Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-39406387

RESUMO

The post-weaning period in pigs is a critical window where nutritional interventions are implemented to prevent post-weaning diarrhea (PWD) and antibiotic use. One common strategy is feeding of low protein diets immediately following weaning. This intervention may reduce protein fermentation and pathogen proliferation, therefore decreasing the incidence of post-weaning diarrhea. These effects may also be mitigated by providing dietary fiber. However, studies examining the role of protein and fiber on gastrointestinal microbiota and metabolism are complicated by the presence of other substrates, including polyphenols and antinutritional factors in complex ingredients. In this study, semi-purified diets formulated to meet nutrient requirements were fed to 40 weaned pigs (n = 10/diet) to examine the effects of high protein (HP), high fiber (HF), or both (HFHP) compared to a control (CON) diet with industry standard crude protein and fiber content. Critical alterations in host metabolism and cecal transcriptome were identified in response to the CON diet. Diets with lower protein levels (CON and HF) induced alteration in transcripts from the serine synthesis pathways and integrated stress response in cecal tissue alongside systemic increases in metabolic pathways related to lysine degradation. High protein diets did not induce increases in gastrointestinal pathogen abundance. These results challenge the practice of feeding low protein diets post-weaning, by demonstrating a detrimental effect on intestinal cell function and muscle accretion. This suggests that with careful ingredient selection, increased dietary protein post-weaning could better pig health and growth compared to a standard diet.

2.
Appl Environ Microbiol ; 89(11): e0097723, 2023 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-37902410

RESUMO

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.


Assuntos
Micobioma , Animais , Suínos , Longevidade , Animais Selvagens , Sus scrofa , Fezes/microbiologia
3.
Appl Environ Microbiol ; 88(24): e0159322, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36448784

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Micobioma , Animais , Feminino , Combinação Amoxicilina e Clavulanato de Potássio/farmacologia , Antibacterianos/farmacologia , Fungos , RNA Ribossômico 16S/genética , Suínos
4.
Microb Pathog ; 173(Pt A): 105873, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36371065

RESUMO

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.


Assuntos
Colite , Disenteria , Infecções por Bactérias Gram-Negativas , Microbiota , Infecções por Spirochaetales , Doenças dos Suínos , Suínos , Animais , Doenças dos Suínos/patologia , Disenteria/veterinária , Disenteria/patologia , Imunidade Inata , Infecções por Bactérias Gram-Negativas/patologia
5.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R303-R316, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34259034

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Microbioma Gastrointestinal/fisiologia , Glucagon/efeitos dos fármacos , Glucagon/metabolismo , Insulina/sangue , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Suínos
6.
Br J Nutr ; 125(2): 129-138, 2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-32684173

RESUMO

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.


Assuntos
Kefir/microbiologia , Obesidade/metabolismo , Animais , Colesterol/sangue , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Alimentos Fermentados/microbiologia , Lactobacillus/metabolismo , Metabolismo dos Lipídeos/genética , Lipídeos/sangue , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/microbiologia , Leveduras/metabolismo
7.
Appl Environ Microbiol ; 83(17)2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28667114

RESUMO

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.


Assuntos
Antibacterianos/administração & dosagem , Bactérias/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Intestinos/microbiologia , Metronidazol/administração & dosagem , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Escherichia coli/fisiologia , Feminino , Humanos , Intestinos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Simbiose/efeitos dos fármacos
8.
J Nutr ; 147(11): 2031-2040, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28954838

RESUMO

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.


Assuntos
Fibras na Dieta/análise , Microbioma Gastrointestinal , Íleo/microbiologia , Amido/análise , Grãos Integrais/química , Amilose/administração & dosagem , Amilose/análise , Ração Animal , Animais , Dieta/veterinária , Fibras na Dieta/administração & dosagem , Fezes/química , Fezes/microbiologia , Fermentação , Firmicutes/isolamento & purificação , Firmicutes/metabolismo , Hordeum/química , Íleo/metabolismo , Lactobacillaceae/isolamento & purificação , Lactobacillaceae/metabolismo , Masculino , Análise de Componente Principal , RNA Ribossômico 16S/isolamento & purificação , Ruminococcus/isolamento & purificação , Ruminococcus/metabolismo , Amido/administração & dosagem , Suínos , Triticum/química , beta-Glucanas/administração & dosagem , beta-Glucanas/análise
9.
J Nutr ; 147(1): 29-36, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27798343

