ABSTRACT

The microbiota plays a vital role in maintaining gut health and influences the overall performance of chickens. Most gut microbiota-related studies have been performed in broilers, which have different microbial communities compared to those of layers. The normal gut microbiota of laying chickens is dominated by Proteobacteria, Firmicutes, Bacteroidetes, Fusobacteria, and Actinobacteria at the phylum level. The composition of the gut microbiota changes with chicken age, genotype, and production system. The metabolites of gut microbiota, such as short-chain fatty acids, indole, tryptamine, vitamins, and bacteriocins, are involved in host-microbiota cross talk, maintenance of barrier function, and immune homeostasis. Resident gut microbiota members also limit and control the colonization of foodborne pathogens. In-feed supplementations of prebiotics and probiotics strengthen the gut microbiota for improved host performance and colonization resistance to gut pathogens, such as Salmonella and Campylobacter. The mechanisms of action of prebiotics and probiotics come through the production of organic acids, activation of the host immune system, and production of antimicrobial agents. Probiotic candidates, including Lactobacillus, Bifidobacterium, Bacillus, Saccharomyces, and Faecalibacterium isolates, have shown promising results toward enhancing food safety and gut health. Additionally, a range of complex carbohydrates, including mannose oligosaccharides, fructo-oligosaccharides, and galacto-oligosaccharides, and inulin are promising candidates for improving gut health. Here, we review the potential roles of prebiotics and probiotics in the reshaping of the gut microbiota of layer chickens to enhance gut health and food safety.

INTRODUCTION

The gut microbiota is a complex community of hundreds of diverse microorganisms. The gut microbiota influences the host, playing a role in the modulation of the immune system, nutrient digestion, and regulation of intestinal function. These modulatory effects are mediated by the complex microbial interactions and metabolites produced by the microbial community members or derived from the transformation of host molecules or diet (1, 2). The microbial metabolites involved in host and microbiota cross talk include short-chain fatty acids (SCFAs), tryptamine, conjugated linoleic acids, indole and its derivatives, and bile acids transformed by the gut microbiota (1). Broilers and layers have different genotypes, very different lifespans in normal commercial production, and are reared in different conditions with different dietary requirements. Therefore, the composition of the gut microbiota in these two lines is different (3). The microbiota differences can differentially influence bird responses to stimuli and challenges. For example, differences in the gut immune response to Campylobacter jejuni have been correlated with different patterns of microbiota composition between broilers and layers (2). The laying chicken gut microbial communities are influenced by multiple factors, such as flock age, production system, disease, diet, and antibiotics (4).
The gut microbiota composition can be enhanced and strengthened with the use of prebiotic and probiotic supplements in the feed. Prebiotics are most commonly complex oligosaccharides that are not digested by host enzymes and, hence, end up in the lower gut where they promote the growth and multiplication of resident gut microbiota. Therefore, prebiotics are fed to enhance the growth of beneficial resident gut bacteria. Examples of prebiotics are inulin, galacto-oligosaccharides, fructo-oligosaccharides, xylo-oligosaccharides, pectin, beta-glucans, and resistant starch. Probiotics are viable bacteria that, when delivered in sufficient quantity, can improve host health. Apart from a range of mostly Gram-positive bacteria, some yeasts and molds have also been used as probiotics. The bacterial genera most commonly used as probiotics include Bacillus, Lactobacillus, Enterococcus, Bifidobacterium, and Streptococcus. Probiotics influence the gut by one or a combination of mechanisms, including that they modulate the host immune system, provide energy via SCFA production, and influence gut structure, integrity, and function. Probiotics also directly influence other bacteria, including pathogens, by production of metabolites and antimicrobial compounds, occupation of ecological niches within the gut to competitively exclude colonization of other bacteria, and by lowering the luminal pH. Some probiotic bacteria attach to receptors on enterocytes and activate Toll-like receptors (TLRs), leading to the induction of cytokine expression (5).
An important application of prebiotics and probiotics in layers is to reduce the colonization of pathogenic bacteria. Gut pathogens, such as Salmonella and Campylobacter, cause clinical diseases in many animals and humans. To trigger an inflammatory response, Salmonella internalizes into enterocytes and survives within macrophages and M cells (6). Laying chicks infected with Salmonella show higher viable counts in the ileum, cecum, and colon than in the crop (7). Unlike mammals, adult laying chickens colonized with nontyphoidal Salmonella spp. generally show no clinical signs; although mucoid and blood-tinged feces may be occasionally present (8). However, poultry-specific Salmonella serotypes, such as Salmonella enterica serovar Gallinarum and Salmonella enterica serovar Pullorum cause clinical diseases in chickens (9). The three main species of Campylobacter that cause health and food safety problems in poultry are Campylobacter jejuni, Campylobacter coli, and Campylobacter hepaticus. Among them, C. hepaticus has attracted recent attention because it has been found to cause spotty liver disease in laying chickens (10). In C. hepaticus, gene clusters associated with stress response, sialic acid modification, glucose utilization, and hydrogen metabolism are implicated in its pathogenicity (11). Chickens infected with C. jejuni and C. coli are often asymptomatic but still cause a potential threat to public health. In vitro study suggests that Campylobacter jejuni establishes its niche in epithelial cells via mechanisms involving serine protease HtrA (12). In laying chickens, prebiotics and probiotics have been used to reduce Salmonella and Campylobacter colonization in the gut (1315). Therefore, the positive manipulation of the gut microbiota is a useful approach to improve food safety and control avian diseases such as spotty liver disease. Even though researchers and consumers generally accept the health benefit claims of prebiotics and probiotics, the underlying molecular mechanisms of action are not fully understood. An in-depth investigation of the underlying principles of the action of prebiotics and probiotics will provide confidence to use such supplements for therapeutic purposes. The objective of this review is to survey and summarize findings from the existing literature on gut microbiota in laying chickens as well as the use of prebiotics and probiotics for improving gut health and food safety in laying chickens and make recommendations for some of the key areas of focus for future work.

The composition of chicken gut microbiota and its role in gastrointestinal health.

The composition of gut microbiota in laying chickens varies among the functionally different segments of the gastrointestinal tract, reflecting their different physiochemical microenvironments (Fig. 1). The compartment pH, redox potential, growth substrates, antibacterial secretions, and metabolites from host and microbiota influence the colonization efficiency of microbes in the gut segments. The proximal segments of the gut are characterized by low pH, which strongly selects acid-tolerant bacteria and limits the growth of most pathogens (16). The crop is dominated by Blautia, Lactobacillus, Bacillus, Pseudomonas, Enterococcus, and Staphylococcus, while in ceca, in addition to the above, other bacteria such as Faecalibacterium, Bifidobacterium, Clostridium, and Ruminococcus are also abundant (17, 18). In the ceca of mature laying chickens, the representative microbial communities at the phylum level, in order of their typical abundance, are Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Deferribacteres, Fusobacteria, Verrucomicrobia, Synergistetes, and Lentisphaerae (19).
FIG 1
FIG 1 Composition and diversity of gut microbiota vary in different segments of the gut in chickens. The microbial composition and diversity are affected by multiple factors including host genetics, age, diet, and rearing conditions. The most common microbial genera depicted here may not be complete, as there is not enough information on individual gut segment microbiota in laying chickens. The information regarding genera present within each segment was collected from a range of relevant literature cited in the text.
Gut microbial communities produce a range of metabolites that are involved in host functions, such as energy sources, cell to cell communication, and immune system regulation (Table 1). SCFAs and tryptophan catabolites affect host-microbiota cross talk (1). Microbial communities are able to metabolize dietary tryptophan into indole and its derivatives. Many indole derivatives, such as indole-3-acetaldehyde, indoleacrylic acid, indole-3-acid-acetic, and indole-3-aldehyde, act as aryl hydrocarbon receptor (AhR) ligands and modulate local and distant host functions that include epithelial barrier physiology and immune system homeostasis (1). The Clostridiaceae, Ruminococcaceae, and Lachnospiraceae contain diverse gene complements that encode enzymes involved in carbohydrate metabolism (20). The Ruminococcaceae are enriched in xylanase and cellulase genes, while both the Ruminococcaceae and Lachnospiraceae produce α-glucosidases and both α- and β-galactosidases (20). Members of the Lachnospiraceae and Ruminococcaceae families can cleave cellulose and hemicellulose to release sugars for utilization by both microbes and host; therefore, Lachnospiraceae and Ruminococcaceae may perform better than Clostridiaceae in degrading plant materials for the production of organic acids that are used by the host as energy sources (20). Future research could focus on investigating the hypothesis that the formation of gut microbial metabolites shapes the integrity of the epithelium and can be manipulated to improve gut barrier function in laying chickens.
TABLE 1
TABLE 1 Microbial metabolites and their functions in host gut and systemic healtha
Gut metabolite Microbe(s) involved Target(s) of metabolite Effect(s) on gut health Reference(s)
Acetate Faecalibacterium, Anaerostipes, Eubacterium, Lactobacillus Activates GLP1, GPR41 and GPR43; activates MAPK pathway; energy substrate Provides energy for enterocytes; cosubstrate for butyrate production; increases mineral absorption; inhibits growth of pathogens; regulates T cells in ceca; attenuates inflammasome activation 78, 79
Propionate Escherichia coli, Propionibacterium, Roseburia inulinivorans, Negativicutes, Bacteroides, Selenomonas ruminantium, Lactobacillus, Akkermansia Activates GLP1, GPR41, GPR43; upregulates leptin, peptide YY, and glucagon-like peptide Antilipogenic and anti-inflammatory; provides energy for enterocytes; increases satiety 79, 80
Butyrate Lachnospiraceae, Ruminococcus, Bacteroides, Lactobacillus, Faecalibacterium, Eubacterium Activates GLP1; inhibits histone deacetylase; suppresses NF-κB pathway Inhibits histone deacetylase; modulates immune system 79, 81
Succinate Prevotella copri, Veillonella parvula, Phascolarctobacterium succinatutens Activates SUCNR1 Proinflammatory; intermediate in propionate production 82
Tryptamine Bacteroides thetaiotaomicron, Ruminococcus gnavus, Clostridium sporogenes Activates epithelial 5-HT4R to increase cAMP level in gut Barrier functions; immune system modulation 83
Indole Eubacterium hallii, Clostridium bartlettii, Lactobacillus, Bacteroides fragilis, Parabacteroides distasonis, Bifidobacterium longum, Escherichia coli Modulates glucagon-like peptide 1 secretion; activates AhR and PXR in gut Improves host metabolism; maintains host-microbe homeostasis; reduces inflammation and improves mucosal barrier function 84, 85
10-Hydroxy-cis-12-octadecenoate Lactobacillus plantarum Activates Nrf2 and GPR40; inhibits ERK phosphorylation Anti-inflammatory; maintains intestinal barrier function; reduces obesity 86
Conjugated linoleic acid Lactobacillus casei Modulates cytokine production; activates PPARα and PPARγ; inhibits cyclooxygenase and lipoxygenase Anti-inflammatory; reduces pathogen growth; reduces obesity 87
Bacteriocins Multiple bacteria Targets cell wall of pathogens; inhibits pathogen DNA gyrase-mediated DNA supercoiling; blocks aminoacyl-tRNA binding to the 50S ribosome Inhibits the growth of pathogenic bacteria 70, 88
Vitamins K2 and B-complex Multiple species of bacteria, including Lactobacillus, Bifidobacterium, Bacteroides, Enterobacter, Serratia, Enterococcus, Lactococcus K2 acts as a cofactor for γ-glutamyl carboxylase; B-complex acts as a cofactor for multiple enzymes Cell metabolism; role in clotting; anticoccidiostate; regulates immune system 8991
a
The gut metabolites mentioned in this table have been studied in nonchicken models. In chickens, mainly SCFAs have been studied, showing various dietary effects on their production. The molecular targets and functions of the gut microbiota metabolites in chickens need to be investigated for the broader role of beneficial microbes in health and disease. It should also be noted that gut effects reported in the table at a genus level refer to unidentified species of the genus and do not imply that the effect is existing in all species of that genus. AhR, aryl hydrocarbon receptor; ERK, extracellular signal-regulated kinase; GLP1, glucagon-like peptide 1; GPR41, G protein-coupled receptor 41; MAPK, mitogen-activated protein kinase; Nrf2, nuclear factor erythroid 2-related factor 2; PPARα, peroxisome proliferator-activated receptor alpha.

Rearing conditions and flock age affect the gut microbiota.

Rearing conditions and host-related factors, such as production system, sex, age, breed, and feed, may have profound effects on the development and composition of gut microbiota. However, rigorous analysis cannot be made, as there is not much literature available on the effects of these conditions on gut microbiota composition and diversity in layers. Correlations have been noted between gender, genotype, age, and body composition and the abundance of a number of microbial genera. For example, Lactobacillus, Lactococcus, and Bifidobacterium were found to be more abundant in low-body-weight laying chickens (21). The gut microbiota develops rapidly from day 1 to 3, and around day 7, most of the organisms that are found in the mature microbiota are already present, although the relative numbers tend to fluctuate for several weeks before stabilizing (22). After 2 weeks posthatch, Ruminococcus and Oscillospira increase substantially while the representation of Enterococcus is reduced (22). Compared with week 8 of chicken age, at week 30 Firmicutes and Bacteroidetes become more abundant in the gut (23). Assessing the effect of age (week 1 to 60) on the composition of gut microbiota in laying chickens, Proteobacteria, Firmicutes, and Bacteroidetes formed the vast majority of microbiota across all age categories (24). This shows that Gram-negative bacteria dominate the gut at an early age while Firmicutes become more prominent in the later age of the laying cycle of hens.
As chickens get older, the integrity of the gut mucosal system is compromised due to changes in the composition of the gut microbiota. The production system affects the development and composition of gut microbiota with a higher abundance of microbial genera involved in amino acid and glycan metabolic pathways in free-range compared with that in cage laying chickens (25). The abundance of Bacteroidetes is lower in cage birds, while the abundance of Firmicutes is higher in free-range birds (25). This shows that rearing conditions shape the composition of the gut microbiota. Further study of the natural development of gut microbiota in layers should be aimed at analysis of the effects on growth performance, egg production traits, and resistance to pathogen infection. Given that the production of free-range eggs is increasing globally, future studies on the development of gut microbiota should include a comparative analysis of hens raised in cage, free-range, and barn systems and the role of range soil microbiota in the modulation of chicken gut health. It could be hypothesized that the gut pathogens, once introduced, will persist for a long period, thereby affecting the composition and diversity of gut microbiota in hens during production.

Difference in the composition and diversity of gut microbiota in broilers and layers.

The phylogenetic composition of microbiota typically found in various intestinal segments of broilers is well characterized (reviewed in references 26 and 27). However, in layers, there is only limited literature available on the composition of microbiota in the gut. The environmental conditions and physiological functions of the gut vary along its length, and these differences are reflected in different populations of microbial communities in various segments of the gut. The gut microbiota is more complex and richer in layers than in broilers (28). Fundamental differences in the composition of gut microbiota between broilers and layers have been reported. For example, using a >0.1% abundance threshold for microbiota analysis, Cytophaga, Thermobaculum, Geobacillus, Desulfovibrio, Cyclobacterium, Caldicellulosiruptor, and Caulobacter were present in the ceca of indigenous Indian layers but were not detected in broilers (3). It is evident from literature that many bacterial genera are common between broilers and layers; however, differences exist in the abundance and richness of individual genera due to differences in the energy requirements of broilers and layers. Given that broilers and layers have major differences in their physiology, husbandry, feeding practices, and life span, it is difficult to compare their gut microbiota. Moreover, the endocrine changes at the onset of lay may potentially influence the gut microbiota in laying hens. Future research could test the hypothesis that differences in the composition of gut microbiota in each gut segments exist between broilers and layers reared in the same or different production systems and could explore the functional consequences of different microbiota compositions.

Campylobacter and Salmonella colonization in the chicken gut.

Egg-based products are among the leading causes of foodborne outbreaks of Salmonella infection. Gut dysbiosis is a microbial imbalance that results in the overgrowth of pathogenic bacteria and can lead to systemic infection (Fig. 2). The resident gut microbiota produces metabolites that inhibit the colonization of pathogenic bacteria. The efficient utilization of available nutrients by microbiota depletes the metabolic niches for pathogenic bacteria, such as Salmonella and Campylobacter. The resident gut microbiota can outcompete pathogens by saturating binding sites on gut epithelium that result in competitive exclusion. The host epithelial cells sense pathogen-associated molecular patterns (PAMPs) and thus boost the secretion of mucus, immunoglobulins, and antimicrobial peptides (AMPs). However, through the use of hyb hydrogenase enzymes, Salmonella can grow by consuming molecular H2 secreted by microbiota (29). Once Salmonella establishes a colonization niche, it regulates the virulence genes vital for multiplication in the lumen and invasion into the host cells. The mechanisms of different host gut colonization by Salmonella and Campylobacter are different, as unlike Campylobacter (type IV secretory system [T4SS]), Salmonella employs a type III secretory system (T3SS) for establishing a niche and internalization into organs. Both Salmonella and Campylobacter can translocate via the transcellular or paracellular routes by breaking down tight junctions. The pathogens also release effector proteins and toxins that facilitate their colonization and invasion.
FIG 2
FIG 2 Generalized mechanisms of colonization by foodborne pathogens in the gut. Pathogens use various metabolic pathways to overcome the resident gut microbiota for establishing a niche in the gut. AMPs, antimicrobial peptides; cdtA, cytolethal distending toxin subunit A; hybA, hydrogenase-2 electron transfer unit; frdA, fumarate reductase subunit; T4SS, type IV secretory system.
Campylobacter is a microaerophilic pathogen that requires O2, H2, and CO2 for its growth; however, under anaerobic conditions, Campylobacter spp. express several virulence factors (30) necessary for cell invasion. Campylobacter jejuni and Campylobacter coli can colonize chickens, usually without serious pathogenic effects; however, they cause clinical disease in humans. C. jejuni is transmitted mainly horizontally (31) in hens; however, globally, there is limited evidence for the association of Campylobacter-contaminated table eggs with human campylobacteriosis. The prevalence of C. jejuni in the gut is affected by production system, with its incidence higher in free-range chickens (32), suggesting that a different pattern of host gut microbiota composition may influence its colonization. C. hepaticus causes spotty liver disease in laying chickens with great economic losses. Spotty liver disease is more common in free-range chickens, and the pathogen is present in different segments of the gut of the infected birds (33). Genes that encode effector molecules required for niche adaptation, virulence, gut colonization, and invasion have been tentatively identified in C. hepaticus (33). One interesting avenue worth exploring in the future is to investigate interactions within the gut microbiota and how Salmonella may influence the shedding levels of Campylobacter, possibly via the production of gut microbial metabolites, as in a mouse model, coinfection of Salmonella enterica serovar Typhimurium increased the virulence of C. jejuni (34).
The complex interactions among gut microbiota, Salmonella, Campylobacter, and host are not completely understood in chickens. In the nutrient-limited environment caused by the intestinal microbiota, Salmonella uses specific metabolic traits for the utilization of compounds that are not metabolized by gut microbiota (Fig. 3). For example, Salmonella utilizes 1,2-propanediol, a product released during the fermentation of l-fucose. Most of the Salmonella enterica serovars (35) and Campylobacter jejuni (36) contain the fucose utilization operons that provide them a competitive advantage for the colonization of the host gut. Other bacteria, such as Escherichia coli and Lactobacillus rhamnosus GG, contain genes for fucose fermentation; however, their interactions with Salmonella and Campylobacter for consumption of fucose need to be investigated. It appears that inflamed epithelial cells present adhesion receptor sites that are exploited only by pathogenic bacteria. Reactive oxygen species generated by neutrophils during inflammation can react with endogenous thiosulfate to form tetrathionate. The ttrRSBCA locus on Salmonella pathogenicity island 2 confers Salmonella Typhimurium the ability to use tetrathionate as a terminal electron acceptor in anaerobic respiration (6). This confers a growth advantage to Salmonella, as it can use ethanolamine as a carbon source in the presence of tetrathionate (37). Under anaerobic conditions and in the presence of tetrathionate, 1,2-propanediol can serve as an energy source for Salmonella Typhimurium (35).
FIG 3
FIG 3 Generalized model shows that intestinal inflammation triggered by Salmonella Typhimurium results in its proliferation. Salmonella Typhimurium, once in the lumen of the gut, upregulates SPI1 to trigger intestinal inflammation. As a result, the released chemokines trigger the release of reactive nitrogen species (RNS), reactive oxygen species (ROS), and lipocalin-2 from the enterocytes. In turn, the lipocalin-2 blocks resident gut microbiota growth leading to a burst of Salmonella Typhimurium. This growth is further helped by the Salmonella Typhimurium nitrate and tetrathionate respiration strategies maintaining its continuous division in the lumen and tissue invasion. To protect itself from the inducible nitric oxide synthase encoded by the infected macrophages, Salmonella Typhimurium upregulates the membrane-bound narGHJI, narZYWV, and periplasmic nap nitrate reductases that could use NO3 as a respiratory substrate. Inside the host cell in a Salmonella-containing vacuole, Salmonella faces various stresses induced by acidic pH, RNS, ROS, reduced concentrations of phosphate, magnesium and iron, and cationic antimicrobial peptides (CAMPs). In response, Salmonella upregulates various regulons vital for its survival inside the vacuole. In chickens, some of these pathways may differ as Salmonella infection does not always result in the development of the clinical disease.
With the proliferation of Salmonella Typhimurium, the T3SS triggers inflammatory host responses that shift competition in favor of the pathogen (38); however, the exact mechanism in chicken is not known. The Salmonella Typhimurium Tat (twin-arginine translocation) system contributes to intestinal infection by facilitating colonization of the gut of mammals (39); however, its role, if any, in the gut of chickens needs to be confirmed, as Tat-deficient mutants of Salmonella enterica serovar Enteritidis did not influence cecal colonization in Leghorn chickens (40). In this system, two Tat-exported enzymes, peptidoglycan amidase AmiA and AmiC, are responsible for the Tat-dependent colonization. Salmonella employs a wide range of metabolic strategies for surviving and establishing a niche in the host gut that seems to be different between mammals and chickens. Future research could focus on understanding the role of resident gut microbiota and microbial metabolites in response to Salmonella infection in chickens, as they do not always develop clinical disease.

The use of probiotics and prebiotics in layer chickens for gut health.

Diets supplemented with probiotics have been reported to significantly improve bird performance in terms of egg production and egg quality (41, 42). Probiotics improve the ecosystem of the gut in layers by balancing many of the microbial genera. For example, using culture medium as a method of quantitation, Bacillus subtilis increased the counts of bifidobacteria and lactobacilli and decreased clostridia and coliforms (41). Probiotic and synbiotic supplementation restored the gut ecosystem disrupted by Salmonella Typhimurium and increased the production of butyrate (43, 44). A Pediococcus acidilactici strain reduced the cholesterol level in egg yolk and improved tibial bone mineralization (42). An Enterococcus faecium strain and fructo-oligosaccharides significantly reduced serum cholesterol level in chickens and improved egg quality (45). Some probiotics have shown to lower pathogenic bacterial load, improve gut microbiota balance, and enhance the gut mucosal immune system (Table 2).
TABLE 2
TABLE 2 Probiotics used for gut health in layer chickens
Probiotic Model system Disease type or model Effect on gut microbiota Function(s) Reference(s)
Lactobacillus reuteri LM1 Layer Brachyspira pilosicoli-induced gut inflammation Microbiota was not investigated Reduced colonization of B. pilosicoli 92
Lactobacillus salivarius CTC2197 Layer S. Enteritidis-induced gut inflammation Microbiota was not investigated Reduced colonization of Salmonella 65
Multistrain probiotica Layer S. Enteritidis-induced gut inflammation Increased the population of Pediococcus and Lactobacillus Reduced colonization of Salmonella in ceca 93
Lactobacillus suspension or Lactobacillus plantarum Layer C. jejuni- and S. Enteritidis-induced infections Microbiota was not investigated Gut colonization resistance to C. jejuni enhanced; no significant effect on Salmonella load reduction in organs; upregulated IL-6, IL-10, and TLR4 in ileum 94, 95
Bacillus cereus var. toyoi (Toyocerin) Layer/broiler S. Enteritidis-induced enteritis Microbiota was not investigated Reduction in cecal Salmonella load 96
Bacillus subtilis PY79 SPF chicks S. Enteritidis- and Clostridium perfringens-induced enteritis Microbiota was not investigated Reduction in bacterial load in gut 97
Citrobacter diversus, Klebsiella pneumoniae, and Escherichia coli Layer C. jejuni colonization Microbiota was not investigated Reduced C. jejuni load in ceca 98
Cecal culture Layer C. jejuni colonization Microbiota was not investigated Reduced C. jejuni load in ceca 15
Bacillus subtilis B2A Layer S. Gallinarum challenge No significant effect on Lactobacillus and Escherichia populations in gut Reduced Salmonella load in gut 99
Bacillus subtilis and Bacillus amyloliquefaciens Layer S. Typhimurium challenge Restored abundance levels of Christensenellaceae R7 group, Lachnospiraceae UCG010, and Ruminococcaceae UCG005 Reduced overall load of Salmonella in feces 43
Multistrain probiotica Layer S. Typhimurium challenge Increased the abundance of Ruminococcus, Trabulsiella, Bifidobacterium, Holdemania, and Oscillospira No significant reduction in Salmonella load in ceca 44
a
Multistrain probiotics contain prebiotic component(s). There are not many research reports focused on the use of probiotics for their mechanistic effects on gut integrity and gut barrier functions in layers. Future research should focus on the strategic use of probiotics in layers for control of pathogens at different points of the production cycle. It is essential to select commercial probiotics that are shelf-stable, cost-effective, and feed-stable. There is substantial literature on the use of probiotics to control enteric pathogens in chicken meat birds; however, the long-term trials with laying chickens are limited due to costs and labor. Developing probiotic strains that can be used in place of in-feed antibiotic growth promoters for production of safer food products should be the target of future research. Characterization of Faecalibacterium, Propionibacterium, Saccharomyces, and Roseburia for improving gut health in laying chickens may be rewarding areas for future research. The increased abundance of Faecalibacterium in the gut of Salmonella Typhimurium-challenged laying hens that turned negative for Salmonella (43) suggests its potential role to be characterized as a probiotic candidate for gut health. The use of Saccharomyces in broilers has shown promising results on gut health. Propionibacterium exhibits anti-inflammatory properties and is used as a starter in the preparation of dairy products.
The use of prebiotics in layers has shown promising results for improving the population of certain beneficial bacterial genera in the gut. For example, a prebiotic increased the abundance levels of Lactobacillus and Olsenella and the expression of genes in microbial communities associated with propanoate and butanoate metabolism in the gut of layers (46). The prebiotics used for the control of Salmonella and Campylobacter in layer production are summarized in Table 3. Table 3 shows that the prebiotics produced variable results in terms of reducing pathogen load in the gut and priming the host immune system.
TABLE 3
TABLE 3 The use of prebiotics for gut health in laying chickensa
Prebiotic Model system Disease type or model Microbiota affected Function(s) Reference(s)
Inulin Chicken macrophage HD11 cell line S. Enteritidis-induced phagocytosis Not applicable Decreased viable cells of S. Enteritidis and production of IL-1β 100
Mannose-oligosaccharides Layer S. Enteritidis challenge Increased Bifidobacterium and Lactobacillus in ceca Reduced S. Enteritidis colonization in ceca 101
Fructo-oligosaccharides Layer S. Enteritidis challenge No significant change in Lactobacillus, Escherichia, Bifidobacterium, and Bacteroides populations in ceca Reduced S. Enteritidis colonization in ceca and feces; upregulated TLR4 and IFN-γ in ileum 102, 103
Galacto-oligosaccharides Layer S. Enteritidis/S. Typhimurium challenge Increased abundance of Lactobacillus and Clostridium No significant effect on Salmonella load in ceca 14
Galacto-oligosaccharides Layer S. Typhimurium challenge Increased abundance of Lactobacillus and Christensenellaceae Increased clearance of S. Typhimurium from gut 104
Fructo-oligosaccharides/lactose/mannose Layer Campylobacter jejuni challenge Did not investigate Reduced C. jejuni load in ceca 98
a
A range of prebiotics can be used to improve gut health in laying chickens. As a result, food safety can be improved when there is less Salmonella and Campylobacter contamination in egg or egg products. Research is needed to investigate the effectiveness of these and other prebiotics for control of Campylobacter hepaticus, which is causing significant economic losses in the layer sector. It could be hypothesized that the continuous feeding of prebiotics improves host digestive functions and resistance to the colonization of pathogens in different gut segments.

Mechanisms of action of probiotics.

The functions of probiotics are achieved through bacteria-bacteria and host-bacteria interactions (Fig. 4). The bacteria-bacteria interactions result in the production of SCFAs, modification of redox potential, the production of antimicrobial compounds, competition for epithelial receptors, quorum sensing, and production of ecosystems unsuitable for pathogen colonization. Probiotic strains of bacteria have cell walls that contain components, such as capsular polysaccharide, peptidoglycan, teichoic acids, and lipoproteins (47). These molecular components represent microbe-associated molecular patterns (MAMPs) that are recognized by specific host intestinal mucosal pattern-recognition receptors (PRRs) that function to prime the immune system to suppress pathogens (47).
FIG 4
FIG 4 Overview of the mechanisms of action of probiotics and prebiotics. Prebiotics increase the population and functionality of certain resident gut microbiota that in turn competitively exclude pathogenic bacteria by mechanisms that include the production of microbial metabolites, mucin production, and modulation of the host immune system. Unlike prebiotics, probiotics are directly available in the gut for the desired functions. During the microbial fermentation, short-chain fatty acids (SCFAs) are produced that exert beneficial effects on the host involving various mechanisms. Regulation of the host immune system is influenced by the increased biomass and cell wall components of the bacteria. Depending upon the compositions of prebiotics and probiotics, they may result in the increase of certain genera of gut microbiota, leading to decreased microbial diversity. The decreased microbial diversity may or may not be useful depending upon the nature of the microbial community. Future research needs to address the mechanistic understanding of the interactions of prebiotics and probiotics with the composition of gut microbiota and its metabolites in laying chickens in different production systems.

Immunomodulatory action.

Probiotics influence immune functions of the host by several pathways. In vitro studies have shown that maturation of human dendritic cells and production of interleukin-10 (IL-10) can be induced by the binding of Lactobacillus reuteri and Lactobacillus casei to CD209 (48). Other probiotics, such as Lactobacillus acidophilus NCFM, L. rhamnosus GG, and Lactobacillus plantarum WCFS1 have the potential to induce signaling via Toll-like receptors (TLRs) through the production of lipoteichoic acid that contains di-acyl or tri-acyl glycolipids (47, 49). Generally, the interactions between probiotics and host cells lead to the production of natural and antigen-specific antibodies, signal induction via TLRs, and regulation of T cells and cytokines. For example, in an in vitro study of chicken splenic and cecal tonsil cells, L. acidophilus and Lactobacillus salivarius induced Th1 and cytokine anti-inflammatory responses, respectively (50). Lactobacillus DNA induced STAT2, STAT4, IL-18, IFN-γ, MyD88, and IFN-α gene expression in chicken cecal tonsil cells (51). In broiler chicks challenged with Salmonella Typhimurium, a Lactobacillus-based probiotic lowered the expression levels of IL-1β, IL-6, and IFN-γ and increased the expression level of IL-10 in cecal tonsils (52). Gene expression of IFN-γ was significantly reduced following probiotic feeding of chickens infected with Salmonella (53). There seems to be a synergy between vaccines and probiotics on the gut immune system that can modulate the clearance of pathogens, as coadministration of L. reuteri and Anaerosporobacter mobilis with an N-glycan-based vaccine resulted in lower gut colonization by Campylobacter jejuni and improved immune response (serum IgY antibodies) and gut microbiota composition in broilers and specific-pathogen-free (SPF) leghorns (54). These results show that probiotics can be used both in prophylactic and therapeutic ways to prime cytokine expression to modulate the host immune system against pathogens. Future research needs to focus on a mechanistic approach to understand the roles of probiotics in regulating NF-κB and mitogen-activated protein kinase (MAPK) pathways in disease conditions in laying chickens. Future research should expand on the finding that the immune response elicited by probiotic bacteria varies with the bacterial strains, and, therefore, there is a need to identify a probiotic strain suitable for boosting the host immune response in the presence of live vaccine strains of gut pathogens, such as Salmonella aroA-based vaccines.

Regulation of tight junction proteins.

The integrity of the gut epithelium is maintained by the formation of tight junction protein complexes between cells. Tight junctions are multiprotein complexes that regulate the movement of ions and inhibit the translocation of pathogenic bacteria between gut epithelial cells. Changes in the tight junction proteins disrupt the intestinal mucosal barrier, thereby allowing the movement of pathogens across the epithelia. Probiotics influence barrier function by the direct inhibition of pathogens, by enhancing the synthesis of tight junction proteins, or by rearrangement of tight junction protein conformation (47). Studies in various animals have shown the beneficial effects of probiotics on the regulation of tight junction proteins in health and disease conditions. For example, Bifidobacterium infantis in mice suffering from necrotizing enterocolitis stabilized tight junctions by increasing the expression of claudin 2, 4, and 7 and occludin in gut tissue (55). In broiler chickens injected with lipopolysaccharide (LPS) from Escherichia coli, Bacillus subtilis probiotic upregulated the expression levels of JAM2, occludin, ZO1, and MUC2 (56). The stabilized tight junction proteins increased host epithelial barrier protection against pathogen invasion into internal organs. We suggest that research should focus on confirming the role of various probiotic strains in stabilizing the regulation of tight junction proteins in laying chickens challenged with foodborne pathogens, as laying cycles induce stress that may result in the increased chances of pathogens translocation in the gut.

Mucin production.

The gut epithelium consists of enterocytes, Paneth cells, goblet cells, M cells, and neuroendocrine cells, while the lamina propria contains immune cells, such as lymphocytes, macrophages, plasma cells, and dendritic cells. The enterocytes are mainly involved in nutrient absorption; the Paneth cells secrete AMPs, while the goblet cells produce mucin. Mucin is a site for bacterial adhesion with subsequent competition between commensal and pathogenic bacteria. The gut microbiota interacts with mucin on several different levels; it influences mucosal cell proliferation and mucin synthesis and degradation (57). Some probiotics promote the development of goblet cells and increase the production of mucin (58). In a mouse model, it has been shown that Bifidobacterium adheres to the intestinal mucus and secretes γ-aminobutyric acid as a metabolite that upregulates MUC2 for modulating the goblet cell functions with a net increase in mucin production (59). In broilers, supplementation of Lactobacillus-, Bifidobacterium-, and Enterococcus-based probiotics increased the goblet cell cup area in the gut and significantly upregulated the expression of MUC and increased the production of mucin glycoprotein (58). It appears that the induction of mucin production in the gut is strain specific, as in broilers, diet supplemented with Bacillus subtilis resulted in higher expression of MUC2 and increased goblet cell density and jejunal villi height; however, the Enterococcus-, Bifidobacterium-, and Lactobacillus-based supplemented diet resulted only in increased goblet cell density and jejunal villi height (60). Alteration of the composition of gut microbiota can result in mucin degradation during infection. For example, in mice infected with Citrobacter rodentium, the microbiota was dominated by bacterial species that degrade mucins (61). Future research in layers could investigate the hypothesis that strategic feeding of probiotics and prebiotics can restore the disruption of mucin production by gut pathogens, such as Salmonella and Campylobacter.

Competition for adhesion sites.

Some probiotic bacteria adhere to the apical brush borders of enterocytes, possibly thorough proteinaceous adhesion-promoting factors present in probiotic bacteria. Competitive exclusion due to inhibition of adhesion of pathogens in the gut has been studied using cell culture. For example, L. acidophilus inhibited the adhesion of S. Typhimurium to enterocyte-like Caco-2 cells (62). Some probiotic bacteria have strong aggregation properties that prevent pathogens from attachment to enterocytes. For example, in human uroepithelium, lipoteichoic acid of Lactobacillus inhibited the adherence of uropathogens (63). In in vitro conditions, multistrain probiotic bacteria competitively inhibited the attachment of Salmonella to human intestinal mucosa (64). L. salivarius CTC2197 seems to function through competitive exclusion to reduce Salmonella Enteritidis colonization in laying chickens (65). Although the competitive exclusion properties of probiotics have been studied and tested in vitro, their applicability and efficiency in colonization resistance to gut pathogens in chicken models are yet to be established. The most efficacious competitive exclusion products are complex bacterial mixtures derived from the ceca of healthy birds. Such products are difficult to standardize and quality control and, hence, are not acceptable in some markets. It would be desirable to find defined probiotic strains of bacteria that could perform as well as some of the undefined competitive exclusion products in reliably excluding foodborne pathogens and, thus, improve food safety in layers.

Bacterial metabolites.

Probiotics produce a range of metabolites that include SCFAs, indole, tryptamine, bacteriocins, and vitamins (Table 1). The three most common SCFAs produced by gut microbiota are propionate, butyrate, and acetate. The SCFAs are an important energy source for enterocytes and induce the production of host AMPs. The AMPs are produced as a result of binding of SCFAs to G protein-coupled receptors (e.g., GPR41 and GPR43) to stimulate β-defensins and RegIIIγ (66). Propionate can restrict the growth of Salmonella Typhimurium through the disruption of intracellular pH homeostasis (67). SCFAs inhibit the growth of Salmonella when present in the dissociated form. For example, in a coculture at pH 5.8, SCFAs inhibited the growth of Salmonella Enteritidis (68). SCFAs also modify the expression of Salmonella virulence genes. For example, the expressions of SPI1 gene regulators (hilA, hilD, and invF) in Salmonella Typhimurium were significantly reduced by propionyl-coenzyme A (propionyl-CoA), a product of propionate metabolism (69). As a range of microbes produce SCFAs, we suggest that further investigation of the efficacy of specific probiotic strains for production of gut metabolites in the presence and absence of pathogens would be desirable.

Bacteriocins production.

Bacteriocins are ribosomally synthesized peptides with antimicrobial properties produced mainly by Gram-positive bacteria; however, certain Gram-negative bacteria can also produce them. The widespread occurrence of bacteriocin peptides in bacterial species of the gut microbiota (70) suggests their regulatory role in population dynamics of gut pathogens as shown for the bacteriocin produced by L. salivarius UCC118 in Listeria-infected mice (71). A potent bacteriocin-producing probiotic, Enterococcus faecium KH 24, resulted in low shedding levels of Salmonella Enteritidis in a mouse model (72). None of the available probiotic strains for feed supplementation in laying chickens have been tested for the production of bacteriocins in vitro and in vivo. Therefore, the prominent role that bacteriocin production may play in the effectiveness of some probiotics in laying chickens needs to be confirmed. Enhancing their synthesis through strategic feeding of probiotics in the rearing phase of layer production might confer resistance to pathogenic bacterial colonization soon after transportation to production housing systems.

Mechanisms of action of prebiotics.

Prebiotics in feed specifically alter the abundance of bacteria within the gut microbiota. The action of prebiotics is exerted via these altered bacterial populations and the metabolites that are produced (Fig. 4). Therefore, we have avoided writing subsections on the mechanisms of action of prebiotics.
As nondigestible by host enzymes, the prebiotics reach the lower gut where they are available to the resident gut microbiota as nutrient. Genomic analysis of bifidobacteria and lactobacilli shows carbohydrate metabolic gene repertoires that are involved in fermentation (73). Fermentation releases sugars and SCFAs that lower the gut luminal pH (Fig. 4). Applying these mechanisms, xylo-oligosaccharides increase the number of lactobacilli in colon and Clostridium cluster XIVa in ceca (74). To enhance the growth of certain gut microbial community members, prebiotics act as a carbon and energy source for the growth of microbes, such as Bifidobacterium longum, Bifidobacterium adolescentis, Lactobacillus fermentum, and Lactobacillus brevis (75). Certain prebiotics inhibit the growth of pathogens in the gut by manipulating the mechanisms of pathogenicity. For example, Salmonella can bind to mannose via type 1 fimbriae, leading to colonization inhibition (76). The competitive exclusion is mainly achieved through the increased population of resident gut microbiota by saturating the available receptors on enterocytes; however, this property of the prebiotics has not been thoroughly investigated. Research is needed to understand the role of prebiotics in the development, composition, and diversity of microbial communities in different segments of the gut (including crop and gizzard) in the presence of pathogenic bacteria, as the current notion is that prebiotics are fermented mainly in ceca and colon. Research in laying chickens has confirmed that prebiotics possess immunomodulatory properties (Table 3) and may increase calcium transport in the gut for improving egg quality (77). Future research should focus on investigating the hypothesis that prebiotics improve shell quality and cuticle cover through the modulation of gut microbiota for increased mineral transport.

Conclusions and recommendations.

The gut microbiota is associated with the health and performance of birds. The available literature suggests that the gut microbiota in newly hatched chicks passes through different stages of maturation. However, further research is required to explicitly understand the effects of age, rearing conditions, and stress factors on the development and maturation of microbiota in various segments of the gut of layers. The commercial life span of layers is substantially longer than that of broilers. Hence, studies are required to understand the development and maturation of microbiota in different segments of the gut in laying chickens during their commercial life span. Establishment of a successful niche by Salmonella and Campylobacter in a chicken’s gut highlights their ability to switch onto various metabolic pathways that help them to overcome the host gut microbiota. Moreover, while Salmonella and Campylobacter are the main pathogens of concern in food safety, it is likely that prebiotics and probiotics will continue to play a role in the control of these pathogens. The microbiota can be modified by in-feed supplementation of prebiotics and probiotics for improving gut health. We suggest that future research should focus on understanding the mechanistic interactions between prebiotics/probiotics, gut segment microbiota, and pathogens in order to improve avian health and reduce the use of in-feed antibiotic growth promoters.

ACKNOWLEDGMENTS

This study was financially supported by Australian Eggs.
We declare no competing or financial competing interests.

REFERENCES

1.
Agus A, Planchais J, Sokol H. 2018. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 23:716–724.
2.
Han Z, Willer T, Pielsticker C, Gerzova L, Rychlik I, Rautenschlein S. 2016. Differences in host breed and diet influence colonization by Campylobacter jejuni and induction of local immune responses in chicken. Gut Pathog 8:56.
3.
Pandit RJ, Hinsu AT, Patel NV, Koringa PG, Jakhesara SJ, Thakkar JR, Shah TM, Limon G, Psifidi A, Guitian J, Hume DA, Tomley FM, Rank DN, Raman M, Tirumurugaan KG, Blake DP, Joshi CG. 2018. Microbial diversity and community composition of caecal microbiota in commercial and indigenous Indian chickens determined using 16s rDNA amplicon sequencing. Microbiome 6:115.
4.
Kers JG, Velkers FC, Fischer EA, Hermes GD, Stegeman JA, Smidt H. 2018. Host and environmental factors affecting the intestinal microbiota in chickens. Front Microbiol 9:235.
5.
Tejada-Simon MV, Pestka JJ. 1999. Proinflammatory cytokine and nitric oxide induction in murine macrophages by cell wall and cytoplasmic extracts of lactic acid bacteria. J Food Prot 62:1435–1444.
6.
Winter SE, Thiennimitr P, Winter MG, Butler BP, Huseby DL, Crawford RW, Russell JM, Bevins CL, Adams LG, Tsolis RM, Roth JR, Bäumler AJ. 2010. Gut inflammation provides a respiratory electron acceptor for Salmonella. Nature 467:426–429.
7.
Barrow PA, Simpson JM, Lovell MA. 1988. Intestinal colonisation in the chicken by food‐poisoning Salmonella serotypes; microbial characteristics associated with faecal excretion. Avian Pathol 17:571–588.
8.
Pande VV, Devon RL, Sharma P, McWhorter AR, Chousalkar KK. 2016. Study of Salmonella Typhimurium infection in laying hens. Front Microbiol 7:203.
9.
Berchieri A, Jr, Murphy CK, Marston K, Barrow PA. 2001. Observations on the persistence and vertical transmission of Salmonella enterica serovars Pullorum and Gallinarum in chickens: effect of bacterial and host genetic background. Avian Pathol 30:221–231.
10.
Van TTH, Elshagmani E, Gor MC, Scott PC, Moore RJ. 2016. Campylobacter hepaticus sp. nov., isolated from chickens with spotty liver disease. Int J Syst Evol Microbiol 66:4518–4524.
11.
Van TTH, Lacey JA, Vezina B, Phung C, Anwar A, Scott PC, Moore RJ. 2019. Survival mechanisms of Campylobacter hepaticus identified by genomic analysis and comparative transcriptomic analysis of in vivo and in vitro derived bacteria. Front Microbiol 10:107.
12.
Harrer A, Bücker R, Boehm M, Zarzecka U, Tegtmeyer N, Sticht H, Schulzke JD, Backert S. 2019. Campylobacter jejuni enters gut epithelial cells and impairs intestinal barrier function through cleavage of occludin by serine protease HtrA. Gut Pathog 11:4.
13.
Wang L, Li L, Lv Y, Chen Q, Feng J, Zhao X. 2018. Lactobacillus plantarum restores intestinal permeability disrupted by Salmonella infection in newly-hatched chicks. Sci Rep 8:2229.
14.
Hughes R-A, Ali RA, Mendoza MA, Hassan HM, Koci MD. 2017. Impact of dietary galacto-oligosaccharide (gos) on chicken’s gut microbiota, mucosal gene expression, and Salmonella colonization. Front Vet Sci 4:192.
15.
Schoeni JL, Doyle MP. 1992. Reduction of Campylobacter jejuni colonization of chicks by cecum-colonizing bacteria producing anti-C. jejuni metabolites. Appl Environ Microbiol 58:664–670.
16.
Thompson JL, Hinton M. 1997. Antibacterial activity of formic and propionic acids in the diet of hens on Salmonellas in the crop. Br Poult Sci 38:59–65.
17.
Janczyk P, Halle B, Souffrant WB. 2009. Microbial community composition of the crop and ceca contents of laying hens fed diets supplemented with Chlorella vulgaris. Poult Sci 88:2324–2332.
18.
Saxena S, Saxena VK, Tomar S, Sapcota D, Gonmei G. 2016. Characterisation of caecum and crop microbiota of Indian indigenous chicken targeting multiple hypervariable regions within 16S rRNA gene. Br Poult Sci 57:381–389.
19.
Dong XY, Azzam MMM, Zou XT. 2017. Effects of dietary threonine supplementation on intestinal barrier function and gut microbiota of laying hens. Poult Sci 96:3654–3663.
20.
Biddle A, Stewart L, Blanchard J, Leschine S. 2013. Untangling the genetic basis of fibrolytic specialization by Lachnospiraceae and Ruminococcaceae in diverse gut communities. Diversity 5:627–640.
21.
Zhao L, Wang G, Siegel P, He C, Wang H, Zhao W, Zhai Z, Tian F, Zhao J, Zhang H, Sun Z, Chen W, Zhang Y, Meng H. 2013. Quantitative genetic background of the host influences gut microbiomes in chickens. Sci Rep 3:1163.
22.
Ballou AL, Ali RA, Mendoza MA, Ellis JC, Hassan HM, Croom WJ, Koci MD. 2016. Development of the chick microbiome: how early exposure influences future microbial diversity. Front Vet Sci 3:2.
23.
Cui Y, Wang Q, Liu S, Sun R, Zhou Y, Li Y. 2017. Age-related variations in intestinal microflora of free-range and caged hens. Front Microbiol 8:1310.
24.
Videnska P, Sedlar K, Lukac M, Faldynova M, Gerzova L, Cejkova D, Sisak F, Rychlik I. 2014. Succession and replacement of bacterial populations in the caecum of egg laying hens over their whole life. PLoS One 9:e115142.
25.
Xu Y, Yang H, Zhang L, Su Y, Shi D, Xiao H, Tian Y. 2016. High-throughput sequencing technology to reveal the composition and function of cecal microbiota in Dagu chicken. BMC Microbiol 16:259.
26.
Shang Y, Kumar S, Oakley B, Kim WK. 2018. Chicken gut microbiota: importance and detection technology. Front Vet Sci 5:254.
27.
Stanley D, Hughes RJ, Moore RJ. 2014. Microbiota of the chicken gastrointestinal tract: influence on health, productivity and disease. Appl Microbiol Biotechnol 98:4301–4310.
28.
Ocejo M, Oporto B, Hurtado A. 2019. 16S rRNA amplicon sequencing characterization of caecal microbiome composition of broilers and free-range slow-growing chickens throughout their productive lifespan. Sci Rep 9:2506.
29.
Maier L, Vyas R, Cordova CD, Lindsay H, Schmidt TSB, Brugiroux S, Periaswamy B, Bauer R, Sturm A, Schreiber F, von Mering C, Robinson MD, Stecher B, Hardt W-D. 2013. Microbiota-derived hydrogen fuels Salmonella Typhimurium invasion of the gut ecosystem. Cell Host Microbe 14:641–651.
30.
Lee H, Ma R, Grimm MC, Riordan SM, Lan R, Zhong L, Raftery M, Zhang L. 2014. Examination of the anaerobic growth of Campylobacter concisus strains. Int J Microbiol 2014:476047.
31.
Hannah JF, Wilson JL, Cox NA, Richardson LJ, Cason JA, Bourassa DV, Buhr RJ. 2011. Horizontal transmission of Salmonella and Campylobacter among caged and cage-free laying hens. Avian Dis 55:580–587.
32.
Novoa Rama E, Bailey M, Jones DR, Gast RK, Anderson K, Brar J, Taylor R, Oliver HF, Singh M. 2018. Prevalence, persistence, and antimicrobial resistance of Campylobacter spp. from eggs and laying hens housed in five commercial housing systems. Foodborne Pathog Dis 15:506–516.
33.
Van TTH, Gor M-C, Anwar A, Scott PC, Moore RJ. 2017. Campylobacter hepaticus, the cause of spotty liver disease in chickens, is present throughout the small intestine and caeca of infected birds. Vet Microbiol 207:226–230.
34.
Wang G, He Y, Jin X, Zhou Y, Chen X, Zhao J, Zhang H, Chen W. 2018. The effect of co-infection of food-borne pathogenic bacteria on the progression of Campylobacter jejuni infection in mice. Front Microbiol 9:1977.
35.
Staib L, Fuchs TM. 2015. Regulation of fucose and 1, 2-propanediol utilization by Salmonella enterica serovar Typhimurium. Front Microbiol 6:1116.
36.
Stahl M, Friis LM, Nothaft H, Liu X, Li J, Szymanski CM, Stintzi A. 2011. l-Fucose utilization provides Campylobacter jejuni with a competitive advantage. Proc Natl Acad Sci U S A 108:7194–7199.
37.
Thiennimitr P, Winter SE, Winter MG, Xavier MN, Tolstikov V, Huseby DL, Sterzenbach T, Tsolis RM, Roth JR, Bäumler AJ. 2011. Intestinal inflammation allows Salmonella to use ethanolamine to compete with the microbiota. Proc Natl Acad Sci U S A 108:17480–17485.
38.
Stecher B, Denzler R, Maier L, Bernet F, Sanders MJ, Pickard DJ, Barthel M, Westendorf AM, Krogfelt KA, Walker AW, Ackermann M, Dobrindt U, Thomson NR, Hardt W-D. 2012. Gut inflammation can boost horizontal gene transfer between pathogenic and commensal Enterobacteriaceae. Proc Natl Acad Sci U S A 109:1269–1274.
39.
Fujimoto M, Goto R, Hirota R, Ito M, Haneda T, Okada N, Miki T. 2018. Tat-exported peptidoglycan amidase-dependent cell division contributes to Salmonella Typhimurium fitness in the inflamed gut. PLoS Pathog 14:e1007391.
40.
Mickael CS, Lam P-K, Berberov EM, Allan B, Potter AA, Köster W. 2010. Salmonella enterica serovar Enteritidis tatB and tatC mutants are impaired in Caco-2 cell invasion in vitro and show reduced systemic spread in chickens. Infect Immun 78:3493–3505.
41.
Abdelqader A, Al-Fataftah A-R, Daş G. 2013. Effects of dietary Bacillus subtilis and inulin supplementation on performance, eggshell quality, intestinal morphology and microflora composition of laying hens in the late phase of production. Anim Feed Sci Technol 179:103–111.
42.
Mikulski D, Jankowski J, Naczmanski J, Mikulska M, Demey V. 2012. Effects of dietary probiotic (Pediococcus acidilactici) supplementation on performance, nutrient digestibility, egg traits, egg yolk cholesterol, and fatty acid profile in laying hens. Poult Sci 91:2691–2700.
43.
Khan S, Chousalkar KK. 2020. Salmonella Typhimurium infection disrupts but continuous feeding of Bacillus based probiotic restores gut microbiota in infected hens. J Anim Sci Biotechnol 11:29.
44.
Khan S, Chousalkar KK. 2020. Short-term feeding of probiotics and synbiotics modulates caecal microbiota during Salmonella Typhimurium infection but does not reduce shedding and invasion in chickens. Appl Microbiol Biotechnol 104:319–334.
45.
Abdel-Wareth AA. 2016. Effect of dietary supplementation of thymol, synbiotic and their combination on performance, egg quality and serum metabolic profile of Hy-Line Brown hens. Br Poult Sci 57:114–122.
46.
Pineda-Quiroga C, Borda-Molina D, Chaves-Moreno D, Ruiz R, Atxaerandio R, Camarinha-Silva A, García-Rodríguez A. 2019. Microbial and functional profile of the ceca from laying hens affected by feeding prebiotics, probiotics, and synbiotics. Microorganisms 7:123.
47.
Bron PA, Van Baarlen P, Kleerebezem M. 2011. Emerging molecular insights into the interaction between probiotics and the host intestinal mucosa. Nat Rev Microbiol 10:66–78.
48.
Smits HH, Engering A, van der Kleij D, de Jong EC, Schipper K, van Capel TMM, Zaat BAJ, Yazdanbakhsh M, Wierenga EA, van Kooyk Y, Kapsenberg ML. 2005. Selective probiotic bacteria induce IL-10–producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell–specific intercellular adhesion molecule 3–grabbing nonintegrin. J Allergy Clin Immunol 115:1260–1267.
49.
Jang K-S, Baik JE, Han SH, Chung DK, Kim B-G. 2011. Multi-spectrometric analyses of lipoteichoic acids isolated from Lactobacillus plantarum. Biochem Biophys Res Commun 407:823–830.
50.
Brisbin JT, Gong J, Parvizi P, Sharif S. 2010. Effects of lactobacilli on cytokine expression by chicken spleen and cecal tonsil cells. Clin Vaccine Immunol 17:1337–1343.
51.
Brisbin JT, Zhou H, Gong J, Sabour P, Akbari MR, Haghighi HR, Yu H, Clarke A, Sarson AJ, Sharif S. 2008. Gene expression profiling of chicken lymphoid cells after treatment with Lactobacillus acidophilus cellular components. Dev Comp Immunol 32:563–574.
52.
Chen C-Y, Tsen H-Y, Lin C-L, Yu B, Chen C-S. 2012. Oral administration of a combination of select lactic acid bacteria strains to reduce the Salmonella invasion and inflammation of broiler chicks. Poult Sci 91:2139–2147.
53.
Haghighi HR, Abdul-Careem MF, Dara RA, Chambers JR, Sharif S. 2008. Cytokine gene expression in chicken cecal tonsils following treatment with probiotics and Salmonella infection. Vet Microbiol 126:225–233.
54.
Nothaft H, Perez-Muñoz ME, Gouveia GJ, Duar RM, Wanford JJ, Lango-Scholey L, Panagos CG, Srithayakumar V, Plastow GS, Coros C, Bayliss CD, Edison AS, Walter J, Szymanski CM. 2017. Coadministration of the Campylobacter jejuni N-glycan-based vaccine with probiotics improves vaccine performance in broiler chickens. Appl Environ Microbiol 83:e01523-17.
55.
Bergmann KR, Liu SXL, Tian R, Kushnir A, Turner JR, Li H-L, Chou PM, Weber CR, De Plaen IG. 2013. Bifidobacteria stabilize claudins at tight junctions and prevent intestinal barrier dysfunction in mouse necrotizing enterocolitis. Am J Pathol 182:1595–1606.
56.
Gadde UD, Oh S, Lee Y, Davis E, Zimmerman N, Rehberger T, Lillehoj HS. 2017. Dietary Bacillus subtilis-based direct-fed microbials alleviate LPS-induced intestinal immunological stress and improve intestinal barrier gene expression in commercial broiler chickens. Res Vet Sci 114:236–243.
57.
Variyam EP, Hoskins LC. 1981. Mucin degradation in human colon ecosystems: degradation of hog gastric mucin by fecal extracts and fecal cultures. Gastroenterology 81:751–758.
58.
Smirnov A, Perez R, Amit-Romach E, Sklan D, Uni Z. 2005. Mucin dynamics and microbial populations in chicken small intestine are changed by dietary probiotic and antibiotic growth promoter supplementation. J Nutr 135:187–192.
59.
Engevik MA, Luk B, Chang-Graham AL, Hall A, Herrmann B, Ruan W, Endres BT, Shi Z, Garey KW, Hyser JM, Versalovic J. 2019. Bifidobacterium dentium fortifies the intestinal mucus layer via autophagy and calcium signaling pathways. mBio 10:e01087-19.
60.
Aliakbarpour HR, Chamani M, Rahimi G, Sadeghi AA, Qujeq D. 2012. The Bacillus subtilis and lactic acid bacteria probiotics influences intestinal mucin gene expression, histomorphology and growth performance in broilers. Asian Australas J Anim Sci 25:1285–1293.
61.
Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA, Kitamoto S, Terrapon N, Muller A, Young VB, Henrissat B, Wilmes P, Stappenbeck TS, Núñez G, Martens EC. 2016. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 167:1339–1353.
62.
Coconnier MH, Bernet MF, Kernéis S, Chauvière G, Fourniat J, Servin AL. 1993. Inhibition of adhesion of enteroinvasive pathogens to human intestinal Caco‐2 cells by Lactobacillus acidophilus strain LB decreases bacterial invasion. FEMS Microbiol Lett 110:299–305.
63.
Chan R, Reid G, Irvin R, Bruce A, Costerton J. 1985. Competitive exclusion of uropathogens from human uroepithelial cells by Lactobacillus whole cells and cell wall fragments. Infect Immun 47:84–89.
64.
Collado MC, Meriluoto J, Salminen S. 2007. In vitro analysis of probiotic strain combinations to inhibit pathogen adhesion to human intestinal mucus. Food Res Int 40:629–636.
65.
Pascual M, Hugas M, Badiola JI, Monfort JM, Garriga M. 1999. Lactobacillus salivarius CTC2197 prevents Salmonella Enteritidis colonization in chickens. Appl Environ Microbiol 65:4981–4986.
66.
Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ, Pike NB, Strum JC, Steplewski KM, Murdock PR, Holder JC, Marshall FH, Szekeres PG, Wilson S, Ignar DM, Foord SM, Wise A, Dowell SJ. 2003. The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem 278:11312–11319.
67.
Jacobson A, Lam L, Rajendram M, Tamburini F, Honeycutt J, Pham T, Van Treuren W, Pruss K, Stabler SR, Lugo K, Bouley DM, Vilches-Moure JG, Smith M, Sonnenburg JL, Bhatt AS, Huang KC, Monack D. 2018. A gut commensal-produced metabolite mediates colonization resistance to Salmonella infection. Cell Host Microbe 24:296–307.
68.
van der Wielen PW, Biesterveld S, Lipman LJ, van Knapen F. 2001. Inhibition of a glucose-limited sequencing fed-batch culture of Salmonella enterica serovar Enteritidis by volatile fatty acids representative of the ceca of broiler chickens. Appl Environ Microbiol 67:1979–1982.
69.
Hung CC, Garner CD, Slauch JM, Dwyer ZW, Lawhon SD, Frye JG, McClelland M, Ahmer BM, Altier C. 2013. The intestinal fatty acid propionate inhibits Salmonella invasion through the post‐translational control of HilD. Mol Microbiol 87:1045–1060.
70.
Cotter PD, Ross RP, Hill C. 2013. Bacteriocins—a viable alternative to antibiotics? Nat Rev Microbiol 11:95–105.
71.
Corr SC, Li Y, Riedel CU, O'Toole PW, Hill C, Gahan CGM. 2007. Bacteriocin production as a mechanism for the antiinfective activity of Lactobacillus salivarius UCC118. Proc Natl Acad Sci U S A 104:7617–7621.
72.
Bhardwaj A, Gupta H, Kapila S, Kaur G, Vij S, Malik RK. 2010. Safety assessment and evaluation of probiotic potential of bacteriocinogenic Enterococcus faecium KH 24 strain under in vitro and in vivo conditions. Int J Food Microbiol 141:156–164.
73.
Goh YJ, Klaenhammer TR. 2015. Genetic mechanisms of prebiotic oligosaccharide metabolism in probiotic microbes. Annu Rev Food Sci Technol 6:137–156.
74.
De Maesschalck C, Eeckhaut V, Maertens L, De Lange L, Marchal L, Nezer C, De Baere S, Croubels S, Daube G, Dewulf J, Haesebrouck F, Ducatelle R, Taminau B, Van Immerseel F. 2015. The effects of xylo-oligosaccharides on performance and microbiota in broiler chickens. Appl Environ Microbiol 81:5880–5888.
75.
Moura P, Barata R, Carvalheiro F, Gírio F, Loureiro-Dias MC, Esteves MP. 2007. In vitro fermentation of xylo-oligosaccharides from corn cobs autohydrolysis by Bifidobacterium and Lactobacillus strains. LWT-Food Sci Technol 40:963–972.
76.
Oyofo BA, DeLoach JR, Corrier DE, Norman JO, Ziprin RL, Mollenhauer HH. 1989. Prevention of Salmonella Typhimurium colonization of broilers with d-mannose. Poult Sci 68:1357–1360.
77.
Li DD, Ding XM, Zhang KY, Bai SP, Wang JP, Zeng QF, Su ZW, Kang L. 2017. Effects of dietary xylooligosaccharides on the performance, egg quality, nutrient digestibility and plasma parameters of laying hens. Anim Feed Sci Technol 225:20–26.
78.
Xu M, Jiang Z, Wang C, Li N, Bo L, Zha Y, Bian J, Zhang Y, Deng X. 2019. Acetate attenuates inflammasome activation through GPR43-mediated Ca2+-dependent NLRP3 ubiquitination. Exp Mol Med 51:1.
79.
Zhang J-M, Sun Y-S, Zhao L-Q, Chen T-T, Fan M-N, Jiao H-C, Zhao J-P, Wang X-J, Li F-C, Li H-F, Lin H. 2019. SCFAs-induced GLP-1 secretion links the regulation of gut microbiome on hepatic lipogenesis in chickens. Front Microbiol 10:2176.
80.
Priyamvada S, Anbazhagan A, Chatterjee I, Alrefai W, Dudeja P, Borthakur A. 2015. Gut bacterial metabolite propionate upregulates intestinal epithelial Kruppel-like factor 4 expression via a PPAR-γ-dependent mechanism. FASEB J 29:854.4.
81.
Inan MS, Rasoulpour RJ, Yin L, Hubbard AK, Rosenberg DW, Giardina C. 2000. The luminal short-chain fatty acid butyrate modulates NF-κB activity in a human colonic epithelial cell line. Gastroenterology 118:724–734.
82.
Connors J, Dawe N, Van Limbergen J. 2019. The role of succinate in the regulation of intestinal inflammation. Nutrients 11:25.
83.
Bhattarai Y, Williams BB, Battaglioli EJ, Whitaker WR, Till L, Grover M, Linden DR, Akiba Y, Kandimalla KK, Zachos NC, Kaunitz JD, Sonnenburg JL, Fischbach MA, Farrugia G, Kashyap PC. 2018. Gut microbiota-produced tryptamine activates an epithelial G-protein-coupled receptor to increase colonic secretion. Cell Host Microbe 23:775–785.
84.
Bansal T, Alaniz RC, Wood TK, Jayaraman A. 2010. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci U S A 107:228–233.
85.
Roager HM, Licht TR. 2018. Microbial tryptophan catabolites in health and disease. Nat Commun 9:3294.
86.
Miyamoto J, Mizukure T, Park S-B, Kishino S, Kimura I, Hirano K, Bergamo P, Rossi M, Suzuki T, Arita M, Ogawa J, Tanabe S. 2015. A gut microbial metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, ameliorates intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. J Biol Chem 290:2902–2918.
87.
Peng M, Tabashsum Z, Patel P, Bernhardt C, Biswas D. 2018. Linoleic acids overproducing Lactobacillus casei limits growth, survival, and virulence of Salmonella Typhimurium and enterohaemorrhagic Escherichia coli. Front Microbiol 9:2663.
88.
Guinane CM, Piper C, Draper LA, O'Connor PM, Hill C, Ross RP, Cotter PD. 2015. Impact of environmental factors on bacteriocin promoter activity in gut-derived Lactobacillus salivarius. Appl Environ Microbiol 81:7851–7859.
89.
Cooke G, Behan J, Costello M. 2006. Newly identified vitamin K-producing bacteria isolated from the neonatal faecal flora. Microb Ecol Health Dis 18:133–138.
90.
Liu Y, Van Bennekom EO, Zhang Y, Abee T, Smid EJ. 2019. Long-chain vitamin K2 production in Lactococcus lactis is influenced by temperature, carbon source, aeration and mode of energy metabolism. Microb Cell Fact 18:129.
91.
Yoshii K, Hosomi K, Sawane K, Kunisawa J. 2019. Metabolism of dietary and microbial vitamin B family in the regulation of host immunity. Front Nutr 6:48.
92.
Mappley LJ, Tchórzewska MA, Nunez A, Woodward MJ, Bramley PM, La Ragione RM. 2013. Oral treatment of chickens with Lactobacillus reuteri LM1 reduces Brachyspira pilosicoli-induced pathology. J Med Microbiol 62:287–296.
93.
Luoma A, Markazi A, Shanmugasundaram R, Murugesan G, Mohnl M, Selvaraj R. 2017. Effect of synbiotic supplementation on layer production and cecal Salmonella load during a Salmonella challenge. Poult Sci 96:4208–4216.
94.
Kobierecka PA, Wyszyńska AK, Aleksandrzak-Piekarczyk T, Kuczkowski M, Tuzimek A, Piotrowska W, Górecki A, Adamska I, Wieliczko A, Bardowski J, Jagusztyn-Krynicka EK. 2017. In vitro characteristics of Lactobacillus spp. strains isolated from the chicken digestive tract and their role in the inhibition of Campylobacter colonization. MicrobiologyOpen 6:e00512.
95.
Adhikari P, Lee CH, Cosby DE, Cox NA, Kim WK. 2019. Effect of probiotics on fecal excretion, colonization in internal organs and immune gene expression in the ileum of laying hens challenged with Salmonella Enteritidis. Poult Sci 98:1235–1242.
96.
Vilà B, Fontgibell A, Badiola I, Esteve-Garcia E, Jiménez G, Castillo M, Brufau J. 2009. Reduction of Salmonella enterica var. Enteritidis colonization and invasion by Bacillus cereus var. toyoi inclusion in poultry feeds. Poult Sci 88:975–979.
97.
La Ragione RM, Woodward MJ. 2003. Competitive exclusion by Bacillus subtilis spores of Salmonella enterica serotype Enteritidis and Clostridium perfringens in young chickens. Vet Microbiol 94:245–256.
98.
Schoeni JL, Wong AC. 1994. Inhibition of Campylobacter jejuni colonization in chicks by defined competitive exclusion bacteria. Appl Environ Microbiol 60:1191–1197.
99.
Hosseindoust A, Mohammadi M, Yao ZP, Jung M, Kim IH. 2018. Dietary Bacillus subtilis B2A strain in laying hens challenged with Salmonella Gallinarum: effects on egg production, egg quality, blood haptoglobin and targeted intestinal Salmonella shedding. J Appl Anim Res 46:512–517.
100.
Babu US, Sommers K, Harrison LM, Balan KV. 2012. Effects of fructooligosaccharide-inulin on Salmonella-killing and inflammatory gene expression in chicken macrophages. Vet Immunol Immunopathol 149:92–96.
101.
Fernandez F, Hinton M, Gils BV. 2002. Dietary mannan-oligosaccharides and their effect on chicken caecal microflora in relation to Salmonella Enteritidis colonization. Avian Pathol 31:49–58.
102.
Fukata T, Sasai K, Miyamoto T, Baba E. 1999. Inhibitory effects of competitive exclusion and fructooligosaccharide, singly and in combination, on Salmonella colonization of chicks. J Food Prot 62:229–233.
103.
Adhikari P, Cosby DE, Cox NA, Franca MS, Williams SM, Gogal RM, Ritz CW, Kim WK. 2018. Effect of dietary fructooligosaccharide supplementation on internal organs Salmonella colonization, immune response, ileal morphology, and ileal immunohistochemistry in laying hens challenged with Salmonella Enteritidis. Poult Sci 97:2525–2533.
104.
Azcarate-Peril MA, Butz N, Cadenas MB, Koci M, Ballou A, Mendoza M, Ali R, Hassan H. 2017. An attenuated Salmonella enterica serovar Typhimurium strain and galacto-oligosaccharides accelerate clearance of Salmonella infections in poultry through modifications to the gut microbiome. Appl Environ Microbiol 84:e02526-17.

Information & Contributors

Information

Published In

cover image Applied and Environmental Microbiology
Applied and Environmental Microbiology
Volume 86Number 1317 June 2020
eLocator: e00600-20
Editor: Danilo Ercolini, University of Naples Federico II
PubMed: 32332137

History

Published online: 17 June 2020

Permissions

Request permissions for this article.

Keywords

  1. gut microbiota
  2. laying chicken
  3. enteropathogens
  4. feed supplements
  5. gut metabolites

Contributors

Authors

School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, South Australia, Australia
RMIT University, School of Science, Bundoora, Victoria, Australia
Central Queensland University, Institute for Future Farming Systems, Rockhampton, Queensland, Australia
School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, South Australia, Australia

Editor

Danilo Ercolini
Editor
University of Naples Federico II

Notes

Address correspondence to Kapil K. Chousalkar, [email protected].

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy