Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Appl Environ Microbiol ; 89(11): e0097723, 2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-37902410

RESUMEN

IMPORTANCE: This work provides evidence that early-life fungal community composition, or host genetics, influences long-term mycobiome composition. In addition, this work provides the first comparison of the feral pig mycobiome to the mycobiome of intensively raised pigs.


Asunto(s)
Micobioma , Animales , Porcinos , Longevidad , Animales Salvajes , Sus scrofa , Heces/microbiología
2.
FEMS Microbiol Ecol ; 99(7)2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37349964

RESUMEN

Fecal microbiota transplantation (FMT) is an emerging technique for modulating the pig microbiota, however, donor variability is one of the major reasons for inconsistent outcomes across studies. Cultured microbial communities may address some limitations of FMT; however, no study has tested cultured microbial communities as inocula in pigs. This pilot study compared the effects of microbiota transplants derived from sow feces to cultured mixed microbial community (MMC) following weaning. Control, FMT4X, and MMC4X were applied four times, while treatment FMT1X was administered once (n = 12/group). On postnatal day 48, microbial composition was modestly altered in pigs receiving FMT in comparison with Control (Adonis, P = .003), mainly attributed to reduced inter-animal variations in pigs receiving FMT4X (Betadispersion, P = .018). Pigs receiving FMT or MMC had consistently enriched ASVs assigned to genera Dialister and Alloprevotella. Microbial transplantation increased propionate production in the cecum. MMC4X piglets showed a trend of higher acetate and isoleucine compared to Control. A consistent enrichment of metabolites from amino acid metabolism in pigs that received microbial transplantation coincided with enhanced aminoacyl-tRNA biosynthesis pathway. No differences were observed among treatment groups for body weight or cytokine/chemokine profiles. Overall, FMT and MMC exerted similar effects on gut microbiota composition and metabolite production.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Porcinos , Animales , Femenino , Trasplante de Microbiota Fecal , Destete , Proyectos Piloto , Heces , Metaboloma
3.
Appl Environ Microbiol ; 88(24): e0159322, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36448784

RESUMEN

Early-life antibiotic exposure is associated with diverse long-term adverse health outcomes. Despite the immunomodulatory effects of gastrointestinal fungi, the impact of antibiotics on the fungal community (mycobiome) has received little attention. The objectives of this study were to determine the impact of commonly prescribed infant antibiotic treatments on the microbial loads and structures of bacterial and fungal communities in the gastrointestinal tract. Thirty-two piglets were divided into four treatment groups: amoxicillin (A), amoxicillin-clavulanic acid (AC), gentamicin-ampicillin (GA), and flavored placebo (P). Antibiotics were administered orally starting on postnatal day (PND) 1 until PND 8, except for GA, which was given on PNDs 5 and 6 intramuscularly. Fecal swabs were collected from piglets on PNDs 3 and 8, and sow feces were collected 1 day after farrowing. The impacts of antibiotics on bacterial and fungal communities were assessed by sequencing the 16S rRNA and the internal transcribed spacer 2 (ITS2) rRNA genes, respectively, and quantitative PCR was performed to determine total bacterial and fungal loads. Antibiotics did not alter the α-diversity (P = 0.834) or ß-diversity (P = 0.565) of fungal communities on PND 8. AC increased the ratio of total fungal/total bacterial loads on PND 8 (P = 0.027). There was strong clustering of piglets by litter on PND 8 (P < 0.001), which corresponded to significant differences in the sow mycobiome, especially the presence of Kazachstania slooffiae. In summary, we observed a strong litter effect and showed that the maternal mycobiome is essential for shaping the piglet mycobiome in early life. IMPORTANCE This work provides evidence that although the fungal community composition is not altered by antibiotics, the overall fungal load increases with the administration of amoxicillin-clavulanic acid. Additionally, we show that the maternal fungal community is important in establishing the fungal community in piglets.


Asunto(s)
Microbioma Gastrointestinal , Micobioma , Animales , Femenino , Combinación Amoxicilina-Clavulanato de Potasio/farmacología , Antibacterianos/farmacología , Hongos , ARN Ribosómico 16S/genética , Porcinos
4.
Microb Pathog ; 173(Pt A): 105873, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36371065

RESUMEN

Brachyspira hyodysenteriae, an etiologic agent of swine dysentery (SD), is known for causing colitis. Although some aspects of colonic defenses during infection have been described previously, a more comprehensive picture of the host and microbiota interaction in clinically affected animals is required. This study aimed to characterize multiple aspects of colonic innate defenses and microbiome factors in B. hyodysenteriae-infected pigs that accompany clinical presentation of hemorrhagic diarrhea. We examined colonic mucus barrier modifications, leukocyte infiltration, cathelicidin expression, as well as microbiome composition. We showed that B. hyodysenteriae infection caused microscopic hemorrhagic colitis with abundant neutrophil infiltration in the colonic lamina propria and lumen, with minor macrophage infiltration. Mucus hypersecretion with abundant sialylated mucus in the colon, as well as mucosal colonization by [Acetivibrio] ethanolgignens, Lachnospiraceae, and Campylobacter were pathognomonic of B. hyodysenteriae infection. These findings demonstrate that B. hyodysenteriae produces clinical disease through multiple effects on host defenses, involving alterations of mucosal innate immunity and microbiota. Given that B. hyodysenteriae is increasingly resistant to antimicrobials, this understanding of SD pathogenesis may lead to future development of non-antibiotic and anti-inflammatory alternative therapeutics.


Asunto(s)
Colitis , Disentería , Infecciones por Bacterias Gramnegativas , Microbiota , Infecciones por Spirochaetales , Enfermedades de los Porcinos , Porcinos , Animales , Enfermedades de los Porcinos/patología , Disentería/veterinaria , Disentería/patología , Inmunidad Innata , Infecciones por Bacterias Gramnegativas/patología
5.
J Anim Sci ; 100(11)2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36205053

RESUMEN

Inclusion of enzymes and organic acids in pig diets is an important strategy supporting decreased antibiotic usage in pork production. However, limited knowledge exists about how these additives impact intestinal microbes and their metabolites. To examine the effects of benzoic acid and enzymes on gut microbiota and metabolome, 160 pigs were assigned to one of four diets 7 days after weaning: a control diet or the addition of 0.5% benzoic acid, 0.045% dietary enzymes (phytase, ß-glucanase, xylanase, and α-amylase), or both and fed ad libitum for 21 to 22 d. Individual growth performance and group diarrhea incidence data were collected throughout the experimental period. A decrease of 20% in pen-level diarrhea incidence from days 8 to 14 in pigs-fed both benzoic acid and enzymes compared to the control diet (P = 0.047). Cecal digesta samples were collected at the end of the experimental period from 40 piglets (n = 10 per group) and evaluated for differences using 16S rRNA sequencing and two-dimensional gas chromatography and time-of-flight mass spectrometry (GCxGC-TOFMS). Analysis of cecal microbiota diversity revealed that benzoic acid altered microbiota composition (Unweighted Unifrac, P = 0.047, r2 = 0.07) and decreased α-diversity (Shannon, P = 0.041; Faith's Phylogenetic Diversity, P = 0.041). Dietary enzymes increased fiber-fermenting bacterial taxa such as Prevotellaceae. Two-step feature selection identified 17 cecal metabolites that differed among diets, including increased microbial cross-feeding product 1,2-propanediol in pigs-fed benzoic acid-containing diets. In conclusion, dietary benzoic acid and enzymes affected the gut microbiota and metabolome of weaned pigs and may support the health and resolution of postweaning diarrhea.


Feeding weaned pigs diets containing benzoic acid or supplemental enzymes for 21 d after weaning changed the gut microbiota and metabolome. Benzoic acid increased feed intake, weight gain, and the presence of 1,2-propanediol in cecal digesta, which is an important microbial cross-feeding product. Dietary enzymes altered microbiota composition, increasing the presence of fiber-fermenting microbes including Prevotellaceae. Pigs fed a combination of both benzoic acid and enzymes showed improved resolution of postweaning diarrhea. These differences demonstrate the role of these feed additives in the establishment of gut microbes and metabolic pathways for the degradation of complex dietary components in the weaned pig. This study provides new information about alterations in microbial function and community composition using microbiota sequencing and metabolomic analysis.


Asunto(s)
Alimentación Animal , Ácido Benzoico , Porcinos , Animales , Destete , Alimentación Animal/análisis , Filogenia , ARN Ribosómico 16S/genética , Dieta/veterinaria , Fibras de la Dieta/metabolismo , Ciego/microbiología , Diarrea/veterinaria
6.
J Anim Sci ; 100(6)2022 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-35569054

RESUMEN

Increased fermentable carbohydrates (e.g., ß-glucan, amylose) may increase endogenous losses including for P, and thereby reduce apparent total tract digestibility (ATTD) of P. The present study assessed effects of barley cultivars varying in fermentable starch and fiber on apparent ileal digestibility (AID) and ATTD of P, myo-inositol 1,2,3,4,5,6-hexakis (dihydrogen phosphate; InsP6) and Ca, and standardized total tract digestibility (STTD) of P and the presence of lower inositol phosphates (InsP) compared to wheat. In a 6 (period) × 5 (diet) Youden square, seven ileal-cannulated barrows (initial BW, 27.7 kg) were fed diets containing 80% of one of five cereal grains differing in amylose, ß-glucan, and fiber content: 1) high-fermentable, high-ß-glucan, hull-less barley (HFB); 2) high-fermentable, high-amylose, hull-less barley (HFA); 3) moderate-fermentable, hull-less barley (MFB); 4) low-fermentable, hulled barley (LFB); and 5) low-fermentable, Canadian Western Red Spring wheat (LFW). On dry matter (DM) basis, cereal grains contained between 0.32% to 0.53% total P and 0.24% to 0.50% InsP6-P. The InsP6-2-P was calculated as the sum of all detected InsP-P (InsP6-P to InsP2-P) in the sample. The P release of degraded InsP-P was calculated by using the following equation: sum InsP6-2-Pdiet (g/kg DM) × (AID or ATTD sum InsP6-2-P (%)/ 100). Data were analyzed using a mixed model with diet as fixed effect, and pig and period as random effects. On DM basis, diets contained 41.4% to 50.6% starch, 0.88% to 8.54% ß-glucan, 0.81% to 0.89% total P, and 0.19% to 0.35% InsP6-P. The MFB, LFB, and LFW had greater (P < 0.05) diet AID of P than HFB and HFA, and MFB had greater (P < 0.05) diet ATTD and STTD of P than HFB. The ATTD of InsP6-P was greater (P < 0.05) for HFB than LFB and the ATTD of the sum InsP6-2-P was greater (P < 0.05) for HFB and HFA than LFB. Total tract P release was greater (P < 0.001) for HFB, HFA, and LFW than MFB and LFB. The LFW had greater (P < 0.05) ATTD of Ca than LFB. Diet ß-glucan content was not correlated with STTD of P (R2 = 0.03) or ATTD of InsP6 (R2 = 0.05). In conclusion, cereal grains high in fermentable fiber, e.g., amylose and ß-glucans included in specific hull-less barley cultivars, had lower diet AID, ATTD, and STTD of P, but greater ATTD of InsP6-P and sum InsP6-2-P. Carbohydrate fermentation, thus, results in greater total tract P release from InsP-P hydrolysis.


Increased fermentable carbohydrates (e.g., ß-glucan, amylose) may increase intestinal endogenous phosphorus (P) losses and thereby reduce P digestibility. The study assessed effects of cereal grains varying in fermentable carbohydrates on non-phytate-P and phytate-P. Phytate is the major binding form of P in plant seed and is incompletely degraded. Seven barrows cannulated at the terminal ileum were fed diets containing 80% of one of five cereal grains: 1) high-fermentable, high-ß-glucan, hull-less barley; 2) high-fermentable, high-amylose, hull-less barley; 3) moderate-fermentable, hull-less barley; 4) low-fermentable, hulled barley; and 5) low-fermentable wheat. Diet ileal digestibility of P was greater for low- and moderate-fermentable grain than high-fermentable grain, and diet total tract digestibility of P was greater for moderate-fermentable barley than high ß-glucan barley. Total tract digestibility of phytate-P was greater for high ß-glucan than low-fermentable barley. Total tract P release was greater for high-fermentable barley, and wheat than moderate- and low-fermentable barley. In conclusion, cereal grains high in fermentable fiber had lower diet ileal and total tract digestibility of P resulting in greater excretion of P, but greater total tract digestibility of phytate-P. Carbohydrate fermentation, thus, increases total tract P release from phytate-P degradation.


Asunto(s)
Hordeum , Fósforo Dietético , beta-Glucanos , Amilosa , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Canadá , Dieta/veterinaria , Fibras de la Dieta/metabolismo , Digestión , Grano Comestible/química , Hordeum/metabolismo , Fósforo/metabolismo , Fósforo Dietético/metabolismo , Almidón/metabolismo , Porcinos , Triticum/metabolismo
7.
JPEN J Parenter Enteral Nutr ; 46(8): 1828-1838, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35383975

RESUMEN

BACKGROUND: Infants and children with short bowel syndrome (SBS) are presumed to be at risk of gut microbial dysbiosis with potential sequelae of bacterial overgrowth that include sepsis, d-lactic acidosis, mucosal inflammation, and malabsorption. In neonatal piglets with SBS, we compared intestinal microbial composition, short-chain fatty acids (SCFAs), and adaptation given probiotic (PRO) treatment (Lactobacillus and Bifidobacterium spp) vs oral metronidazole (MET). METHODS: Following 75% distal small intestinal resection, piglets were allocated to PRO (500 mg twice a day, n = 7), MET (15 mg/kg twice a day, n = 8), and placebo (PLA) (500 mg twice a day, n = 8). After 10 days of parenteral and enteral nutrition, 16S ribosomal RNA gene amplicon sequencing (colon tissue and stool) was undertaken and SCFA analysis (stool and colon effluent) was performed using gas chromatography. RESULTS: In colon, Shannon diversity was higher for PRO compared with MET and PLA (P = 0.002). PRO and PLA increased abundance of Bacteroidetes species (eg, Bacteroides fragilis) compared with MET (P < 0.001). PRO, compared with PLA, increased abundance of Firmicutes species (eg, Lactobacillus fermentum) (P < 0.001). MET increased abundance of Proteobacteria members, predominately Enterobacteriaceae, compared with PRO (P = 0.004). In stool, microbial findings were similar and SCFA (butyrate) concentrations were highest for PRO (P = 0.003) compared with MET. CONCLUSION: In pediatric SBS, the empiric use of oral antibiotics, such as MET, is common for presumed clinical consequences of microbial dysbiosis. In this study of SBS piglets, that approach was associated with decreased microbial diversity and increased abundance of potentially inflammatory Proteobacteria. In contrast, a PRO treatment using Lactobacillus and Bifidobacterium spp increased both diversity and SCFAs.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Probióticos , Síndrome del Intestino Corto , Animales , Porcinos , Disbiosis/tratamiento farmacológico , Disbiosis/complicaciones , Síndrome del Intestino Corto/tratamiento farmacológico , Síndrome del Intestino Corto/complicaciones , Antibacterianos/uso terapéutico , Microbioma Gastrointestinal/genética , Heces/microbiología , Probióticos/uso terapéutico , Ácidos Grasos Volátiles , Lactobacillus , Proteobacteria , Poliésteres
8.
Microorganisms ; 9(12)2021 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-34946118

RESUMEN

High-fat diets (HFD) have been shown to induce substantial shifts in intestinal microbial community composition and activity which are associated with adverse metabolic outcomes. Furthermore, changes in microbial composition are affected by fatty acid composition; saturated, monounsaturated (MUFA), and industrial trans fats (iTFA) adversely affect microbial diversity while polyunsaturated fats (PUFA) have been shown to have neutral effects. The effects of naturally occurring trans fats on gut microbial composition are unknown. Vaccenic acid (VA) is the most abundant naturally occurring trans fat (abundant in meat and dairy), can be elevated by altering a cow's diet, and has been shown to have hypolipidemic effects. The aim of this study was to determine how variations of VA content in beef fat affect gut microbial composition, insulin resistance, and lipid metabolism in pigs. Low birth weight (LBW) and control pigs were fed a control or high-fat, high-carbohydrate (HFHC) diet supplemented with beef fat containing either high or low VA levels for 7 weeks. An adapted modified oral glucose tolerance test and fat challenge test were performed at 9 weeks of age following implantation of jugular catheters. Impacts on microbial composition were assessed using 16S rRNA gene amplicon sequencing. The HFHC diet containing beef fat rich in VA had a mild insulin sensitizing effect (p < 0.05, slope of curve), increased plasma HDL cholesterol (p < 0.05, +28%), reduced postprandial plasma TG (p < 0.05), and showed protection from HFHC-induced changes to gut microbial composition in LBW pigs as compared to HFHC diet containing standard beef fat. This is the first study to show effects of natural trans fats on gut dysbiosis; further studies are needed to elucidate mechanisms.

9.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R303-R316, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34259034

RESUMEN

Neonatal antibiotics administered to human infants initiate gut microbiota dysbiosis that may have long-term effects on body weight and metabolism. We examined antibiotic-induced adaptations in pancreatic islets of the piglet, a well-accepted model of human infant microbiota and pancreas development. Neonatal piglets randomized to amoxicillin [30 mg/kg body wt/day; n = 7, antibiotic (ANTI)] or placebo [vehicle control; n = 7, control (CON)] from postnatal day (PND)0-13 were euthanized at PND7, 14, and 49. The metabolic phenotype along with functional, immunohistological, and transcriptional phenotypes of the pancreatic islets were studied. The gut microbiome was characterized by 16S rRNA gene sequencing, and microbial metabolites and microbiome-sensitive host molecules were measured. Compared with CON, ANTI PND7 piglets had elevated transcripts of genes involved in glucagon-like peptide 1 ((GLP-1) synthesis or signaling in islets (P < 0.05) coinciding with higher plasma GLP-1 (P = 0.11), along with increased tumor necrosis factor α (Tnf) (P < 0.05) and protegrin 1 (Npg1) (P < 0.05). Antibiotic-induced relative increases in Escherichia, Coprococcus, Ruminococcus, Dehalobacterium, and Oscillospira of the ileal microbiome at PND7 normalized after antibiotic withdrawal. In ANTI islets at PND14, the expression of key regulators pancreatic and duodenal homeobox 1 (Pdx1), insulin-like growth factor-2 (Igf2), and transcription factor 7-like 2 (Tcf7l2) was downregulated, preceding a 40% reduction of ß-cell area (P < 0.01) and islet insulin content at PND49 (P < 0.05). At PND49, a twofold elevated plasma insulin concentration (P = 0.07) was observed in ANTI compared with CON. We conclude that antibiotic treatment of neonatal piglets elicited gut microbial changes accompanied by phasic alterations in key regulatory genes in pancreatic islets at PND7 and 14. By PND49, reduced ß-cell area and islet insulin content were accompanied by elevated nonfasted insulin despite normoglycemia, indicative of islet stress.


Asunto(s)
Antibacterianos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Animales , Microbioma Gastrointestinal/fisiología , Glucagón/efectos de los fármacos , Glucagón/metabolismo , Insulina/sangre , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Porcinos
10.
JPGN Rep ; 2(4): e127, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37206457

RESUMEN

The objectives of this pilot study were to examine fecal microbiota composition of pediatric patients with celiac disease (CD) before and after a 1-year gluten-free diet (GFD) and to determine the association with symptoms and anti-tissue transglutaminase (aTTG) antibody. Methods: Stool samples were obtained from pediatric patients with CD and from healthy controls. Patients were classified by the presence (diarrhea, abdominal pain, weight loss) or absence (asymptomatic, headache, fatigue, etc.) of typical CD gastrointestinal symptoms and by aTTG normalization post-GFD intervention (< 7 U/mL). Fecal microbial composition was measured using 16S ribosomal RNA gene amplicon sequencing of the V3-V4 region. Results: At diagnosis, 13 of 22 patients with CD had typical gastrointestinal symptoms, the remaining patients having atypical or asymptomatic presentations. After a 1-year GFD, all symptomatic patients improved and 9 of 19 had normalized aTTG. Prior to GFD, no distinct microbial signature was observed between patients and controls (P = 0.39). Post-GFD, patients with CD had a unique microbial signature with reductions in known fiber-degrading bacteria, including Blautia, Dorea, Lactobacillus, and Prevotella compared with controls. Within the patients with CD, microbial composition was not associated with reported symptom presentation or aTTG normalization. Conclusions: Pediatric patients with CD only had a unique microbial signature compared with healthy controls when placed on the GFD. These results suggest that pediatric patients with CD may not have a unique fecal microbial signature indicative of inherent dysbiosis, in contrast to that suggested for older patients. In children with CD, diet may play a role in shaping microbial composition more so than disease status.

11.
Br J Nutr ; 125(2): 129-138, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-32684173

RESUMEN

Kefir consumption has been demonstrated to improve lipid and cholesterol metabolism; however, our previous study identified that benefits vary between different commercial and traditional kefir. Here, we investigate the ability of pitched culture kefir, that is, kefir produced by a small number of specific strains, to recapitulate health benefits of a traditional kefir, in a diet-induced obesity mouse model, and examine how microbial composition of kefir impacts these benefits. Eight-week-old female C57BL/6 mice were fed a high-fat diet (40 % energy from fat) supplemented with one of five kefir varieties (traditional, pitched, pitched with no Lactobacillus, pitched with no yeast and commercial control) at 2 ml in 20 g of food for 8 weeks prior to analysis of plasma and liver lipid profiles, and liver gene expression profiles related to lipid metabolism. Both traditional and pitched kefir lowered plasma cholesterol by about 35 % (P = 0·0005) and liver TAG by about 55 % (P = 0·0001) when compared with commercial kefir despite no difference in body weight. Furthermore, pitched kefir produced without either yeast or Lactobacillus did not lower cholesterol. The traditional and pitched kefir with the full complement of microbes were able to impart corresponding decreases in the expression of the cholesterol and lipid metabolism genes encoding 3-hydroxy-3-methylglutaryl-coenzyme A reductase, PPARγ and CD36 in the liver. These results demonstrate that traditional kefir organisms can successfully be utilised in a commercial process, while highlighting the importance of microbial interactions during fermentation in the ability of fermented foods to benefit host health.


Asunto(s)
Kéfir/microbiología , Obesidad/metabolismo , Animales , Colesterol/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Alimentos Fermentados/microbiología , Lactobacillus/metabolismo , Metabolismo de los Lípidos/genética , Lípidos/sangre , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/microbiología , Levaduras/metabolismo
12.
PLoS One ; 15(4): e0231942, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32339193

RESUMEN

Salmonella enterica serovar Typhimurium is an animal welfare and public health concern due to its ability to parasite livestock and potentially contaminate pork products. To reduce Salmonella shedding and the risk of pork contamination, antibiotic therapy is used and can contribute to antimicrobial resistance. Here we hypothesized that immune system education by the microbiota can play a role in intestinal resilience to infection. We used amoxicillin (15mg/Kg) to modulate the intestinal microbiome of 10 piglets, paired with same age pigs that received a placebo (n = 10) from 0 to 14 days of age. Animals were euthanized at 4-weeks old. Each pig donated colon sections for ex vivo culture (n = 20 explants/pig). Explants were inoculated with S. Typhimurium, PBS or LPS (n = 6 explants/pig/group, plus technical controls). The gut bacteriome was characterized by sequencing of the 16S rRNA at 7, 21 days of age, and upon in vitro culture. Explants response to infection was profiled through high-throughput mRNA sequencing. In vivo antibiotic treatment led to ß-diversity differences between groups at all times (P<0.05), while α-diversity did not differ between amoxicillin and placebo groups on day 21 and at euthanasia (P<0.03 on day 7). Explant microbiomes were not different from in vivo. In vitro challenge with S. Typhimurium led to lower necrosis scores in explants from amoxicillin-treated pigs, when compared to explants placebo-treated pigs (P<0.05). This was coupled with the activation of immune-related pathways in explants from amoxicillin-treated pigs (IL-2 production, NO production, BCR activation), when compared to placebo-treated pigs. In addition, several DNA repair and intestinal wound healing pathways were also only activated in explants from amoxicillin-treated pigs. Taken together, these findings suggest that immune education by the amoxicillin-disturbed microbiota may have contributed to mitigate intestinal lesions following pathogen exposure.


Asunto(s)
Antibacterianos/farmacología , Epigénesis Genética/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Salmonella typhimurium/patogenicidad , Amoxicilina/farmacología , Animales , Animales Recién Nacidos , Bacterias/genética , Bacterias/aislamiento & purificación , Colon/citología , Colon/microbiología , Colon/patología , Regulación hacia Abajo/efectos de los fármacos , Heces/microbiología , Análisis de Componente Principal , ARN Ribosómico 16S/química , ARN Ribosómico 16S/metabolismo , Salmonelosis Animal/inmunología , Porcinos , Enfermedades de los Porcinos/inmunología , Regulación hacia Arriba/efectos de los fármacos
13.
Front Immunol ; 10: 2059, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552023

RESUMEN

Antibiotic exposure during neonatal development may result in transient or persistent alterations of key microbes that are vital for normal development of local and systemic immunity, potentially impairing immune competence later in life. To further elucidate the relationship between antibiotic exposure and immune development, newborn pigs were exposed to a therapeutic pediatric dose (30 mg/kg/day) of amoxicillin (AB) or placebo (PL) from post-natal day (PND) 0-14. Subsequently, immune cell phenotype, microbial composition, and immune response to an intraperitoneal (IP) challenge with Salmonella enterica serovar Typhimurium were evaluated. AB exposure caused significant changes in fecal microbial composition on PND 3 (P = 0.025). This stemmed from a 2-fold increase in Enterobacteriaceae with live cecal coliforms on PND 7 indicating at 10-fold increase (P = 0.036). Alterations in microbial composition were transient, and successional patterns were normalizing by PND 14 (P = 0.693). Differences in PBMC (peripheral blood mononuclear cell) immune cell subtypes were detected, with the percentage of CD3+CD4+ T cells among the broader T cell population (CD3+CD4+/CD3+) being significantly higher (P = 0.031) in AB pigs and the numbers of CD4+CD45RA+ (naïve) T cells per liter of blood were lower on PND 21 in AB pigs (P = 0.036). Meanwhile, PBMCs from AB pigs produced significantly more IFNγ upon stimulation with a T-cell mitogen on PND 21 and 49 (P = 0.021). When AB pigs were challenged with heat-killed Salmonella (IP) on PND 49, IFNγ gene expression in peripheral blood was upregulated compared to those treated with PL (P = 0.043). Additionally, AB pigs showed stronger activation among neutrophils infiltrating the peritoneal cavity after in vivo immune challenge, based on higher levels of NF-κB nuclear translocation (P = 0.001). Overall, our results indicate that early life treatment with a therapeutically relevant dose of a commonly prescribed antibiotic has a programming effect on the immune system. Despite antibiotics only causing a transient disruption in gut-associated microbial communities, implications were long-term, with antibiotic treated pigs mounting an upregulated response to an immune challenge. This research adds to the growing body of evidence indicating adverse immune outcomes of early life antibiotic exposures.


Asunto(s)
Amoxicilina/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Animales , Animales Recién Nacidos , Antibacterianos/farmacología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Ciego/efectos de los fármacos , Ciego/microbiología , Células Cultivadas , Heces/microbiología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Salmonella typhi/inmunología , Porcinos , Factores de Tiempo
14.
Front Immunol ; 10: 1802, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31447837

RESUMEN

The gastrointestinal tract microbiome plays a critical role in regulating host innate and adaptive immune responses against pathogenic bacteria. Disease associated dysbiosis and environmental induced insults, such as antibiotic treatments can lead to increased susceptibility to infection, particularly in a hospital setting. Dietary intervention is the greatest tool available to modify the microbiome and support pathogen resistance. Some dietary components can maintain a healthy disease resistant microbiome, whereas others can contribute to an imbalanced microbial population, impairing intestinal barrier function and immunity. Characterizing the effects of dietary components through the host-microbe axis as it relates to gastrointestinal health is vital to provide evidence-based dietary interventions to mitigate infections. This review will cover the effect of dietary components (carbohydrates, fiber, proteins, fats, polyphenolic compounds, vitamins, and minerals) on intestinal integrity and highlight their ability to modulate host-microbe interactions as to improve pathogen resistance.


Asunto(s)
Infecciones Bacterianas/inmunología , Dieta , Interacciones Microbiota-Huesped , Mucosa Intestinal/inmunología , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Fibras de la Dieta/administración & dosificación , Proteínas en la Dieta/administración & dosificación , Resistencia a la Enfermedad , Microbioma Gastrointestinal , Humanos , Fitoquímicos/administración & dosificación
15.
Front Vet Sci ; 6: 48, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30886850

RESUMEN

Animal performance, feed efficiency, and overall health are heavily dependent on gut health. Changes in animal production systems and feed regulations away from the use of antibiotic growth promoters (AGP) have necessitated the identification of strategies to optimize gut health in novel and effective ways. Among alternatives to AGP, the inclusion of dietary fibers (DF) in monogastric diets has been attempted with some success. Alternative feedstuffs and coproducts are typically rich in fiber and can be used in the diets to reduce feed costs and optimize gut health. DF are naturally occurring compounds with a diverse composition and are present in all plant-based feedstuffs. DF stimulate the growth of health-promoting gut bacteria, are fermented in the distal small intestine and large intestine to short-chain fatty acids and have beneficial effects on the immune system. Maternal DF supplementation is one novel strategy suggested to have a beneficial programming effect on the microbial and immune development of their offspring. One mechanism by which DF improves gut health is through maintenance of an anaerobic intestinal environment that subsequently prevents facultative anaerobic pathogens from flourishing. Studies with pigs and poultry have shown that fermentation characteristics and their beneficial effects on gut health vary widely based on type, form, and the physico-chemical properties of the DF. Therefore, it is important to have information on the different types of DF and their role in optimizing gut health. This review will provide information and updates on different types of DF used in monogastric nutrition and its contribution to gut health including microbiology, fermentation characteristics, and innate and adaptive immune responses.

16.
Vaccine ; 37(13): 1743-1755, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30808565

RESUMEN

We investigated gene expression patterns in whole blood and fecal microbiota profile as potential predictors of immune response to vaccination, using healthy M. hyopneumoniae infection free piglets (n = 120). Eighty piglets received a dose of prophylactic antibiotics during the first two days of life, whereas the remaining 40 did not. Blood samples for RNA-Seq analysis were collected on experimental Day 0 (D0; 28 days of age) just prior to vaccination, D2, and D6 post-vaccination. A booster vaccine was given at D24. Fecal samples for microbial 16SrRNA sequencing were collected at 7 days of age, and at D0 and D35 post-vaccination. Pigs were ranked based on the levels of M. hyopneumoniae-specific antibodies in serum samples collected at D35, and groups of 'high' (HR) and 'low' (LR) responder pigs (n = 15 each) were selected. Prophylactic antibiotics did not influence antibody titer levels and differential expression analysis did not reveal differences between HR and LR at any time-point (FDR > 0.05); however, based on functional annotation with Ingenuity Pathway Analysis, D2 post-vaccination, HR pigs were enriched for biological terms relating to increased activation of immune cells. In contrast, the immune activation decreased in HR, 6 days post-vaccination. No significant differences were observed prior to vaccination (D0). Two days post-vaccination, multivariate analysis revealed that ADAM8, PROSER3, B4GALNT1, MAP7D1, SPP1, HTRA4, and ENO3 genes were the most promising potential biomarkers. At D0, OTUs annotated to Prevotella, CF21, Bacteroidales and S24-7 were more abundant in HR, whereas Fibrobacter, Paraprevotella, Anaerovibrio, [Prevotella], YRC22, and Helicobacter positively correlated with the antibody titer as well as MYL1, SPP1, and ENO3 genes. Our study integrates gene differential expression and gut microbiota to predict vaccine response in pigs. The results indicate that post-vaccination gene-expression and early-life gut microbiota profile could potentially predict vaccine response in pigs, and inform a direction for future research.


Asunto(s)
Vacunas Bacterianas/inmunología , Heces/microbiología , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Mycoplasma hyopneumoniae/inmunología , Neumonía Porcina por Mycoplasma/prevención & control , Transcriptoma , Animales , Vacunas Bacterianas/administración & dosificación , Porcinos , Vacunación
17.
Nutrients ; 9(11)2017 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-29137145

RESUMEN

Several studies have demonstrated the beneficial impact of dried peas and their components on glucose tolerance; however, the role of gut microbiota as a potential mediator is not fully examined. In this study, we investigated the effect of dietary supplementation with raw and cooked pea seed coats (PSC) on glucose tolerance, microbial composition of the gut, select markers of intestinal barrier function, and short chain fatty acid profile in glucose intolerant rats. Male Sprague Dawley rats were fed high fat diet (HFD) for six weeks to induce glucose intolerance, followed by four weeks of feeding PSC-supplemented diets. Cooked PSC improved glucose tolerance by approximately 30% (p < 0.05), and raw and cooked PSC diets reduced insulin response by 53% and 56% respectively (p < 0.05 and p < 0.01), compared to HFD (containing cellulose as the source of dietary fiber). 16S rRNA gene sequencing on fecal samples showed a significant shift in the overall microbial composition of PSC groups when compared to HFD and low fat diet (LFD) controls. At the family level, PSC increased the abundance of Lachnospiraceae and Prevotellaceae (p < 0.001), and decreased Porphyromonadaceae (p < 0.01) compared with HFD. This was accompanied by increased mRNA expression of mucin genes Muc1, Muc2, and Muc4 in ileal epithelium (p < 0.05). Serum levels of acetate and propionate increased with raw PSC diet (p < 0.01). These results indicate that supplementation of HFD with PSC fractions can improve glycemia and may have a protective role against HFD-induced alterations in gut microbiota and mucus layer.


Asunto(s)
Bacterias/crecimiento & desarrollo , Glucemia/metabolismo , Fibras de la Dieta/administración & dosificación , Suplementos Dietéticos , Ácidos Grasos Volátiles/sangre , Microbioma Gastrointestinal , Intolerancia a la Glucosa/dietoterapia , Íleon/metabolismo , Íleon/microbiología , Mucinas/metabolismo , Pisum sativum , Animales , Bacterias/clasificación , Bacterias/genética , Biomarcadores/sangre , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/microbiología , Insulina/sangre , Masculino , Mucinas/genética , Ocludina/metabolismo , Ratas Sprague-Dawley , Factores de Tiempo , Receptores Toll-Like/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
18.
J Nutr ; 147(11): 2031-2040, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28954838

RESUMEN

Background: Changes in whole-grain chemical composition can affect the site of nutrient digestion, which may alter substrate availability and gut microbiota composition.Objective: This study elucidated the function of whole-grain fermentable fiber composition on ileal substrate flow, hindgut substrate availability, and subsequent gut microbial profiles in pigs.Methods: Five whole grains-1) high-fermentability, high-ß-glucan hull-less barley (HFB); 2) high-fermentability, high-amylose hull-less barley (HFA); 3) moderate-fermentability hull-less barley (MFB); 4) low-fermentability hulled barley (LFB); or 5) low-fermentability hard red spring wheat (LFW)-were included at 800 g/kg into diets fed to ileal-cannulated growing pigs for 9 d in a 6 (periods) × 5 (diets) Youden square. Digesta were analyzed for nutrient flow and microbial composition via 16S ribosomal RNA gene sequencing.Results: The consumption of fermentable whole grains, HFB, and HFA increased (P < 0.05) ileal starch flow by 69% and dry matter flow by 37% compared with LFB and LFW intakes. The consumption of HFB and HFA increased (P < 0.05) fecal Firmicutes phylum abundance by 26% and 21% compared with LFB intake and increased (P < 0.05) fecal Dialister genus abundance, on average, by 98% compared with LFB and LFW intakes. Fecal Sharpea and Ruminococcus genera abundances increased (P < 0.05) with HFB intake compared with LFB and LFW intakes. In contrast, the consumption of LFB increased (P < 0.05) fecal Bacteroidetes phylum abundance by 43% compared with MFB intake. Ileal starch flow and fecal Firmicutes abundance were positively correlated and determined by using principal components analysis.Conclusions: Increasing dietary fermentable fiber from whole grains can increase ileal substrate flow and hindgut substrate availability, shifting the fecal microbiota toward Firmicutes phylum members. Thus, digesta substrate flow is important to shape gut microbial profiles in pigs, which indicates that the manipulation of substrate flow should be considered as a tool to modulate gut microbiota composition.


Asunto(s)
Fibras de la Dieta/análisis , Microbioma Gastrointestinal , Íleon/microbiología , Almidón/análisis , Granos Enteros/química , Amilosa/administración & dosificación , Amilosa/análisis , Alimentación Animal , Animales , Dieta/veterinaria , Fibras de la Dieta/administración & dosificación , Heces/química , Heces/microbiología , Fermentación , Firmicutes/aislamiento & purificación , Firmicutes/metabolismo , Hordeum/química , Íleon/metabolismo , Lactobacillaceae/aislamiento & purificación , Lactobacillaceae/metabolismo , Masculino , Análisis de Componente Principal , ARN Ribosómico 16S/aislamiento & purificación , Ruminococcus/aislamiento & purificación , Ruminococcus/metabolismo , Almidón/administración & dosificación , Porcinos , Triticum/química , beta-Glucanos/administración & dosificación , beta-Glucanos/análisis
19.
Appl Environ Microbiol ; 83(17)2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28667114

RESUMEN

Antibiotics are important for treating bacterial infection; however, efficacies and side effects of antibiotics vary in medicine and experimental models. A few studies have correlated microbiota composition variations with health outcomes in response to antibiotics; however, no study has demonstrated causality. We had noted variation in colonic expression of C-type lectins, regenerating islet-derived protein 3ß (Reg3ß) and Reg3γ, after metronidazole treatment in a mouse model. To investigate the effects of specific variations in the preexisting microbiome on host response to antibiotics, mice harboring a normal microbiota were allocated to 4 treatments in a 2-by-2 factorial arrangement with or without commensal Escherichia coli and with or without metronidazole in drinking water. E. coli colonized readily without causing a notable shift in the microbiota or host response. Metronidazole administration reduced microbiota biodiversity, indicated by decreased Chao1 and Shannon index values, and altered microbiota composition. However, the presence of E. coli strongly affected metronidazole-induced microbiota shifts. Remarkably, this single commensal bacterium in the context of a complex population led to variations in host responses to metronidazole treatment, including increased expression of antimicrobial peptides Reg3ß and Reg3γ and intestinal inflammation indicated by tumor necrosis factor alpha levels. Similar results were obtained from 2-week antibiotic exposure and with additional E. coli isolates. The results of this proof-of-concept study indicate that even minor variations in initial commensal microbiota can drive shifts in microbial composition and host response after antibiotic administration. As well as providing an explanation for variability in animal models using antibiotics, the findings encourage the development of personalized medication in antibiotic therapies.IMPORTANCE This work provides an understanding of variability in studies where antibiotics are used to alter the gut microbiota to generate a host response. Furthermore, although providing evidence only for the one antibiotic, the study demonstrated that initial gut microbial composition is a key factor driving host response to antibiotic administration, creating a compelling argument for considering personalized medication based on individual variations in gut microbiota.


Asunto(s)
Antibacterianos/administración & dosificación , Bacterias/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/microbiología , Metronidazol/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Escherichia coli/fisiología , Femenino , Humanos , Intestinos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Simbiosis/efectos de los fármacos
20.
Front Microbiol ; 8: 848, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28553275

RESUMEN

The rumen microbiome is integral to efficient production in cattle and shows strong host specificity, yet little is known about what host factors shape rumen microbial composition. Secretory immunoglobulin A (SIgA) is produced in large amounts in the saliva, can coat both commensal and pathogenic microbes within the gut, and presents a plausible mechanism of host specificity. However, the role salivary SIgA plays in commensal bacteria selection in ruminants remains elusive. The main objectives of this study were to develop an immuno-affinity benchtop method to isolate SIgA-tagged microbiota and to determine if salivary SIgA preferentially binds selected bacteria. We hypothesized that SIgA-tagged bacteria would differ from total bacteria, thus supporting a potential host-derived mechanism in commensal bacterial selection. Whole rumen (n = 9) and oral secretion samples (n = 10) were incubated with magnetic beads conjugated with anti-secretory IgA antibodies to enrich SIgA-tagged microbiota. Microbial DNA from the oral secretion, whole rumen, SIgA-tagged oral secretion, and SIgA-tagged rumen was isolated for amplicon sequencing of V1-V3 region of 16S rDNA genes. Whole rumen and oral secretion had distinctive (P < 0.05) bacterial compositions indicated by the non-parametric multidimensional scaling plot using Euclidean distance metrics. The SIgA-tagged microbiota from rumen and oral secretion had similar abundance of Bacteroidetes, Actinobacteria, Fibrobacter, candidate phyla TM7, and Tenericutes and are clustered tightly. Composition of SIgA-tagged oral secretion microbiota was more similar to whole rumen microbiota than whole oral secretion due to enrichment of rumen bacteria (Lachnospiraceae) and depletion of oral taxa (Streptococcus, Rothia, Neisseriaceae, and Lactobacillales). In conclusion, SIgA-tagged oral secretion microbiota had an increased resemblance to whole rumen microbiota, suggesting salivary SIgA-coating may be one host-derived mechanism impacting commensal colonization. Further studies, to explore the variations in antibody affinity between different animals as a driver of microbial composition are warranted.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...