RESUMO

BACKGROUND: Variant chemical composition and physical structure of whole grains may change the site of energy digestion from the small to the large intestine. OBJECTIVE: We determined the site of nutrient digestion, standardized ileal digestibility (SID) of amino acids (AAs), and net energy (NE) value of barley cultivars that vary in nutrient composition compared with wheat. METHODS: Ileal-cannulated barrows (27.7 kg initial body weight) were fed diets containing 800 g whole grains/kg alongside a basal and nitrogen-free diet for calculations in a 6 (period) × 7 (diet) Youden square. Diets included 1 of 5 whole grains-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, hard red spring wheat (LFW). Intestine nutrient flow and whole-body energy utilization were tested and explained by using whole-grain and digesta confocal laser scanning. RESULTS: Starch apparent ileal digestibility was 14-29% lower (P < 0.05) in HFB and HFA than in MFB, LFB, and LFW due to the unique embedding of starch within the protein-fiber matrix of HFB and the high amylose content in HFA. Starch hindgut fermentation was 50-130% higher (P < 0.05) in HFB and HFA than in MFB, LFB, and LFW. The SID of indispensable AAs was lower (P < 0.05) in HFB and HFA than in MFB, LFB, and LFW. NE value was 18% higher (P < 0.05) for HFB than for HFA and was not different from MFB, LFB, and LFW. CONCLUSIONS: Whole grains high in fermentable carbohydrates shifted digestion from the small intestine to the hindgut. NE value depended on the concentration of fermentable fiber and starch and digestible protein, ranging from 2.12-1.76 Mcal/kg in barley to 1.94 Mcal/kg in wheat. High-fiber whole grains may be used as energy substrates for pigs; however, the reduced SID of AAs requires titration of indispensable AAs to maintain growth.


Assuntos
Aminoácidos/metabolismo , Fibras na Dieta/análise , Digestão/efeitos dos fármacos , Íleo/efeitos dos fármacos , Suínos/fisiologia , Grãos Integrais , Ração Animal/análise , Fenômenos Fisiológicos da Nutrição Animal , Animais , Dieta/veterinária , Fibras na Dieta/farmacologia , Digestão/fisiologia , Metabolismo Energético/fisiologia , Íleo/metabolismo , Masculino
10.
J Nutr ; 145(11): 2464-70, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26377761

RESUMO

BACKGROUND: Dietary amylose resists enzymatic digestion, thereby providing a substrate for microbial fermentation that stimulates proliferation of beneficial microbiota and production of short-chain fatty acids (SCFAs) in the large intestine of pigs and humans. However, the effect of increasing dietary amylose in pigs immediately postweaning on growth, nutrient digestibility and flow, and intestinal microbial and SCFA profiles has not been studied and can be used as a model for newly weaned human infants. OBJECTIVE: We studied the effects of increasing dietary amylose on growth, nutrient digestibility, and intestinal microbial and metabolite profiles in weaned pigs. METHODS: Weaned pigs (n = 32) were randomly allocated to 1 of 4 diets containing 67% starch with 0%, 20%, 28%, or 63% amylose for 21 d. Subsequently, pigs were killed to collect feces and digesta for measuring starch digestion and microbial and metabolite profiles. RESULTS: Feeding weaned pigs 63% compared with 0%, 20%, and 28% amylose decreased (P < 0.05) feed intake by 5% and growth by ≥ 12%. Ileal digestibility of dry matter decreased (P < 0.05) by 10% and starch by 9%, thereby increasing (P < 0.05) hindgut fermentation, cecal and colonic total SCFAs, and colonic Bacteroides, and lowering (P < 0.01) ileal, cecal, and colonic pH in pigs consuming 63% compared with 0%, 20%, and 28% amylose. Cecal and colonic Bifidobacteria spp. increased by 14-30% (P < 0.05) and Clostridium clusters IV and XIVa were decreased (P < 0.01) in pigs consuming 63% compared with 0%, 20%, and 28% amylose. CONCLUSION: Increasing dietary amylose in pigs immediately postweaning stimulated hindgut fermentation and Bifidobacteria spp., thereby manipulating the gut environment, but also reduced intake and growth. An optimum dietary amylose concentration should be determined, which would maintain desired growth rate and gut environment in weaned pigs.


Assuntos
Amilose/administração & dosagem , Ceco/metabolismo , Fermentação , Íleo/metabolismo , Amido/administração & dosagem , Desmame , Amilose/química , Ração Animal/análise , Animais , Bacteroides/isolamento & purificação , Bifidobacterium/isolamento & purificação , Ceco/microbiologia , DNA Bacteriano/genética , Dieta/veterinária , Digestão , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Microbioma Gastrointestinal , Dosagem de Genes , Genoma Bacteriano , Genômica , Concentração de Íons de Hidrogênio , Íleo/microbiologia , Amido/química , Suínos
11.
Microbiol Spectr ; 12(10): e0084324, 2024 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-39162552

RESUMO

The microbiome of wild animals is believed to be co-evolved with host species, which may play an important role in host physiology. It has been hypothesized that the rigorous hygienic practices in combination with antibiotics and diets with simplified formulas used in the modern swine industry may negatively affect the establishment and development of the gut microbiome. In this study, we evaluated the fecal microbiome of 90 domestic pigs sampled from nine farms in Canada and 39 wild pigs sampled from three different locations on two continents (North America and Europe) using 16S rRNA gene amplicon sequencing. Surprisingly, the gut microbiome in domestic pigs exhibited higher alpha-diversity indices than wild pigs (P < 0.0001). The wild pig microbiome showed a lower Firmicutes-to-Bacteroidetes ratio and a higher presence of bacterial phyla Elusimicrobiota, Verrucomicrobiota, Cyanobacteria, and Fibrobacterota when compared to their domestic counterparts. At the genus level, the wild pig microbiome had enriched genera that were known for fiber degradation and short-chain fatty acid production. Interestingly, the phylum Fusobacteriota was only observed in domestic pigs. We identified 31 ASVs that were commonly found in the pig gut microbiome, regardless of host sources, which could be recognized as members of the core gut microbiome. Interestingly, we found five ASVs missing in domestic pigs that were prevalent in wild ones, whereas domestic pigs harbored 59 ASVs that were completely absent in wild pigs. The present study sheds light on the impact of domestication on the pig gut microbiome, including the gain of new genera, which might provide the basis to identify novel targets to manipulate the pig gut microbiome for improved health. IMPORTANCE: The microbiome of pigs plays a crucial role in shaping host physiology and health. This study sought to identify if domestication and current rearing practices have resulted in a loss of co-evolved bacterial species by comparing the microbiome of wild boar and conventionally raised pigs. It provides a comparison of domestic and wild pigs with the largest sample sizes and is the first to examine wild boars from multiple sites and continents. We were able to identify core microbiome members that were shared between wild and domestic populations, and on the contrary to expectation, few microbes were identified to be lost from wild boar. Nevertheless, the microbiome of wild boars had a lower abundance of important pathogenic genera and was distinct from domestic pigs. The differences in the microbial composition may identify an opportunity to shift the microbial community of domestic pigs towards that of wild boar with the intent to reduce pathogen load.


Assuntos
Bactérias , Fezes , Microbioma Gastrointestinal , RNA Ribossômico 16S , Sus scrofa , Animais , Suínos/microbiologia , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , RNA Ribossômico 16S/genética , Fezes/microbiologia , Sus scrofa/microbiologia , Animais Selvagens/microbiologia , Canadá , Europa (Continente) , Filogenia , Microbiota , Biodiversidade
12.
FEMS Microbiol Ecol ; 99(7)2023 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-37349964

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Microbiota , Suínos , Animais , Feminino , Transplante de Microbiota Fecal , Desmame , Projetos Piloto , Fezes , Metaboloma
13.
J Anim Sci ; 100(6)2022 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-35569054

RESUMO

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.


Assuntos
Hordeum , Fósforo na Dieta , beta-Glucanas , Amilose , Ração Animal/análise , Fenômenos Fisiológicos da Nutrição Animal , Animais , Canadá , Dieta/veterinária , Fibras na Dieta/metabolismo , Digestão , Grão Comestível/química , Hordeum/metabolismo , Fósforo/metabolismo , Fósforo na Dieta/metabolismo , Amido/metabolismo , Suínos , Triticum/metabolismo
14.
JPEN J Parenter Enteral Nutr ; 46(8): 1828-1838, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35383975

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Microbiota , Probióticos , Síndrome do Intestino Curto , Animais , Suínos , Disbiose/tratamento farmacológico , Disbiose/complicações , Síndrome do Intestino Curto/tratamento farmacológico , Síndrome do Intestino Curto/complicações , Antibacterianos/uso terapêutico , Microbioma Gastrointestinal/genética , Fezes/microbiologia , Probióticos/uso terapêutico , Ácidos Graxos Voláteis , Lactobacillus , Proteobactérias , Poliésteres
15.
J Anim Sci ; 100(11)2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36205053

RESUMO

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.


Assuntos
Ração Animal , Ácido Benzoico , Suínos , Animais , Desmame , Ração Animal/análise , Filogenia , RNA Ribossômico 16S/genética , Dieta/veterinária , Fibras na Dieta/metabolismo , Ceco/microbiologia , Diarreia/veterinária
16.
JPGN Rep ; 2(4): e127, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37206457

RESUMO

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.

17.
Microorganisms ; 9(12)2021 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-34946118

RESUMO

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.

18.
PLoS One ; 15(4): e0231942, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32339193

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Epigênese Genética/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Salmonella typhimurium/patogenicidade , Amoxicilina/farmacologia , Animais , Animais Recém-Nascidos , Bactérias/genética , Bactérias/isolamento & purificação , Colo/citologia , Colo/microbiologia , Colo/patologia , Regulação para Baixo/efeitos dos fármacos , Fezes/microbiologia , Análise de Componente Principal , RNA Ribossômico 16S/química , RNA Ribossômico 16S/metabolismo , Salmonelose Animal/imunologia , Suínos , Doenças dos Suínos/imunologia , Regulação para Cima/efeitos dos fármacos
19.
Front Immunol ; 10: 1802, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31447837

RESUMO

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.


Assuntos
Infecções Bacterianas/imunologia , Dieta , Interações entre Hospedeiro e Microrganismos , Mucosa Intestinal/imunologia , Carboidratos da Dieta/administração & dosagem , Gorduras na Dieta/administração & dosagem , Fibras na Dieta/administração & dosagem , Proteínas Alimentares/administração & dosagem , Resistência à Doença , Microbioma Gastrointestinal , Humanos , Compostos Fitoquímicos/administração & dosagem
20.
Front Vet Sci ; 6: 48, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30886850

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA