Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
mBio ; 15(2): e0260323, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38179939

RESUMEN

Diet composition plays a large role in regulating gut health and enteric infection. In particular, synthetic "Western-style" diets may predispose to disease, while whole-grain diets containing high levels of crude fiber are thought to promote gut health. Here, we show that, in contrast to this paradigm, mice fed with unrefined chow are significantly more susceptible to infection with Trichuris muris, a caecum-dwelling nematode, than mice fed with refined, semi-synthetic diets (SSDs). Moreover, mice fed with SSD supplemented with inulin, a fermentable fiber, developed chronic T. muris burdens, whereas mice fed with SSD efficiently cleared the infection. Diet composition significantly impacted infection-induced changes in the host gut microbiome. Mice infected with the bacterium Citrobacter rodentium were also more susceptible to pathogen colonization when fed with either chow or inulin-enriched SSD. However, transcriptomic analysis of tissues from mice fed with either SSD or inulin-enriched SSD revealed that, in contrast to T. muris, increased C. rodentium infection appeared to be independent of the host immune response. Accordingly, exogenous treatment with interleukin (IL)-25 reduced T. muris burdens in inulin-fed mice, whereas IL-22 treatment was unable to restore resistance to C. rodentium colonization. Diet-mediated effects on pathogen burden were more pronounced for large intestine-dwelling pathogens, as effects on small the intestinal helminth (Heligmosomoides polygyrus) were less evident, and protozoan (Giardia muris) infection burdens were equivalent in mice fed with chow, inulin-enriched SSD, or SSD, despite higher cyst excretion in chow-fed mice. Collectively, our results point to a tissue- and pathogen-restricted effect of dietary fiber levels on enteric infection intensity.IMPORTANCEEnteric infections induce dysbiosis and inflammation and are a major public health burden. As the gut environment is strongly shaped by diet, the role of different dietary components in promoting resistance to infection is of interest. While diets rich in fiber or whole grain are normally associated with improved gut health, we show here that these components predispose the host to higher levels of pathogen infection. Thus, our results have significance for interpreting how different dietary interventions may impact on gastrointestinal infections. Moreover, our results may shed light on our understanding of how gut flora and mucosal immune function is influenced by the food that we eat.


Asunto(s)
Intestino Delgado , Inulina , Ratones , Animales , Dieta/métodos , Inflamación , Membrana Mucosa , Fibras de la Dieta
2.
Sci Rep ; 13(1): 21931, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-38081984

RESUMEN

Brown seaweeds have a rich bioactive content known to modulate biological processes, including the mucosal immune response and microbiota function, and may therefore have the potential to control enteric pathogens. Here, we tested if dietary seaweed (Saccharina latissima) supplementation could modulate pig gut health with a specific focus on parasitic helminth burdens, gut microbiota composition, and host immune response during a five week feeding period in pigs co-infected with the helminths Ascaris suum and Oesophagostomum dentatum. We found that inclusion of fermented S. latissima (Fer-SL) at 8% of the diet increased gut microbiota α-diversity with higher relative abundances of Firmicutes, Tenericutes, Verrucomicrobia, Spirochaetes and Elusimicrobia, and lower abundance of Prevotella copri. In the absence of helminth infection, transcription of immune-related genes in the intestine was only moderately influenced by dietary seaweed. However, Fer-SL modulated the transcriptional response to infection in a site-specific manner in the gut, with an attenuation of infection-induced gene expression in the jejunum and an amplification of gene expression in the colon. Effects on systemic immune parameters (e.g. blood lymphocyte populations) were limited, indicating the effects of Fer-SL were mainly localized to the intestinal tissues. Despite previously documented in vitro anti-parasitic activity against pig helminths, Fer-SL inclusion did not significantly affect parasite egg excretion or worm establishment. Collectively, our results show that although Fer-SL inclusion did not reduce parasite burdens, it may modify the gut environment during enteric parasite infection, which encourages continued investigations into the use of seaweeds or related products as novel tools to improve gut health.


Asunto(s)
Microbioma Gastrointestinal , Animales , Porcinos , Dieta , Oesophagostomum , Suplementos Dietéticos , Inmunidad
3.
BMC Biol ; 21(1): 138, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37316905

RESUMEN

BACKGROUND: The influence of diet on immune function and resistance to enteric infection and disease is becoming ever more established. Highly processed, refined diets can lead to inflammation and gut microbiome dysbiosis, whilst health-promoting dietary components such as phytonutrients and fermentable fibres are thought to promote a healthy microbiome and balanced mucosal immunity. Chicory (Cichorium intybus) is a leafy green vegetable rich in fibres and bioactive compounds that may promote gut health. RESULTS: Unexpectedly, we here show that incorporation of chicory into semisynthetic AIN93G diets renders mice susceptible to infection with enteric helminths. Mice fed a high level of chicory leaves (10% dry matter) had a more diverse gut microbiota, but a diminished type-2 immune response to infection with the intestinal roundworm Heligmosomoides polygyrus. Furthermore, the chicory-supplemented diet significantly increased burdens of the caecum-dwelling whipworm Trichuris muris, concomitant with a highly skewed type-1 immune environment in caecal tissue. The chicory-supplemented diet was rich in non-starch polysaccharides, particularly uronic acids (the monomeric constituents of pectin). In accordance, mice fed pectin-supplemented AIN93G diets had higher T. muris burdens and reduced IgE production and expression of genes involved in type-2 immunity. Importantly, treatment of pectin-fed mice with exogenous IL-25 restored type-2 responses and was sufficient to allow T. muris expulsion. CONCLUSIONS: Collectively, our data suggest that increasing levels of fermentable, non-starch polysaccharides in refined diets compromises immunity to helminth infection in mice. This diet-infection interaction may inform new strategies for manipulating the gut environment to promote resistance to enteric parasites.


Asunto(s)
Dieta , Infecciones por Nematodos , Animales , Ratones , Polisacáridos , Suplementos Dietéticos , Pectinas
4.
J Infect Dis ; 227(12): 1428-1432, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36932044

RESUMEN

Infection with intestinal whipworms (Trichuris spp.) causes widespread morbidity and may alter responses to enteric and extraintestinal coinfections. Here, we show that Trichuris muris infection in mice increases coinfection with 2 evolutionary divergent enteric pathogens, the bacterium Citrobacter rodentium and the helminth Heligmosomoides polygyrus. Coinfection caused reduced weight gain and promoted type 1-biased inflammation. In contrast, T. muris-infected mice were more resistant to migrating Ascaris suum larvae in the lungs. Our results highlight the divergent nature of pathogen interactions and suggest that whipworm infection is a risk factor for coinfections with other pathogens within the gastrointestinal tract.


Asunto(s)
Coinfección , Tricuriasis , Animales , Ratones , Ascaris , Intestinos , Trichuris , Pulmón
5.
Parasite Immunol ; 45(4): e12965, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36571323

RESUMEN

Helminths are large multicellular parasites responsible for widespread chronic disease in humans and animals. Intestinal helminths live in close proximity with the host gut microbiota and mucosal immune network, resulting in reciprocal interactions that closely influence the course of infections. Diet composition may strongly regulate gut microbiota composition and intestinal immune function and therefore may play a key role in modulating anti-helminth immune responses. Characterizing the multitude of interactions that exist between different dietary components (e.g., dietary fibres), immune cells, and the microbiota, may shed new light on regulation of helminth-specific immunity. This review focuses on the current knowledge of how metabolism of dietary components shapes immune response during helminth infection, and how this information may be potentially harnessed to design new therapeutics to manage parasitic infections and associated diseases.


Asunto(s)
Helmintiasis , Helmintos , Microbiota , Animales , Humanos , Intestinos , Dieta
6.
FASEB J ; 36(4): e22256, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35333423

RESUMEN

Proanthocyanidins (PAC) are dietary polyphenols with putative anti-inflammatory and immunomodulatory effects. However, whether dietary PAC can regulate type-2 immune function and inflammation at mucosal surfaces remains unclear. Here, we investigated if diets supplemented with purified PAC modulated pulmonary and intestinal mucosal immune responses during infection with the helminth parasite Ascaris suum in pigs. A. suum infection induced a type-2 biased immune response in lung and intestinal tissues, characterized by pulmonary granulocytosis, increased Th2/Th1 T cell ratios in tracheal-bronchial lymph nodes, intestinal eosinophilia, and modulation of genes involved in mucosal barrier function and immunity. Whilst PAC had only minor effects on pulmonary immune responses, RNA-sequencing of intestinal tissues revealed that dietary PAC significantly enhanced transcriptional responses related to immune function and antioxidant responses in the gut of both naïve and A. suum-infected animals. A. suum infection and dietary PAC induced distinct changes in gut microbiota composition, primarily in the jejunum and colon, respectively. Notably, PAC consumption substantially increased the abundance of Limosilactobacillus reuteri. In vitro experiments with porcine macrophages and intestinal epithelial cells supported a role for both PAC polymers and PAC-derived microbial metabolites in regulating oxidative stress responses in host tissues. Thus, dietary PAC may have distinct beneficial effects on intestinal health during infection with mucosal pathogens, while having a limited activity to modulate naturally-induced type-2 pulmonary inflammation. Our results shed further light on the mechanisms underlying the health-promoting properties of PAC-rich foods, and may aid in the design of novel dietary supplements to regulate mucosal inflammatory responses in the gastrointestinal tract.


Asunto(s)
Ascaris suum , Proantocianidinas , Animales , Antioxidantes , Ascaris suum/fisiología , Colon , Dieta , Inflamación , Pulmón , Proantocianidinas/farmacología , Porcinos
7.
Mol Nutr Food Res ; 66(7): e2101004, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35107883

RESUMEN

SCOPE: Garlic is a source of bioactive phytonutrients that may have anti-inflammatory or immunomodulatory properties. The mechanism(s) underlying the bioactivity of these compounds and their ability to regulate responses to enteric infections remains unclear. METHODS AND RESULTS: This study investigates if a garlic-derived preparation (PTSO-PTS) containing two organosulfur metabolites, propyl-propane thiosulfonate (PTSO), and propyl-propane thiosulfinate (PTS), regulate inflammatory responses in murine macrophages and intestinal epithelial cells (IEC) in vitro, as well as in a model of enteric parasite-induced inflammation. PTSO-PTS decreases lipopolysaccharide-induced secretion of TNFα, IL-6, and IL-27 in macrophages. RNA-sequencing demonstrates that PTSO-PTS strongly suppresses pathways related to immune and inflammatory signaling. PTSO-PTS induces the expression of a number of genes involved in antioxidant responses in IEC during exposure to antigens from the parasite Trichuris muris. In vivo, PTSO-PTS does not affect T. muris establishment or intestinal T-cell responses but significantly alters cecal transcriptomic responses. Notably, a reduction in T. muris-induced expression of Tnf, Saa2, and Nos2 is observed. CONCLUSION: Garlic-derived organosulfur compounds exert anti-inflammatory effects in macrophages and IEC, and regulate gene expression during intestinal infection. These compounds and related organic molecules may thus hold potential as functional food components to improve gut health in humans and animals.


Asunto(s)
Ajo , Animales , Antiinflamatorios/farmacología , Antioxidantes , Inflamación/tratamiento farmacológico , Macrófagos , Ratones
8.
J Nutr Biochem ; 100: 108887, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34655757

RESUMEN

Phytonutrients such as cinnamaldehyde (CA) have been studied for their effects on metabolic diseases, but their influence on mucosal inflammation and immunity to enteric infection are not well documented. Here, we show that consumption of CA in mice significantly down-regulates transcriptional pathways connected to inflammation in the small intestine, and alters T-cell populations in mesenteric lymph nodes. During infection with the enteric helminth Heligomosomoides polygyrus, CA treatment attenuated infection-induced changes in biological pathways connected to cell cycle and mitotic activity, and tended to reduce worm burdens. Mechanistically, CA did not appear to exert activity through a prebiotic effect, as CA treatment did not significantly change the composition of the gut microbiota. Instead, in vitro experiments showed that CA directly induced xenobiotic metabolizing pathways in intestinal epithelial cells and suppressed endotoxin-induced inflammatory responses in macrophages. Collectively, our results show that CA down-regulates inflammatory pathways in the intestinal mucosa and can limit the pathological response to enteric infection. These properties appear to be largely independent of the gut microbiota, and instead connected to the ability of CA to induce antioxidant pathways in intestinal cells. Our results encourage further investigation into the use of CA and related phytonutrients as functional food components to promote intestinal health in humans and animals.


Asunto(s)
Acroleína/análogos & derivados , Suplementos Dietéticos , Inflamación/inmunología , Intestino Delgado/metabolismo , Fitoquímicos/administración & dosificación , Infecciones por Strongylida/inmunología , Acroleína/administración & dosificación , Acroleína/farmacología , Animales , Células Cultivadas , Femenino , Microbioma Gastrointestinal , Inmunidad Mucosa , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Ganglios Linfáticos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nematospiroides dubius , Fitoquímicos/farmacología , Linfocitos T/inmunología , Transcripción Genética , Transcriptoma , Xenobióticos/metabolismo
9.
BMC Vet Res ; 17(1): 62, 2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514383

RESUMEN

Increasing evidence suggests that nutritional manipulation of the commensal gut microbiota (GM) may play a key role in maintaining animal health and production in an era of reduced antimicrobial usage. Gastrointestinal helminth infections impose a considerable burden on animal performance, and recent studies suggest that infection may substantially alter the composition and function of the GM. Here, we discuss the potential interactions between different bioactive dietary components (prebiotics, probiotics and phytonutrients) and helminth infection on the GM in livestock. A number of recent studies suggest that host diet can strongly influence the nature of the helminth-GM interaction. Nutritional manipulation of the GM may thus impact helminth infection, and conversely infection may also influence how the GM responds to dietary interventions. Moreover, a dynamic interaction exists between helminths, the GM, intestinal immune responses, and inflammation. Deciphering the mechanisms underlying the diet-GM-helminth axis will likely inform future helminth control strategies, as well as having implications for how health-promoting feed additives, such as probiotics, can play a role in sustainable animal production.


Asunto(s)
Dieta , Enfermedades Gastrointestinales/veterinaria , Microbioma Gastrointestinal/fisiología , Helmintiasis Animal/patología , Animales , Enfermedades Gastrointestinales/parasitología , Helmintos , Parasitosis Intestinales , Ganado/microbiología , Ganado/parasitología , Prebióticos , Probióticos
10.
Front Immunol ; 12: 793260, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069576

RESUMEN

Dietary probiotics may enhance gut health by directly competing with pathogenic agents and through immunostimulatory effects. These properties are recognized in the context of bacterial and viral pathogens, but less is known about interactions with eukaryotic pathogens such as parasitic worms (helminths). In this study we investigated whether two probiotic mixtures (comprised of Bacillus amyloliquefaciens, B. subtilis, and Enterococcus faecium [BBE], or Lactobacillus rhamnosus LGG and Bifidobacterium animalis subspecies Lactis Bb12 [LB]) could modulate helminth infection kinetics as well as the gut microbiome and intestinal immune responses in pigs infected with the nodular worm Oesophagostomum dentatum. We observed that neither probiotic mixture influenced helminth infection levels. BBE, and to a lesser extent LB, changed the alpha- and beta-diversity indices of the colon and fecal microbiota, notably including an enrichment of fecal Bifidobacterium spp. by BBE. However, these effects were muted by concurrent O. dentatum infection. BBE (but not LB) significantly attenuated the O. dentatum-induced upregulation of genes involved in type-2 inflammation and restored normal lymphocyte ratios in the ileo-caecal lymph nodes that were altered by infection. Moreover, inflammatory cytokine release from blood mononuclear cells and intestinal lymphocytes was diminished by BBE. Collectively, our data suggest that selected probiotic mixtures can play a role in maintaining immune homeostasis during type 2-biased inflammation. In addition, potentially beneficial changes in the microbiome induced by dietary probiotics may be counteracted by helminths, highlighting the complex inter-relationships that potentially exist between probiotic bacteria and intestinal parasites.


Asunto(s)
Bacillus/inmunología , Enterococcus faecium/inmunología , Microbioma Gastrointestinal/inmunología , Esofagostomiasis , Oesophagostomum/inmunología , Probióticos/farmacología , Enfermedades de los Porcinos , Animales , Femenino , Masculino , Esofagostomiasis/inmunología , Esofagostomiasis/microbiología , Esofagostomiasis/veterinaria , Porcinos , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/parasitología
11.
Br J Nutr ; 126(3): 375-382, 2021 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-33106192

RESUMEN

Hyperprolific sows rear more piglets than they have teats, and to accommodate this, milk replacers are often offered as a supplement. Milk replacers are based on bovine milk, yet components of vegetable origin are often added. This may reduce growth, but could also accelerate maturational changes. Therefore, we investigated the effect of feeding piglets a milk replacer with gradually increasing levels of wheat flour on growth, gut enzyme activity and immune function compared with a diet based entirely on bovine milk. The hypothesis tested was that adding a starch component (wheat flour) induces maturation of the mucosa as measured by higher digestive activity and improved integrity and immunity of the small intestines (SI). To test this hypothesis, piglets were removed from the sow at day 3 and fed either a pure milk replacer diet (MILK) or from day 11 a milk replacer diet with increasing levels of wheat (WHEAT). The WHEAT piglets had an increased enzyme activity of maltase and sucrase in the proximal part of the SI compared with the MILK group. There were no differences in gut morphology, histopathology and gene expression between the groups. In conclusion, the pigs given a milk replacer with added wheat displayed immunological and gut mucosal enzyme maturational changes, indicatory of adaptation towards a vegetable-based diet. This was not associated with any clinical complications, and future studies are needed to show whether this could improve responses in the subsequent weaning process.


Asunto(s)
Dieta , Sustitutos de la Leche/administración & dosificación , Porcinos , Triticum , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Dieta/veterinaria , Harina , Intestino Delgado/enzimología , Intestino Delgado/inmunología , Leche , Sacarasa/metabolismo , Porcinos/crecimiento & desarrollo , Porcinos/inmunología , Destete , alfa-Glucosidasas/metabolismo
12.
J Immunol ; 204(11): 3042-3055, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32284331

RESUMEN

Fermentable dietary fibers promote the growth of beneficial bacteria, can enhance mucosal barrier integrity, and reduce chronic inflammation. However, effects on intestinal type 2 immune function remain unclear. In this study, we used the murine whipworm Trichuris muris to investigate the effect of the fermentable fiber inulin on host responses to infection regimes that promote distinct Th1 and Th2 responses in C57BL/6 mice. In uninfected mice, dietary inulin stimulated the growth of beneficial bacteria, such as Bifidobacterium (Actinobacteria) and Akkermansia (Verrucomicrobia). Despite this, inulin prevented worm expulsion in normally resistant mice, instead resulting in chronic infection, whereas mice fed an equivalent amount of nonfermentable fiber (cellulose) expelled worms normally. Lack of expulsion in the mice fed inulin was accompanied by a significantly Th1-skewed immune profile characterized by increased T-bet+ T cells and IFN-γ production in mesenteric lymph nodes, increased expression of Ido1 in the cecum, and a complete absence of mast cell and IgE production. Furthermore, the combination of dietary inulin and high-dose T. muris infection caused marked dysbiosis, with expansion of the Firmicutes and Proteobacteria phyla, near elimination of Bacteroidetes, and marked reductions in cecal short-chain fatty acids. Neutralization of IFN-γ during infection abrogated Ido1 expression and was sufficient to restore IgE production and worm expulsion in inulin-fed mice. Our results indicate that, whereas inulin promoted gut health in otherwise healthy mice, during T. muris infection, it exacerbated inflammatory responses and dysbiosis. Thus, the positive effects of fermentable fiber on gut inflammation appear to be context dependent, revealing a novel interaction between diet and infection.


Asunto(s)
Fibras de la Dieta/metabolismo , Inflamación/inmunología , Inulina/metabolismo , Células TH1/inmunología , Células Th2/inmunología , Tricuriasis/inmunología , Trichuris/fisiología , Animales , Progresión de la Enfermedad , Disbiosis , Fermentación , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Ratones , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
13.
Front Microbiol ; 11: 312, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194529

RESUMEN

The gut microbiota (GM) displays a profound ability to adapt to extrinsic factors, such as gastrointestinal pathogens and/or dietary alterations. Parasitic worms (helminths) and host-associated GM share a long co-evolutionary relationship, exerting mutually modulatory effects which may impact the health of the host. Moreover, dietary components such as prebiotic fibers (e.g. inulin) are capable of modulating microbiota toward a composition often associated with a healthier gut function. The effect of helminth infection on the host microbiota is still equivocal, and it is also unclear how parasites and prebiotic dietary components interact to influence the microbiota and host health status. Some helminths, such as Trichuris suis (porcine whipworm), also exhibit strong immunomodulatory and anti-inflammatory effects. We therefore explored the effects of T. suis, alone and in interaction with inulin, both in fecal microbiota during the infection period and luminal microbiota across four intestinal segments at the end of a 4-week infection period. We observed that T. suis generally had minimal, but mainly positive, effects on the microbiota. T. suis increased the relative abundance of bacterial genera putatively associated with gut health such as Prevotella, and decreased bacteria such as Proteobacteria that have been associated with dysbiosis. Interestingly, dietary inulin interacted with T. suis to enhance these effects, thereby modulating the microbiota toward a composition associated with reduced inflammation. Our results show that administration of T. suis together with the consumption of prebiotic inulin may have the potential to positively affect gut health.

14.
Mol Immunol ; 121: 127-135, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32200170

RESUMEN

Consumption of fermentable dietary fibres, such as inulin, or administration of helminth products (e.g. Trichuris suis ova) have independently been shown to alleviate inflammation in vivo. We recently found that dietary inulin and T. suis infection in pigs co-operatively suppressed type-1 inflammatory responses in the gut, suggesting the potential of dietary components to augment anti-inflammatory responses induced by certain helminths. Here, we explored whether T. suis antigens and inulin could directly suppress inflammatory responses in vitro in a cooperative manner. T. suis soluble products (TsSP) strongly suppressed lipopolysaccharide (LPS)-induced IL-6 and TNF-α secretion from murine macrophages and induced an anti-inflammatory phenotype as evidenced by transcriptomic and gene pathway analyses. Inulin regulated the expression of a small number of genes and transcriptional pathways in macrophages after exposure to LPS, but did not enhance the suppressive activity of TsSP, either directly or in co-culture experiments with intestinal epithelial cells. Culture of macrophages with short-chain fatty acids, the products of microbial fermentation of inulin, did however appear to enhance TsSP-mediated inhibition of TNF-α production. Our results confirm a direct role for helminth products in suppressing inflammatory responses in macrophages. In contrast, inulin had little capacity to directly modulate LPS-induced responses. Our results suggest distinct mode-of-actions of T. suis and inulin in regulating inflammatory responses, and that the role of inulin in modulating the response to helminth infection may be dependent on other factors such as production of metabolites by the gut microbiota.


Asunto(s)
Antígenos Helmínticos/farmacología , Inflamación/terapia , Inulina/farmacología , Macrófagos/efectos de los fármacos , Trichuris/inmunología , Animales , Antígenos Helmínticos/inmunología , Antígenos Helmínticos/uso terapéutico , Células Cultivadas , Técnicas de Cocultivo , Fibras de la Dieta/farmacología , Células Epiteliales , Ácidos Grasos Volátiles/farmacología , Humanos , Inflamación/inmunología , Interleucina-6/inmunología , Interleucina-6/metabolismo , Mucosa Intestinal/citología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
15.
Vet Immunol Immunopathol ; 211: 6-9, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31084896

RESUMEN

Helminth parasites are highly prevalent in swine production, causing chronic infections and considerable morbidity due to growth retardation. Moreover, helminths actively modulate host immune responses to other pathogens and/or vaccines. Here, we investigated the modulatory effects of Ascaris suum adult body fluid (ABF) and Trichuris suis Soluble Products (TsSP) on the cytokine response in porcine peripheral blood mononuclear cells (PBMCs) and the intestinal epithelial cell line IPEC-J2. In PBMCs, TsSP induced the secretion of IL-6, IL-10 and IL-1ß, but not TNF-α. Moreover, TsSP significantly enhanced the production of bacterial lipopolysaccharide (LPS)-induced IL-6 and IL-10 but suppressed the production of LPS-induced TNF-α. ABF did not induce cytokine secretion from PBMC, but suppressed LPS-induced secretion of TNF-α and IL-6. ABF did not have any effect on cytokine production in IPEC-J2 cells. In contrast, TsSP selectively induced the secretion of IL-6, and enhanced the IL-6 response induced by LPS. The IL-6 response appeared to be a conserved response to T. suis products, as significant secretion was also observed in alveolar macrophages. Thus, T. suis products have diverse modulatory effects on cytokine secretion in vitro, with IL-6 production a consistent feature of the innate host response.


Asunto(s)
Antígenos Helmínticos/inmunología , Ascaris suum/inmunología , Citocinas/metabolismo , Células Epiteliales/inmunología , Leucocitos Mononucleares/inmunología , Enfermedades de los Porcinos/parasitología , Trichuris/inmunología , Animales , Ascariasis/inmunología , Ascariasis/parasitología , Ascariasis/veterinaria , Citocinas/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/parasitología , Femenino , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/parasitología , Masculino , Porcinos/inmunología , Porcinos/parasitología , Enfermedades de los Porcinos/inmunología , Tricuriasis/inmunología , Tricuriasis/parasitología , Tricuriasis/veterinaria
16.
Front Immunol ; 9: 2557, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30473696

RESUMEN

Diet composition may play a crucial role in shaping host immune responses and commensal gut microbiota populations. Bioactive dietary components, such as inulin, have been extensively studied for their bioactive properties, particularly in modulating gut immune function and reducing inflammation. It has been shown that colonization with gastrointestinal parasitic worms (helminths) may alleviate chronic inflammation through promotion of T-helper cell type (Th) 2 and T-regulatory immune responses and alterations in the gut microbiome. In this study, we investigated if dietary inulin could modulate mucosal immune function in pigs during colonization with the porcine whipworm Trichuris suis. T. suis infection induced a typical Th2-biased immune response characterized by transcriptional changes in Th2- and barrier function-related genes, accompanied by intestinal remodeling through increased epithelial goblet and tuft cell proliferation. We observed that inulin also up-regulated Th2-related immune genes (IL13, IL5), and suppressed Th1-related pro-inflammatory genes (IFNG, IL1A, IL8) in the colon. Notably, inulin augmented the T. suis-induced responses with increased transcription of key Th2 and mucosal barrier genes (e.g., IL13, TFF3), and synergistically suppressed pro-inflammatory genes, such as IFNG and CXCL9. 16S rRNA sequencing of proximal colon digesta samples revealed that inulin supplementation reduced the abundance of bacterial phyla linked to inflammation, such as Proteobacteria and Firmicutes, and simultaneously increased Actinobacteria and Bacteroidetes. Interestingly, pigs treated with both inulin and T. suis displayed the highest Bacteroidetes: Firmicutes ratio and the lowest gut pH, suggesting an interaction of diet and helminth infection that stimulates the growth of beneficial bacterial species. Overall, our data demonstrate that T. suis infection and inulin co-operatively enhance anti-inflammatory immune responses, which is potentially mediated by changes in microbiota composition. Our results highlight the intricate interactions between diet, immune function and microbiota composition in a porcine helminth infection model. This porcine model should facilitate further investigations into the use of bioactive diets as immunomodulatory mediators against inflammatory conditions, and how diet and parasites may influence gut health.


Asunto(s)
Mucosa Intestinal/inmunología , Inulina/inmunología , Porcinos/inmunología , Porcinos/parasitología , Células Th2/inmunología , Tricuriasis/inmunología , Trichuris/inmunología , Animales , Colon/inmunología , Colon/parasitología , Dieta/métodos , Femenino , Microbioma Gastrointestinal/inmunología , Inflamación/inmunología , Inflamación/parasitología , Mucosa Intestinal/parasitología , Masculino , Membrana Mucosa/inmunología , Membrana Mucosa/parasitología , ARN Ribosómico 16S/inmunología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/parasitología , Tricuriasis/parasitología , Tricuriasis/veterinaria
18.
Sci Rep ; 6: 23458, 2016 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-26996237

RESUMEN

Lymphatic filariasis and onchocerciasis are parasitic helminth diseases, which cause severe morbidities such as elephantiasis, skin disease and blindness, presenting a major public health burden in endemic communities. The anti-Wolbachia consortium (A·WOL: http://www.a-wol.com/) has identified a number of registered antibiotics that target the endosymbiotic bacterium, Wolbachia, delivering macrofilaricidal activity. Here we use pharmacokinetics/pharmacodynamics (PK/PD) analysis to rationally develop an anti-Wolbachia chemotherapy by linking drug exposure to pharmacological effect. We compare the pharmacokinetics and anti-Wolbachia efficacy in a murine Brugia malayi model of minocycline versus doxycycline. Doxycycline exhibits superior PK in comparison to minocycline resulting in a 3-fold greater exposure in SCID mice. Monte-Carlo simulations confirmed that a bi-daily 25-40 mg/Kg regimen is bioequivalent to a clinically effective 100-200 mg/day dose for these tetracyclines. Pharmacodynamic studies showed that minocycline depletes Wolbachia more effectively than doxycycline (99.51% vs. 90.35%) after 28 day 25 mg/Kg bid regimens with a more potent block in microfilarial production. PK/PD analysis predicts that minocycline would be expected to be 1.7 fold more effective than doxycycline in man despite lower exposure in our infection models. Our findings warrant onward clinical investigations to examine the clinical efficacy of minocycline treatment regimens against lymphatic filariasis and onchocerciasis.


Asunto(s)
Antibacterianos/administración & dosificación , Filariasis Linfática/prevención & control , Minociclina/administración & dosificación , Wolbachia/efectos de los fármacos , Animales , Antibacterianos/farmacocinética , Brugia Malayi/efectos de los fármacos , Brugia Malayi/parasitología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Doxiciclina/administración & dosificación , Doxiciclina/farmacocinética , Filariasis Linfática/parasitología , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Minociclina/farmacocinética , Wolbachia/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...