-infected mice. In conclusion, we postulate that eosinophils play a central role in mediating host innate and humoral immune responses by controlling parasite burden, tissue inflammation, and remodeling during Ascaris suum infection. Furthermore, we suggest that the use of probiotics can induce eosinophilia and SIgA production and contribute to controlling parasite burden and morbidity of helminthic diseases with pulmonary cycles.


Asunto(s)
Ascariasis/inmunología , Ascaris suum/inmunología , Eosinófilos/fisiología , Inmunoglobulina A Secretora/metabolismo , Neumonía/prevención & control , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Ascariasis/metabolismo , Ascariasis/parasitología , Femenino , Inmunoglobulina A Secretora/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neumonía/inmunología , Neumonía/parasitología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética
2.
Exp Parasitol ; 221: 108049, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33307097

RESUMEN

Globally, ascariasis ranks as the second leading intestinal helminth infection. However, progress in developing better control strategies, such as vaccines, remains slow-paced. This study aims to measure antibody production and parasite load in male BALB/c mice immunized with crude Ascaris suum intestinal tract homogenate. Thirty-two (32) mice were randomized into: (1) unvaccinated, uninfected (UU); (2) unvaccinated, infected (UI); (3) vaccinated, uninfected (VU); and (4) vaccinated, infected (VI) groups. A 100-µL vaccine containing 50 µg of homogenized A. suum intestines and Complete Freund's Adjuvant (1:1) were introduced intraperitoneally. Immunizations were done on days 0, 10, and 20. Oral gavage with 1000 embryonated eggs was done on day 30. Blood was obtained at day 40. To measure serum IgG levels, indirect ELISA was done. Microtiter plates were coated with 100 µg larval homogenate, and HRP-conjugated anti-mouse IgG was used as secondary antibody. Parasite load was measured in lung and liver tissues. Tukey's HSD of signal to cut-off ratios of absorbance readings obtained in indirect ELISA procedure for the 1:200 serum dilution showed statistically significant difference between the UU and VI (p = 0.026) as well as between UI and VI (p = 0.003) groups. No statistically significant difference in parasite load was observed in the lungs (p = 0.074), liver (p = 0.130), and both lungs and liver (p = 0.101). Immunization elicited a significant larva-directed IgG production. However, there is no significant difference in parasite loads in either lung or liver tissues across all treatment groups as the larval counts obtained from the study were very low and may not be indicative of the actual parasite load in mice.


Asunto(s)
Anticuerpos Antihelmínticos/biosíntesis , Antígenos Helmínticos/biosíntesis , Ascaris suum/inmunología , Inmunoglobulina G/biosíntesis , Análisis de Varianza , Animales , Anticuerpos Antihelmínticos/inmunología , Antígenos Helmínticos/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunización/métodos , Inmunoglobulina G/inmunología , Intestinos/parasitología , Larva/inmunología , Hígado/parasitología , Pulmón/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Carga de Parásitos , Distribución Aleatoria
3.
Immunology ; 159(3): 322-334, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31705653

RESUMEN

Dendritic cells (DCs) are essential for generating T-cell-based immune responses through sensing of potential inflammatory and metabolic cues in the local environment. However, there is still limited insight into the processes defining the resultant DC phenotype, including the type of early transcriptional changes in pro-inflammatory cues towards regulatory or type 2 immune-based cues induced by a variety of exogenous and endogenous molecules. Here we compared the ability of a selected number of molecules to modulate the pro-inflammatory phenotype of lipopolysaccharide (LPS) and interferon-γ (IFN-γ)-stimulated human monocyte-derived DCs towards an anti-inflammatory or regulatory phenotype, including Ascaris suum body fluid [helminth pseudocoelomic fluid (PCF)], the metabolites succinate and butyrate, and the type 2 cytokines thymic stromal lymphopoietin and interleukin-25. Our data show that helminth PCF and butyrate treatment suppress the T helper type 1 (Th1)-inducing pro-inflammatory DC phenotype through induction of different transcriptional programs in DCs. RNA sequencing indicated that helminth PCF treatment strongly inhibited the Th1 and Th17 polarizing ability of LPS + IFN-γ-matured DCs by down-regulating myeloid differentiation primary response gene 88 (MyD88)-dependent and MyD88-independent pathways in Toll-like receptor 4 signaling. By contrast, butyrate treatment had a strong Th1-inhibiting action, and transcripts encoding important gut barrier defending factors such as IL18, IL1B and CXCL8 were up-regulated. Collectively, our results further understanding of how compounds from parasites and gut microbiota-derived butyrate may exert immunomodulatory effects on the host immune system.


Asunto(s)
Ascaris suum/inmunología , Líquidos Corporales/inmunología , Células Dendríticas/inmunología , Mediadores de Inflamación/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Ascaris suum/metabolismo , Ascaris suum/patogenicidad , Bacterias/inmunología , Bacterias/metabolismo , Bacterias/patogenicidad , Líquidos Corporales/metabolismo , Butiratos/farmacología , Comunicación Celular , Citocinas/metabolismo , Citocinas/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Microbioma Gastrointestinal , Interacciones Huésped-Parásitos , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Células TH1/metabolismo , Células Th17/metabolismo , Receptor Toll-Like 4/metabolismo
4.
Parasite Immunol ; 42(7): e12728, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32394439

RESUMEN

Helminth infection represents a major health problem causing approximately 5 million disability-adjusted life years worldwide. Concerns that repeated anti-helminthic treatment may lead to drug resistance render it important that vaccines are developed but will require increased understanding of the immune-mediated cellular and antibody responses to helminth infection. IL-4 or antibody-activated murine macrophages are known to immobilize parasitic nematode larvae, but few studies have addressed whether this is translatable to human macrophages. In the current study, we investigated the capacity of human macrophages to recognize and attack larval stages of Ascaris suum, a natural porcine parasite that is genetically similar to the human helminth Ascaris lumbricoides. Human macrophages were able to adhere to and trap A suum larvae in the presence of either human or pig serum containing Ascaris-specific antibodies and other factors. Gene expression analysis of serum-activated macrophages revealed that CCL24, a potent eosinophil attractant, was the most upregulated gene following culture with A suum larvae in vitro, and human eosinophils displayed even greater ability to adhere to, and trap, A suum larvae. These data suggest that immune serum-activated macrophages can recruit eosinophils to the site of infection, where they act in concert to immobilize tissue-migrating Ascaris larvae.


Asunto(s)
Ascariasis/inmunología , Ascaris suum/inmunología , Quimiocina CCL24/metabolismo , Eosinófilos/inmunología , Macrófagos/inmunología , Animales , Anticuerpos Antihelmínticos/sangre , Formación de Anticuerpos , Ascaris lumbricoides/inmunología , Humanos , Sueros Inmunes/farmacología , Larva/inmunología , Recuento de Leucocitos , Ratones , Porcinos , Enfermedades de los Porcinos/inmunología , Vacunas/inmunología
5.
J Infect Dis ; 217(2): 310-319, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29136163

RESUMEN

Ascaris suum is a helminth parasite of pigs closely related to its human counterpart, A. lumbricoides, which infects almost 1 billion people. Ascaris is thought to modulate host immune and inflammatory responses, which may drive immune hyporesponsiveness during chronic infections. Using transcriptomic analysis, we show here that pigs with a chronic A. suum infection have a substantial suppression of inflammatory pathways in the intestinal mucosa, with a broad downregulation of genes encoding cytokines and antigen-processing and costimulatory molecules. A. suum body fluid (ABF) suppressed similar transcriptional pathways in human dendritic cells (DCs) in vitro. DCs exposed to ABF secreted minimal amounts of cytokines and had impaired production of cyclooxygengase-2, altered glucose metabolism, and reduced capacity to induce interferon-gamma production in T cells. Our in vivo and in vitro data provide an insight into mucosal immune modulation during Ascaris infection, and show that A. suum profoundly suppresses immune and inflammatory pathways.


Asunto(s)
Ascariasis/patología , Ascaris suum/inmunología , Células Dendríticas/inmunología , Tolerancia Inmunológica , Mucosa Intestinal/patología , Animales , Ascariasis/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Mucosa Intestinal/inmunología , Modelos Biológicos , Porcinos
6.
Am J Respir Cell Mol Biol ; 58(1): 55-65, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28850259

RESUMEN

S28463 (S28), a ligand for Toll-like receptor 7/8, has been shown to have antiinflammatory properties in rodent models of allergic asthma. The principle goal of this study was to assess whether these antiinflammatory effects can also be observed in a nonhuman primate (NHP) model of allergic asthma. NHPs were sensitized then challenged with natural allergen, Ascaris suum extract. The animals were treated with S28 orally before each allergen challenge. The protective effect of S28 in NHPs was assessed by measuring various asthma-related phenotypes. We also characterized the metabolomic and proteomic signatures of the lung environment and plasma to identify markers associated with the disease and treatment. Our data demonstrate that clinically relevant parameters, such as wheal and flare response, blood IgE levels, recruitment of white blood cells to the bronchoalveolar space, and lung responsiveness, are decreased in the S28-treated allergic NHPs compared with nontreated allergic NHPs. Furthermore, we also identified markers that can distinguish allergic from nonallergic or allergic and drug-treated NHPs, such as metabolites, phosphocreatine and glutathione, in the plasma and BAL fluid, respectively; and inflammatory cytokines, IL-5 and IL-13, in the bronchoalveolar lavage fluid. Our preclinical study demonstrates that S28 has potential as a treatment for allergic asthma in primate species closely related to humans. Combined with our previous findings, we demonstrate that S28 is effective in different models of asthma and in different species, and has the antiinflammatory properties clinically relevant for the treatment of allergic asthma.


Asunto(s)
Alérgenos/toxicidad , Ascaris suum/química , Asma , Proteínas del Helminto/toxicidad , Receptor Toll-Like 7 , Receptor Toll-Like 8 , Animales , Ascaris suum/inmunología , Asma/inducido químicamente , Asma/inmunología , Asma/patología , Interleucina-13/inmunología , Interleucina-5/inmunología , Macaca fascicularis , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 8/agonistas , Receptor Toll-Like 8/inmunología
7.
Parasite Immunol ; 40(5): e12525, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29574798

RESUMEN

Giardia duodenalis is a common intestinal protozoan parasite known to modulate host immune responses, including dendritic cell (DC) function. Coinfections of intestinal pathogens are common, and thus, DCs may be concurrently exposed to antigens from multiple parasites. Here, we investigated the effects of G. duodenalis products on human monocyte-derived DC function independently and in combination with helminth antigens (Ascaris suum and Trichuris suis). All antigens individually induced an anti-inflammatory phenotype in DCs, reducing lipopolysaccharide (LPS)-induced interleukin (IL)-6, IL-12p70 and tumour necrosis factor (TNF)-α secretion. G. duodenalis and T. suis products also consistently upregulated IL-10 production. Despite a similar modulation of cytokine secretion, additive effects between Giardia and helminth products were not observed, indicating a dominant effect of a single parasite stimulus and limited interactive effects on DC function. G. duodenalis trophozoites induced rapid apoptosis in DCs, which was not observed with the helminth antigens suggesting that the modulatory effects of G. duodenalis may override that of A. suum and T. suis. Thus, G. duodenalis modulates DC activity by modulating cytokine secretion and/or inducing apoptosis, which may be a parasite-driven mechanism to dampen host immunity and establish chronic infections. The differential mechanisms of DC modulation by intestinal parasites warrant further attention.


Asunto(s)
Antígenos Helmínticos/inmunología , Ascaris suum/inmunología , Células Dendríticas/inmunología , Giardia lamblia/inmunología , Giardiasis/inmunología , Trichuris/inmunología , Animales , Apoptosis/inmunología , Células Cultivadas , Giardiasis/parasitología , Giardiasis/patología , Humanos , Subunidad p35 de la Interleucina-12/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
8.
Parasite Immunol ; 39(9)2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28703913

RESUMEN

Helminth infections have the ability to modulate host's immune response through mechanisms that allow the chronic persistence of the worms in the host. Here, we investigated the mechanisms involved on the suppressive effect of Ascaris suum infection using a murine experimental model of LPS-induced inflammation. We found that infection with A. suum markedly inhibited leucocyte influx induced by LPS into air pouches, suppressed secretion of pro-inflammatory cytokines (IL-1ß, TNF-α and IL-6) and induced high levels of IL-10 and TGF-ß. Augmented frequency of CD4+ CD25high Foxp3+ T cells was observed in the mesenteric lymph nodes of infected mice. Adoptive transfer of purified CD4+ CD25+ T cells to recipient uninfected mice demonstrated that these cells were able to induce a suppressive effect in the LPS-induced inflammation in air pouch model. In addition, adoptive transfer of CD4+ CD25+ T cells derived from IL-10 knockout mice suggests that this suppressive effect of A. suum infection involves IL-10 cytokine. In conclusion, our results demonstrated that A. suum experimental infection was capable of suppressing LPS-induced inflammation by mechanisms, which seem to be dependent on responses of CD4+ CD25+ T cells and secretion of IL-10 cytokine.


Asunto(s)
Ascariasis/inmunología , Ascaris suum/inmunología , Traslado Adoptivo , Animales , Ascariasis/parasitología , Antígenos CD4/metabolismo , Citocinas/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Inflamación/inducido químicamente , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Parasitol Res ; 115(12): 4701-4705, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27604123

RESUMEN

Ascarid nematodes, Ascaris suum, Toxocara canis and Toxocara cati, are the most important causative species of larva migrans syndrome (LMS) in humans. Although the diagnosis of ascarid LMS is generally based on serological tests, specific serological tests for A. suum infection have not been fully developed. In the present study, the sensitivity and specificity of three A. suum antigen preparations, i.e., the somatic adult worm antigen (As-SWAP), larval excretory-secretory (ES) antigens derived from infective L3 (AsiL3-ES) and larval ES from tissue migratory L3 (AsmL3-ES), were evaluated for the serodiagnosis of A. suum infection in enzyme-linked immunosorbent assay (ELISA). We found that all A. suum antigen preparations showed positive reaction to all sera from A. suum-infected mice, while only AsmL3-ES obtained 100 % detection of anti-A. suum antibodies in human visceral ascarosis patients. Comparing the reactivity of each A. suum antigen, sera from both A. suum-infected mice and human patients bound to AsiL3-ES significantly weaker than As-SWAP and AsmL3-ES. Moreover, the OD450 values of ELISA with the A. suum antigen preparations and T. canis larval ES antigen (TciL3-ES) were compared in order to discriminate between ascarosis and toxocarosis. Linear discriminant analysis showed that diagnosis based on TciL3-ES and AsmL3-ES ELISA gave the most reliable result for the discrimination of infecting species. In conclusion, the application of AsmL3-ES antigen in ELISA can be recommended for the serodiagnosis of A. suum infection in humans.


Asunto(s)
Antígenos Helmínticos/inmunología , Ascariasis/diagnóstico , Ascaris suum , Ensayo de Inmunoadsorción Enzimática/métodos , Animales , Ascaris suum/inmunología , Femenino , Humanos , Larva/inmunología , Ratones , Ratones Endogámicos C57BL , Pruebas Serológicas
10.
Parasitol Res ; 115(3): 1263-74, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26728072

RESUMEN

In multicellular parasites (e.g., nematodes and protozoa), proteins and glycolipids have been found to be decorated with phosphorylcholine (PC). PC can provoke various effects on immune cells leading to an immunomodulation of the host's immune system. This immunomodulation allows long-term persistence but also prevents severe pathology due to downregulation of cellular immune responses. PC-containing antigens have been found to interfere with key proliferative signaling pathways in B and T cells, development of dendritic cells and macrophages, and mast cell degranulation. These effects contribute to the observed modulated cytokine levels and impairment of lymphocyte proliferation. In contrast to glycosphingolipids, little is known about the PC-epitopes of proteins. So far, only a limited number of PC-modified proteins from nematodes have been identified. In this project, PC-substituted proteins and glycolipids in Ascaris suum have been localized by immunohistochemistry in specific tissues of the body wall, intestine, and reproductive tract. Subsequently, we investigated the PCome of A. suum by 2D gel-based proteomics and detection by Western blotting using the PC-specific antibody TEPC-15. By peptide-mass-fingerprint matrix-assisted laser-desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS), we could identify 59 PC-substituted proteins, which are in involved multiple cellular processes. In addition to membrane proteins like vitellogenin-6, we found proteins with structural (e.g., tubulins) and metabolic (e.g., pyruvate dehydrogenase) functions or which can act in the defense against the host's immune response (e.g., serpins). Initial characterization of the PC-epitopes revealed a predominant linkage of PC to the proteins via N-glycans. Our data form the basis for more detailed investigations of the PC-epitope structures as a prerequisite for comprehensive understanding of the molecular mechanisms of immunomodulation.


Asunto(s)
Antígenos Helmínticos/química , Ascaris suum/química , Epítopos/química , Proteínas del Helminto/química , Fosforilcolina/química , Animales , Antígenos Helmínticos/inmunología , Ascaris suum/inmunología , Western Blotting , Electroforesis en Gel Bidimensional , Epítopos/inmunología , Femenino , Proteínas del Helminto/inmunología , Inmunomodulación , Modelos Biológicos , Fosforilcolina/inmunología , Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
11.
Parasitol Res ; 115(3): 1213-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26637313

RESUMEN

The pig roundworm, Ascaris suum, is commonly found in domestic pigs all over the world. The transmission to humans takes place by ingestion of infective A. suum eggs present in soil because pig manure is widely used as fertilizer. The possible role of A. suum in the human visceral larva migrans (VLM) syndrome has been discussed controversially during past decades, even though various case reports, particularly from Japan document pulmonal, hepatic and even cerebral symptoms caused by migrating A. suum larvae after ingestion of infected row meat (liver) or contaminated vegetables. We examined 4481 sera by A. suum immunoblot (As-IB) and 5301 sera by Toxocara-ELISA from patients with symptoms associated with the VLM syndrome during three consecutive years (2012-2014). The incidence of A. suum-specific antibodies was 13.2 %, the incidence of T. canis specific antibodies 12.9 % and from a part of the As-IB positive sera (n = 417) additional Toxocara serology was performed to demonstrate the specificity of our tests. Only 56 out of the 417 (13.4 %) sera showed antibodies to both helminth species demonstrating that double infections exist. Interestingly the age distribution of the patients showed that 2.8 % of the Ascaris-positive patients were younger than 21 years, while in the Toxocara-positive group 13.4 % were <21 years. These results are in accordance with a Dutch study suspecting different ways of transmission as cause for this interesting age distribution. Due to the fact that large amounts of untreated pig manure are used as fertilizer and that the expulsion of adult A. suum worms causing intestinal ascariosis is extremely rare in Central European countries, the zoonotic potential of A. suum is considerably underestimated. We suggest that the performance of reliable immunoserological tests, in all industrialized countries where pigs are raised and their manure is used as fertilizer, could help to assess the actual potential of A. suum as causative agent of the VLM syndrome in humans.


Asunto(s)
Anticuerpos Antihelmínticos/sangre , Ascaris suum/inmunología , Larva Migrans Visceral/epidemiología , Adolescente , Adulto , Distribución por Edad , Anciano , Anciano de 80 o más Años , Animales , Austria/epidemiología , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina G/sangre , Incidencia , Lactante , Larva/inmunología , Larva Migrans Visceral/diagnóstico , Larva Migrans Visceral/inmunología , Masculino , Estiércol/parasitología , Persona de Mediana Edad , Sensibilidad y Especificidad , Distribución por Sexo , Suelo/parasitología , Toxocara canis/inmunología , Toxocariasis/epidemiología , Adulto Joven
12.
Parasitol Res ; 114(1): 305-10, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25367210

RESUMEN

Visceral larva migrans (VLM) syndrome caused by Toxocara canis larvae was first described in the 1950s. The role of other nematode larvae, i.e. the pig roundworm Ascaris suum as a causative agent of visceral larva migrans-associated symptoms like general malaise, cough, liver dysfunction, hypereosinophilia with hepatomegaly and/or pneumonia, was discussed controversially during the last decades. Recent serological screening studies for specific A. suum antibodies carried out in the Netherlands and Sweden yielded remarkable high seroprevalences, while a number of case reports from Japan report pulmonal, hepatic and cerebral symptoms caused by A. suum larvae after ingestion of infected raw meat (liver) or contaminated vegetables. We present here a sensitive and specific larval excretory-secretory (E/S) antigen-based immunoblot (As-IB) for the serodiagnosis of A. suum-infected patients suffering from symptoms associated to the VLM syndrome. In total, 34 sera from patients with hypereosinophilia and other clinical symptoms associated to the VLM syndrome tested negative for Toxocara sp. antibodies but positive in our newly established As-IB, 30 sera from healthy volunteers, 53 sera from patients with clinically and serologically confirmed toxocarosis and other helminthoses as well as 3 sera from patients with intestinal ascariosis due to Ascaris lumbricoides were included in the study. When evaluated with 30 sera from healthy volunteers and 53 sera from patients suffering from different helminthoses, the calculated specificity of our new As-IB is 95%. Problems hampering the establishment of simple serological screening tests for specific A. suum antibodies, like extensive antigenic similarities between the nematodes Ascaris and Toxocara or the absence of suitable experimental animals, are discussed. We assume that specific serological testing for antibodies of A. suum is very important for the treatment of individual patients on one hand and seroepidemiological investigations will help to clarify routes of transmission on the other hand. Further studies will be necessary to learn more about the extent of A. suum as a causative agent of the VLM syndrome and the role of pigs and their manure as the main source of human Ascaris infections in Austria and other industrialized countries.


Asunto(s)
Anticuerpos Antihelmínticos/sangre , Ascariasis/diagnóstico , Ascaris suum/inmunología , Immunoblotting , Larva Migrans Visceral/diagnóstico , Adulto , Animales , Ascariasis/inmunología , Austria , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Larva/inmunología , Larva Migrans Visceral/inmunología , Larva Migrans Visceral/parasitología , Masculino , Sensibilidad y Especificidad , Estudios Seroepidemiológicos , Porcinos , Toxocara canis/inmunología , Toxocariasis/diagnóstico , Toxocariasis/inmunología
13.
J Helminthol ; 89(3): 367-74, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-24703095

RESUMEN

Helminths use several strategies to evade and/or modify the host immune response, including suppression or inactivation of the host antigen-specific response. Several helminth immunomodulatory molecules have been identified. Our studies have focused on immunosuppression induced by the roundworm Ascaris suum and an A. suum-derived protein named protein 1 from A. suum (PAS-1). Here we assessed whether PAS-1 is an excretory/secretory (E/S) protein and whether it can suppress lipopolysaccharide-induced inflammation. Larvae from infective eggs were cultured in unsupplemented Dulbecco's modified Eagle medium (DMEM) for 2 weeks. PAS-1 was then measured in the culture supernatants and in adult A. suum body fluid at different time points by enzyme-linked immunosorbent assay (ELISA) with the monoclonal antibody MAIP-1. Secreted PAS-1 was detected in both larval culture supernatant and adult body fluid. It suppressed lipopolysaccharide (LPS)-induced leucocyte migration and pro-inflammatory cytokine production, and stimulated interleukin (IL)-10 secretion, indicating that larval and adult secreted PAS-1 suppresses inflammation in this model. Moreover, the anti-inflammatory activity of PAS-1 was abolished by treatment with MAIP-1, a PAS-1-specific monoclonal antibody, confirming the crucial role of PAS-1 in suppressing LPS-induced inflammation. These findings demonstrate that PAS-1 is an E/S protein with anti-inflammatory properties likely to be attributable to IL-10 production.


Asunto(s)
Ascaris suum/fisiología , Proteínas del Helminto/inmunología , Proteínas del Helminto/metabolismo , Tolerancia Inmunológica , Inmunosupresores/metabolismo , Animales , Ascaris suum/química , Ascaris suum/inmunología , Movimiento Celular/efectos de los fármacos , Medios de Cultivo/química , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Interacciones Huésped-Patógeno , Larva/química , Larva/inmunología , Larva/fisiología , Leucocitos/metabolismo , Leucocitos/fisiología , Factores de Tiempo
14.
Parasitology ; 141(14): 1904-11, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24775944

RESUMEN

Ascariasis is one of the most common parasitic diseases in both humans and pigs. It has been shown to cause growth deficits in both species and to impair cognitive development in children. Notwithstanding its substantial impact on pig economy and public health, diagnosis of ascariasis has mostly relied on the detection of eggs in stool and further development of novel, more sensitive methods has been limited or non-existent. Here, we discuss the currently available techniques for the diagnosis of ascariasis in pigs, their caveats, and the implications of a new serological detection technique for the evaluation of both pig and human ascariasis.


Asunto(s)
Ascariasis/diagnóstico , Ascaris suum/inmunología , Enfermedades de los Porcinos/diagnóstico , Animales , Ascariasis/parasitología , Ascaris suum/aislamiento & purificación , Ensayo de Inmunoadsorción Enzimática/veterinaria , Heces/parasitología , Femenino , Humanos , Hígado/parasitología , Pulmón/parasitología , Masculino , Recuento de Huevos de Parásitos/veterinaria , Porcinos , Enfermedades de los Porcinos/parasitología
15.
Curr Protoc ; 4(6): e1074, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38923794

RESUMEN

Ascariasis, caused by both Ascaris lumbricoides and Ascaris suum, is the most prevalent parasitic disease worldwide, affecting both human and porcine populations. However, due to the difficulties of assessing the early events of infection in humans, most studies of human ascariasis have been restricted to the chronic intestinal phase. Therefore, the Ascaris mouse model has become a fundamental tool for investigating the immunobiology and pathogenesis of the early infection stage referred to as larval ascariasis because of the model's practicality and ability to replicate the natural processes involved. The Ascaris mouse model has been widely used to explore factors such as infection resistance/susceptibility, liver inflammation, lung immune-mediated pathology, and co-infections and, notably, as a pivotal element in preclinical vaccine trials. Exploring the immunobiology of larval ascariasis may offer new insights into disease development and provide a substantial understanding of key components that trigger a protective immune response. This article focuses on creating a comprehensive guide for conducting Ascaris experimental infections in the laboratory as a foundation for future research efforts. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Acquisition and embryonation of Ascaris suum eggs from adult females Alternate Protocol: Cleaning and purification of Ascaris suum from female A. suum uteri Basic Protocol 2: Preparation of Ascaris suum eggs and murine infection Basic Protocol 3: Measurement of larval burden and Ascaris-larva-induced pathogenesis Basic Protocol 4: In vitro hatching and purification of Ascaris L3 larvae Support Protocol: Preparation of crude antigen from Ascaris infectious stages Basic Protocol 5: Ultrastructure-expansion microscopy (U-ExM) of Ascaris suum larval stages.


Asunto(s)
Ascariasis , Ascaris suum , Modelos Animales de Enfermedad , Larva , Ascariasis/parasitología , Ascariasis/inmunología , Animales , Ratones , Ascaris suum/inmunología , Larva/inmunología , Femenino , Ascaris/inmunología , Ascaris/patogenicidad , Humanos
16.
Int J Parasitol ; 54(10): 523-533, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38677398

RESUMEN

Coprological and serological diagnostic tests were compared to define the status of a pig farm with regard to Ascaris suum. On each of the 100 farms in France visited for the study, 10 blood samples were taken from pigs at the end of fattening (at least 22 weeks old) and 20 to 30 faecal samples were taken, depending on the category of animals present on the farm (10 sows, 10 piglets aged 10 to 12 weeks and 10 pigs at the end of fattening, aged at least 22 weeks). A SERASCA® ELISA test (Laboratory of Parasitology, Ghent University) was performed on each blood sample (cut-off 0.5) and a coprological analysis on each faecal sample. A Bayesian approach was used to estimate the sensitivity and specificity of the coprological and serological tests. A farm was considered positive if at least one A. suum egg was observed in the faecal samples. With regard to the serological test, various hypotheses were tested in order to define the number of seropositive animals required to consider a farm positive for A. suum. The coprological test has very good specificity in the search for A. suum, whether 20 or 30 samples are taken per farm. However, even with an increase in the number of samples, the sensitivity of this diagnostic approach is very low (less than 30%). On the other hand, the serological diagnostic method, which consists of taking blood samples from 10 animals at the end of fattening, has good sensitivity and seems better suited to defining the status of a farm with regard to A. suum, provided that a farm is considered seropositive only if two out of 10 samples are positive.


Asunto(s)
Ascariasis , Ascaris suum , Teorema de Bayes , Heces , Sensibilidad y Especificidad , Enfermedades de los Porcinos , Animales , Ascaris suum/aislamiento & purificación , Ascaris suum/inmunología , Enfermedades de los Porcinos/diagnóstico , Enfermedades de los Porcinos/parasitología , Heces/parasitología , Heces/química , Porcinos , Ascariasis/diagnóstico , Ascariasis/veterinaria , Ascariasis/parasitología , Pruebas Serológicas/veterinaria , Pruebas Serológicas/métodos , Ensayo de Inmunoadsorción Enzimática/veterinaria , Ensayo de Inmunoadsorción Enzimática/métodos , Francia , Granjas , Anticuerpos Antihelmínticos/sangre
17.
Front Immunol ; 15: 1396446, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38799456

RESUMEN

Ascaris spp. undergo extensive migration within the body before establishing patent infections in the small intestinal tract of humans and pigs. However, whether larval migration is critical for inducing efficient type 2 responses remains poorly understood. Therefore, we investigated systemic versus local adaptive immune responses along the hepato-tracheal migration of Ascaris suum during primary, single infections in conventionally raised pigs. Neither the initial invasion of gut tissue nor migration through the liver resulted in discernable Th2 cell responses. In contrast, lung-stage larvae elicited a Th2-biased pulmonary response, which declined after the larvae had left the lungs. In the small intestine, we observed an accumulation of Th2 cells upon the arrival of fourth-stage larvae (L4) to the small intestinal lumen. In parallel, we noticed robust and increasing Th1 responses in circulation, migration-affected organs, and draining lymph nodes. Phenotypic analysis of CD4+ T cells specifically recognizing A. suum antigens in the circulation and lung tissue of infected pigs confirmed that the majority of Ascaris-specific T cells produced IL-4 (Th2) and, to a much lesser extent, IL-4/IFN-g (Th2/1 hybrids) or IFN-g alone (Th1). These data demonstrate that lung-stage but not the early liver-stage larvae lead to a locally restricted Th2 response. Significant Th2 cell accumulation in the small intestine occurs only when L4 complete the body migration. In addition, Th2 immunity seems to be hampered by the concurrent, nonspecific Th1 bias in growing pigs. Together, the late onset of Th2 immunity at the site of infection and the Th1-biased systemic immunity likely enable the establishment of intestinal infections by sufficiently large L4 stages and pre-adult worms, some of which resist expulsion mechanisms.


Asunto(s)
Ascariasis , Ascaris suum , Células TH1 , Células Th2 , Animales , Ascaris suum/inmunología , Ascariasis/inmunología , Ascariasis/parasitología , Células Th2/inmunología , Porcinos , Células TH1/inmunología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/parasitología , Pulmón/inmunología , Pulmón/parasitología , Larva/inmunología , Citocinas/metabolismo
18.
Sci Rep ; 14(1): 14586, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918457

RESUMEN

Natural killer (NK) cells play a key role in defense against Salmonella infections during the early phase of infection. Our previous work showed that the excretory/secretory products of Ascaris suum repressed NK activity in vitro. Here, we asked if NK cell functionality was influenced in domestic pigs during coinfection with Ascaris and Salmonella enterica serotype Typhimurium. Ascaris coinfection completely abolished the IL-12 and IL-18 driven elevation of IFN-γ production seen in CD16 + CD8α + perforin + NK cells of Salmonella single-infected pigs. Furthermore, Ascaris coinfection prohibited the Salmonella-driven rise in NK perforin levels and CD107a surface expression. In line with impaired effector functions, NK cells from Ascaris-single and coinfected pigs displayed elevated expression of the inhibitory KLRA1 and NKG2A receptors genes, contrasting with the higher expression of the activating NKp46 and NKp30 receptors in NK cells during Salmonella single infection. These differences were accompanied by the highly significant upregulation of T-bet protein expression in NK cells from Ascaris-single and Ascaris/Salmonella coinfected pigs. Together, our data strongly indicate a profound repression of NK functionality by an Ascaris infection which may hinder infected individuals from adequately responding to a concurrent bacterial infection.


Asunto(s)
Ascariasis , Coinfección , Células Asesinas Naturales , Enfermedades de los Porcinos , Animales , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ascariasis/inmunología , Ascariasis/veterinaria , Ascariasis/parasitología , Coinfección/inmunología , Coinfección/microbiología , Coinfección/parasitología , Porcinos , Enfermedades de los Porcinos/parasitología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Salmonelosis Animal/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Ascaris suum/inmunología , Interferón gamma/metabolismo , Perforina/metabolismo , Interleucina-12/metabolismo , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Interleucina-18/metabolismo
19.
mSphere ; 9(9): e0047824, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39140728

RESUMEN

Ascaris is one of the most widespread helminth infections, leading to chronic morbidity in humans and considerable economic losses in pig farming. In addition, pigs are an important reservoir for the zoonotic salmonellosis, where pigs can serve as asymptomatic carriers. Here, we investigated the impact of an ongoing Ascaris infection on the immune response to Salmonella in pigs. We observed higher bacterial burdens in experimentally coinfected pigs compared to pigs infected with Salmonella alone. The impaired control of Salmonella in the coinfected pigs was associated with repressed interferon gamma responses in the small intestine and with the alternative activation of gut macrophages evident in elevated CD206 expression. Ascaris single and coinfection were associated with a rise of CD4-CD8α+FoxP3+ Treg in the lymph nodes draining the small intestine and liver. In addition, macrophages from coinfected pigs showed enhanced susceptibility to Salmonella infection in vitro and the Salmonella-induced monocytosis and tumor necrosis factor alpha production by myeloid cells was repressed in pigs coinfected with Ascaris. Hence, our data indicate that acute Ascaris infection modulates different immune effector functions with important consequences for the control of tissue-invasive coinfecting pathogens.IMPORTANCEIn experimentally infected pigs, we show that an ongoing infection with the parasitic worm Ascaris suum modulates host immunity, and coinfected pigs have higher Salmonella burdens compared to pigs infected with Salmonella alone. Both infections are widespread in pig production and the prevalence of Salmonella is high in endemic regions of human Ascariasis, indicating that this is a clinically meaningful coinfection. We observed the type 2/regulatory immune response to be induced during an Ascaris infection correlates with increased susceptibility of pigs to the concurrent bacterial infection.


Asunto(s)
Ascariasis , Ascaris suum , Coinfección , Salmonelosis Animal , Enfermedades de los Porcinos , Animales , Ascariasis/inmunología , Ascariasis/veterinaria , Porcinos , Coinfección/inmunología , Coinfección/microbiología , Coinfección/parasitología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/parasitología , Ascaris suum/inmunología , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Macrófagos/inmunología , Linfocitos T Reguladores/inmunología , Ganglios Linfáticos/inmunología , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Intestino Delgado/parasitología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Hígado/inmunología , Hígado/parasitología
20.
Am J Respir Cell Mol Biol ; 49(6): 1085-92, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23889698

RESUMEN

Spleen tyrosine kinase (SYK) is a key activator of signaling pathways downstream of multiple surface receptors implicated in asthma. SYK function has been extensively studied in mast cells downstream of the high-affinity IgE receptor, FcεR1. Preclinical studies have demonstrated a role for SYK in models of allergic inflammation, but a role in airway constriction has not been demonstrated. Here, we have used a potent and selective pharmacological inhibitor of SYK to determine the role of SYK in allergen-mediated inflammation and airway constriction in preclinical models. Attenuation of allergic airway responses was evaluated in a rat passive anaphylaxis model and rat and sheep inhaled allergen challenge models, as well as an ex vivo model of allergen-mediated airway constriction in rats and cynomolgus monkeys. Pharmacological inhibition of SYK dose-dependently blocked IgE-mediated tracheal plasma extravasation in rats. In a rat ovalbumin-sensitized airway challenge model, oral dosing with an SYK inhibitor led to a dose-dependent reduction in lung inflammatory cells. Ex vivo analysis of allergen-induced airway constriction in ovalbumin-sensitized brown Norway rats showed a complete attenuation with treatment of a SYK inhibitor, as well as a complete block of allergen-induced serotonin release. Similarly, allergen-mediated airway constriction was attenuated in ex vivo studies from nonhuman primate lungs. Intravenous administration of an SYK inhibitor attenuated both early- and late-phase allergen-induced increases in airway resistance in an Ascaris-sensitive sheep allergen challenge model. These data support a key role for SYK signaling in mediating allergic airway responses.


Asunto(s)
Alérgenos/administración & dosificación , Asma/prevención & control , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Ascaris suum/inmunología , Asma/etiología , Asma/fisiopatología , Broncoconstricción/efectos de los fármacos , Broncoconstricción/inmunología , Broncoconstricción/fisiología , Degranulación de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Macaca fascicularis , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Ovalbúmina/inmunología , Proteínas Tirosina Quinasas/fisiología , Ratas , Ratas Endogámicas BN , Ratas Sprague-Dawley , Ovinos , Transducción de Señal/efectos de los fármacos , Quinasa Syk
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA